Gallbladder and Biliary Tract Diseases
Gastroenterology and Hepatology Executive Editor J. Thomas LaMont, M.D. Chi...
126 downloads
1100 Views
12MB Size
Report
This content was uploaded by our users and we assume good faith they have the permission to share this book. If you own the copyright to this book and it is wrongfully on our website, we offer a simple DMCA procedure to remove your content from our site. Start by pressing the button below!
Report copyright / DMCA form
Gallbladder and Biliary Tract Diseases
Gastroenterology and Hepatology Executive Editor J. Thomas LaMont, M.D. Chief, Division of Gastroenterology Beth Israel Hospital Boston, Massachusetts and Charlotte F. and Irving W. Rabb Professor of Medicine Harvard Medical School Boston, Massachusetts 1. Crohn's Disease, edited by Cosimo Prantera and Burton I. Korelitz 2. Clinical Gastroenterology in the Elderly, edited by Alvin M. Gelb 3. Biliary and Pancreatic Ductal Epithelia: Pathobiology and Pathophysiology, edited by Alphonse E. Sirica and Daniel S. Longnecker 4. Viral Hepatitis: Diagnosis • Treatment • Prevention, edited by Richard A. Willson 5. Gastrointestinal Infections: Diagnosis and Management, edited by J. Thomas LaMont 6. Gastroesophageal Reflux Disease, edited by Roy C. Orlando 7. Gallbladder and Biliary Tract Diseases, edited by Nezam H. Afdhal ADDITIONAL VOLUMES IN PREPARATION
Page i
Gallbladder and Biliary Tract Diseases edited by Nezam H. Afdhal Beth Israel Deaconess Medical Center Harvard Medical School Boston, Massachusetts
Page ii
ISBN: 0824703111 This book is printed on acidfree paper. Headquarters Marcel Dekker, Inc. 270 Madison Avenue, New York, NY 10016 tel: 2126969000; fax: 2126854540 Eastern Hemisphere Distribution Marcel Dekker AG Hutgasse 4, Postfach 812, CH4001 Basel, Switzerland tel: 41612618482; fax: 41612618896 World Wide Web http://www.dekker.com The publisher offers discounts on this book when ordered in bulk quantities. For more information, write to Special Sales/Professional Marketing at the headquarters address above. Copyright © 2000 by Marcel Dekker, Inc. All Rights Reserved. Neither this book nor any part may be reproduced or transmitted in any form or by any means, electronic or mechanical, including photocopying, microfilming, and recording, or by any information storage and retrieval system, without permission in writing from the publisher. Current printing (last digit): 10 9 8 7 6 5 4 3 2 1 PRINTED IN THE UNITED STATES OF AMERICA
Page iii
In Memoriam
This book is dedicated to the fond memory of Edward Weldon Moore, M.D., Ph.D. Ed died on Christmas Eve, 1999, following a long illness during which every system failed except his infectious sense of humor. Ed was a peerless collaborator and close friend to most of the contributors to this book, and his uniquely creative mind originated or stimulated many of the concepts presented here. Ed was born in Covington, Kentucky, and graduated with top honors from Vanderbilt University Medical School in 1955. From 1955 to 1960, he underwent residency training on the Harvard Medical Service at Boston City Hospital and Lemuel Shattuck Hospital in Boston, and spent 2 years as a Clinical Associate at the National Cancer Institute in Bethesda, MD. In 1960, he returned to the Shattuck Hospital as an NIH Research Fellow with Dr. Thomas Chalmers. From 1965 to 1970, he was Chief of Gastroenterology at the Shattuck Hospital and recipient of a Research Career Development Award from the NIH, the first of his many NIH research grants. During the 1960s, he was a member of Dr. Franz Ingelfinger's famous GI Journal Club at Boston University. In 1970, Ed moved permanently to the Medical College of Virginia in Richmond. There he became Director of Gastrointestinal Research and Professor of Medicine, Pathology, Physiology, and Biophysics, ultimately becoming Emeritus in 1998.
Page iv
Ed was a most vibrant, inquisitive person, who never lost his boyish excitement about the mysteries of the universe, natural phenomena, electronics, animals, and people. He loved gadgets and was codeveloper of the calciumion electrode; his basement was filled with one of the largest private collections of miniature electric trains in the United States, which he built entirely by himself. Ed's innate charm, warmth, and unbounded enthusiasm always made him fun and stimulating to be with, rendering him a consummate teacher. Besides his many original publications and insightful review articles and book chapters, Ed was the heart and soul of the Undergraduate Teaching Project of the American Gastroenterological Association for 2 years. This group produced over 30 sets of outstanding visual aids to assist medical faculty in the education of students and trainees in fundamental pathophysiological concepts of gastrointestinal and liver diseases. In 1997, the AGA honored him for his many contributions by presenting him with its Distinguished Educator Award. We mourn the loss of an irreplaceable colleague whose diverse scientific contributions—many of which are included in this book—will long outlive him. J. Donald Ostrow, M.D. It is a pleasure and an honor to be invited to join my longstanding friend and colleague Don Ostrow in dedicating this timely volume to Ed Moore, a uniquely creative and charismatic medical investigator who died after a long battle with aspergillosis. In this book, Nid Afdhal has assembled the thoughts of the "best and the brightest" to survey our current knowledge (and ignorance) of the pathophysiology of the biliary tract and gallbladder. If Ed had recovered from his debilitating illness, he surely would have contributed a chapter that contained dazzling new insights. Some 15 years ago, Ed Moore, Don Ostrow, and I created a collaborative research program aimed at developing principles of calcium precipitation in bile. Over the next decade, we met regularly in Ed's palatial country home to develop strategy and to discuss our latest research findings. We were supported by a large NIH grant that Ed had orchestrated. Ed studied calcium binding to bile acid monomers and micelles, and worked on calcium entry into bile. Don joined forces with Pasupati Mukerjee, a superbly talented colloid chemist, to define the ionization properties of bilirubin and the solubility product of the calcium salt relevant to pigment stone formation. My laboratory studied calcium entry into bile as well as measuring the solubility products of the calcium salts of the natural conjugated and unconjugated bile acids. This work was performed in collaboration with Karol Mysels, a legendary physical chemist, now deceased, and led to the development of the idea of "calcium sensitive ions" and calcium precipitation. Our trio soon became a quartet when Bob Rege joined the group. Ed acted as a theoretician and Bob did the experiments in the dog. Together they showed that hepatic bile was supersaturated in calcium (carbonate), and that gallbladder mucosal acidification rendered bile unsaturated by increasing the bicarbonate/carbonate ratio. Ed also revisited his longstanding interest in the Donnan equilibrium and, with Bob, showed that the concentration of calcium in bile could be explained by application of this wellknown physicochemical principle. Thus, the idea gradually emerged that because the paracellular junctions of both the canaliculus and the biliary ductule were permeable to calcium, the activity of biliary calcium was rather constant. If this were the case, calcium precipitation in bile had to be explained predominantly by an increase in the activity of the calciumsensitive anion. Reno Vlahcevic, Ed's chief at Medical College of Virginia, made the classic finding of a diminished exchangeable bile acid pool in gallstone patients. He encouraged Ed to share his longstanding interest in cholesterol gallstone formation. Ed created the classic Venn diagram that suggested that cholesterol gallstone formation results from the simul
Page v
taneous occurrence of cholesterol supersaturation, impaired gallbladder motility, and an excess of nucleation factors. He coined the term "beyond supersaturation," popularizing an idea that Tom Holzbach had advanced many years before. Ed stimulated Mitch Shiffman to perform detailed analyses of biliary lipid and electrolyte composition in obese patients undergoing rapid weight loss after gastric bypass surgery. Ed developed the idea that the mucus layer played a key role in cholesterol gallstone formation and began to collaborate with Nid Afdhal and his colleagues, most notably Gwynneth Offner, who were cloning the mucin genes and defining mucin function. He also began experiments with cultured bile duct cells and gallbladder epithelial cells. Using the latter, he observed phospholipid transport perhaps by lateral diffusion and raised the question as to the extent to which the gallbladder mucosa absorbed phospholipid as well as cholesterol. Ed's health began to decline about 5 years ago, and his productive hours became fewer each day. When he could work, his energies were absorbed in analyzing his hospital bills, whose complexity and inscrutability often defied scientific logic. Abstracts were not translated into papers. His last grant application is replete with ideas that need experimental verification. Ed's mission was to understand, to make the world laugh, and to debunk scientific pretense. His origins in rural Kentucky made him love the common man and detest arrogance in any form. He had a deep knowledge of solution chemistry, electrophysiology, and transport phenomena, and he applied this unusual background to advance biliary physiology. Ed's diverse contributions advanced the field greatly and stimulated many of us. He enriched our lives with his charm, warmth, and humor. We shall all miss him deeply. Alan Hofmann, M.D., Ph.D.
Page vii
Preface The gallbladder and biliary tract are the "orphan organs" of the digestive system, falling between the realms of the solidorgan liver specialist and the holloworgan intestinal expert. This orphan status has led to the inclusion of the gallbladder in texts of both gastroenterology and hepatology, but no major comprehensive text has been devoted to disorders of the gallbladder. With the recent advances in both the basic pathophysiology of gallbladder disease and the introduction of many new treatment modalities, the time has come for the gallbladder to claim its place as a major organ in the digestive tract and to have a comprehensive text of its own. In addition, biliary tract disease remains the most expensive of all digestive diseases, with a total health care cost in the United States of over $7 billion. Gallstones are the major cause of biliary disease, and the incidence of symptomatic stones is rising with our aging population. Major advances have occurred in our understanding of the epidemiology and pathogenesis of gallstone disease; these can potentially be used as novel strategies for gallstone prevention. Such advances in the basic sciences have been slow to translate into real clinical therapies, perhaps because of the advent and rapid rise of laparoscopic cholecystectomy. However, the management of gallbladder and biliary disease is truly multidisciplinary, involving gastroenterologists, surgeons, endoscopists, and radiologists. Wherever possible, the team approach to management and the concept of biliary centers are promoted within this book. In this book we have also attempted to translate advances in basic science into clinically relevant treatment and to bridge the gap between clinical disciplines. Parts I and II focus on important physiological and pathophysiological principles, with a special emphasis on gallstones. The contributors represent the leading researchers in gallbladder physiology, smooth muscle function, and lipid metabolism. Each chapter is written with a focus on new advances in our understanding of basic mechanisms, with illustrations of how these may translate into clinical treatment for gallstones. In Parts III to V we focus on clinical disorders of the gallbladder and biliary tree with input on management from surgeons, endoscopists, and radiologists. Where appropriate, a chapter on combined management highlights the areas in which a team approach involving all disciplines is mandatory, as in the treatment of common duct strictures. New imaging techniques, such as magnetic resonance cholangiography and endoscopic ultrasound, are discussed from both the radiologist's and endoscopist's perspective and their role in disease management is defined. Finally, this book will serve as a comprehensive text for both basic researchers and clinicians involved in the management of biliary tract disease. I hope it will spark
Page viii
further research and collaboration that will lead to better clinical treatment modalities for biliary diseases. When I was first approached about putting together a book on gallbladder and biliary diseases, I felt that there was little place for yet another clinical text in this field. I envisioned a more ambitious volume, one that would integrate basic research with clinical advances and be relevant to multiple disciplines involved in biliary tract disease. I would like to thank the many contributors who helped me create such a unique book with broad appeal. I apologize for all my harassing emails and phone calls, and I hope you will all agree that the end product is worthy of your efforts. I would like to thank my teachers Tom LaMont and Diarmuid O'Donoghue, whose advice, friendship, and direction have helped me immeasurably in my career. I am grateful to my father, who made me the physician and person I am today, and to my wife, Clare, who manages not only my career but also her own and those of our two children, Sophie and Mo. Without all of your help and understanding, this book would not have become a reality. NEZAM H. AFDHAL, M.D.
Page ix
Contents Preface
vii
Contributors
xiii
Part I. The Normal Biliary System
1. Neurobiology of the Gallbladder Gary M. Mawe and Lee Jennings
1
2. Gallbladder Mucosal Function J. Henriette Klinkspoor and Sum P. Lee
21
3. Gallbladder Smooth Muscle Function and Its Dysfunction in Cholesterol Gallstone Disease Piero Portincasa and Gerard P. vanBergeHenegouwen
39
4. Canalicular Lipid Secretion James M. Crawford
65
5. Bile Ductal Secretion and Its Regulation Won Kyoo Cho
99
Part II. Pathogenesis of Gallstones
127
7. Pigment Gallstones Roger D. Soloway, Nyingi M. Kemmer, and Jinguang Wu
147
8. Hepatic Metabolism of Cholesterol, Bile Salts, and Phospholipids Douglas M. Heuman and Z. Reno Vlahcevic
165
6. Epidemiology, Risk Factors, and Pathogenesis of Gallstones Nezam H. Afdhal
Page x
9. Cholesterol Crystallization in Bile Fred M. Konikoff and Joanne M. Donovan
185
10. Gallbladder Mucin Gwynneth D. Offner
211
11. Role of Proteins in Cholesterol Crystallization in Bile A. Andre van den Berg and Albert K. Groen
235
12. Normal Gallbladder Motor Functions R. P. Jazrawi
251
13. Gallbladder Motility and Gallstones Ralph R. S. H. Greaves and Luke J. D. O'Donnell
275
14. The Role of Intestinal Transit R. Hermon Dowling
297
15. Calcium Salt Precipitation in Bile and Biomineralization of Gallstones Huguette Lafont and J. Donald Ostrow
317
16. Prevention of Gallstones Mitchell L. Shiffman
361
17. The Gallbladder and Biliary Tree in Cystic Fibrosis Michael P. Curry and John E. Hegarty
387
Part III. Management of Clinical Gallstone Disease
407
19. Endoscopic Ultrasound of the Gallbladder and Bile Ducts Brian R. Stotland
437
20. The Silent Gallstone William R. Brugge
447
21. Biliary Crystals, Microlithiasis, and Sludge Dieter Jüngst and Christoph von Ritter
455
22. Biliary Colic and Acute Cholecystitis Robert V. Rege
471
23. Laparoscopic Cholecystectomy A. C. T. Wan and A. Darzi
491
24. Overview of Nonsurgical Therapy of Gallstones Dominique E. Howard and Hans Fromm
521
25. Biliary Lithotripsy M. W. Neubrand and Tilman Sauerbruch
527
18. Selected Advances in Imaging of the Gallbladder and Bile Ducts Matthew Barish, Michael Blake, and Joseph T. Ferrucci
Page xi
26. Topical Contact Dissolution of Gallbladder Stones Salam F. Zakko
547
27. Common Bile Duct Stones Tony C. K. Tham and David R. Lichtenstein
567
Part IV. Gallbladder Disease
28. Acalculous Cholecystitis David Nunes
593
29. Gallbladder Cancer R. Montague Beazley
625
Part V. Diseases of the Bile Ducts
639
31. Primary Sclerosing Cholangitis John M. Vierling and Thomas D. Amankonah
659
32. Vanishing Bile Duct Syndrome P. Aiden McCormick and Niamh Nolan
705
33. Cholangiocarcinoma Steven A. Ahrendt and Henry A. Pitt
725
34. Ampullary Tumors Keith D. Lillemoe
755
35. Infections of the Bile Ducts Andrew P. Keaveny
773
36. Bile Duct Injuries Noel N. Williams and Daniel Kreisel
823
37. The Management of Benign and Malignant Biliary Strictures Laurence S. Bailen and Eric D. Libby
843
Index
875
30. Congenital and Cystic Diseases of the Biliary Tree David McAneny and James M. Becker
Page xiii
Contributors Nezam H. Afdhal, M.D., F.R.C.P.(I) Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts Steven A. Ahrendt, M.D., F.A.C.S. Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin Thomas D. Amankonah, M.D. Department of Medicine, CedarsSinai Medical Center and UCLA School of Medicine, Los Angeles, California Laurence S. Bailen, M.D. Division of Gastroenterology, New England Medical Center, Boston, Massachusetts Matthew Barish, M.D. Department of Radiology, Boston Medical Center, Boston, Massachusetts R. Montague Beazley, M.D., F.A.C.S. Section of Surgical Oncology and Endocrine Surgery, Department of Surgery, Boston University School of Medicine, Boston, Massachusetts James M. Becker, M.D. Department of Surgery, Boston University School of Medicine, Boston, Massachusetts Michael Blake, M.D., F.R.C.P.(C) Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts William R. Brugge, M.D. Gastrointestinal Unit, Massachusetts General Hospital, Boston, Massachusetts Won Kyoo Cho, M.D. Department of Medicine—GI/Hepatology, Indiana University School of Medicine and The Richard L. Roudebush VA Medical Center, Indianapolis, Indiana James M. Crawford, M.D., Ph.D. Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida
Page xiv
Michael P. Curry, M.D. Liver Unit, St. Vincent's University Hospital, Dublin, Ireland A. Darzi, M.D., F.R.C.S., F.R.C.S.I., F.A.C.S. Academic Surgical Unit, Imperial College School of Medicine at St. Mary's, London, England Joanne M. Donovan, M.D., Ph.D. Department of Medicine, Harvard Medical School and Boston VA Medical Center, Boston, Massachusetts R. Hermon Dowling, M.D. Academic Gastroenterology Unit, The Guy's, King's College and St. Thomas's Medical and Dental School, London, England Joseph T. Ferrucci, M.D. Department of Radiology, Boston Medical Center, Boston, Massachusetts Hans Fromm, M.D. Department of Medicine, The George Washington University Medical Center, Washington, D.C. Ralph R. S. H. Greaves, M.B., B.S., M.R.C.P. St. Bartholomew's and the Royal London School of Medicine and Dentistry, London, England Albert K. Groen, Ph.D. Department of Gastroenterology, Academic Medical Center, Amsterdam, The Netherlands John E. Hegarty, M.D., F.R.C.P. Liver Unit, St. Vincent's University Hospital, Dublin, Ireland Douglas M. Heuman, M.D. Department of Medicine, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia Dominique E. Howard, M.D. Department of Gastroenterology, The George Washington University Medical Center, Washington, D.C. R. P. Jazrawi, M.B., Ch.B., M.Sc., Ph.D. Department of Gastroenterology, Endocrinology and Metabolism, St. George's Hospital Medical School, London, England Lee Jennings, Ph.D. Department of Clinical Services, Ilex Oncology, San Antonio, Texas Dieter Jüngst, M.D. Department of Medicine II, Klinikum Grosshadern, LudwigMaximiliansUniversity, Munich, Germany Andrew P. Keaveny, M.B., M.R.C.P.I. Section of Gastroenterology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts Nyingi M. Kemmer, M.D. Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas J. Henriette Klinkspoor, Ph.D. Gastroenterology Section, Veterans Affairs Medical Center, Seattle, Washington Fred M. Konikoff, M.D., M.Sc. Department of Gastroenterology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
Page xv
Daniel Kreisel, M.D. Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania Huguette Lafont, Ph.D. INSERM U476, Marseilles, France Sum P. Lee, M.D. Gastroenterology Section, Veterans Affairs Medical Center, Seattle, Washington Eric D. Libby, M.D. Division of Gastroenterology, New England Medical Center, Boston, Massachusetts David R. Lichtenstein, M.D., F.A.C.G. Section of Gastroenterology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts Keith D. Lillemoe, M.D. Department of Surgery, The Johns Hopkins Medical Institutions, Baltimore, Maryland Gary M. Mawe, Ph.D. Department of Anatomy and Neurobiology, The University of Vermont, Burlington, Vermont David McAneny, M.D. Department of Surgery, Boston University School of Medicine, Boston, Massachusetts P. Aiden McCormick, M.D., F.R.C.P. Liver Unit, St. Vincent's University Hospital, Dublin, Ireland M. W. Neubrand, M.D. Medizinische Klinik and Poliklinik, Rheinische FriedrichWilhelmsUniversity, Bonn, Germany Niamh Nolan, F.R.C.Path. Department of Pathology, St. Vincent's University Hospital, Dublin, Ireland David Nunes, M.B., B.Ch., F.R.C.P.I. Section of Gastroenterology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts Luke J. D. O'Donnell, M.D., F.R.C.P.I. Department of Gastroenterology, Mayo General Hospital, Castlebar, Ireland Gwynneth D. Offner, Ph.D. Section of Gastroenterology, Boston University Medical Center, Boston, Massachusetts J. Donald Ostrow, M.D. Department of GI/Hepatology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands Henry A. Pitt, M.D., F.A.C.S. Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin Piero Portincasa, M.D., Ph.D. Department of Internal Medicine, University Medical School, Bari, Italy Robert V. Rege, M.D. Department of Surgery, University of Texas, Southwestern Medical Center, Dallas, Texas
Page xvi
Tilman Sauerbruch, M.D. Medizinische Klinik and Poliklinik, Rheinische FriedrichWilhelmsUniversity, Bonn, Germany Mitchell L. Shiffman, M.D. Hepatology Section, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia Roger D. Soloway, M.D. Division of Gastroenterology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas Brian R. Stotland, M.D. Endoscopic Ultrasound, Section of Gastroenterology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts Tony C. K. Tham, M.D., F.R.C.P. Division of Medicine, Ulster Hospital, Belfast, Northern Ireland Gerard P. vanBergeHenegouwen, M.D. Gastroenterology, University Hospital Utrecht, Utrecht, The Netherlands A. Andre van den Berg, M.D. Department of Gastroenterology, Academic Medical Center, Amsterdam, The Netherlands John M. Vierling, M.D., F.A.C.P. Department of Medicine, CedarsSinai Medical Center and UCLA School of Medicine, Los Angeles, California Z. Reno Vlahcevic, M.D. Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia Christoph von Ritter, M.D., Ph.D. Department of Medicine I, Vinzentimm Ruhpolding Hospital, Ruhpolding, Germany A. C. T. Wan, M.B., B.Ch., F.R.C.S.I. Academic Surgical Unit, Imperial College School of Medicine at St. Mary's, London, England Noel N. Williams, M.D., M.Ch., FR.C.S.I., F.R.C.S. Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania Jinguang Wu Department of Chemistry, Peking University, Beijing, People's Republic of China Salam F. Zakko, M.D., F.A.C.P. Division of Gastroenterology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut
Page 1
1— Neurobiology of the Gallbladder Gary M. Mawe The University of Vermont, Burlington, Vermont Lee Jennings Ilex Oncology, San Antonio, Texas I— Introduction Motility and absorption by the gallbladder are known to be influenced by circulating hormones such as cholecystokinin (CCK), secretin, gastrin, and pancreatic polypeptide (1). Nevertheless, the gallbladder contains a welldefined ganglionated plexus, which lies at the outer surface of the muscularis, as well as two extensive axonal plexuses, which lie within the muscularis and in the submucosa. In the bowel, motor and absorptive functions are influenced to a great extent by the enteric nervous system (ENS), the intrinsic ganglionated plexuses of the gut (2–4); therefore, since the gallbladder develops as an extension of the fetal bowel, it seems probable that the ganglionated neural plexus of the gallbladder plays a significant role in the motor and absorptive functions of the gallbladder. In order to understand the physiology of the gallbladder, it is thus of great importance to determine how its nervous system is organized, what types of neurons it contains, the degree to which it receives synaptic input from extrinsic sources (such as the central nervous system, prevertebral sympathetic ganglia, and the bowel), and to what extent, if any, the effects of circulating hormones and inflammatory mediators on the gallbladder are transmitted by intrinsic neurons. Several lines of evidence indicate that the nervous system does indeed play a crucial role in the emptying and filling of the gallbladder. As described in this chapter, CCK appears to act within the ganglia to cause the gallbladder to contract. Prostaglandins released during gallbladder inflammation can act pre and postsynaptically in gallbladder ganglia, and the ganglia of the gallbladder are equipped with multiple neuronal subtypes, based on their expression of multiple neurotransmitters. Furthermore, disruption of the neural input to the organ can lead to gallbladder malfunction. Clinical and experimental reports indicate that vagotomy results in an increased incidence of gallstone formation, changes in the lithogenicity of bile, an increase in the resting volume of the gallbladder, hypotonia of the gallbladder with biliary stasis, and impairment of gallbladder emptying following fatty meals (5). Until recently, the properties and roles of the neurons in the gallbladder wall were evaluated indirectly by using approaches such as measuring the effects of vagotomy on gallbladder function and testing the effects of neural blockers on gallbladder emptying or gallbladder muscle strip tension. Over the past decade, efforts in this and a few other laboratories have concentrated on elucidating the properties of the neurons that directly innervate the tissues of the gallbladder. This chapter includes a summary of the morphological, neurochemical, and electrophysiological findings of the past few years. Furthermore, it includes a discussion of how the ganglionic transmission can be modulated by neural, hormonal, and immunemediated signals, and how this may influence gallbladder function in health and disease.
Page 2
II— Properties of Gallbladder Ganglia A— Morphological and Neurochemical Features of Gallbladder Ganglia 1— General Morphology of Gallbladder Ganglia The gallbladder has a relatively simple structure consisting of a serosal layer, a layer of muscle (the muscularis), and a mucosal layer with an underlying lamina propria. Each of these layers of the gallbladder has its own neural plexus interconnected with those of the other layers. The most obvious and welldefined plexus is the one lying within the serosa. These neural plexuses of the gallbladder have been studied in many species including human, rhesus monkey, pig, dog, cat, marmoset, guinea pig, North American opossum (Didelphis virginiana and Monodelphis domestica), Australian brushtailed possum (Trichosurus vultecula), and mouse (6–15). The neural plexus of the serosa consists of a ganglionated network of small, irregularly shaped ganglia—reminiscent of the ganglia of the submucosal plexus of the intestine—connected by bundles of unmyelinated axons (Fig. 1A). These bundles of unmyelinated axons are contiguous with perivascular nerve bundles that follow the extensive vascular network in this layer. The neural plexus of the muscularis is prominent in species such as the dog and human but is rather sparse in other species such as the guinea pig and opossum. This plexus comprises interconnected nerve bundles that travel parallel to the direction of smooth muscle bundles. A rich network of nerve fibers lies in the lamina propria, with branching nerve fibers that pass through the mucosa and are often in close apposition with epithelial cells. In some species,
Figure 1 Human gallbladder ganglia. A. Photomicrograph of cholinesterase histochemical staining in a wholemount preparation of the human gallbladder. B. Immunoreactivity for neuropeptide Y in a ganglion that was attached to an interganglionic connective by a stem of nerve fibers, many of which are NPYimmunoreactive. C and D. Photomicrographs of a single field demonstrating immunoreactivity for vasoactive intestinal peptide and substance P (SP). Note that most neurons are immunoreactive for both of these compounds but that some neurons are immunoreactive for VIP but not for SP (arrows) and one of the neurons is not immunoreactive for either peptide but is innervated by immunoreactive nerve fibers (arrowhead). Scale bars = 50 m.
Page 3
including human and Australian possum, this plexus contains occasional small ganglia that are ovoid or triangular in shape. The structure of gallbladder neurons has been studied by intracellular staining of individual neurons with horseradish peroxidase and neurobiotin (15–17). In guinea pig and opossum, these neurons consist of a soma with one or two long processes, but no appreciable dendritic arborization. The axons of these neurons often pass from their ganglion of origin into interganglionic connectives, where they travel for some distance before they terminate. Processes that surround or travel through adjacent ganglia or run parallel with smooth muscle bundles often exhibit varicosities, suggesting of the possibility of interganglionic and neuromuscular communication. However, these processes do not seem to project to para or perivascular networks, possibly indicating that gallbladder ganglion neurons do not play a major role in modulating vascular tone within the organ. 2— Ultrastructural Features of Gallbladder Ganglia The ultrastructural properties of the ganglionated plexus of the guinea pig gallbladder have been studied in ultrathin sections from conventional preparations and from preparations containing horseradish peroxidasefilled neurons that were visualized with a diamino benzidine histochemical reaction (16). Neurons, glial cells, and a compact neuropil (usually displaced to the periphery of the ganglion) are all surrounded by a connective tissue sheath and an outer layer of basal lamina to form a gallbladder ganglion. Like the interiors of enteric ganglia, gallbladder ganglia are notably free of collagen, basal laminae, intercellular spaces, or blood vessels. The neuropil consists of glial processes, unmyelinated axons, and nerve terminals that contain clear spherical and dense core vesicles. Despite a common lineage and the ultrastructural similarities described above, the ganglia and the interganglionic connectives of the gallbladder have significant differences with those of the enteric nervous system. The neurons of the gallbladder have a similar morphology to parasympathetic neurons and are much simpler and less diverse than those of the enteric nervous system. Furthermore, the unmyelinated axons in interganglionic connectives are individually ensheathed by Schwann cell processes in a similar manner to nonenteric peripheral nerve bundles. 3— Chemical Coding of Gallbladder Neurons In order to understand how gallbladder neurons can influence the physiological activity of a given organ, it is useful to identify the neurotransmitters, produced by the neurons, that may potentially be released at several sites to drive, inhibit, and/or modify gallbladder actions. The chemical coding of gallbladder neurons has been studied in several species, including the human, dog, guinea pig, and opossum. Certain characteristic trends as well as interspecies differences have emerged from these studies. A diagram summarizing the expression patterns of neuroactive compounds in the gallbladder ganglion neurons of various species is shown in Figure 2. It is likely that all gallbladder neurons in all species are cholinergic since all express the essential biosynthetic precursor enzyme for acetylcholine, choline acetyltransferase (ChAT) (Figs. 2 and 3A) (14). However, immunohistochemical studies have shown that gallbladder neurons synthesize additional neuroactive compounds (see Figs. 1 to 3). To date, gallbladder neurons have been shown to express immunoreactivity for tachykinins, vasoactive intestinal peptide (VIP), nitric oxide synthase (NOS), and either neuropeptide Y (NPY) in the guinea pig and human or, in the dog and opossum, galanin (14,18–20). Furthermore, pituitary adenylate cyclaseactivating peptide (PACAP) has also been reported in most neurons of the human gallbladder (21). Despite this melange of neuroactive compounds expressed by gallbladder neurons, some species specificity does appear to be involved. Immunoreactivity for VIP has been demonstrated in human, dog, and guinea pig gallbladder neurons, although the distribution is slightly different among the species. In the human
Page 4
Figure 2 Chemical coding in neurons in ganglia of the human, dog, and guinea pig gallbladder. Note the coexpression of neuroactive compounds that have excitatory (+) and inhibitory () effects on gallbladder smooth muscle. ACh, acetylcholine; VIP, vasoactive intestinal peptide; NPY, neuropeptide Y; SP, substance P; NOS, nitric oxide synthase. (Modified from Ref. 14.)
and the dog, most of the neurons are immunoreactive for VIP, but only a small subset are VIPpositive in the guinea pig. Antibodies to substance P (SP) have illustrated immunoreactivity in most gallbladder neurons of the human, dog, and guinea pig. Human and guinea pig gallbladder neurons express immunoreactivity for NPY (Fig. 1B) in most neurons, but this pattern seems to be replaced with galanin in the dog (14,18,19). Gallbladder ganglia also comprise neurons that express nitric oxide synthase (NOS), the synthetic enzyme for nitric oxide. In all species that have been studied to date—including the human (13,22), monkey (22), dog (13), opossom (13), guinea pig (13,23,24), gerbil (13), and mouse (25), neurons in the gallbladder express NOS and/or NADPHdiaphorase (NADPHDA) activity. In guinea pig gallbladder neurons, there is a onetoone correlation between NOS immunoreactivity and NADPHDA activity (26), and these neurons also express VIP (Fig. 3C and D) (13). In the human, NOS immunoreactivity is expressed by a population of neurons that is distinct from the VIP immunoreactive neurons (21) (Mawe and Talmage, unpublished observations). In addition to the diversity of neurotransmitter expression in neurons of gallbladder ganglia, there is also coexpression of neuropeptides in extrinsic sensory fibers. Immunohistochemical studies in the human (13,22), dog (14), pig (27), guinea pig (12,18,28), Monodelphis domesticus opossum (14), and toad (29) failed to identify neurons in the gallbladder wall that expressed immunoreactivity for calcitonin generelated peptide (CGRP) but did describe abun
Page 5
Figure 3 Guinea pig gallbladder ganglia. A and B. Photomicorgraphs of a single field demonstrating immunoreactivities for choline acetyltransferase and nitric oxide synthase in a guinea pig gallbladder ganglion. Immunoreactivities in this doublestained preparation illustrates the coexpression of ChAT and NOS in a subset of gallbladder neurons. C and D. Photomicrographs of a single field demonstrating immunoreactivities for nitric oxide synthase (NOS) and vasoactive intestinal peptide (VIP) in a guinea pig gallbladder ganglion. All NOSimmunoreactive neurons in the guinea pig gallbladder also express VIP. E and F. In the wall of the guinea pig gallbladder, there is an abundance of extrinsic nerve fibers that are immunoreactive for both calcitonin generelated peptide (CGRP) and SP. The photomicrographs in E and F demonstrate immunoreactivities for SP and CGRP in the same field. Note the abundance of doublelabeled varicose nerve fibers in he ganglion. Doublelabeled immunoreactive fibers are found in the paravascular plexus passing along a blood vessel in the bottom of the field. These fibers are believed to be extrinsic sensory fibers, since CGRPimmunoreactivity has never been demonstrated in gallbladder neurons. Scale bars = 50 m. (Figures C and D were adapted from Ref. 26.)
Page 6
dant CGRPimmunoreactive nerve fibers in the ganglionated plexus and in the paravascular plexus. In the human, dog, opossum, Australian possum, guinea pig, and toad gallbladders, these fibers have been shown to coexpress SPimmunoreactivity (12,14,28) and they probably originate in sensory ganglia (Fig. 3E and F). Despite the wealth of information on neuroactive expression patterns in gallbladder ganglia, it remains difficult to explain the coexpression in terms of neural regulation. In isolation, each of the putative neurotransmitters has been demonstrated to induce a response in the gallbladder. For instance, acetylcholine (1), SP (30–33), and NPY (34) are known to contract gallbladder smooth muscle. On the other hand, VIP has been shown to relax precontracted gallbladder muscle (35–37). Nitric oxide can relax the gallbladder (38,39) and, further, may play a role in modulating CGRPinduced relaxation (40). Together with the knowledge that all of the gallbladder neurons are cholinergic, these patterns of expression of neuroactive compounds are somewhat perplexing, as they suggest the coexpression of excitatory and inhibitory transmitters in single neurons and/or nerve fibers of all of these species. The question remains: How does coexpression of these various neuroactive compounds contribute to gallbladder function? Despite many years of investigation, it is relatively unclear which neuroactive compounds are released during activation of the local gallbladder neuronal network. Electrical field stimulation of gallbladder muscle strips induces contraction, suggesting that excitatory neuroactive compounds are released. However, this same mode of excitation in the presence of atropine elicits relaxation or a reduction in tone, suggesting the concomitant release of inhibitory compounds. The preponderance of excitation over inhibition in these studies may be related to the role of neural output to the muscle from the ganglia. Two major theories have been propose to explain how the gallbladder fills. One theory suggests that the gallbladder undergoes a passive filling between meals (1,41); the other suggests that the gallbladder actively expands to draw hepatic bile into its lumen, much as a bellows draws in air as it is expanded (42,43). In order for the gallbladder to act as a bellows, a potent inhibitory output from the ganglia of the gallbladder would be necessary to induce an active relaxation. The ostensibly opposing outputs from individual gallbladder neurons that express excitatory and inhibitory compounds could be the following theoretical scenarios: (a) excitatory and inhibitory neuroactive compounds are separately released from a given neuron in response to distinct inputs; (b) compounds with opposing actions are released onto the same target sequentially, with one acting as a physiological antagonist of the other; or (c) both sets of compounds are coreleased but act on different targets. For example, if acetylcholine, SP, and VIP were released together, acetylcholine and SP may act on the muscle to elicit a contraction, while VIP may act on epithelial cells. Neuroactive compounds may also act on adjacent nerve terminals to modulate further release. Additional studies will be required to test these models. B— Physiological Properties of Gallbladder Neurons 1— Electrical Properties of Gallbladder Neurons The electrical properties of guinea pig, North American opossum, and human gallbladder neurons have been investigated using the intracellular recording technique (15,17,44). Unlike the neurons that reside in the bowel, these neurons have relatively simple properties. Based on electrical properties, there appears to be only one type of neuron in these ganglia, and it is fairly inexcitable. During long periods of recording, these neurons very rarely exhibit spontaneous action potentials. This indicates that gallbladder neurons must receive excitatory inputs in order to release neurotransmitter onto their target tissues, which include smooth muscle and epithelium. Under normal recording conditions, both guinea pig and human gallbladder neurons are relatively inexcitable, since they fire only one to three action potentials at the onset of a
Page 7
depolarizing current pulse (Fig. 4A). Of the species studied to date, the properties of the action potential in guinea pig gallbladder neurons have been investigated most extensively (17). The upstroke of the action potential is due to a tetrodotoxinsensitive sodium conductance, although there is a contribution from an inward calcium current that is revealed as a calcium spike in the presence of tetrodotoxin (TTX) and tetraethylammonium (TEA). The action potential is followed by an afterhyperpolarization (AHP) of about 15 mV, which lasts about 170 ms and can be divided into an early and a late phase (Fig. 4B). The early and late phases of the AHP are mediated by two different sequential calciumactivated potassium conductances. The early phase of the AHP is attenuated in the presence of TEA and is likely to result from activation of largeconductance potassium channels (BK), whereas the latter phase of the AHP is sensitive to apamin and probably results from activation of smallconductance potassium channels (SK). The late phase of the AHP contributes to the rapid adaptation of guinea pig gallbladder neurons,
Figure 4 Active electrical properties of guinea pig gallbladder neurons. A. Four consecutive overlapping traces showing the response of a gallbladder neuron to a prolonged depolarizing current pulse (250 ms, 0.2 nA). As is typical of guinea pig gallbladder neurons, the cell generated only one action potential at the onset of each current pulse. B. Response of a gallbladder neuron to a brief depolarizing current pulse (2 ms, 0.2 nA), illustrating a typical afterhyperpolarization (AHP). The AHP of gallbladder neurons is composed of early and late phases, indicated by arrows, that are sensitive to tetraethyl ammonium and apamin, respectively. C. Elimination of the last phase of the AHP by application of apamin (100 nM) to the bath converts gallbladder neurons from a phasic state, firing one action potential in response to a depolarizing current pulse, to a tonic state, generating a burst of action potentials throughout the duration of a depolarizing current pulse. Resting membrane potentials: A, mV; B, 52 mV; C, 52mV. (From Ref. 26.)
Page 8
since suppression of this component of the AHP with apamin causes the cells to fire action potentials repetitively throughout the duration of a depolarizing current pulse (Fig. 4C) (17). The properties of neurons of the North American opossum gallbladder are classified into two groups (15): adaptive neurons, responding to intracellular current pulses with a short burst of action potentials, and rapidly adaptive neurons, responding to current pulses with a single action potential. The adaptive cells are more numerous, making up about 70% of the population. Action potentials of neurons in these ganglia are tetrodotoxinsensitive and are followed by a brief AHP lasting about 30 ms. 2— Synaptic Events in Gallbladder Ganglia In the wholemount preparations used to study the electrical properties of gallbladder neurons, synaptic inputs can be activated by stimulating interganglionic fiber bundles or the nerve bundles that pass toward the gallbladder along the cystic duct. Synaptic inputs have been studied in guinea pig, opossum, and human gallbladder preparations (15,17,44). Lowfrequency stimulation of interganglionic nerve bundles results in the production of fast excitatory postsynaptic potentials (EPSPS) in gallbladder neurons (15,17,44,45). Although subthreshold spontaneous fast synaptic events can occasionally be seen in recordings from guinea pig gallbladder neurons, fast EPSPs typically require active stimulation, and the conversion from subthreshold to suprathreshold is dependent upon the magnitude of the stimulus. Fast EPSPs in the gallbladder are blocked by the nicotinic receptor antagonist hexamethonium and are abolished when calcium is removed from the bathing solution. Highfrequency stimulation (10 to 20 Hz) of interganglionic connectives results in the appearance of slow EPSPs in about 30% of guinea pig gallbladder neurons and 20% of neurons of the North American opossum (15,17). Slow EPSPs can also be significantly reduced with lowcalcium/highmagnesium solutions and with antagonists of the neurokinin 3 (NK3) receptor, indicating the involvement of tachykinins in this response (46). A residual component of the slow EPSP in the presence of an NK3 antagonist may indicate the involvement of CGRP in the response (47). The slow EPSP is likely to involve an activation of a nonselective cation conductance, since it has an estimated reversal potential near 0 mV. III— Neural Inputs to Gallbladder Ganglia A— Vagal Preganglionic Input to Gallbladder Ganglia Parasympathetic and enteric ganglia of the abdomen and pelvis receive preganglionic innervation from neurons located in the vagal motor complex or the sacral spinal cord. Although these preganglionic neurons provide nicotinic, fast synaptic input to essentially all parasympathetic ganglion cells, most neurons of the enteric nervous system—especially those in the jejunum, ileum, and colon—lack direct input from the central nervous system (48–50). As the intramural neurons of gallbladder ganglia have a common lineage with those of the gut tube, it could not be taken for granted that vagal efferent nerve fibers represent the major source of the fast synaptic inputs that are received by all gallbladder neurons. Stimulation of vagus nerves in vivo elicits gallbladder contraction or, in the presence of atropine, gallbladder relaxation (1,51,52). Complementary to these data, morphological evidence has been obtained suggesting vagal innervation of the gallbladder. First, following retrograde dye application to the gallbladder, neurons of the dorsal motor nucleus of the vagus become labeled (12). Second, immunohistochemical staining for choline acetyltransferase (ChAT), the biosynthetic enzyme of acetylcholine, in wholemount preparations of the gallbladder shows immunoreactive nerve fibers surrounding intramural neurons and in the paravascular plexus, suggesting extrinsic cholinergic innervation (14). Although the evidence cited
Page 9
above does indeed suggest that there is a potent preganglionic parasympathetic innervation of the gallbladder, it verifies that gallbladder neurons typically receive vagal input and that vagal efferent axons represent the principal source of fast excitatory synaptic input to these cells. In order to address this question, nerve bundles passing to the gallbladder along the cystic duct were stimulated while recording from gallbladder neurons in order to activate synaptic inputs to gallbladder neurons that arise from sources outside of the gallbladder (45). Experiments were conducted in control animals and in those following vagotomy. Electrical stimulation of the cystic nerves in control animals resulted in the appearance of fast EPSPs in all neurons that were sensitive to hexamethonium and low calcium. In vagotomized animals, no responses to cystic nerve stimulation were detected. Therefore, fast synaptic input to gallbladder neurons, unlike the enteric nervous system, is primarily from extrinsic fibers from the vagus nerves. However, stimulation of intrinsic nerve fiber bundles in these vagotomized animals did result in the appearance of some fast EPSPS, indicating that interganglionic communication among gallbladder neurons does exist. B— Sympathetic Postganglionic Input to Gallbladder Ganglia Postganglionic sympathetic innervation of the gallbladder probably arises from the celiac ganglion, which in turn has preganglionic sympathetic innervation from neurons in the thoracic spinal cord. Evidence that the sympathetic postganglionic neurons lie in the celiac ganglion has been obtained from retrograde tracer studies in which a tracer injected into the gallbladder wall has been found to be transported to cell bodies lying in this ganglion (12). In order to confirm the presence of sympathetic nerve fibers in the gallbladder, whole mount preparations have been examined for several sympathetic markers. Catecholamine histofluorescence (11,12,53), in addition to imunoreactivity for tyrosine hydroxylase and dopamine hydroxylase (12) can be observed in nerve fibers that are abundant in gallbladder ganglia can be seen in peri and paravascular nerve fibers that follow blood vessels. Stimulation of sympathetic innervation of the gallbladder leads to a decrease in contractile tone (54–58). Interestingly, when sympathetic innervation is stimulated at a subthreshold level, at which there is no effect on motility per se, there is a reduction in contraction in response to concurrent parasympathetic stimulation (58). This observation suggests that norepinephrine released from sympathetic fibers has an inhibitory effect in gallbladder ganglia. In order to test this, electrical recordings have been obtained from gallbladder neurons while exposing them to norepinephrine. Within gallbladder ganglia, the sympathetic postganglionic fibers have a presynaptic inhibitory effect on the release of acetylcholine (ACh) from vagal preganglionic terminals (Fig. 5B) (45,59). Norepinephrine decreases the amplitude of fast EPSPs in a concentrationdependent manner, and it mediates this effect by acting on 2 adrenoreceptors. The action of norepinephrine is mimicked by the 2 adrenoreceptor agonist clonidine and is suppressed by the 2 adrenoreceptor antagonist yohimbine. Release of endogenous catecholamine stores, by tyramine application or by electrical stimulation of the vascular plexus, also causes a yohimbinesensitive decrease in fast synaptic activity. Therefore, the decrease in gallbladder tone that can be elicited by stimulation of the splanchnic nerves may be the result of a presynaptic inhibitory effect of sympathetic nerves on the vagal terminals in gallbladder ganglia. On the basis of present data, it is possible that the decrease in gallbladder tone observed upon stimulation of splanchnic nerves is the result of norepinephrine modulation of ACh release from vagal terminals in gallbladder ganglia. The physiological role for norepinephrine acting in this manner may be to facilitate gallbladder filling by reducing tone and therefore increasing volume.
Page 10
Figure 5 Synaptic responses elicited by electrical stimulation of vagal inputs to gallbladder neurons are enhanced by CCK and are inhibited by norepinephrine. A. Subthreshold EPSPs are converted to suprathreshold events in the presence of CCK. B. EPSPsevoked by cystic nerve stimulation are reversibly suppressed in the presence of norepinephrine (NE). Each condition is represented by single even (A) or by an average of five consecutive events (B). (From Ref. 45.)
C— Sensory Axon Reflexes in Gallbladder Ganglia Sensory innervation of the gallbladder has been examined primarily in the cat and guinea pig. In the guinea pig, retrograde tracing has established afferent cell bodies in the nodose ganglion of the vagus and in dorsal root ganglia at the thoracic spinal level (12). In the cat, retrograde tracing has also demonstrated that afferent cell bodies were found in dorsal root ganglia at the thoracocolumbar level (T2L2) and in the nodose ganglion (60). In addition, the latter study indicated that the projections of the afferent neurons from the dorsal root ganglia to the gallbladder probably travel in the lesser and greater splanchnic nerves, as splanchnicotomy or removal of the celiac ganglion ipsilaterally decreased retrograde staining on the same side but not on the contralateral side. Immunoreactivity for SP and CGRP, indicative of afferent neurotransmitters, is present in nerves that pass along blood vessels in the gallbladder and in varicose axons that are particularly abundant in the ganglia of the gallbladder (Fig. 3E and F) (12,14,28,32). Since neurons within the ganglionated plexus of the gallbladder do not exhibit immunoreactivity for CGRP, the most likely source of the CGRP/SP immunoreactive fibers are the sensory neurons of the dorsal root and nodose ganglia (12,14,18,28). Though the primary role of sensory afferent axons in the periphery may be to relay sensory information to the spinal cord and central areas, there is mounting evidence that these nerves can also release neuropeptides from their peripheral processes to initiate reflex activity in response to local sensory stimuli. In the gallbladder, it has been shown that application of capsaicin causes release of the sensory transmitters SP and CGRP, resulting in increased muscle tension (32). This may indicate that, under certain conditions, afferent fibers can release neuroactive compounds, which might initiate gallbladder action in an attempt to maintain homeostasis. Alternatively, it is possible that afferent neurotransmitter release may contribute to gallbladder inflammation, since release of neuroactive peptides from extrinsic sensory fibers is one of the initial steps in toxin Ainduced inflammation in the bowel (61). Both SP and CGRP have the ability to modulate the action of gallbladder smooth muscle. Calcitonin generelated peptide activates an ATPdependent potassium channel that causes a hyperpolarization of the resting membrane potential (62) and a consequent relaxation of the muscle (63). Substance P, on the other hand, elicits a dosedependent contraction of gallbladder smooth muscle (30–33). In electrophysiological studies of guinea pig gallbladder neurons, application of tachykinins, CGRP, or capsaicin caused a prolonged depolarization together with an enhancement
Page 11
Figure 6 Substance P causes a prolonged depolarization of guinea pig gallbladder neurons that is associated with an increase in excitability. A. Response of a gallbladder neuron to a brief pressure microejection of SP (0.1 mM; 500 ms; 10 PSI). Note the similarity between this response and the slow excitatory postsynaptic potential shown in Fig. 2B. B. Following application of SP, gallbladder neurons exhibit an increase in excitability, which is demonstrated by the generation of a burst of action potential during a depolarizing current pulse. Resting membrane potentials: A, 54 mV; B, 52 mV. (From Refs. 26 and 46.)
in excitability similar to the slow EPSP recorded in these preparations (Fig. 6) (46,64). This slow depolarization is caused by the activation of a nonselective cation conductance. The effects of tachykinins on these intramural neurons have been studied in considerable detail. Rankorder potency of tachykinins in this preparation is neurokinin B > neurokinin A > SP, which is suggestive of the activation of NK3 receptors. Confirmation of this is provided by evidence that senktide, the NK3 receptor agonist, is more potent than any of the naturally occurring tachykinins in eliciting a depolarization and that [Trp7, bAla8]NKA (4–10), an antagonist of NK 3 receptors, shifts the concentrationeffect curve for SP to the right and depresses both capsaicininduced depolarizations and stimulusevoked slow excitatory postsynaptic potentials (46). Therefore, it is likely that tachykinins, and possibly CGRP released from sensory nerve terminals, mediate the slow EPSPs in gallbladder ganglia. When sensory fibers are activated by extreme pressure or inflammatory agents, they could act locally as the afferent limb of a local reflex to facilitate ganglionic transmission. D— The Sphincter of Oddi Evidence seems to suggest a neuronal link between the gallbladder and the sphincter of Oddi (SO). In dogs, cats, and humans, in vivo distention or electrical stimulation of the gallbladder results in a decreased motility, or flow resistance, in the SO (65–68). In the cat, this response was eliminated by TTX or application of local anesthetics to the bile ducts (65). The mechanisms for the reflex between the gallbladder and the SO are unknown. Authors of these reports have suggested that the reflex may involve a direct neural link between the gallbladder and the SO; however, these studies have not ruled out the possibility of a vagal reflex mechanism.
Page 12
E— The Duodenum Early this century, DuBois and Kistler (69) reported that stimulation of the duodenal ampulla resulted in contraction of the gallbladder. Transection of the common bile duct resulted in the elimination of the gallbladder contraction, but gallbladder responses to vagal stimulation and to the cut end of the bile duct persisted. On the basis of these findings, it was proposed that a direct neural connection exists between the gut and the gallbladder and that these axons pass along the cystic duct via the common bile duct. More recently, experiments involving retrograde tracing of dyes injected into the wall of the gallbladder, in the guinea pig and Australian possum, demonstrated that neurons of the duodenal myenteric plexus and the ganglia of the SO project to the gallbladder (12,70). These data indicate that, in addition to being regulated the central nervous system, the circuitry exists for the gallbladder to receive direct inputs from the bowel. The existence of a potential neural communication network between the gut and the gallbladder has not been investigated further. Questions remain about the targets and origin of the gutgallbladder projections and of the physiological relevance. At this point it is not known whether the projections from the gut have targets on gallbladder epithelia, smooth muscle, or intramural ganglia. In the case of intramural ganglia, it is unlikely that these gutgallbladder projections contribute to fast EPSPs because these events are largely absent following vagotomy (45). It is possible that a gutgallbladder projection could involve serotonin. There is a precedent for this in terms of serotoninergic projections from the gut to the pancreas (71). In addition, nerve fibers immunoreactive for serotonin have been observed in gallbladder ganglia (12), and serotonin has been demonstrated to cause a slow depolarization in gallbladder neurons that is associated with an increase in excitability (17). One potential physiological role of an excitatory neural pathway from the gut to the gallbladder would be to activate the gallbladder contractions that occur in coordination with the migrating myoelectric complex. During the interdigestive period, the gallbladder undergoes periods of increased intraluminal pressure, in phase with the migrating myoelectric complex, accompanied by a delivery of bile from the gallbladder to the lumen of the duodenum (1,72). It is thought that the migrating myoelectric complex serves a ''housekeeping function." According to this model, increased motor activity would advance undigested food from the proximal bowel toward the large intestines, and the associated delivery of bile into the intestinal lumen could facilitate the overall digestive process. IV— Hormonal and ImmuneMediated Modulation of Gallbladder Neurotransmission A— Cholecystokinin It is now many years since cholecystokinin (CCK) was identified as the principal contractile agent of the gallbladder following the ingestion of a meal (73). Since that time, many studies have been carried out in an attempt to precisely identify the cellular target(s) of CCK in the gallbladder. Initial studies proposed that CCK acted on gallbladder smooth muscle to initiate contraction, but is soon became apparent that a neuronal component was involved in this process. In recent years, evidence has accumulated suggesting that, under physiological conditions, CCK acts in gallbladder ganglia to increase excitatory ganglionic output to smooth muscle and hence cause contraction. Many studies have contributed to the concept that the action of CCK on the gallbladder involves a neural component. In in vivo experiments, it has been demonstrated that gallbladder contractions elicited by feeding or by injection of postprandial concentrations of CCK can be attenuated by atropine, hexamethonium, TTX, and vagal blockade (74–85). Similar observations have been reported in in vitro gallbladder experiments (86), and application of CCK to
Page 13
gallbladder strips in motility experiments can result in the release of ACh, presumably of neuronal origin (87–90). This body of work further reinforces the concept that CCK acts through neural mechanisms as its principal mode of action. It is clear that CCK receptors do exist on gallbladder smooth muscle cells, but these receptors may not normally play a role in gallbladder emptying. It is plausible that the neural effect described was a result of actions of CCK on vagal afferent fibers and/or vagal terminals within gallbladder ganglia, since hexamethonium and vagal blockade disrupt the meal and CCKinduced gallbladder contraction (76,85). Electrophysiological studies of gallbladder wholemount preparations support this view. Direct studies of the actions of CCK in gallbladder ganglia have been conducted in the guinea pig (45,91) and the opossum (15) using intracellular electrophysiological recording techniques. In both of these species, CCK has a profound presynaptic facilitatory effect on ganglionic transmission but does not have a direct effect on the gallbladder neurons. Upon application of CCK, the amplitude of cholinergic fast EPSPs is increased, usually converting subthreshold EPSPs to suprathreshold EPSPs (Fig. 5A) (15,91). CCK increases the quantal content (the amount of ACh released) by threefold without altering quantal size, but it does not alter the sensitivity of these neurons to exogenously applied acetylcholine, indicating that CCK acts through a presynaptic mechanism (91). Most importantly, it has been shown that CCK is quite potent in its ability to promote the release of ACh (Fig. 7). The concentrationeffect relationship for CCK in gallbladder ganglia peaks at 1.0 nM and has a half maximal effective concentration (EC50) of 33 pM; the EC50 for the direct contractile effect of CCK on gallbladder muscle is 10 nM (Fig. 7) (91). In the presence of 10 pM CCK, which is within the range of postprandial serum levels of CCK (85), the peptide increases synaptic currents by about 20%. The nerve terminals that are sensitive to CCK are from the vagus nerve, since synaptic responses to cystic nerve stimulation are sensitive to CCK and these inputs are eliminated following vagotomy (45). B— Prostaglandin E2 Prostaglandins, particularly prostaglandin E2 (PGE2), have been shown to be intimately associated with pathology of the gallbladder (92). Early studies employing diseased human gallbladders demonstrated that both the mucosa and the muscularis of the organ produce high levels of PGE2 (94). Furthermore, a correlation between severity of inflammation and PGE concentrations has been observed (93). In animal model studies, PGE2 has been shown to have two major effects: a dose dependent contraction of the tissue (94) and a significant reversal in net fluid movement from absorption to secretion, including an increase in mucin secretion (95,96). Several investigators have noted indirectly that PGE2 probably exerts its effects, at least in part, through the neural network resident within the organ (97–99). In order to investigate this aspect further, intracellular recordings have been obtained from gallbladder neurons during PGE2 application (100). Prostaglandin E2 acted directly on gallbladder neurons to elicit a complex triphasic change in the resting membrane potential, and a decrease in the duration and amplitude of the AHP. Each component of the triphasic response was concentrationdependent, was associated with a change in input resistance, and changed in amplitude when the membrane potential was electronically increased or decreased. The predominant aspect of the triphasic change in resting membrane potential was a longlasting hyperpolarization. In addition to the direct effects of PGE2 on gallbladder neurons, PGE2 acted presynaptically to attenuate both fast and slow excitatory synaptic responses. Longlasting hyperpolarization of the neurons and suppression of fast and slow excitatory input would significantly decrease ganglionic output, since gallbladder neurons need to be synaptically driven to generate action potentials. In human tissue (94,101) as well as animal models (102), gallbladder muscle becomes desensitized to prostaglandins. With the contractile effect of prostaglandin diminished with time, chronic prostaglandin pro
Page 14
Figure 7 The concentration of CCK present in the serum following a meal is high enough to act in gallbladder ganglia but too low to directly activate gallbladder muscle. Graphs represent the CCKinduced increase in synaptic current in the guinea pig gallbladder and CCKinduced increase in tension of a guinea pig gallbladder muscle strip. Traces demonstrate the concentrationdependence of the CCKinduced facilitation of fast synaptic currents, elicited by fiber tract stimulation, in a guinea pig gallbladder ganglion. Each trace represents an average of five consecutive events. [From Ref. 103 and the data of Mawe (91), Harrington et al. (104), and Takahashi et al. (85).]
duction may contribute to gallbladder stasis by decreasing ganglionic output, therefore effectively denervating the tissue. V— Concluding Remarks The ganglionated plexus of the gallbladder is a target of modulatory inputs to the gallbladder, including nerves, hormones, and immune mediators. A schematic diagram depicting modulatory
Page 15
events in gallbladder ganglia is shown in Fig. 8. To release their neuroactive compounds onto the effector tissues of the organ, gallbladder neurons must be stimulated to fire action potentials, and the major source of excitatory input to these cells is vagal preganglionic fibers. Modulating inputs that can up or downregulate the efficacy of this nicotinic ganglionic transmission include CCK and norepinephrine, which have presynaptic excitatory and inhibitory effects on vagal terminals, respectively, and sensory fibers that can release SP and CGRP in gallbladder ganglia to depolarize and increase the excitability of gallbladder neurons. Immune mediators such as prostaglandin E2 may contribute to gallbladder hypomotility by hyperpolarizing gallbladder neurons and attenuating synaptic input. To be resolved in future studies is how gallbladder neurons, which express acetylcholine plus an assortment of excitatory and inhibitory neuromodulators, conduct clear signals to the smooth muscle and epithelial cells of the gallbladder.
Figure 8 Schematic illustration of the modulatory events that occur in the ganglia of the gallbladder. Vagal preganglionic inputs provide the main driving force to gallbladder neurons by activating nicotinic receptors to elicit fast EPSPS. The efficacy of this connection can be up or downregulated by CCK and sympathetic inputs, respectively, which act on presynaptic CCKA and 2 receptors to alter the amount of ACh released by the vagus nerves. An axon reflex exists in gallbladder ganglia in the form of sensory fibers that can release tachykinins and CGRP directly onto gallbladder neurons, resulting in their depolarization and increased excitability. Substance P and CGRP have both been shown to elicit prolonged depolarizations in gallbladder neurons, and slow EPSPs in gallbladder ganglia have been shown to be involve the release of tachykinins and the activation of neurokinin3 receptors. Prostaglandin E2 causes a prolonged hyperpolarization of gallbladder neurons as well as presynaptic inhibition of fast and slow EPSPS. Abbreviations: NE, norepinephrine; ACh, acetylcholine; CCK, cholecystokinin; CGRP, calcitonin generelated peptide; NKA, neurokinin A; PGE2, prostaglandin E2. Numbers in circles represent references that from which the information is derived. (Modified from Ref. 46.)
Page 16
Acknowledgments The studies performed in the Mawe laboratory have been supported by NIH grants DK 45410 and NS 26995. We thank the alumni members of the Green Mountain Gallbag Company, including Erin Talmage, Wendy Pouliot, Ellen Cornbrooks, David Wells, Lei Zhang, Kirk Hillsley, Audra Kennedy, and Alex Gokin for their contributions to many of the studies that have been described here. References 1. JP Ryan. Motility of the gallbladder and biliary tree. In: L. R. Johnson, ed. Physiology of the Gastrointestinal Tract. New York: Raven Press, 1987, pp 695–721. 2. M Costa, SJH Brookes. The enteric nervous system. Am J Gastroenterol 89:S129–S137, 1994. 3. MD Gershon, AL Kirchgessner, PR Wade. Functional anatomy of the enteric nervous system. In: L. R. Johnson, ed. Physiology of the Gastrointestinal Tract. New York: Raven Press, 1994, pp 381–422. 4. JD Wood. Physiology of the enteric nervous system. In: L. R. Johnson, ed. Physiology of the Gastrointestinal Tract. New York: Raven Press, 1994, pp 423–482. 5. PA Pellegrini, MG Patti. Motility of the Gallbladder and Bile Ducts and the Kinetics of Bile Flow. Philadelphia: Saunders, 1981. 6. WF Alexander. The innervation of the biliary system. J Comp Neurol 72:357–370, 1940. 7. W Burnett, FW Gairns, P Bacsich. Some observations on the innervation of the extrahepatic biliary system in man. Ann Surg 159:8–26, 1964. 8. SD Sutherland. The intrinsic innervation of the gallbladder in Macaca rhesus and Cavia porcellus. J Anat 100:261–268, 1966. 9. SD Sutherland. The neurons of the gallbladder and gut. J Anat 101:701–709, 1967. 10. K Kyösola. Cholinesterases of the gall bladder. Histochemistry 50:337–346, 1977. 11. W Cai, G Gabella. Innervation of the gallbladder and biliary pathways in the guinea pig. J Anat 136:97–109, 1983. 12. GM Mawe, MD Gershon. Structure, afferent innervation, and transmitter content of ganglia of the guinea pig gallbladder: relationship to the enteric nervous system. J Comp Neurol 283:374–390, 1989. 13. EK Talmage, GM Mawe. NADPHdiaphorase and VIP are colocalized in neurons of gallbladder ganglia. J Auton Nerv Syst 43:83–90, 1993. 14. EK Talmage, WA Pouliot, M Schemann, GM Mawe. Structure and chemical coding of human, canine and opossum gallbladder ganglia. Cell Tissue Res 284:289–302, 1996. 15. AJ Bauer, M Hanani, TC Muir, JH Szurszewski. Intracellular recordings from gallbladder ganglia of opossums. Am J Physiol 260:G299–G306, 1991. 16. EB Cornbrooks, WA Pouliot, GM Mawe. The structure of neurons and ganglia of the guinea pig gallbladder: light and electron microscopic studies. J Comp Neurol 317:31–44, 1992. 17. GM Mawe. Intracellular recording from neurones of the guineapig gallbladder. J Physiol (Lond) 429:323–338, 1990. 18. EK Talmage, WA Pouliot, EB Cornbrooks, GM Mawe. Transmitter diversity in ganglion cells of the guinea pig gallbladder: an immunohistochemical study. J Comp Neurol 317: 45–56, 1992. 19. RD De Giorgia, TT Zittel, JE Parodi, JM Becker, FC Brunicardi, VLW Go, NC Brecha, C Sternini. Peptide immunoreactivities in the ganglionated plexuses and nerve fibers innervating the human gallbladder. J Auton Nerv Sys 51:37–47, 1995. 20. JR Keast, JB Furness, M Costa. Distribution of certain peptide containing nerve fibres and endocrine cells in the gastrointestinal mucosa of five mammalian species. J Comp Neurol 236:403–422, 1985.
Page 17
21. S Uemura, S Pompolo, JB Furness, KJ Hardy. Nitric oxide synthase in neurons of the human gallbladder and its colocalization with neuropeptides. J Gastroenterol Hepatol 12:257–265, 1997. 22. RD De Giorgio, JE Parodi, NC Brecha, FC Brunicardi, JM Becker, VLW Go, C Sternini. Nitric oxide producing neurons in the monkey and human digestive system. J Comp Neurol 342:619–627, 1994. 23. GPS Siou, A Belai, G Burnstock. A developmental study of the localization of NADPHdiaphorase in the ganglionated plexus of the guineapig gallbladder. Cell Tissue Res 276:61–68, 1994. 24. Z Grozdanovic, B Mayer, HG Baumgarten, G Bruning. Nitric oxide synthasecontaining nerve fibres and neurones in the gall bladder and biliary pathways of the guineapig. Neuroreport 5:837–840, 1994. 25. Z Grozdanovic, HG Baumgarten, G Bruning. Histochemistry of NADPHdiaphorase, a marker for neuronal nitric oxide synthase, in the periopheral autonomic nervous system of the mouse. Neuroscience 48:225–235, 1992. 26. GM Mawe, EK Talmage, EB Cornbrooks, AP Gokin, L Zhang, LJ Jennings. Innervation of the gallbladder: structure, neurochemical coding, and physiological properties of guinea pig gallbladder ganglia. Microsc Res Tech 39:1–13, 1997. 27. J Sand, H Tainio, I Nordback. Neuropeptides in pig sphincter of Oddi, bile duct, gallbladder, and duodenum. Dig Dis Sci 38:694–700, 1993. 28. LE Goehler, C Sternini, NC Brecha. Calcitonin generelated peptide immunoreactivity in the biliary pathway and liver of the guineapig: distribution and colocalization with substance P. Cell Tissue Res 253:145–150, 1988. 29. PJ Davies, G Campbell. The distribution and colocalization of neuropeptides and catecholamines in nerves supplying the gall bladder of the toad, Bufo marinus. Cell Tissue Res 277:169–175, 1994. 30. LA Meldrum, JC Bojarski, J Calam. Effects of substance P and other neuropeptides on guineapig gallbladder muscle. Digestion 37:193–199, 1987. 31. C Dahlstrand, S Bjorck, R Edin, A Dahlstrom, H Ahlman. Substance P in the control of extrahepatic biliary motility in the cat. Regulatory Peptide 20:11–24, 1988. 32. CA Maggi, P Sannticioli, D Renzi, R Patacchini, C Surrenti, A Meli. Release of substance P and calcitonin generelated peptidelike immunoreactivity and motor response of the isolated guinea pig gallbladder to capsaicin. Gastroenterology 96:1093–1101, 1989. 33. YS Guo, P Singh, G Gomez, S Rajaraman, JC Thompson. Contractile response of canine gallbladder and sphincter of Oddi to substance P and related peptides in vitro. Dig Dis Sci 34:812–817, 1989. 34. KD Lillemoe, TH Webb, HA Pitt. Neuropeptide Y: a candidate neurotransmitter for biliary motility. J Surg Res 45:254–260, 1988. 35. J Ryan, S Cohen. Effect of vasoactive intestinal polypeptide on basal and cholecystokinin induced gallbladder pressure. Gastroenterology 73:870–872, 1977. 36. JP Ryan, S Ryave. Effect of vasoactive intestinal polypeptide on gallbladder smooth muscle in vitro. Am J Physiol 234:E44–E46, 1978. 37. C Dahlstrand, A Dahlström, H Ahlman. Adrenergic and VIPergic relaxatory mechanisms of the feline extrahepatic biliary tree. J Auton Nerv Syst 26:97–106, 1989. 38. M Mourelle, F Guarner, X Molero, S Moncada, J Malagelada. Regulation of gall bladder motility by the argininenitric oxide pathway in guinea pigs. Gut 34:911– 915, 1993. 39. ML McKirdy, HC McKirdy, CD Johnson. Nonadrenergic noncholinergic inhibitory innervation shown by electrical field stimulation of isolated strips of human gall bladder muscle. Gut 35:412–416, 1994. 40. LW Kline, PKT Pang. Nitric oxide modulates the calcitonin generelated peptideinduced relaxation in guinea pig gallbladder strips in vitro. Regul Pept 50:207– 212, 1994. 41. EA Shaffer. Gallbladder Disease. In: W. A. Walker and P. R. Dude, eds. Paediatric Gastroenterology. Philadelphiha: BC Becker, 1991, pp 1152–1170.
Page 18
42. RP Jazrawi, ML Petroni, N Prandini, C Paul, JA Adam, S Gullini, TC Northfield. Postporandial gallbladder motor function: refilling and turnover of bile in health and in cholelithiasis. Gastroenterology 109:582–591, 1995. 43. A Lanzini, RP Jazrawi, TC Northfield. Simultaneous quantitative measurements of absolute gallbladder storage and emptying during fasting and eating in humans. Gastroenterology 92:852–861, 1987. 44. K Hillsley, LJ Jennings, GM Mawe. Neural control of the gallbladder: an intracellular study of human gallbladder neurons. Digestion 59:125–129, 1998. 45. GM Mawe, AP Gokin, DG Wells. Actions of cholecystokinin and norepinephrine on vagal inputs to ganglionic cells in guinea pig gallbladder. Am J Physiol 267:G1146–G1151, 1994. 46. GM Mawe. Tachykinins as mediators of slow EPSPs in guineapig gallbladder ganglia: Involvement of neurokinin3 receptors. J Physiol (Lond) 485:513–524, 1995. 47. AP Gokin, LJ Jennings, GM Mawe. Actions of calcitonin generelated peptide in guinea pig gallbladder ganglia. Am J Physiol 34:G876–G883, 1996. 48. M Costa, S Brooks, S Waterman, R Mayo. Enteric neuronal circuitry and transmitters controlling intestinal motor function. In: G. E. Holle and J. D. Wood, eds. Advances in the Innervation of the Gastrointestinal Tract. Amsterdam: Elsevier Science, 1992, pp 115–121. 49. JB Furness, M Costa. The Enteric Nervous System. New York: ChurchillLivingstone, 1987. 50. JD Wood. Electrical and synaptic behavior of enteric neurons. In: S. G. Schultz, ed. Handbook of Physiology. Bethesda, MD: American Physiological Society, 1989, pp 465–518. 51. C Dahlstrand. The vagal nerves and peptides in the control of extrahepatic biliary motility. Acta Physiol Scand 139:1–52, 1990. 52. WJ Dodds, WJ Hogan, JE Geenen. Motility of the biliary system. In: S. G. Schultz, ed. Handbook of Physiology. Bethesda, MD: American Physiological Society, 1989, pp 1055–1101. 53. Cai G Gabella. Catecholaminecontaining cells in the nerve plexus of the guinea pig gallbladder. Acta Anat 119:10–17, 1984. 54. B Pallin, S Skoglund. Neural and humeral control of gallbladder emptying mechanism in the cat. Acta Physiol Scand 60:358–362, 1964. 55. CGA Persson. Adrenoceptor functions in the cat choledochoduodenal junction in vitro. Br J Pharmacol 42:447–461, 1971. 56. CGA Persson. Adrenergic, cholecystokinetic and morphine induced effects on extrahepatic biliary motility. Acta Physiol Scand (Suppl) 383:1–32, 1972. 57. CGA Persson. Dual effects of the sphincter of Oddi and gallbladder induced by stimulation of the right splanchnic nerves. Acta Physiol Scand 87:334–343, 1973. 58. T Yamasato, S Nakayama. Participation of the parasympathetic and sympathetic nerves in regulation of gallbladder motility in the dog. Acta Med Okayama 44:79–86, 1990. 59. GM Mawe. Noradrenaline acts as a presynaptic inhibitory neurotransmitter in ganglia of the guineapig gallbladder. J Physiol (Lond) 461:378–402, 1993. 60. GA Iwamoto, TG Waldrop, JC Longhurst, GA Ordway. Localization of the cells of origin for primary afferent fibers supplying the gallbladder of the cat. Exp Neurol 84:709–714, 1984. 61. CR Mantyh, TN Pappas, JA Lapp, MK Washington, LM Neville, JR Ghilardi, SD Rogers, PW Mantyh, SR Vigna. Substance P activation of enteric neurons in response to intraluminal clostridium difficile toxin a in the rat ileum. Gastroenterol 111:1272–1280, 1996. 62. L Zhang, AD Bonev, GM Mawe, MT Nelson. Protein kinase A mediates activation of ATPsensitive K+ currents by CGRP in gallbladder smooth muscle. Am J Physiol 267: G494–G499, 1994.
Page 19
63. LW Kline, PKT Pang. Calcitonin gene related peptide relaxes cholecystokinininduced contraction in guinea pig gallbladder strips in vitro. Can J Physiol Pharmacol 70:1571–1575, 1992. 64. AP Gokin, LJ Jennings, GM Mawe. Actions of calcitonin generelated peptide (CGRP) in guinea pig gallbladder ganglia. Am J Physiol 271:G876–G883, 1996. 65. A Thune, E Thorness, J Svanvik. Reflex regulation of flow resistance in the feline sphincter of Oddi by hydrostatic pressure in the biliary tract. Gastroenterology 91:1364–1369, 1986. 66. A Thune, L Jivegård, J Svanvik. Flow resistance in the feline choledochoduodenal sphincter as studied by constantpressure and constantperfusion techniques. Acta Physiol Scand 135:279–284, 1989. 67. A Thune, GTP Saccone, JP Scicchitano, J Toouli. Distension of the gall bladder inhibits sphincter of Oddi motility in humans. Gut 32:690–693, 1991. 68. AP Wyatt. The relationship of the sphincter of Oddi to the stomach, duodenum and gallbladder. J Physiol 193:225–243, 1967. 69. FS DuBois, GH Kistler. Concerning the mechanism of contraction of the gallbladder in the guinea pig. Proc Soc Exp Biol Med 30:1178–1180, 1933. 70. RTA Padbury, JB Furness, RA Baker, J Toouli, JP Messenger. Projections of nerve cells from the duodenum to the sphincter of Oddi and gallbladder of the Australian possum. Gastroenterology 104:130–136, 1993. 71. AL Kirchgessner, JE Pintar. Guinea pig pancreatic ganglia: projections, transmitter content, and the typespecific localization of monoamine oxidase. J Comp Neurol 305:613–631, 1991. 72. Z Itoh, I Takahashi, M Nakaya, T Suzuki. Interdigestive function of the gallbladder in the dog. In: W. Y. Chey, ed. Functional Disorders of the Digestive Tract. New York: Raven Press, 1983, pp 259–265. 73. AC Ivy, E Oldberg. A hormone mechanism for gallbladder contraction and evacuation. Am J Physiol 86:599–613, 1928. 74. KM Strah, TN Pappas, RL Melendez, HT Debas. Anticholinergic influence on exogenous and endogenous stimulation of gallbladder contraction. Gastroenterology 88:1601A, 1985. 75. L Marzio, AM DiGiammarco, M Neri, F Cuccurollo, P Malfertheiner. Atropine antagonizes cholecystokinin and cerulein induced gallbladder evacuation in man: a real time ultrasonographic study. Am J Gastroenterol 80:1–4, 1985. 76. N Hanyu, WJ Dodds, RD Layman, I Takahashi. Mechanism of cholecystokinininduced contraction of the opossum gallbladder. Gastroenterology 98:1299– 1306, 1990. 77. MI Grossman. Gastrointestinal hormones: Spectrums of Actions and StructureActivity Relations. Thorofare, NJ: Slack, 1975. 78. J Behar, P Biancani. Pharmacologic characterization of excitatory and inhibitory cholecystokinin receptors of the cat gallbladder and sphincter of Oddi. Gastroenterology 92: 764–770, 1987. 79. J Behar, P Biancani. The effect of cholecystokinin and the octapeptide of cholecystokinin on the feline sphincter of Oddi and gallbladder. J Clin Invest 66:1231– 1239, 1980. 80. RS Fisher, E Rock, LS Malmud. Cholinergic effects on gallbladder emptying in humans. Gastroenterology 89:716–722, 1985. 81. L Gullo, L Bolondi, P Priori, P Casanova, G Labo. Inhibitory effect of atropine on cholecystokinininduced gallbladder contraction in man. Digestion 29:209–213, 1984. 82. MJ Pozo, GM Salida, JA Madrid. Cholyecystokinininduced gallbladder contraction is influenced by nicotinic and muscarinic receptors. 97:403–408, 1989. 83. KM Strah, TN Pappras, RL Melendez, HT Debas. Contrasting cholinergic dependence of pancreatic and gallbladder responses to cholecystokinin. Am J Physiol 250:G665–G669, 1986.
Page 20
84. I Takahashi, T Suzuki, I Aizawa, Z Itoh. Comparison of gallbladder contraction induced by motilin and cholecystokinin in dogs. Gastroenterology 82:419–424, 1982. 85. T Takahashi, D May, C Owyang. Cholinergic dependence of gallbladder response to cholecystokinin in the guinea pig in vitro. Am J Physiol 261:G565–G569, 1991. 86. EA Brotschi, J Pattavino, LF Williams Jr. Intrinsic nerves affect gallbladder contraction in the guinea pig. Gastroenterology 99:826–830, 1990. 87. T Yamamura, T Takahashi, M Kusunoki, M Kantoh, Y Ishikawa, J Utsunomiya. Cholecystokinin octapeptideevoked [3H] acetylcholine release from guineapig gallbladder. Neurosci Lett 65:167–170, 1986. 88. WM Yau, ML Youther. Modulation of gallbladder motility by intrinsic cholinergic neurons. Am J Physiol 247:G662–G666, 1984. 89. A Rakovska, K Milenov, A Bocheva. Effect of cholecystokinin octapeptide and somatostatin on the motility of guinea pig and canine gallbladder. Comp Biochem Physiol 94C: 649–653, 1989. 90. JJ Galligan, PP Bertrand. ATP mediates fast synaptic potentials in enteric neurons. J Neurosci 14:7563–7571, 1994. 91. GM Mawe. The role of cholecystokinin in ganglionic transmission in the guineapig gallbladder. J Physiol (Lond) 439:89–102, 1991. 92. SI Myers, L Bartula. Human cholecystitis is associated with increased gallbladder prostaglandin I2 and prostaglandin E2 synthesis. Hepatology 16:1176–1179, 1992. 93. DL Kaminski, Y Deshpande, L Thomas, W Blank. Evaluation of the role of prostaglandins E and F in human cholecystitis. Prost Leukot Med 16:109–120, 1984. 94. JR Wood, SH Saverymuttu, AB Ashbrooke, IF Stamford. Effects of various prostanoids on gallbladder muscle. Adv Prost Thromb Res 8:1569–1571, 1980. 95. E Thornell, J Svanvik, JR Wood. Effects of intraarterial prostaglandin E2 on gallbladder fluid transport, motility, and hepatic bile flow in the cat. Gastroenterology 16:1083–1088, 1981. 96. SP Lee, JT LaMont, MC Carey. Role of gallbladder mucus hypersecretion in the evolution of cholesterol gallstones: studies in the prairie dog. J Clin Invest 67:1712–1723, 1981. 97. L Jivegard, A Thune, J Svanvik. Intraluminal prostaglandin E2 affects gallbladder function by activation of intramural nerves in the anesthetized cat. Acta Physiol Scand 132: 549–555, 1988. 98. K Nakata, Y Osumi, M Fujiwara. Prostaglandins and the contractility of the guinea pig biliary system. Pharmacology 22:24–30, 1981. 99. K Nakata, K Ashida, K Nakazawa, M Fujiwara. Effects of indomethacin on prostaglandin synthesis and on contractile response of the guinea pig gallbladder. Pharmacology 23:95–101, 1981. 100. LJ Jennings, GM Mawe. PGE2 hyperpolarizes gallbladder neurons and inhibits synaptic potentials in gallbladder ganglia. Am J Physiol 274:G493–G502, 1998. 101. CA Kotwall, AS Clanachan, HP Baer, GW Scott. Effects of prostaglandins on motility of gallbladders removed from patients with gallstones. Arch Surg 119:709–712, 1984. 102. WC Chapman, GA Peterkin, WW LaMorte, LF Williams. Alterations in biliary motility correlate with increased gallbladder prostaglandin synthesis in early cholelithiasis in prairie dog. Dig Dis Sci 34:1420–1424, 1989. 103. GM Mawe. Nerves and hormones interact to regulate gallbladder function. News in Physiological Sciences 13:34–90, 1998. 104. K Harrington, A Bomzon, KA Sharkey, JS Davison, EA Shaffer. Differential sensitivities of the sphincter of Oddi and gallbladder to cholecystokinin in the guinea pig: their role in transsphincteric bile flow. Can J Physiol Pharmacol 70:1336–1341, 1992.
Page 21
2— Gallbladder Mucosal Function J. Henriette Klinkspoor and Sum P. Lee Veterans Affairs Medical Center, Seattle, Washington I— Introduction The association between bile and the gallbladder has been appreciated ever since the seventeenth century, when Diemerbroek wrote that bile enters the gallbladder to "aquire greater strength and digestive power." This observation adequately expresses some of the main functions of the gallbladder. A great part of the hepatic bile secretion enters the gallbladder, where it is stored and concentrated between meals. While in the gallbladder, the composition of hepatic bile is changed by absorption and secretion by the gallbladder mucosa. Because of these storage and concentration functions, the gallbladder is one of several organs that are lined with an epithelium whose proper function requires a lumen solution markedly different in composition from extracellular fluid. This bile solution is derived at considerable metabolic expense by transport processes that take place in the hepatocytes of the liver. Bile is then concentrated in the gallbladder by an additional energyconsuming process. Bile is likely to remain in the gallbladder for several hours until it is evacuated to the intestine, where it participates in digestive events. Because of the very low level of most organic bile constituents in the fluids of the body, it is difficult to assess the concentration gradient of these constituents across the gallbladder wall. However, gradients of at least 10,000:1 have been estimated. To maintain these gradients, the gallbladder wall must represent a barrier of considerable integrity. In addition to restricting transepithelial diffusion of many substances, the gallbladder wall is also designed to facilitate net absorption of other substances in the concentrative process. This requires high selectivity of the epithelium to passage of the individual bile constituents. The primary barrier regulating gallbladder permeability is located in the epithelium rather than in the thicker subepithelial layers. Diamond (2) reviewed the early methods of studying gallbladder properties in detail. These early studies on the gallbladder epithelium established that it is a relatively leaky epithelium. The range and the relevance of the physiological functions that the gallbladder serves are not clearly known. The contractile or motor function of the smooth muscle in response to intestinal peptides, such as cholecystokinin, and regulation of hydrostatic pressure in the biliary tract are other important functions of the gallbladder. Several other properties of the gallbladder mucosa have been investigated and discussed. It is thought to have a role in the acidification of bile by means of the secretion of hydrogen ions, the absorption of cholesterol and other biliary lipids, as well as the absorption of bile pigments, amino acids, and sugars. In addition, the gallbladder secretes mucous glycoproteins, immunoglobulins, fluid, and electrolytes when suitably stimulated. This chapter describes the role of the gallbladder mucosa in modifying hepatic bile composition by means of its absorptive and secretory functions. Electrolyte and
Page 22
water absorption and secretion by the gallbladder will be discussed, as well as the regulation of this transport. Our current knowledge on the changes that occur in the absorptive functions of the gallbladder during gallstone formation and other biliary diseases is summarized. Secretion and absorption of biliary lipids by the gallbladder are discussed and, finally, the secretion of proteins and mucous glycoproteins by the gallbladder epithelium is examined. II— Gallbladder Morphology Although the biliary apparatus is lined by the same columnar epithelium from a common embryonic origin, there are morphological and functional differences among the terminal, intrahepatic, and extrahepatic cholangiocytes and the gallbladder epithelial cells. They also have different immunological markers (2). This single layer of epithelial cells is able to subserve vectorial transport of water, electrolytes, and macromolecules between the internal milieu and the biliary lumen. It is usually assumed that the transport properties of biliary epithelial cells vary according to their anatomic location. Intrahepatic biliary epithelial cells are mainly committed to secretion, whereas gallbladder epithelial cells are thought to be specialized in absorption. However, it is now known that ductal and gallbladder epithelial cells are capable of absorbing and secreting fluid and electrolytes. In addition, extrahepatic biliary epithelial cells are able to synthesize and secrete mucins by directed exocytosis. The different transporting capacities of biliary epithelial cells are not associated with significant structural differences. Ultrastructural studies have shown that, apart from their progressive increase in size along the biliary tract, all biliary epithelial cells exhibit similar but not identical morphological features. The most characteristic trait of gallbladder epithelial cells is their high degree of structural polarization, evidenced by the asymmetrical distribution of their intracellular organelles and by the polarized organization of their plasma membrane, divided into two distinct domains, respectively apical and basolateral. The apical membrane (facing the lumen) contains microvilli, which are more or less prominent in different species. The basolateral membrane (facing the serosa) has a basal region that is anchored to the basement membrane by hemidesmosomes and a lateral region that is frequently interdigitated with the membranes of the neighboring cells, lining a convoluted lateral intercellular space. The two membrane domains are separated by tight junctions. Tight or occluding junctions join epithelial cells together to form a sheet, enabling them to have a selective permeability barrier and preventing watersoluble molecules from leaking between the cells. Tight junctions also serve to prevent the entry of apical membrane structures and proteins into the basolateral domain of the cell membrane, and vice versa (3). The subepithelial layer of the gallbladder contains connective tissue, smooth muscle, blood vessels, and the serosa. Transport across the epithelium in vivo occurs into or from the underlying capillaries; the blood compartment functions as a sink for the transported water or solutes. Altered cell biological behavior of the gallbladder epithelium is believed to contribute to the pathogenesis of gallstones, hepatobiliary disease, cystic fibrosis, and various other diseases including carcinogenesis. However, despite extensive research into the physiology and pathophysiology of the biliary tract, little is known about the role of the gallbladder epithelium in the pathogenesis of biliary tract disease. In vitro techniques are required for the further study of the pathological and physiological properties of the gallbladder wall and epithelium. Epithelial cell cultures represent one of the useful in vitro approaches. Several methods of monolayer and threedimensional cell culture of biliary epithelial cells from normal and pathological biliary tract are now available. In a recent review (4), several representative models for the preparation and isolation of gallbladder epithelial cells and their use in monolayer and threedimensional cell culture were discussed. Successful primary and longterm culture of canine (5), mouse (6–8), bovine (9), and human (10–13) gallbladder epithelial cells has been reported. Tissue culture using explants, an interesting alternative to cell culture, has mainly been used
Page 23
for physiological studies of the gallbladder, such as studies on the transport of water and electrolytes. III— Water and Electrolyte Transport A— NaCl Transport: Na+/H+ and Cl/HCO3 Exchange The main transport function of the gallbladder epithelium is the absorption of NaCl and water in nearisosmotic proportions. This results in concentration of the impermeant components of bile in the lumen of the gallbladder. This involves apical membrane entry of Na+ and Cl and basolateral membrane extrusion of both ions. The presence of Na+ in the lumen is necessary for fluid absorption, since bathing of the gallbladder in vitro in a balanced buffer solution that contains K+ substituted for Na+, resulted in the complete cessation of net volume and electrolyte absorption. However, the requirement for Cl is not absolute. The precise mechanisms of net entry of Na+ and Cl at the apical membrane are not fully agreed on, although it is accepted that entry is electroneutral, involving carriermediated cotransport or countertransport. The following possibilities have been proposed: (a) NaCl cotransport; (b) NaKCl2 cotransport; (c) double Na+/H+, anion exchanger was recently demonstrated in the apical membrane of gallbladder epithelial cells, where it is possibly involved in the regulation of bicarbonate secretion into bile (15). Another recent study demonstrated the presence of the CLC3 Cl channel, a member of the CLC family of chloride channels, which includes several molecular isoforms with tissuespecific distributions, in human gallbladder epithelium. Alterations in CLC3 activity may alter epithelial Cl permeability and influence gallbladder electrolyte and fluid transport (16). Although quantitatively less important than NaCl absorption, the gallbladder epithelium also secretes K+ and H+. For a long time it has been known that the pH in bile declines in the gallbladder lumen. Acid secretion by the human gallbladder has important implications for gallstone formation, because the majority of gallstones contain calcium carbonate and changes in the pH of bile are of critical importance in influencing the solubility of calcium in bile (17). Acid secretion by the gallbladder epithelium could be due to either absorption of bicarbonate ions or a secretion of hydrogen ions. It has now been demonstrated that the human gallbladder mucosa secretes hydrogen ions by means of an active Na+/H+ pump in the apical membrane. Using the Ussing chamber method, Plevris et al. (18) showed that viable human gallbladder mucosa is capable of acidifying physiological solutions in vitro. The hydrogen concentration on the mucosal side of gallbladder tissue was observed to increase, whereas simultaneously the hydrogen concentration in the serosal compartment decreased, suggesting a transfer of hydrogen ions from the serosal to the mucosal side of the tissue. The concomitant decrease of bicarbonate concentration with increased in the mucosal side, indicates that the acidification was not simply due to an absorption of bicarbonate. Acidification was abolished when
Page 24 +
+
the gallbladder epithelium was exposed to sodiumfree solution or in the presence of high concentrations of amiloride, a specific Na /H transport inhibitor, in the mucosal compartment, suggesting that hydrogen secretion in the human gallbladder depends on Na+/H+ antiport. The actual presence of a Na+/H+ exchanger in the gallbladder epithelium has now been demonstrated. Mucosa of human gallbladder was found to contain mRNA of the NHE3 isoform of the Na+/H+ exchanger (19). Subsequent studies failed to demonstrate the presence of mRNA for the NHE2 isoform in human gallbladder. In situ hybridization experiments demonstrated that the NHE3 mRNA was strictly localized to the gallbladder epithelial cells. Synthesis of the NHE3 isoform of the Na+/H+ exchanger suggests that this isoform plays an important role in water and electrolyte absorption by the gallbladder (20). Results showing that cAMP inhibits sodium absorption by the gallbladder (21) are consistent with a major role of the NHE3 isoform, since it has been demonstrated that, unlike the NHE1 and NHE2 isoforms, NHE3 activity is decreased by an increase in intracellular cAMP. Basolateral membrane Na+ exit is mediated by the Na+, K+activated ATPase. The importance of Na+ transport was demonstrated by the use of ouabain to inhibit Na+, K+ATPase, which prevented net water flow. The activity of the Na+, K+ATPase pump correlates directly with the rate of fluid transport. The basolateral membrane also has a K+ conductive pathway, across which part of the K+ transported inward by the pump is recycled. The mechanism of Cl transport from cell to basolateral solution has not been fully resolved but appears to result from both conductive transport and electroneutral KCl cotransport (14,22–24). A schematic representation of gallbladder electrolyte and fluid transport is shown in Fig. 1. B— Other Electrolyte Transporters Hepatobiliary complications in cystic fibrosis result predominantly from lesions of the biliary epithelium. These abnormalities affect the intrahepatic as well as the extrahepatic bile ducts and the gallbladder. The protein cystic fibrosis transmembrane conductance regulator (CFTR), the gene product defective in cystic fibrosis, functions as a cAMP activated chloride channel in the plasma membrane. As such, it may represent an important driving force for fluid transport across the gallbladder epithelium. The CFTR protein was detected by immunolocalization in human gallbladder tissue sections and was found to be predominantly localized to the apical membrane of the epithelial cells. RTPCR was used to demonstrate the presence of CFTR mRNA in freshly isolated and cultured human gallbladder epithelial cells. High levels of CFTR protein were maintained in gallbladder epithelial cells, as demonstrated by Western blotting and immunoprecipitation. Chloride efflux in these cells could be stimulated by Ca2+dependent pathways and more intensely by cAMPdependent pathways. Vasoactive intestinal peptide and secretin stimulated chloride efflux in vitro. The cAMPmediated chloride efflux was inhibited by chloride channel inhibitors (25). Subsequent studies, using Western blotting, RTPCR, and Ussingchamber experiments, revealed the presence of CFTR protein in normal cultured mouse gallbladder epithelial cells (6,8). However, gallbladders from CFTR( /) knockout mice lacked the cAMP induced chloride current observed in normal gallbladders. In fluid transport measurements, normal and CFTR knockout gallbladders were equally active in basal resorption. The addition of forskolin, which activates CFTR anion channel activity through the cAMP system, resulted in net fluid secretion in the normal mouse gallbladders. In contrast, CFTR (/) gallbladders were unable to secrete fluid while a complete inhibition of resorption by forskolin was observed. Therefore it was concluded that in normal mouse gallbladder epithelium, cAMPinduced fluid secretion involves simultaneous inhibition of apical sodium chloride resorption and activation of CFTR (6,7). These findings indicate a potential role for CFTR in the pathophysiology of the gallbladder epithelium. In the small intestine and the airway epithelium, CFTR has been shown to regulate sodium absorption, and CFTR (/) was associated with hyperabsorption of sodium (26). Whether this applies to the gallbladder epithelium has not been studied.
Page 25
Figure 1 Ion transport systems in gallbladder epithelium. A model for isosmotic fluid resorption in the gallbladder as suggested by Reuss et al. for various species (23). Direction of fluxes observed in steady state are depicted. Apical membrane NaCl entry is via parallel Na+/H+ and dissociates as CO2 and H2O. Basolateral Na+ extrusion is via the Na+, K+ATPase pump. Basolateral Cl exit is by KCl cotransport and a Cl conductive pathway, and apical Cl exit can occur via the cystic fibrosis transmembrane conductance regulator (CFTR). There are selective K+ channels in both membranes, but K+ recycles mostly at the basolateral border, which has a much higher K+ conductance.
A recent immunohistochemical study revealed that besides the basolateral Na+, K+ATPase membrane transporter and the apically located CFTR transporter, human gallbladder epithelial cells also express the MDR1 transporter on their apical membranes. Involvement of the MDR1 transporter in fluid transport has been postulated. Interestingly, CFTR and MDR1 were found to be colocalized in the apical domain of all biliary cells, including gallbladder epithelial cells. This finding contrasts with the fact that in most other epithelia, CFTR and MDR1 show complementary patterns of expression. CFTR and MDR1 are expressed predominantly in secretory and absorptive cells, respectively. Their coexpression in the gallbladder, therefore, supports the notion that the gallbladder is capable of both secretory and absorptive transport (15). C— Water Transport Concentration within the gallbladder is a consequence of the removal of water from the lumen, and it is now accepted that this concentration of hepatic bile secreted by the liver is a result of one of the highest rates of water absorption reported. The rate of fluid absorption has been
Page 26 2
measured in several species and ranges from 5 to 80 l/cm per hour. Transport rates are higher in mammals than in other vertebrates. Water is absorbed in isosmotic proportions, and this can occur against its electrochemical gradient. Water absorption by the gallbladder is always coupled to salt transport. The precise mechanism of water transport is not known, but the predominant view is that the water flow is osmotically coupled to NaCl transport. It is believed that salt transport causes small osmotic gradients across both cell membranes, making the cell interior hyperosmotic to the mucosal solution and hypoosmotic to the fluid in the lateral intercellular spaces. The resulting elevation of the hydrostatic pressure in the lateral spaces causes the solution to flow towards the subepithelial space (22–24). D— Regulation of Electrolyte and Water Transport Since many of the transport mechanisms accounting for baseline electrolyte and water transport in the gallbladder epithelium have now been characterized, recent studies have focused on the regulation of transport. The rate of transepithelial ion absorption is the result of integrated activity of transporters in the apical and basolateral cell membranes. A variety of mediators affect the rate of electrolyte and water transport by the gallbladder. Some of these could act to regulate gallbladder water and electrolyte transfer under physiological conditions by modification of NaCl influx, active Na+ extrusion, and/or junctional permeability. In gallbladder epithelium, the best understood regulatory mechanisms involve intracellular factors such as pH, Ca2+, and cAMP. Changes in intracellular levels of these and other agents mediate the effects of peptides, hormones, and neurotransmitters on salt transport. For example, secretin, glucagon, and vasoactive intestinal peptide inhibit fluid absorption by the gallbladder mucosa, whereas cholecystokinin and gastrin are without effect. Of the regulatory peptides mentioned above, only VIP and secretin are able to modify gallbladder fluid transport at physiological concentrations. Other peptides, however, may act to potentiate or inhibit the effects of other regulatory factors. As discussed in detail below, prostaglandins play an important role in the regulation of water and electrolyte transport by the gallbladder epithelium. They have also been shown to alter the normal process of water absorption and induce net secretion. Elevating intracellular cAMP levels decreases the rate of fluid absorption by the gallbladder and, in some species, can elicit net secretion. A number of agents including prostaglandins, secretin, vasoactive intestinal peptide, bradykinin, and vasopressin elevate cAMP levels, which suggests that this mechanism is responsible for the effect of these agents on salt and water transport. The predominant effects of cAMP on gallbladder epithelial cells are exerted at the apical membrane and consist of activation and/or insertion of Cl channels, such as CFTR and inhibition of the apical Na+/H+ exchanger and the apical secretion. The current information available on the regulation of specific transporters and the means by which regulation is integrated to determine the overall rate of fluid transport has been extensively reviewed (14,22–24,27). E— Electrolyte and Water Transport during Gallstone Formation Recent studies suggest that increased absorption of electrolytes and water across the gallbladder epithelium occurs during the early stages of experimentally induced gallstone formation. Moreover, altered transepithelial absorption has been implicated in the pathogenesis of both cholesterol and pigment gallstones. Increased gallbladder absorption may play an important role, contributing to gallstone formation by increasing the concentration of absorbable constituents of bile. These constituents may serve as either components of gallbladder sludge, a precursor of gallstones, or as pronucleating factors promoting cholesterol nucleation and growth.
Page 27
Ionic calcium has been suggested to serve as an important mediator of intestinal absorption. Moreover, calcium channel antagonists have also been documented to alter intestinal water and electrolyte flux. Until recently, however, little information was available with respect to alterations in extracellular calcium or the effect of calcium channel antagonists on gallbladder absorption. Gallbladder bile concentrations of total and ionized calcium have been reported to be increased in human beings with gallstones and increased biliary calcium levels have been found in numerous experimental models of both cholesterol and pigment gallstones. Biliary calcium has been suggested both to play a structural role in the formation of gallstones and to contribute to the nucleation of cholesterol crystals by serving as a pronucleating agent. Therefore, Scheeres et al. (28) investigated whether extracellular calcium modulates gallbladder absorption. They demonstrated that in the rabbit gallbladder, alterations in serosal extracellular calcium concentration did not significantly affect gallbladder absorption. The calcium channel antagonist verapamil, however, significantly decreased gallbladder absorption, suggesting that calcium channels may mediate this process in the gallbladder. Changes in extracellular calcium were demonstrated to affect ion transport across the gallbladder epithelium of the prairie dog, which has emerged as an important animal model for the study of human cholesterol gallstone disease. Prairie dog hepatic and gallbladder bile compositions are similar to those of humans. Furthermore, prairie dogs maintained on a cholesterolrich diet develop cholesterol gallstones in a manner that recapitulates events known to occur in humans with cholelithiasis. The gallbladder epithelium of the prairie dog is electrogenic and resembles that of humans. However, in contrast to gallbladders of most other species, the prairie dog gallbladder epithelium simultaneously but independently absorbs Na+ and secretes Cl (29). Cates et al. demonstrated—by exposing gallbladders to dantrolene, which traps calcium within intracellular organelles, and nickel, which prevents influx of extracellular calcium—that the effects of extracellular calcium on prairie dog gallbladder ion transport are mediated by changes in intracellular calcium (30). Recently, apical and basolateral Ca2+ channels were demonstrated to be present in the gallbladder epithelium (31). Further investigation into the pathophysiology of these cation channels should yield useful information regarding the control of calcium flux. Subsequent studies were aimed at elucidating the mechanism by which changes in intracellular calcium regulates gallbladder ion transport. Prairie dog gallbladders, mounted in Ussing chambers were exposed to trifluoperazine, a potent agonist of Ca2+calmodulin, a receptor protein in the Ca2+ messenger system. In addition, the ion transport effects of increased extracellular calcium were determined in the presence of calmodulin inhibition. Inhibition of calmodulin resulted in an increase in net Na+ and water absorption. The effects of trifluoperazine could be reversed by increasing luminal Ca2+; therefore it was concluded that Ca2+calmodulin regulates basal gallbladder absorption in the prairie dog gallbladder (32) (Fig. 2A). However, when the intracellular calcium concentration in prairie dog gallbladder was increased by exposing the gallbladders to the calcium ionophore A23187, mucosaltoserosal Cl flux was inhibited and serosaltomucosal flux of Na+ was stimulated, resulting in increased net Cl secretion and decreased net Na+ absorption. A23187 converted gallbladder water absorption to secretion. The effect of A23187 was delayed by pretreatment with indomethacin, suggesting a prostaglandindependent mechanism (33). This is the first study demonstrating that increased intracellular calcium converts the gallbladder from its normal absorptive state to a secretory one. Next, the authors investigated whether protein kinase C is also essential in the regulation of gallbladder ion transport in this model. Activation of protein kinase C by phorbol esters resulted in inhibition of Na+ absorption and a stimulation of Cl secretion by the gallbladder epithelium. Similar effects were seen with serotonin, a hormone mediating its effects through the phosphoinositide/PKC pathway. Pretreatment of the tissues with H7, a very potent inhibitor of protein kinase C, blocked the inhibitory effect of phorbol esters on ion transport. Due to the changes in Na+ and Cl transport, mucosaltoserosal water flux was diminished, while serosaltomucosal flux remained essentially the same, causing a change from net water absorption to secretion (34) (Fig. 2B). It is apparent from these studies that intracellular calcium mediates its regulation of gallbladder ion transport through the Ca2+ calmodulin complex and
Page 28
Figure 2 Role of Ca2+ in the regulation of ion transport in gallbladder epithelium. Regulation of basal gallbladder absorption by calmodulin (CaM) in prairie dog gallbladder as suggested0160by Moser et al. (32). Effects of biliary Ca2+ are mediated by changes in intracellular Ca2+ ([Ca2+]i). In the presence of free Ca2+, calmodulin binds to inactive calmodulindependent protein kinase (CaMPK), converting it to the active Ca2+calmodulinbound form. Active calmodulindependent protein kinase phosphorylates mucosal Na+/H+ and exchange mechanisms, downregulating Na+ and Cl absorption (Fig. 2A). Mechanism of protein kinase C (PKC)induced inhibition of ion transport in prairie dog gallbladder epithelium as proposed by Cates et al. (34). Agonist binds to its receptor and together they activate guanine nucleotidebinding protein (G protein), which stimulates phospholipase C (PLC). Phospholipase C hydrolyzes membranebound phosphatidylinositol 4,5bisphosphate (PIP2) to produce inositol 1,4,5triphosphate (IP3) and diacylglycerol (DAG). 1,4,5Triphosphate then mobilizes Ca2+ from intracellular stores. Diacylglycerol binds to the regulatory subunit of inactive protein kinase C, increasing its affinity for Ca2+ and converting it to active protein kinase C. Active protein kinase C phosphorylates mucosal Na+/H+ and exchangers, resulting in inhibition of mucosaltoserosal fluxes of Na+ and Cl (Fig. 2B).
Page 29
protein kinase C, the two major systems through which intracellular calcium carries out its secondmessenger functions in many cell types. The results suggest that Ca2+calmodulin regulates gallbladder ion transport at basal intracellular calcium levels, whereas protein kinase C regulates transport at elevated calcium levels. Therefore it can be hypothesized that changes in biliary calcium during gallstone formation might result in alterations in gallbladder absorption. This was investigated in a study that sought to correlate gallbladder Na+ and Cl fluxes with biliary lipid composition during the various stages of gallstone formation. Prairie dogs were fed a standard or cholesterolrich diet for 4 to 21 days. Hepatic and gallbladder bile was analyzed for lipids and Ca2+. Animals were designated either precrystal, crystal, or gallstone based on the absence or presence of crystals or gallstones, respectively. Gallbladders were then mounted in Ussing chambers and unidirectional Na+ and Cl fluxes were measured. Na+ absorption was found to be increased during the precrystal stage but decreased during the gallstone stage due to increased serosato mucosa flux and mucosatoserosa flux, respectively. Increased serosatomucosa flux of both Na+ and Cl characterized the crystal stage. Biliary lipids (total bile acids, phospholipids, and total lipids) increased progressively during various stages of gallstone formation and correlated positively with fluxes of Na+ and Cl. Although these correlations do not indicate the causeandeffect relationships between biliary lipids and gallbladder ion transport, these relationships suggest that individual biliary moieties may play an important role in regulating transport of specific ions during gallstone formation. Also a significant positive correlation between Na+ and Cl fluxes and Ca2+ concentration was observed. It is possible that the increased concentrations of biliary lipids and Ca2+ directly modulate gallbladder ion transport, thereby promoting gallstone formation (35). Transepithelial Na+ transport in prairie dog gallbladder occurs via Na+/H+ exchange at the apical membrane followed by extrusion at the basolateral membrane via Na+, K+ATPase. Gallstone formation is accompanied by a significant decrease in net Na+ absorption, due primarily to the increase in serosatomucosa Na+ flux, with loss of the normal Na+ gradient. This finding is consistent with the described downregulation of Na+, K+ATPase in gallstone animals (29). F— Absorption versus Secretion The gallbladder is conventionally regarded as an absorptive organ. However, studies by Igimi et al. (36), showed that the gallbladder secretes water and electrolytes into the lumen during periods of digestion. They studied 35 patients who had recovered from a percutaneous trans
Page 30
hepatic gallbladder drainage performed for acute cholecystitis. After an overnight fast, gallbladder bile was dark brown in color and had a wide scatter in the lipid composition. Two hours after a meal, the gallbladder bile was opalescent white in color and had the composition of an extracellular fluid. This phenomenon was uniformly observed in all patients. Using normal dog gallbladder epithelial cells, Igimi et al. studied sodium flux. Control cells were demonstrated to absorb sodium, but the addition of secretin resulted in a reversal of sodium flux and net sodium secretion. The investigators concluded that secretion is a physiological function of the gallbladder mucosa. They suggested that after feeding, neural and humoral factors induce active de novo secretion, thus producing a gallbladder bile that is opalescent white with no lipids. These findings also challenge the accepted view of the origin of the pathological ''white bile." It is believed that white bile is the result of impaction of gallstones into the cystic duct, with the pigments, lipids, and bile salts absorbed by the mucosa, leaving behind a whitish fluid. This study suggests that white bile might be the result of active secretion and not the remnants of selective reabsorption. These observations were made in patients with patent bile ducts communicating with the gallbladder. Theoretically, the fluid could have come from the bile ducts, and therefore the interpretation that this is de novo gallbladder secretion has been challenged (37). In a subsequent study, Glickerman et al. (38) investigated a patient with multiple bile duct strictures whose gallbladder was excluded from the extrahepatic ducts. The patient required separate drainage of his gallbladder and common hepatic duct, thus allowing separate yet simultaneous analysis of gallbladder and hepatic secretions. In doing so, the authors were able to confirm their previous observation that the gallbladder produces a clear, colorless fluid. Although hepatic bile flow was continuous, gallbladder drainage was intermittent, occurring only after meals, and the volume was variable. The gallbladder fluid was rich in protein, with mucin accounting for more than 60% of the protein. The fluid had no bilirubin, bile salts, cholesterol, or phospholipids and had the ionic profile of an extracellular fluid. The secretion was found to be alkaline and contained abundant bicarbonate. In addition to demonstrating that the gallbladder can be a secretory organ, these observations raise other issues concerning the pathophysiology of biliary tract disorders. IV— Biliary Lipids An absorption of lipids by the gallbladder has been claimed. Several observations support such a contention. The guinea pig gallbladder was shown to absorb significant amounts of cholesterol, bile acids, and phospholipids. Using model biles, the rate of cholesterol absorption was found to correlate with biliary saturation rather than biliary cholesterol concentration. The absorption showed a linear increase as saturation increased and reached a maximum when model bile became saturated with cholesterol. Metabolic inhibitors and colchicine, a microtubule inhibitor, reduced uptake and serosal secretion, demonstrating that the serosal secretion is not simply a product of passive cholesterol exchange between bile and mucosa and mucosa and serosal fluid (39). The rate of cholesterol absorption exceeds that of the other biliary lipids, which should render the gallbladder contents less saturated. The human gallbladder was also demonstrated to absorb cholesterol. Using in vitro and in vivo experiments, it was shown that radioactive cholesterol from model bile and gallbladder bile enters the mucosal cells of human gallbladder (40,41). Infusion of a mixture of bile acids and cholesterol into the damaged gallbladders of dogs resulted in an increased cholesterol concentration of the mixture and a decrease of the bile acid concentration. And dietary cholesterol alters biliary as well as gallbladder mucosal cholesterol concentration. A selective absorption of cholesterol and phospholipids within the gallbladder has been implied. However, the possible exchange of cholesterol between gallbladder and bile and the net movement of cholesterol in one direction have not been studied. More recent studies suggest a bidirectional flux of cholesterol between biliary cholesterol carriers and gallbladder epithelial cells. Hayashi et al. (42), using cultured dog gallbladder epi
Page 31
thelial cells, demonstrated that cholesterol in the apical membrane bilayer of the epithelial cells exchanged readily with that in bile, but only in the presence of bile salts. The rate of exchange was found to be dependent on the concentration and species of bile salts. The net gain of cholesterol (absorption) or net loss of cholesterol (cytotoxicity) exhibited by the epithelial cells was regulated by the thermodynamic stability of cholesterol and the detergent effect of mixed micelles in bile. Therefore, it cannot be excluded that the physicochemical composition of lipids in bile may modify the cellular function of the gallbladder epithelium by directly influencing the lipid composition in the gallbladder mucosa. A bidirectional traffic of lipids may be the mechanism whereby the gallbladder epithelial cells can sense and respond to a change in the chemical composition of its milieu. A net gain or loss of cholesterol in the membrane of the gallbladder epithelial cells may influence the fluidity of the membrane and perturb intramembrane enzyme or receptor functions. A number of cell biological functions—such as fluid and electrolyte transport and mucin secretion—may therefore be profoundly altered as a result. A recent study on the relative efficiencies of human gallbladder mucosal absorption of fluid and lipids in health and disease demonstrates the possible importance of lipid absorption by the human gallbladder epithelium. Biliary lipids and pigment content were measured in fasting gallbladder bile samples obtained from gallstonefree controls and from four study groups: multiple and solitary cholesterol gallstone patients and morbidly obese subjects with and without gallstones. Bile salts and pigment content were found to be significantly greater in gallstonefree controls than in all other study groups, suggesting a more effective gallbladder mucosal fluid absorption in controls. Correlation plot analyses of biliary lipids showed lower concentrations of phospholipids and cholesterol than expected from the index bile salt concentrations. These findings were more pronounced in gallstonefree controls and were interpreted as evidence of more efficient gallbladder absorption of biliary lipids in controls. The authors concluded that efficient gallbladder mucosal absorption of both fluid and lipids from bile is a normal physiological process that is often seriously impaired during cholesterol gallstone disease (43). V— Bile Pigments Because in normal function the gallbladder maintains a considerable concentration gradient of bile pigments across its wall, the permeability of the mucosa to these organic substances is of some interest. Absorption of unconjugated bilirubin from bile proceeds much faster than absorption of the pigment in conjugated form. Bilirubin absorption is not reduced by metabolic inhibitors even though fluid absorption is reduced. Absorption is linearly related to the concentration of the pigment in the luminal solution, which indicates that loss of this pigment from the gallbladder proceeds by simple diffusion (44). VI— Amino Acids and Sugars It has been demonstrated that there is active absorption of amino acids and sugar by the dog, guinea pig, and human gallbladder. The unidirectional transepithelial flux of glycine from mucosa to serosa is severalfold greater than the oppositely directed flux, and tissue accumulation of glycine follows saturation kinetics. Lysine is accumulated in the mucosa to a concentration 20 times that in bathing media, whereas the presence of another dibasic amino acid, arginine, inhibits the process. Uptake of sugars and amino acids is remarkably reduced by tissue incubation in Nafree bathing solutions or by exposure to metabolic inhibitors or ouabain. The present information suggests that the gallbladder mucosa has the capacity to conserve much of the sugar and amino acid present in hepatic bile by an active transport mechanism (44).
Page 32
VII— Mucins Among the major secretions of the gallbladder epithelium are mucous glycoproteins. Gallbladder mucin is presumed to act as a protective agent against molecules such as the membrane toxic bile acids and lysophosphatidylcholine. However, besides having a protective function, gallbladder mucin has been reported to stimulate cholesterol nucleation, crystal growth, and crystal aggregation. The role of gallbladder mucin in the pathogenesis of gallstones has been the subject of review (45) and is also discussed in Chapter 10 of this volume. During the last few years a vast amount of new knowledge has been acquired about the regulation of mucin secretion by the gallbladder epithelium as well as about the variety of mucin genes that are expressed in the gallbladder mucosa. A lot of this information has been the result of the study of cultured gallbladder epithelial cells derived from different species. Several intracellular signaling pathways seem to be involved in the regulation of mucin secretion. Besides the protein kinase A (cAMP) pathway (46), a calciumdependent pathway, implicating Ca2+calmodulin kinase II and protein kinase C, has been described (47). Although different results have been reported, depending on the species studied, mucin secretion by the gallbladder epithelium seems to be stimulated by a variety of substances. Compounds that cause an increase in intracellular cAMP (VIP, adrenaline, isoproterenol, prostaglandins) have been shown to cause an increase in mucin secretion in the dog (46) and guinea pig gallbladder but not in the prairie dog. Whether an increase in cAMP also stimulates mucin secretion in human gallbladder remains controversial. Prostaglandins, adrenaline, and isoproterenol were demonstrated to cause an increase in cAMP in one culture system (13), but forskolin, secretin, and VIP did not significantly stimulate mucin secretion in another (47). Recently, the CFTR protein has also been implicated in the regulation of mucin secretion by the gallbladder epithelium as well as other epithelia. Using a retroviral vector to overexpress the CFTR protein in cultured dog gallbladder epithelial cells, Kuver et al. (48) demonstrated a fourfold increase in constitutive mucin synthesis and secretion by these cells. However, in a similar mouse model, no evidence for a link between mucin secretion and CFTR activity was found. Agonists of cAMP stimulated chloride efflux in cultured mouse gallbladder epithelial cells, but cAMP, Ca2+, and protein kinase C agonists did not cause an increase in mucin secretion by cultured mouse gallbladder epithelial cells (6). An increase in intracellular calcium stimulated mucin secretion in the guinea pig but not in the dog or prairie dog gallbladder. Bile salts were found to be stimulants of mucin secretion in dog gallbladder (49). More recently, bile salts were demonstrated to also cause an increase in mucin secretion by human gallbladder epithelial cells via a cytosolic calcium increase and Ca2+CaM kinase II. Mucin secretion in these human gallbladder epithelial cells could be stimulated by ionomycin and phorbol ester and was found to be predominantly regulated by a calciumdependent pathway, implicating Ca2+CaM kinase II and protein kinase C. ATP, UTP, UDP, and ADP also stimulated mucin secretion, whereas extracellular adenosine had no effect, implicating P2u purinergic receptors (47). In recent years considerable advances have been made in our knowledge of human mucin genes. Although analysis of their genomic organization is still in progress; the pattern of their expression in different human mucosa is now fairly well established. However, little was known until recently about their expression in the biliary tree. Biliary epithelial cells are subjected to an especially toxic environment; the bile. The role of biliary mucins is likely to ensure cytoprotection of these epithelial cells. Northern blot studies demonstrated the presence of MUC1, MUC3, MUC4, MUC5AC, and MUC5B mRNAs in cultured human gallbladder epithelial cells, whereas MUC2 mRNAs were barely detectable (50). Vandenhoute et al. (51) determined the pattern of expression of the different human mucin genes in gallbladder epithelial cells, intrahepatic bile ducts, and liver by means of Northern blot and in situ hybridization. Biliary epithelial cells showed a strong mRNA expression of MUC3, MUC6, and MUC5B and a weak expression of MUC1, MUC5AC, and MUC2. No expression of MUC4 and MUC7 was observed. Surprisingly, MUC3, which was the gene most strongly expressed in the biliary tree, was also found in hepatocytes. Electron microscopy shows no indication that hepatocytes secrete mucin granules. This result suggests that possibly
Page 33
MUC3 may not only be a secreted mucin but also might be anchored into the cell surface membrane and might have another role apart from mucus gel formation. VIII— Proteins Electron microscopic studies have demonstrated a bloodtobile transport pathway, which could represent a route of entry to the gallbladder lumen for various blood borne macromolecules. For example, horseradish peroxidase was shown to permeate the basement membrane and entered the lateral intercellular space, after which there was a vesicular transport to the lumen. A selective reabsorption of proteins from bile by the gallbladder also has been suggested. An increased interest in the ability of the gallbladder mucosa to absorb and secrete proteins is due to the possible role of proteins as pro or antinucleating factors in cholesterol crystallization. It has been suggested that the nucleating factor for cholesterol in gallbladder bile is a protein other than mucous glycoprotein that is absorbed or degraded in the normal gallbladder but not in the gallbladder harboring stones. On the other hand, the increased cholesterol nucleation might be due to an increased secretion of these pronucleating proteins by the gallbladder mucosa. However, few proteins are synthesized in the biliary tract itself; the majority reflect the composition of serum proteins. Based on the observation that there is delayed onset of cholesterol crystal nucleation in normal human gallbladder bile compared to model biles of similar biliary lipid composition, it has been postulated that there are antinucleating factors in normal gallbladder bile. The role of pro and antinucleating proteins in the pathogenesis of gallstone disease is reviewed in Chapter 9. Whether there is an increased protein content of bile in cholesterol gallstone patients, possibly due to increased protein secretion by the gallbladder, is still a subject for discussion. Some groups have reported an increase in total protein concentration in the bile of gallstone patients, whereas in other studies an increase in the glycoprotein fraction was observed. In contrast, other studies failed to demonstrate an increased protein content of gallstone bile. However, the nucleating or antinucleating action of gallbladder proteins is of much greater interest than protein concentrations. Therefore, studies on whether and how the gallbladder removes or adds pronucleating or antinucleating proteins to the bile are of great importance. But despite its potential importance in stone formation, there are few studies on protein secretion and absorption by the human gallbladder. In an in vitro study on protein absorption and secretion by the human gallbladder, both absorption of albumin and secretion of protein were demonstrated. The secreted proteins might come from serum proteins in the vessels of the gallbladder or from the gallbladder mucosa. Unfortunately, the type of proteins secreted were not studied. No significant differences in water or albumin transport rates or protein secretion rates were observed between gallbladders derived from control patients and cholesterol gallstone patients in this study (52). As mentioned above, little information is available about which specific proteins are synthesized and secreted by the gallbladder epithelium (53). Recently, evidence was presented to support the theory that the gallbladder is the predominant source of human bile IgA. Human bile contains immunoglobulins: IgG, IgM, and different forms of IgA. In some species the liver has been attributed the special function of clearing IgA from the circulation and excreting it with the bile into the intestinal tract. However, in humans the hepatobiliary transport of circulating IgA is much less important. In humans the amount of hepatic bile IgA delivered to the intestine averages less than 1 mg/kg/day and interruption of bile flow does not lead to significant elevations of IgA in plasma. In addition, the biliary tract and gallbladder mucosa contain immunoglobulinproducing cells. Thus, the biliary immunoglobulins are probably a mixture of molecules originating from distant sites and excreted by the liver and immunoglobulins produced in the biliary tract. Indeed, a study on the sedimentation profiles of IgA and secretory component (SC) and the concentrations of IgA, IgG, IgM, SC, and albumin in the gallbladder bile of adult human subjects without hepatobiliary disease, after an overnight fast, revealed an enrichment of bile with IgA and IgM relative to albumin. These results suggest
Page 34
that a significant amount of IgA could have been added to bile during its storage in the gallbladder. Therefore, the gallbladder should be regarded as the predominant source of bile IgA in humans (54). This could be an important finding, since biliary immunoglobulins have been implicated to play a role in the pathogenesis of cholesterol gallstone disease. IX— Arachidonic Acid Metabolites Arachidonic acid metabolites, or eicosanoids, are involved in a wide spectrum of hepatobiliary physiological functions and disease. Arachidonic acid is a polyunsaturated fatty acid found in phospholipids in cell membranes. After inflammatory or other stimuli, it is converted to prostaglandins through the action of the cyclooxygenase enzyme system or to leukotrienes through the action of lypoxygenase enzymes. Prostaglandins and leukotrienes are important mediators of inflammation and are involved in numerous physiological activities and pathological disease states of the gallbladder. The role of arachidonic acid metabolites in biliary physiology and disease has been reviewed (55). Prostanoids alter hepatic bile flow, with certain prostaglandins stimulating bile flow and others inhibiting bile flow. Prostanoids are also involved in gallbladder contraction, they cause gallbladder contraction in some species and relaxation in other species, and they may be mediators of cholecystokinetic hormone action. The inflamed gallbladder secretes rather than absorbs fluid. Prostaglandins have been demonstrated to alter the normal process of water absorption by the gallbladder mucosa and induce net water secretion. For example, prostaglandin E2 inhibits fluid absorption and induces net secretion in some species. However, no information is available to suggest that prostanoids stimulate gallbladder water absorption. Several hormones, such as secretin and vasoactive intestinal peptide, can stimulate gallbladder mucosal water secretion, but we do not know of any hormones that enhance fluid absorption. Also, indomethacin, an inhibitor of cyclooxygenase activity, has no effect on basal gallbladder absorptive function. In cholecystitis, the prostanoids may mediate the distention produced by mucosal fluid secretion and the contraction of the diseased gallbladder. The inflammatory changes produced in various experimental models of cholecystitis can be prevented by cyclooxygenase inhibitors. Normal gallbladder mucosa and muscle tissue from cats, dogs, guinea pigs, opossums, and humans produce prostanoids, suggesting a role for prostaglandins in the physiological functions of gallbladder mucosal water transport and muscle contraction. Human gallbladder mucosa was demonstrated to produce prostaglandin E as well as prostaglandin F. The amount of prostaglandins produced by inflamed gallbladder muscle and mucosal cells was found to increase with the severity of the inflammatory process (56). Cyclooxygenase inhibitors decrease gallbladder prostaglandin formation and are effective in producing relief of some of the symptoms of gallbladder disease. Prostanoids have also been implicated in gallstone formation. Increased cholesterol in bile results in increased prostanoid and lysolecithin formation, leading to the nucleation of cholesterol and subsequent gallstone formation (55). As described above, prostaglandins are also involved in the regulation of mucin secretion by the gallbladder mucosa, which can act as a nidus for stone formation. In some studies, cyclooxygenase inhibitors were reported to prevent the formation of gallstones in experimental animals. In humans, the significance of antiinflammatory drugs in the prevention of gallstone formation is less clear. However, some studies seem to point at a possible inhibitory effect of drugs such as aspirin on mucin secretion by the gallbladder mucosa (45). Most recently, Sterling et al. (57) tried to determine whether chronic use of nonsteroidal antiinflammatory drugs (NSAIDS) was associated with a reduction in the mucin content or affected the lipid composition of human gallbladder bile. They found that patients with gallstones had a significantly greater concentration of gallbladder mucin in their gallbladder bile than patients without gallstones. Among gallstone patients, gallbladder mucin was reduced in those patients with a history of chronic NSAID use. Also, chronic NSAID use was associated with a reduction in the cholesterol/phospholipid ratio of bile in both patient groups.
Page 35
However, more research is needed to determine the exact role of prostaglandins in the pathogenesis of gallstone disease. In addition to prostaglandins, it has now been demonstrated that the gallbladder is able to synthesize and secrete a variety of signal molecules, such as cAMP and cytokines. Mouse gallbladder epithelial cells produce mRNas for several cytokines such as TNF , IL6, RANTES, macrophage inflammatory protein 2 (MIP2) and ICAM (58). Human gallbladder epithelial cells were demonstrated to secrete endothelin1 (11), and both human and dog gallbladder epithelial cells express iNOS (59,60). Taken together these findings suggest that gallbladder cells can serve a paracrineautocrine function. X— Summary In this chapter we have described the different functions served by the gallbladder mucosa. The main function of the gallbladder epithelium is the concentration of hepatic bile. This is achieved by means of active absorption of water and electrolytes. Sodium and chloride are absorbed by the gallbladder by a double exchange mechanism in which hydrogen and bicarbonate are excreted, resulting in acidification of bile. Water absorption occurs owing to osmotic coupling to sodiumchloride transport. Absorption of water and electrolytes by the gallbladder epithelium is influenced by intracellular cAMP and calcium levels. In biliary disease, such as gallstone formation, the transport function of the gallbladder can be dramatically altered, resulting in net secretion instead of absorption. Besides water and electrolytes, the gallbladder also absorbs biliary lipids, such as cholesterol, and biliary pigments as well as proteins, amino acids, and sugars. Recent studies have demonstrated that the gallbladder has a secretory function as well; gallbladder bile composition is modified by means of active fluid and electrolyte secretion by the gallbladder epithelium. The gallbladder epithelium also secretes proteins and mucous glycoproteins. Arachidonic acid metabolites produced locally in the gallbladder have been found to influence several of these absorptive and secretory functions. In conclusion, through its diverse functions of absorption and secretion, the gallbladder mucosa dramatically influences the composition of bile, and, in its turn, the composition of bile can affect gallbladder mucosal function. Alterations in these functions will, therefore, play an important role in the pathogenesis of biliary disease. References 1. Diamond JM. Transport Mechanisms in the Gallbladder. In: Hoffman JF, ed. Handbook of Physiology: Alimentary Canal. Washington DC: American Physiological Society, 1968, pp 2451–2482. 2. Longnecker DS, Terhune PG. Carcinogenesis and pathology of carcinomas in the pancreas: comparison with the biliary tract. In: Sirici AE, Longnecker DS, eds. Biliary and Pancreatic Ductal Epithelia: Pathobiology and Pathophysiology. New York: Marcel Dekker, 1996, pp 527–567. 3. Hopwood D, Ross PE. Biochemical and morphological correlations in human gallbladder with reference to membrane permeability. Microsc Res Tech 1997; 38:631–642. 4. Nakanuma Y, Katayanagi K, Kawamura Y, Yoshida K. Monolayer and threedimensional culture and living tissue culture of gallbladder epithelium. Microsc Res Tech 1997; 39:71–84. 5. Oda D, Lee SP, Hayashi A. Long term culture and partial characterization of dog gallbladder epithelial cells. Lab Invest 1991; 64:682–689. 6. Peters RH, French PJ, van Doorninck JH, Lamblin G. CFTR expression and mucin secretion in cultured mouse gallbladder epithelial cells. Am J Physiol 1996; 271:G1074–1083.
Page 36
7. Peters RH, van Doorninck JH, French PJ, Ratcliff R, Evans MJ, Colledge WH, Bijman J, Scholte BJ. Cystic fibrosis transmembrane conductance regulator mediates the cyclic adenosine monophosphateinduced fluid secretion but not the inhibition of resorption in mouse gallbladder epithelium. Hepatology 1997; 25:270– 277. 8. Kuver R, Savard C, Nguyen TD, Osborne WRA, Lee SP. Isolation and longterm culture of gallbladder epithelial cells from wildtype and CF mice. In Vitro Cell Dev Biol 1997; 33:104–109. 9. Plevris JN, Walker SW, Harrison DJ, Dhariwal A, Hayes PC, Bouchier IAD. Primary culture of bovine gallbladder epithelial cells. Gut 1993; 34:1612–1615. 10. Hoerl BJ, Vroman BT, Kasperbauer JL, LaRusso NF, Scott RE. Biological characteristics of primary cultures of human gallbladder epithelial cells. Lab Invest 1992; 66:243–250. 11. Housset C, Carayon A, Housset B, Legendre C, Hannoun L, Poupon R. Endothelin1 secretion by human gallbladder epithelial cells in primary culture. Lab Invest 1993; 69:750–755. 12. Auth MKH, Keitzer RA, Scholaz M, Blaheta RA, Hottenrot EC, Hermann G, Encke A, Markus BH. Establishment and immunological characterization of human gallbladder epithelial cells. Hepatology 1993; 18:546–555. 13. Oda D, Eng L, Savard CE, Newcomer M, Haigh WG, Lee SP. Organotypic culture of human gallbladder epithelium. Exp Mol Pathol 1995; 63:16–22. 14. Reuss L, Stoddard JS. Role of H+ and HCO3 in salt transport in gallbladder epithelium. Annu Rev Physiol 1987; 49:35–49. 15. Scoazec JY, Bringuier AF, Medina JF, MartinezAnso E, Veissiere D, Feldmann G, Housset C. The plasma membrane polarity of human biliary epithelial cells: in situ immunohistochemical analysis and functional implications. J Hepatol 1997; 26:543–553. 16. Abedin ZR, Morgenstern KE, Roslyn JJ, Moser AJ, Abedin MZ. Chloride channel CLC3 is expressed by gallbladder epithelium: a potential regulator of Cl secretion (abstr). Gastroenterology 1998; 114:A514. 17. Gleeson D, Hood KA, Murphy GM, Dowling RH. Calcium and carbonate ion concentrations in gallbladder and hepatic bile. Gastroenterology 1992; 102:1701– 1716. 18. Plevris JN, Hayes PC, Harrison DJ, Bouchier IAD. Evidence of hydrogen ion secretion from human gallbladder in vitro. Gut 1992; 33:554–559. 19. Colombani V, Silviani V, Marteau C, Lerique B, Cartouzou G, Gerolami A. Presence of the NHE3 isoform of the Na+/H+ exchanger in human gallbladder. Clin Sci Colch 1996; 91:209–212. 20. Silviani V, Gastaldi M, Planells R, Marteau C, Massacrier A, Cohen P, Cau P, Gerolami A. NHE3 isoform of the Na+/H+ exchanger in human gallbladder. Localization of specific mRNA by in situ hybridization. J Hepatol 1997; 26:1281–1286. 21. Petersen KU, Wehner F, Winterhager JM. Na+/H+ exchange at the apical membrane of guinea pig gallbladder epithelium: properties and inhibition by cyclic AMP. Pflugers Arch 1985; 4051:S115–S120. 22. Reuss L. Ion transport across gallbladder epithelium. Physiol Rev 1989; 69:503–545. 23. Reuss L, Segal Y, Altenberg G. Regulation of ion transport across gallbladder epithelium. Annu Rev Physiol 1991; 53:361–373. 24. Reuss L. Salt and water transport by gallbladder epithelium. In: Handbook of Physiology: The Gastrointestinal System. Bethesda, MD: American Physiological Society, 1991, pp. 303–322. 25. DrayCharier N, Paul A, Veissiere D, Mergey M, Scoazec JY, Capeau J, BrahimiHorn C, Housset C. Expression of cystic fibrosis transmembrane conductance regulator in human gallbladder epithelial cells. Lab Invest 1995; 73:828–836. 26. Greger R, Mall M, Bleich M, Ecke D, Warth R, Riedemann N, Kunzelmann K. Regulation of epithelial ion channels by the cystic fibrosis transmembrane conductance regulator. J Mol Med 1996; 74:527–534.
Page 37
27. Svanvik J. Role of gallbladder in modifying hepatic bile composition. In: Tavoloni N, Berk PD, eds. Hepatic Transport and Bile Secretion: Physiology and Pathophysiology. New York: Raven Press, 1993, pp 607–618. 28. Scheeres DE, Magnuson TH, Pitt HA, Bastidas JA, May CA, Lillemoe KD. The effect of calcium on gallbladder absorption. J Surg Res 1990; 48:547–551. 29. Roslyn JJ, Abedin MZ, Saunders KD, Cates JA, Strichartz SD, Alperin M, Fromm, M, Palant CE. Uncoupled basal sodium absorption and chloride secretion in prairie dog (Cynomys ludovicianus) gallbladder. Comp Biochem Physiol 1991; 100A:335–341. 30. Cates JA, Saunders KD, Abedin MZ, Roslyn JJ. Intracellular calcium modulates gallbladder ion transport. J Surg Res 1991; 50:545–551. 31. Giurgiu DI, Karam JA, Madan AK, Roslyn JJ, Abedin MZ. Apical and basolateral Ca2+ channels modulate cytosolic Ca2+ in gallbladder epithelia. J Surg Res 1996; 63:179–184. 32. Moser AJ, Abedin MZ, Abedin ZR, Roslyn JJ. Ca2+ calmodulin regulates basal gallbladder absorption. Surgery 1993; 114:300–307. 33. Moser AJ, Abedin MZ, Cates JA, Giurgiu DI, Karam JA, Roslyn JJ. Converting gallbladder absorption to secretion: the role of intracellular calcium. Surgery 1996; 119:410–416. 34. Cates JA, Abedin MZ, SaundersKirkwood KD, Moser AJ, Giurgiu DI, Roslyn JJ. Protein kinase C regulates prairie dog gallbladder ion transport. Surgery 1995; 117:206–212. 35. Giurgiu DI, SaundersKirkwood KD, Roslyn JJ, Abedin MZ. Sequential changes in biliary lipids and gallbladder ion transport during gallstone formation. Ann Surg 1997; 225:382–390. 36. Igimi H, Yamamoto F, Lee SP. Gallbladder mucosal function: studies in absorption and secretion in humans and in dog gallbladder epithelium. Am J Physiol. 1992; 263:G69–G74. 37. Sweeting JG. Does the gallbladder secrete? Gastroenterology 1993; 104:329–330. 38. Glickerman DJ, Kim MH, Malik R, Lee SP. The gallbladder also secretes. Dig Dis Sci 1997; 42:489–491. 39. Neiderhiser DH, Harmon CK, Roth HP. Absorption of cholesterol by the gallbladder. J Lipid Res 1976; 17:117–124. 40. Ross PE, Azman M, Hopwood D, Shepherd AN, Ransay A, Bouchier IAD. Lipid absorption by human gallbladder. Ann NY Acad Sci 1986; 463:344–346. 41. Ross PE, Butt AN, Gallacher C. Cholesterol absorption by the gallbladder. J Clin Pathol 1990; 43:572–575. 42. Hayashi A, Lee SP. Bidirectional transport of cholesterol between gallbladder epithelial cells and model bile. Am J Physiol 1996; 271:G410–414. 43. GianniCorradini S, Yamashita G, Nuutinen H, Chernosky A, Williams C, Hays L, Shiffman ML, Walsh RM, Svanvik J, DellaGuardia P, Capocaccia L, Holzbach RT. Human gallbladder mucosal function: effects on intraluminal fluid and lipid composition in health and disease. Dig Dis Sci 1998; 43:335–343. 44. Rose RC. Absorptive Functions of the Gallbladder. In: Johnson LR, ed. Physiology of the Gastrointestinal Tract, 2nd ed. New York: Raven Press, 1987, pp 1455–1468. 45. Klinkspoor JH, Tytgat GNJ, Groen AK. Gallbladder mucin and cholesterol gallstones. Eur J Gastroenterol Hepatol 1993; 5:226–234. 46. Kuver R, Savard CE, Oda D, Lee SP. Prostaglandin E generates intracellular cAMP and accelerates mucin secretion by cultured dog gallbladder epithelial cells. Am J Physiol 1994; 267:G998–G1003. 47. DrayCharier N, Paul A, Combettes L, Bouin M, Mergey M, Balladur P, Capeau J, Housset C. Regulation of mucin secretion in human gallbladder epithelial cells: predominant role of calcium and protein kinase C. Gastroenterology 1997; 112:978–990. 48. Kuver R, Ramesh N, Lau S, Savard CE, Lee SP, Osborne WRA. Constitutive mucin secretion linked to CFTR expression. Biochem Biophys Res Comm. 1994; 203:1457–1462.
Page 38
49. Klinkspoor JH, Kuver R, Savard CE, Oda D, Azzouz H, Tytgat GNJ, Groen AK, Lee SP. Model bile and bile salts accelerate mucin secretion by cultured dog gallbladder epithelial cells. Gastroenterology 1995; 109:264–274. 50. Campion JP, Porchet N, Aubert JP, L'Helgoualc'h A, Clement B. UWpreservation of cultured human gallbladder epithelial cells: phenotypic alterations and differential mucin gene expression in the presence of bile. Hepatology 1995; 21:223–231. 51. Vandenhoute B, Buisine MP, Debailleul V, Clement B, Moniaux N, Dieu MC, Degand P, Porchet P, Aubert JP. Mucin gene expression in biliary epithelial cells. J Hepatol 1997; 27:1057–1066. 52. Toth JL, Harvey PRC, Upadyha GA, Strasberg SM. Albumin absorption and protein secretion by the gallbladder in man and in the pig. Hepatology 1990; 12:729–737. 53. LaRusso NF. Proteins in the bile: how they get there and what they do. Am J Physiol 1984; 247:G199–G205. 54. Vuitton DA, Seilles E, Cause P. Gallbladder: the predominant source of bile IgA in man? Clin Exp Immunol 1985; 62:185–192. 55. Kaminski DL. Arachidonic acid metabolites in hepatobiliary physiology and disease. Gastroenterology 1989; 97:781–792. 56. Kaminski DL, Desphande YG, Westfall S, Herbold D. Evaluation of prostacyclin production by human gallbladder. Arch Surg 1989; 124:277–280. 57. Sterling RK, Shiffman ML, Sugerman HJ, Moore EW. Effect of NSAIDs on gallbladder bile composition. Dig Dis Sci 1995; 40:2220–2226. 58. Savard CE, Blinman TA, Pandol SJ, Lee SP. Lipopolysaccharide stimulates cytokine production by mouse gallbladder epithelial cells (abstr). Gastroenterology 1998; 114:A1077. 59. Choi JW, Savard CE, Lee SP. Organotypic cultured dog gallbladder epithelial cells express inducible nitric oxide synthase (iNOS) like activity (abstr). Gastroenterology 1996; 110:A1169. 60. Keavany AP, Offner GD, Afdhal NH. Inducible nitric oxide synthase (iNOS) is the principal isoform expressed in human gallbladder (abstr). Gastroenterology 1998; 114:A526.
Page 39
3— Gallbladder Smooth Muscle Function and Its Dysfunction in Cholesterol Gallstone Disease Piero Portincasa University Medical School, Bari, Italy Gerard P. vanBergeHenegouwen University Hospital Utrecht, Utrecht, The Netherlands I— Introduction At variance with other smooth muscle cells of the gastrointestinal tract, gallbladder musculature belongs to an organ in which a highly concentrated secretion, the bile is collected. During cholesterol gallstone disease, cholesterol and cytotoxic bile salts can considerably increase in bile. Such potentially myotoxic substances are able to interact with muscularis plasma membranes and to induce functional changes. An understanding of the mechanisms of gallbladder smooth muscle contractility in health and disease is therefore of paramount importance to elucidate the steps leading to abnormal gallbladder motility in patients with cholesterol gallstones. The aim of this chapter is therefore to focus on the basic mechanisms of normal gallbladder contractility at a cellular level—its regulation and its inhibition. The cellular abnormalities of smooth muscle cell function that occur during gallstone formation are discussed. II— General Features of the Smooth Muscle Smooth muscle differs from skeletal muscle both structurally and functionally. The term smooth muscle cell indicates that the cell lacks the crossstriation pattern typical for both cardiac and skeletal muscle cells (1). Rather, a main feature of the smooth cell is the presence of intracellular sarcomerelike, diagonally arranged units of actin (thin filaments) and myosin (thick filaments). Smooth muscle cells are rather short in size (60 to 100 m in length by 3 to 10 m in width); compared to skeletal muscle, smooth muscle contains less myosin, much more actin and little if any troponin. Actin, in particular, is the key constituent of thin filaments, whereas myosin constitutes the thick filaments (2). The arrangement of contractile units of smooth muscle cells is shown schematically in Fig. 1 (2–4). An important feature of smooth muscle cells is their extremely slow contraction. Still, smooth musculature can maintain the tone for prolonged periods with little energy expenditure, which, in fact, is 102 to 103 of that of striated muscle cells. This is mainly caused by a low ATPase activity of myosin molecules,
Page 40
Figure 1 A. Diagonal arrangement of contractile units of actin and myosin filaments of the gallbladder smooth muscle cell. B. Contractile unit set at its maximal length. C. Sliding of actin and myosin filaments, which causes shortening of the cell and increase in contractile force. (Modified from Ref. 2.)
while tension is maintained or gives way to a very slow relaxation (so called latch) (2). However, the force developed by smooth musculature (expressed as force per cm2 of fiber cross section) is comparable to that developed by skeletal musculature. The tension built up by smooth muscle cells is a function of the muscle length before stimulation. The velocity of isotonic shortening is dependent on the load that the muscle has to move. Contractility is dependent on these two parameters (5). Functional coupling of smooth muscle cells is accomplished by conjunctions of the membranes in the form of gap junctions or nexuses, which are areas of low resistance for the spread of excitation from one cell to another. Nerve axons enter the muscle bundles and release neurotransmitters from swellings along their length. As these swellings are at some distance from single muscle cells, neurotransmitters probably act on only a few of these cells and their influence is transmitted from one cell to the next. III— Physiological Mechanisms of Gallbladder Smooth Muscle Contraction and Relaxation Several methods are employed to study gallbladder smooth muscle function in relation to contractility, as shown in Table 1. They span from receptor binding studies using radioactive agonists, which act on plasma membranes or histological sections, to light microscopy or tensiometry, which investigate the contraction of isolated muscle cells or muscle strips/whole gallbladder, respectively. Different agonists initiate contraction by interacting at the level of specific receptors in the plasma membrane. A general overview of pathways that mediate agonistinduced contraction and relaxation and are discussed in detail are shown in Table 2.
Page 41 Table 1 Principal Methods to Study Gallbladder Smooth Muscle Cell Function in Vitro in Relation to Contractility Method
Tissue
Main information
Species
References
Radioimmunoassay
Plasma membranes
Receptor binding
Animals, humans
18, 31–33
Autoradiography
Histological section
Receptor binding
Animals, humans
34
Light microscopy
Isolated cells
Contractility (shortening)
Animals, humans
39, 43, 147, 193
Tensiometry
Strips
Spontaneous phasic activity
Animals, humans
11, 34, 36, 53, 145, 152, 179, 194
Contractility and relaxation (agonists, antagonists and electric transmural field stimulation)
Whole gallbladder
Contractility
Animals
78, 195
Figures 2 and 3 schematically show the technique used for tensiometric studies of gallbladder smooth muscle cell strips in vitro and some representative results obtained with two agonists. In general, gallbladder motility is under the neurohormonal control. The ultimate target organ is the smooth muscle cell of the gallbladder wall. The vagal nerve has a stimulatory effect through release of acethylcholine (ACh), acting on muscarinic receptors. The sympathetic nervous system has an inhibitory effect through adrenaline and noradrenaline on the 2adrenergic receptor. In the postprandial period, gallbladder contraction is primarily mediated by the gastrointestinal hormone CCK (6), as also shown by a number of studies using specific CCKreceptor antagonists (6–9). Figures 4 and 5 provide some insights into the pathways of smooth muscle contraction and relaxation at the receptor and plasma membrane level and at the intracellular level. Table 2 reports the most important substances that have been shown to induce gallbladder smooth muscle contraction and relaxation. The role of these substances is discussed in the following paragraphs. A— Contraction The gallbladder smooth muscle exhibits a socalled myogenic tone (2) i.e., phasic spontaneous contractions followed by relaxation. This type of activity is best documented in vitro when gallbladder strips are suspended in the organ bath. The contraction often lasts for seconds and can be maintained even after denervation and even after pharmacological blockage of ganglion cells at the intramural level. At least two different patterns of spontaneous phasic contractions have been described in vitro when smooth muscle strips are suspended in the organ bath: lowamplitude, highfrequency contractions (about 1.2 to 4.0 contractions per minute), and a high amplitude, lowfrequency pattern (0.3 to 0.7 contractions per minute) (10–12). Spontaneous contractility can be detected in up to 90% of viable tissues from both normal specimens as well as strips from gallstonecontaining gallbladders. Gallbladder smooth muscle cells are grouped into branching bundles that are surrounded by connective tissue. In the gallbladder, the tunica muscularis is thick and invested with an interlocked array of longitudinal and spiral smooth muscle fibers. Apparently, no difference in contractility is seen between longitudinal, circular, and oblique axes strips (12). However, the sensitivity to both hormonal and neural stimulation increases from proximal (fundus) to distal (cystic duct) gallbladder musculature, as seen in strips (12) and in isolated muscle cells (13).
Page 42 Table 2 Substances Influencing Gallbladder Contractility in Vitro Effect
Receptor(s)
Mechanism/notes
Contraction Cholecystokinin
CCKA > CCKB
Hormone, intestinal release
Cerulein
Drug (cholecystokininanalogue)
Acetylcholine
Muscarinic M3 >> M1
Vagus
GRP, Substance K, Substance P, ET1, BRP, PACAP
GRP, NK2, SP, ET1, NMB, NANC neuroendocrine and paracrine transmission PACAP1
Histamine, bradykinin, prostaglandins, LTC4,
H1, B2(B1), LTD4
From inflammatory cells
Motilin
Motilide receptors (?)
Hormone, intestinal release
Erythromycin, other ''motilides"
Drugs/motilin analogues
Cisapride
Procholinergic
Drug
Potassium chloride
Influx of extracellular Ca2+ Membrane depolarizing agent
LTD4
Relaxation Acetylcholine
Muscarinic M2
Vagus
Adrenaline, Noradrenaline
badrenergic
Sympathetic nervous system
CGRP, PACAP, VIP, NT, PHI, ST
CGRP, PACAP2, VIP2, NT, PHI
NANC neuroendocrine and paracrine transmission
Histamine
H2
Inflammatory cells
NSAIDs
Inhibition of prostaglandin synthesis
Drugs
Nitric oxide
Intracellular effect
NANC nerves
Bile salts
CCK receptor + cholinergic nerves
Drugs, bile solutes
Key: BRP, bombesinrelated peptides; B, bradykinin; CGRP, calcitoningene related peptide; ET 1, endothelin1; GRP, gastrinreleasing peptide; H, histamine; LTD4, leukotrien D4 receptor; LT, leukotriens; NMB, NMBpreferring receptor; NT, neurotensin; PACAP, pituitary adenylate cyclaseactivating peptide; PHI, peptide histidine isoleucine; VIP, vasoactive intestinal peptide.
These findings indicate that a proximaltodistal biliary gradient exists which is an intrinsic property of smooth muscle cells; it is likely that this gradient acts to facilitate gallbladder emptying in vivo. The complex mechanism of smooth muscle cell excitationcontraction coupling results from the initiation of signal transduction at the level of the plasma membrane and activation of the contractile machinery at the intracellular level. The increase of cytosolic Ca2+ plays a key role in the contraction of smooth muscle cells; it depends on influx of extracellular Ca2+ and/or release from intracellular Ca2+ stores (sarcoplasmatic reticulum). The final pathway is the activation of contractile proteins, actin, and myosin by phosphorylation of myosin light chain, independent of the source of the Ca2+ influx (14). Ca2+ flux is regulated by both voltagedependent Ca2+ channels (Ltype and Ntype Ca2+ channels) and receptoroperated Ca2+ channels. Ltype channels are predominant type in smooth muscle cells, whereas Ntype channels are mainly found in intramural neurons. Both are activated by high
Page 43
Figure 2 Schematic representation of tensiometric studies of gallbladder smooth muscle strips in vitro. Strips are placed in the organ bath containing the plasma replacing solution. The force produced by contraction or the relaxation of strips in the presence agonist alone or with antagonists or in response to transmural electric field stimulation is measured using a transducer linked to a recording system. The dot (•) in the graph indicates the addition of single or cumulative concentrations of agonists in the bath; the symbol "W" indicates the washing time of the strip by fresh plasmareplacing solution.
membrane potentials. By contrast, receptoroperated Ca2+ channels are triggered by receptor binding by agonists rather than by membrane potential. In humans, intrinsic vagal nerve endings release ACh after inflow of Ca2+ through both L and Ntype Ca2+ channels in the nerves (15). CCK induces a potent contraction of smooth muscle strips in vitro. CCK exerts its contractile effects mainly through interaction directly with receptors on gallbladder muscle cells activating receptoroperated Ca2+ channels. Also, CCK can interact with cholinergic nerves (16) and can enhance ongoing nicotinic ganglionic transmission by release of acetylcholine occurring in the serosal layer. It has been shown that gallbladder CCK receptors present in the human and cow are both N linked complex glycoproteins, with different carbohydrate domains and similar protein cores (17). Computer analysis of equilibriumbinding data in the musculature suggest the presence of a single class of binding sites, which are similar in health and disease, with no apparent differences related to age, gender, or body habitus. The molecular weight of human gallbladder muscularis CCK receptor is 85,000 to 95,000 (18). However, the effect of CCK is not only direct on smooth muscle but also involves cholinergic pathways (6,19,20). ACh and the hormone CCK also induce contraction by both intracellular Ca2+ release and influx of extracellular Ca2+ through voltagedependent Ltype Ca2+ channels. The Ca2+channel antagonist verapamil can block this channel, whereas BayK8644, and Ltype channel dihydropyridine agonist, potentiates the action of this channel (21). Influx of extracellular Ca2+ and contraction is observed with the depolarizing agent potassium chloride, which acts on voltageoperated, receptordependent Ca2+ channels (22–24). At variance with the AChinduced contraction, the CCKinduced contraction seems more de
Page 44
Figure 3 Construction of agonist concentrationresponse curves as result of tensiometric studies of gallbladder strips. A representative experiment obtained from one gallbladder of a patient with cholesterol gallstones is shown. In this case force is normalized to 100% maximal response obtained with each agonist. EC50 = concentration of agonist leading to 50% maximal observed response. A. ACh (single concentrations are added to the bath ranging from 1010 M to 104 M. B. CCKOP (cumulative concentrations are added to the bath ranging from 1011 M to 106 M).
pendent on intracellular Ca2+ (22,25). Experiments with histamine and strontium, which inhibit intracellular Ca2+ release from the sarcoplasmatic reticulum, support this hypothesis (21,26,27). Other studies in humans given nifedipine, a dihydropyridine compound which inhibits Ca2+ influx through voltagedependent Ca2+ channels (28), suggest that, at least in part, stimulation with CCK is followed by influx of extracellular Ca2+ (29,30). Furthermore, the utilization of intracellular and extracellular Ca2+ appears to be dosedependent, since, in the guinea pig, high concentrations of CCK use more intracellular Ca2+ for contraction than low CCK concentrations do (22). The whole process of smooth muscle cell activation takes place at the level of the plasma membrane, which therefore plays a pivotal role in the overall mechanism leading to contraction or relaxation. We were the first to isolate smooth muscle cell plasma membranes from human gallbladder homogenates (31,32); these preparations showed a reversible and saturable binding for radioactive CCK. Others have extended this work (17,18,33,34) and shown that CCK binding involves two types of receptors, the CCKA and CCKBreceptors, which seem to have appeared early in the course of evolution (35). Alteration of the CCKA receptor, in particular, is associated with abnormal contractility, which is a known risk factor for cholesterol gallstones (36). Transmural electric field stimulation of gallbladder strips induces frequencydependent contractile responses through stimulation of the cholinergic nerves. A stimulus of 15 V, 1 ms for 10 s, range of 1 to 20 Hz, causes a prompt and sustained increase in tension, which is over as soon as the stimulation is discontinued (24). The mechanism involves Ca2+ influx into nerve terminals and ACh release from nerve endings (37,38). While ACh is released from intrinsic postganglionic cholinergic nerves, the contraction is regulated by L and Ntype Ca2+ channels (15), which can be blocked by nifedipine and conotoxin, respectively. The addition of potent neural blockers such as tetrodotoxin or the cholinergic antagonist atropine abolishes the re
Page 45
Figure 4 Schematic representation of pathways involved in gallbladder smooth muscle cell contraction and relaxation before activation of the intracellular machinery (for this, see Fig. 5). Key: (+), stimulation (contraction); (), relaxation or inhibition of contraction; , increase; , decrease; ACh, acetylcholine; A, adrenaline; cAMP, cyclic adenosinemonophosphate; cGMP, cyclicguanosinemonophosphate; CCK, cholecystokinin; DAG, Diacylglycerol; G, G protein; IP3, inositol1,4,5trisphosphate; M, muscarinic receptor, NANC, nonadrenergicnoncholinergic; NO, nitric oxide; NA, noradrenaline; PIP2, phosphatidylinositol4,5bisphosphate; SR, sarcoplasmatic reticulum; rocc, receptoroperated Ca2+ channels; vdcc, voltagedependent Ca2+ channels.
sponses to electric field stimulation, proving that this type of contraction is indeed neurally mediated (10). The events associated with smooth muscle contraction at the intracellular level are also complex and are summarized in Fig. 5. The signaltransduction pathway involves agonist binding to and activation of receptors linked to a class of membrane proteins, G protein (i.e., guanine nucleotidebinding protein). This step is followed by intracellular hydrolysis of PIP2, (phosphatidylinositol4,5bisphosphate) into IP3, (inositol1,4,5trisphosphate) and DAG (diacylglycerol) (39). These two phosphoinositide metabolites have fundamental functions as intracellular messenger (2,3,39–41). IP3 initiates the release of Ca2+ from the sarcoplasmatic reticulum with the formation of Ca2+calmodulin complex. DAG activates the lipiddependent protein kinase C (PKC); this step is followed by phosphorylation of some intracellular proteins, increased sensitivity of actomyosin to intracellular Ca2+, and activation of Ltype voltagedependent Ca2+ channels (42). The concentration of CCK at the level of plasma membrane receptor seems to modulate the intracellular pathway differently. At least in humans, the IP3 pathway is activated by high concentrations CCK, whereas low concentrations preferentially activate the PKC pathway (39). In the first case, in fact, the effect of IP3 on calmodulin is strongly inhibited by the calmodulin antagonist CGS9343B but not by two PKC inhibitors, H7 and chelerythrine (43).
Page 46
Figure 5 Schematic representation of intracellular pathway leading to contraction of the gallbladder smooth muscle. The key step is the phosphorylation of myosin. Key: aMLCK, activemyosin light chain kinase; ATP, adenosinetriphosphate; CAM, calmodulin; DAG, diacylglycerol; iMLCK, inactivemyosin light chain kinase; IP3, inositol1,4,5trisphosphate; PIP2, phosphatidylinositol4,5bisphosphate; PKC, protein kinase C; SR, sarcoplasmatic reticulum.
The human gallbladder contains both estrogen and progesterone receptors (44). Pregnancy strongly inhibits gallbladder contractility at the level of the Gprotein activation. CCK receptor binding, linked to Gi 3, is inhibited in pregnant guinea pigs, and this is most probably due to the action of progesterone. Oral contraceptives that contain progestagens, however, do not seem to influence gallbladder motility (45). Studies in guinea pigs and in humans showed that the M3, (muscarinic) receptor is the receptor involved in cholinergic neurotransmission of gallbladder directly on smooth muscle cell (46,47). In humans, however, the M1 muscarinic receptor plays a prejunctional facilitatory role in muscle cellreceptor mediated contractility (47,48). Apart from the physiological mediators of gallbladder contractility, CCK and ACh, a number of other hormones and substances have shown to exert contractile properties, at least in vitro. Their ultimate role in vivo, however, remains to be fully elucidated both in health and disease. Gastrinreleasing peptide caused some contraction in guinea pig gallbladder, which was unaffected by the muscarinic blockers atropine or the CCKA receptor antagonist loxiglumide (49). Substance K causes contraction through binding to the type 2 neurokinin receptor (NK2), which activates the PKCdependent intracellular pathway (50). Substance P (51) and endothelin1 (52) both induce guinea pig gallbladder contraction. Bombesinrelated peptides have a contractile effect on guinea pig gallbladder through a GRP and a NMBpreferring receptor (53). The NANCgroup neurotransmitter pituitaryadenylatecyclaseactivatingpeptide (PACAP) causes contraction through the PACAP1 receptor by adenylate cyclase and phospholipase C as intracellular messengers (54). Peptide YY, which is structurally related to pancreatic polypeptide, on the other hand, has not been shown to have any contractile effect in vitro or in vivo (55). Histamine has a stimulating effect on gallbladder contraction through the H1 receptor. The sensitivity of this receptor, however, declines with the severity of the inflammation in the
Page 47
gallbladder (56). Prostaglandins have a direct contractile effect on gallbladder smooth muscle cell in vitro (57). Das et al. showed, however, that aspirin (which inhibits the formation of prostaglandins) can correct the impaired gallbladder motility (58). The bradykinin B2 receptor and possibly also the bradykinin B1 receptor, stimulated by bradykinin agonists, cause contraction in guinea pig gallbladder mediated through a cyclooxygenase pathway. This action is inhibited by indomethacin (59). This contraction is fully dependent on extracellular Ca2+ influx; however, it is not related to PKCdependet pathways (60). Instead, contraction caused by bradykinin activates the intracellular phospholipase C pathway. Both C4 and D4 leukotriene cause contraction of guinea pig smooth muscle of the gallbladder through the LTD4 receptor. The intracellular increase of Ca2+ is fully dependent on intracellular Ca2+ stores (61). Motilin, a 22amino acid peptide which is released from enterochromaffin cells of duodenum and upper jejunum, plays a key role in the regulation of the interdigestive motility of the small intestine (62,63). There is a relation between plasma motilin, gallbladder emptying, and the migrating motor complex because gallbladder volume decreases up to 30% when plasma motilin peaks in the fasting subject (64) and up to 18% after exogenous motilin infusion (65). Interestingly, the macrolide erythromycin behaves as a motilin receptor agonist and stimulates gallbladder motility in vivo possibly through a cholinergic pathway (66–75). In a preliminary study, motilin also showed a direct effect on human gallbladder strips in vitro (76), as did erythromycin (77). Cisapride, a prokinetic agent with procholinergic properties, has been shown to stimulate smooth muscle cells in both in vivo and in vitro studies (78–80). B— Relaxation The essential step in smooth muscle relaxation is the lowering of the intracellular Ca2+ concentration by activation of specific Ca2+ pumps located in the cell membrane and in the sarcoplasmatic reticulum (81). Whereas Ca2+ is sequestered into the sarcoplasmic reticulum by an ATPdriven Ca2+ pump, Ca2+ extrusion out of the cell is achieved by Na+/Ca2+ exchange or by an ATPdependent pump (82). These events are shown in Fig. 4. Cyclic nucleotides play an important role in smooth muscle cell relaxation. Adrenergic inhibition of gallbladder smooth muscle cells is mediated by cAMP, which, in turn, activates a cAMPdependent protein kinase (PKA). This intracellular messenger has several effects that cause relaxation (2). Kline et al. showed that cAMP can relax previously contracted gallbladder in the bullfrog (83). Earlier studies showed that both NO and cAMP relax vascular smooth muscle through a protein kinase related to K+ channels (84). cGMP acts as a second messenger activating cGMPdependent protein kinase (PKG), which has a similar role as PKA, inhibiting the actions of intracellular Ca2+ and stimulating reuptake of Ca2+ (85). The inhibition of biliary flow seems to be mediated by activation of cholinergic interneurons or effector neurons (86). Maintenance of the inhibitory basal tone in the prairie dog gallbladder is mainly mediated by inducible nitric oxide synthase (iNOS) (87). Larginine, a NOdonor, can provoke a very significant increase in gallbladder fasting and residual volumes and impairs gallbladder emptying (68,88). Whereas adrenergic inhibition of contractility cannot be induced by dopamine (89), several other adrenergic substances, like adrenaline and noradrenaline, induce relaxation of the gallbladder through adrenergic receptors (2,90). There are a number of NANC neurotransmitters, which have been shown to inhibit contraction in both animal and human studies. NANC relaxation implies a NO dependent pathway (10,91). Transmural electric field stimulation can also be used to induce relaxation of gallbladder muscle strips; this effect is NANCmodulated and independent of other inhibitory pathways (10). This finding confirms that an important neurally mediated relaxation pathway operates in physiological state in the gallbladder (92). On the other hand, one should take into account that gallbladder motility in cholesterol gallstone patients is modulated by NO and is negatively influenced by a higher degree of scarification (93) due to chronic inflammation.
Page 48
Relaxation is achieved in the presence of calcitonin generelated peptide, which increases cAMP (94). The previously mentioned NANC hormone PACAP can also induce relaxation together with vasoactive intestinal peptide (VIP) through the VIP2/PACAP2 receptor (95). Previous investigators showed that VIP has a relaxing effect on CCKinduced contractions of Oncorhychus mykiss gallbladder, possibly related to adrenergic receptors (96). Neurotensin is another NANC substance that seems to have a negative effect on gallbladder contractility in humans (97), whereas it has a contractile effect in guinea pig gallbladder (97). Peptide histidine isoleucine has a relaxing effect on guinea pig smooth muscle cells (98). Although somatostatin and the synthetic analogue octreotide have an inhibitory effect on gallbladder contraction in vivo (99), these molecules have not been shown to have a direct effect on gallbladder smooth muscle cell in vitro (100). Histamine has a relaxing effect through the H2 receptor, whereas it stimulates contraction through H1 (101). The NSAID indomethacin has been shown to give a decrease in either tone or contractility by inhibiting prostaglandin synthesis both in vitro (102,103). However this is still controversial, since in clinical studies a prokinetic effect of indomethacin has been reported by some (104) but not others (105). IV— Mechanisms of Impaired Smooth Muscle Contractility and Relaxation The primum movens in cholesterol gallstone disease is excess cholesterol in bile due to hepatic hypersecretion (106,107). In predisposed individuals, bile chronically supersaturated with cholesterol leads to precipitation (108,109) and aggregation of cholesterol crystals into stone(s) as well as changes of concentration and/or quality of other biliary solutes (107,110–112). The gallbladder actively participates in these pathological events: it is able to absorb water from the incoming hepatic bile but also biliary lipids from the lumen such as cholesterol (113), phospholipids (114,115), and small amounts of bile salts (116). The gallbladder also has secretory ability and the production of mucin in cholesterol gallstone disease increases (117). Other factors are secreted into bile, such as immunoglobulins (mainly of the IgG class) by inflammatory cells in the wall and aminopeptidase N by the gallbladder (118–120). Some of these events can influence one or several pathways of smooth muscle function, and this can ultimately lead to impaired gallbladder motility (121). The importance of these factors in relation to smooth muscle function is discussed in the following paragraphs. A— Excess Cholesterol in Bile The gallbladder smooth muscle is a primary target of excess biliary cholesterol, which appears to have myotoxic properties. Although there is no significant difference in basal cell length between muscle cells associated with cholesterol stones and muscle cells associated with "control" pigment stones (43), several functional defects of the smooth muscle appear early during cholesterol gallstone disease. In the animal model of cholesterol cholelithiasis (ground squirrel and prairie dog), increased biliary cholesterol saturation and appearance of cholesterol crystals and stones are associated with impaired gallbladder smooth muscle contractility to CCK both in vivo and in vitro (122–124). In cholesterol gallstone patients, gallbladder muscle strips have weaker contractility in response to CCK than "control" strips from pigment stone patients who do not have bile supersaturated with cholesterol (11,25,26,125,126). This is also the case when contractility is measured at microscopy in smooth muscle cells isolated from gallbladders of animals fed a lithogenic diet or from cholesterol gallstone patients (43). By comparing gallbladders from gallstone patients with "normal" gallbladders from organtransplant donors, we found that smooth muscle contracts less even if the thickness of the muscle layer increases, as shown by quantitative morphometry after cholecystectomy (127). In another study, gallbladder
Page 49
motility was first assessed by functional ultrasonography. After cholecystectomy we found that the gallbladder muscle layer was significantly thicker in patients with defective gallbladder motility than in patients with preserved motility (1073 ± 76 m vs. 745 ± 75 m, respectively) (11). Since stone characteristics and inflammation were comparable between the two groups, we speculated that a form of "hypertrophic leiomyopathy" occurs in response to one or more chronic insults in the gallbladder, such as hypercholesterobilia, excess of toxic bile salts (see below), and/or mechanical injury (e.g., the presence of large stones). It must be emphasized that proliferative changes observed in gallbladder smooth cells exposed to excess cholesterol have similarities with the proliferative ability of arterial myocytes during atherogenesis (128). Impaired smooth muscle cell function in cholesterol gallstone disease might account for an important clinical finding—i.e., enlarged fasting and postprandial gallbladder volumes in a subgroup of cholesterol gallstone patients (11,129–133). As discussed by Weisbrodt (5), it is likely that excess cholesterol in bile leads to inadequate strength of the smooth muscle cell at normal length, resulting in less ejected bile and greater residual gallbladder volume. After adaptation occurs, however, the smooth muscle cell stretches to a greater length in order to develop adequate force during gallbladder contraction; this adaptation, however, will ultimately result in a greater fasting volume. This concept is summarized in Fig. 6. The relaxation pathways of the gallbladder smooth muscle might also be impaired in cholesterol gallstone patients. Gallbladder strips and cells isolated from cholesterol stone patients show decreased relaxation to transmural electric field stimulation or relaxing agents, compared to pigment stone patients. The motility defect is associated with decreased cellular cAMP production. Again, the defect seems to reside at the level of plasma membranes, since substances that circumvent the membrane and directly activate intracellular mechanisms elicit similar responses in gallbladders from cholesterol and pigment stone patients (92). Decreased relaxation in vitro might be linked to reduced gallbladder refilling as well as a reduced ente
Figure 6 Putative events linking the impaired smooth muscle contractility in vitro with defective gallbladder motility in vivo in cholesterol gallstone disease.
Page 50
rohepatic circulation observed in vivo in patients at risk for cholesterol gallstones, thereby increasing the possibility of cholesterol crystals to develop and to precipitate (134). Several studies point to the importance of hypercholesterobiliadependent leiomyopathy as one of the important pathogenic factors in cholesterol gallstone disease. Increased cholesterol content of bile leads to progressive impairment of gallbladder motility as seen in healthy subjects (135), in patients with cholesterolosis (25), and in cholesterol gallstone patients (11,129–131,136). Increased gallbladder volume has been noted in early studies in guinea pigs (103) and mice (137,138) fed high cholesterol diet. More recently, increased fasting gallbladder volume was also found in the +/+ strain of inbred mice that are genetically prone to develop supersaturated bile, cholesterol crystals, and gallstones (139,140). Indeed, cholesterol molecules absorbed by the gallbladder mucosa can rapidly diffuse into the muscle layer (107,113,141). Excess cholesterol molecules are removable from smooth muscle cells by esterification and storage; alternatively, they can diffuse back into bile (142), since the gallbladder lacks lipoproteins for cholesterol export into blood (113). In cholesterol gallstone patients, however, backdiffusion of cholesterol is greatly inhibited by chronically supersaturated bile (143). Although cholesterol molecules can be esterified by local cholesterolacyltransferase (ACAT) (144), other cholesterol molecules are incorporated into smooth muscle sarcolemma (145), leading to increased ratio of cholesterol to phospholipids in the membranes (146,147). These events can ultimately affect several cellular functions (148), including, of course, contractility. As recently underlined by Apstein and Carey (107), a key concept is that unesterified cholesterol is toxic to smooth muscle cell plasmalemma by altering the physical state and increasing rigidity (i.e., decreasing fluidity) of membrane phospholipids (149). In an elegant study, Yu et al. (146) showed that feeding a high cholesterol diet to prairie dogs or adding cholesterolenriched liposomes to isolated gallbladder smooth muscle cells increased cholesterol/phospholipid molar ratio by 90%; this change was paralleled by a 58% decrease in contractility in response to CCK. The motility defects were reversed when smooth muscle cells were incubated for few hours with cholesterolfree liposomes (146). Thus, it is possible that, at a very early stage (e.g., before chronic inflammation and fibrosis occur), the "functional" motility defects of the smooth muscle will be reversible. The intriguing question is therefore if and to what extent this reversible process is relevant in humans. In vivo, cholesterol supersaturation of bile can be reduced by oral ursodeoxycholate (120,150,151). We speculated that this would represent a possible way to reverse the smooth muscle defect in cholesterol gallstone disease. Thus, we studied gallbladder strip contractility in a group of gallstone patients treated for 3 weeks preoperatively with ursodeoxycholate in comparison with untreated patients. As expected, treated patients had a decreased cholesterol saturation index in their gallbladder bile. A striking finding in this group, however, was the improved contractility to CCK and cholinergic stimulation by acetylcholine. Despite the fact that degree of inflammation was mild, it was significantly lower in the patients treated with ursodeoxycholate (152). The motility defect of the smooth muscle in the gallbladder seems to be located at the plasmalemma level, somewhere between the receptor site and the intracellular messengers of contraction. A "structural" change or damage to the CCK receptor itself cannot be ruled out (33,36), since the CCK receptor sensitivity of gallbladder strips is either unchanged in cholesterolfed animals compared to controls (124,153) or decreased in cholesterol gallstone patients compared to "control" pigment gallstone patients (11). The intracellular machinery of contraction appears to be preserved if it is activated by intracellular messengers (26,147) or agents that are receptorindependent (e.g., potassium chloride) (36). Moreover, the concentrations of smooth muscle contractile proteins (i.e., actin and myosin) do not change after high cholesterol feeding of prairie dogs (153). Thus, functional defects of gallbladder smooth muscle might reside in the steps before Gprotein activation (43,154,155) as result of cholesterol incorporation and decreased membrane fluidity. This would lead to decreased mobilization of second messengers. It was indeed found that muscle cells from human gallbladders with cholesterol and pigment stones have similar contraction in response to the intracellular messengers IP3 (26), DAG, and the Gprotein
Page 51
activator GTPgS (147,156). By means of receptor binding studies, Xiao et al. recently found that gallbladder smooth muscle from cholesterol stone patients had increased CCK binding affinity (Kd) but decreased binding capacity (Bmax) compared to smooth muscles from pigment stone patients. Again, these defects were reversed by incubating smooth muscle cells with cholesterolfree liposomes, which remove excess cholesterol from the plasmalemma (157). The same impairment was a found for the VIP binding to Gs with another Gprotein subunit (156). As a consequence of excess cholesterol in bile, defects in excitationcontraction coupling of the gallbladder smooth muscle develop, such as impaired Ca2+ release from intracellular stores (143) and influx of extracellular Ca2+ through Ca2+ channels (158). b Recently, Jennings et al. (159) used fixed cells and wholemount preparations of the guinea pig gallbladder to measure cholesterol ester incorporation (via methyl cyclodextrins conjugated to a fluorophore) by confocal microscopy. Interestingly, the incorporation of intracellular cholesterol was associated with alterations in ionic conductance and action potentials as measured by the patchclamp technique; this phenomenon might contribute as well to the genesis of impaired contractility of gallbladder smooth muscle. B— Innervation, Inflammation, and Biliary Bile Salt Composition A general overview of the mechanisms involved is given in Fig. 7. Neural innervation plays an important role in the gallbladder: vagal pathways maintain gallbladder tone during fasting in concert with the gastrointestinal migrating motor complex (160–167), and vagal pathways also play a role postprandially (160). Whereas fasting gallbladder volume increases and gallbladder emptying can deteriorate after vagotomy (168–170), highly selective vagotomy has a minor effect on gallbladder motility (169). Spinal cord injury that abolishes sympathetic innervation of the gallbladder is associated with decreased fasting gallbladder volume but normal contractility (171,172), although the incidence of biliary sludge can be higher. Factors linked to cholelithiasis, such as chronic inflammation of the gallbladder wall (173) or excess toxic bile salts (see below), can adversely affect the intramural neurons of the gallbladder. Inflammatory changes in human gallbladders are associated with a significant reduction in the number of stainable nerve fibers and ganglion cells (174) and with decreased contractility in response to neural stimulation (175). The chronic inflammatory response of the gallbladder in uncomplicated cholesterol gallstone disease is generally mild (11,33). However, absorption of excess cholesterol from bile can produce very early inflammatory changes: in mice, the mucosa reacts to lithogenic diet within 48 h with increased mitotic index. Epithelial hyperplasia is then noted after 2 weeks and before gallstones appear (137,138). As shown in the prairie dog, other early changes at the mucosal side in response to increased lithogenic index of bile include inflammatory infiltrates, decrease in net sodium and water transport (176), and increased mucosal blood flow (177). In humans and pigs, supersaturated bile (with excess deoxycholate) is followed by plasmacellular infiltration of the gallbladder wall and increased secretion of immunoglobulins in bile (mainly IgG) (178). The hydrophobicity of the bile salts is another factor potentially influencing the function of gallbladder smooth muscle cells. Stolk et al. showed that the effect of bile salts on gallbladder smooth muscle contractility correlate significantly with their relative hydrophobicity. Even very low concentrations of the hydrophobic bile salt deoxycholateinhibited AChinduced contractility of human gallbladder strips in vitro. This was not the case with the hydrophilic ursodeoxycholate (179). In the guinea pig gallbladder, Xu and Shaffer (24) found that supraphysiological concentrations of tauroconjugated bile salts directly inhibited stimulation by CCK and the transmural electric field. The rank order of this inhibitory effect was from more to less hydrophobic bile salt (i.e., deoxycholate > chenodeoxycholate > cholate >> m ursodeoxycholate). Most important was that premedication of strips with the hydrophilic ursodeoxycholic acid (50 M), a bile salt used for oral litholysis of cholesterol gallstones, prevented the adverse effect of the
Page 52
Figure 7 Putative factors leading to impaired gallbladder smooth muscle contractility in cholesterol gallstone disease. On the left: excess biliary cholesterol leads to precipitation of cholesterol into crystals of various shapes [i.e., arcs, needles, tubules, spirals and plates (108,109)], damage of the gallbladder mucosa, lamina propria (inflammatory cells) and smooth muscle cells (incorporation of cholesterol into plasma membrane). On the right: effect of the hydrophobic bile salt deoxycholate at the level of the mucosa, smooth muscle cell, and intramural neurons. Under certain circumstances (e.g., litholytic therapy or cholestatic liver diseases), bile salts can also reach the gallbladder wall in high concentrations through the blood supply and serosal side.
other bile salts. This study clearly shows that bile salts have a direct dose and physicochemicaldependent adverse effect on gallbladder smooth muscle response. Since both voltageoperated Ca2+ channels and muscarinic (M3) receptors gave a normal response, the authors could rule out an alteration of both intracellular contractile machinery and intracellular Ca2+ release from sarcoplasmatic reticulum. Instead, the putative mechanisms might involve alterations of the CCK receptor at various levels (e.g., binding, receptor loss, or impaired Gprotein response) as well as defective intramural cholinergic transmission. The inhibitory effect of bile salts might either be at the level of the axon (with decreased conduction of the action potential) or directly on the intrinsic cholinergic nerve endings (180). Interestingly, a direct effect on smooth muscle contractility has also been shown in other organs such as the ileum (181) and in vascular smooth muscle (182). Would this bile saltmediated inhibition of gallbladder smooth muscle function be relevant in vivo? Several studies support this hypothesis. Bile salts could reach the gallbladder smooth muscle from the lumen. This possibility has been shown in the guinea pig by Ostrow, who also showed that mucosal damage was associated with enhanced bile salt luminal absorption (116). High levels of the hydrophobic cytotoxic bile salt deoxycholate are found in cholesterol gallstone patients (109,116,183,184) as result of delayed intestinal transit time and increased bacterial deconjugation of primary bile salts (184–186). Also, bile salt therapy for stone dissolution leads to elevated biliary urso
Page 53
deoxycholate. This has been shown to increase fasting gallbladder volume (132,187,188) and to depress gallbladder emptying in vivo by some (132,188) but not all studies (187,189). It is also possible that bile salts can reach gallbladder smooth muscle cells through the serosal side when serum levels increase (190), as in chronic cholestatic liver diseases, which can be associated with gallstone disease (191) and impaired gallbladder emptying (192). Indeed, as shown by Stolk et al., it appears that physiological amounts of bile salts in the serum as low as 107 M are sufficient to affect adversely gallbladder contractility (179). This possibility, however, seems to be very remote or absent for the hydrophilic ursodeoxycholate (24,152,179). V— Conclusions In recent years a major progress has been made in our understanding of gallbladder motility in human cholesterol cholelithiasis. Several studies in both humans and cholesterolfed animal models have focused on cellular and molecular events during the interaction of luminal signals with the gallbladder wall (mucosa and smooth muscle). Thus, the interplay between contraction and relaxation of the gallbladder muscularis leads to appropriate gallbladder emptying and refilling during fasting and in the postprandial state. The final outcome depends on the interaction between several neurohormonal signals and the muscle plasma membranes, from which regulatory signals originate. Longterm exposure of the gallbladder wall to luminal factors during cholesterol lithogenesis, such as excess cholesterol and the cytotoxic bile salt deoxycholate, will cause chronic inflammation of the mucosa and the lamina propria (178). In this context, the smooth muscle layer might react in several ways that ultimately lead to a form of gallbladder leiomyopathy (11,127) and several functional defects involving the plasma membranes and signal transduction (11,25,26,43,125,126). This, in turn, results in pathological contraction and/or relaxation of smooth musculature, impaired gallbladder motility, and gallbladder stasis, which is a key factor and an early feature in the pathogenesis of cholesterol crystallization and gallstones. Acknowledgments The authors gratefully acknowledge K. J. van Erpecum and N. G. Venneman for helpful discussion during the preparation of this chapter. References 1. Weisbrodt NW. Regulation: nerves and smooth muscle. In: Johnson LR, ed. Gastrointestinal Physiology. 5th ed. St Louis: MosbyYear Book, 1997, pp 325–332. 2. Ruegg JC. Smooth muscle. In: Greger R, Windhorst U, eds. Comprehensive Human Physiology. Berlin, Heidelberg: SpringerVerlag, 1996, pp 895–910. 3. Ruegg JC. Calcium in muscle contraction. 2nd ed. Berlin, Heidelberg, New York: SpringerVerlag, 1992. 4. Small JV, Squire JM. Structural basis of contraction in vertebrate smooth muscle. J Mol Biol 1972; 67:117–119. 5. Weisbrodt NW, Moody FG. Gallbladder contractility [Letter]. Gastroenterology 1992; 102:741–742. 6. Beglinger C, Hildebrand P, Adler G, Werth B, Harvey JR, Toouli J. Postprandial control of gallbladder contraction and exocrine pancreatic secretion in man. Eur J Clin Invest 1992; 22:827–834. 7. Liddle RA, Gertz BJ, Kanayama S, Beccaria L, Coker LD, Turnbull TA, Morita ET. Effects of a novel cholecystokinin (CCK) receptor antagonist, MK329, on gallbladder
Page 54
contraction and gastric emptying in humans. Implications for the physiology of CCK. J Clin Invest 1989; 84:1220–1225. 8. Malesci A, De Fazio C, Festorazzi S, Bonato C, Valentini A, Tacconi M, Rovati L, Setnikar I. Effect of loxiglumide on gallbladder contractile response to cerulein and food in humans. Gastroenterology 1990; 98:1307–1310. 9. Palasciano G, Portincasa P, Belfiore A, Baldassarre S, Albano O. Opposite effects of cholestyramine and loxiglumide on gallbladder dynamics in humans. Gastroenterology 1992; 102:633–639. 10. McKirdy ML, McKirdy HC, Johnson CD. Nonadrenergic noncholinergic inhibitory innervation shown by electrical field stimulation of isolated strips of human gall bladder muscle. Gut 1994; 35:412–416. 11. Portincasa P, Di Ciaula A, Baldassarre G, Palmieri V, Gentile A, Cimmino A, Palasciano G. Gallbladder motor function in gallstone patients: sonographic and in vitro studies on the role of gallstones, smooth muscle function and gallbladder wall inflammation. J Hepatol 1994; 21:430–440. 12. Bird NC, Wegstapel H, ChessWilliams R, Johnson AG. In vitro contractility of stimulated and nonstimulated human gallbladder muscle. Neurogastroenterol Motil 1996; 8:63–68. 13. Severi C, Grider JR, Makhlouf GM. Functional gradients in muscle cells isolated from gallbladder, cystic duct, and common bile duct. Am J Physiol 1988; 255:G647–G652. 14. Washabau RJ, Wang MB, Dorst C, Ryan JP. Role of myosin lightchain phosphorylation in guinea pig gallbladder smooth muscle contraction. Am J Physiol 1994; 266:G469–G474. 15. Parkman HP, Pagano AP, Martin JS, Ryan JP. Electric field stimulationinduced guinea pig gallbladder contractions. Dig Dis Sci 1997; 42:1919–1925. 16. Schjoldager BT. Role of CCK in gallbladder function. Ann NY Acad Sci 1994; 713: 207–218. 17. Schjoldager B, Molero X, Miller LJ. Gallbladder CCK receptors: species differences in glycosylation of similar protein cores. Regul Peptides 1990; 28:265–272. 18. Schjoldager B, Molero X, Miller LJ. Functional and biochemical characterization of the human gallbladder muscularis cholecystokinin receptor. Gastroenterology 1989; 96: 1119–1125. 19. Hanyu N, Dodds WJ, Layman RD, Hogan WJ, Chey WY, Takahashi I. Mechanism of cholecystokinininduced contraction of the opossum gallbladder. Gastroenterology 1990; 98:1299–1306. 20. Brotschi EA, Pattavino J, Williams LF, Jr. Intrinsic nerves affect gallbladder contraction in the guinea pig. Gastroenterology 1990; 99:826–830. 21. Lee JK, Rhee PL, Kang TM, Kim J, Koh KC, Paik SW, Rhee JC, Choi KW. Calcium sources mediating human gallbladder smooth muscle contraction by acetylcholine and cholecystokinin (abstr). Gastroenterology 1998; 114:A528. 22. Ryan JP. Calcium and gallbladder smooth muscle contraction in the guinea pig: effect of pregnancy. Gastroenterology 1985; 89:1279–1285. 23. Parkman HP, Pagano AP, Ringold MA, Ryan JP. Effect of modulating voltagedependent calcium channels on cholecystokinin and acetylcolineinduced contractions of the guinea pig gallbladder. Regul Peptides 1996; 63:31–37. 24. Xu QW, Freedman SM, Shaffer EA. Inhibitory effect of bile salts on gallbladder smooth muscle contractility in the guinea pig in vitro. Gastroenterology 1997; 112:1699–1706. 25. Behar J, Lee KY, Thompson WR, Biancani P. Gallbladder contraction in patients with pigment and cholesterol stones. Gastroenterology 1989; 97:1479–1484. 26. Behar J, Rhim BY, Thompson W, Biancani P. Inositol trisphosphate restores impaired human gallbladder motility associated with cholesterol stones. Gastroenterology 1993; 104:563–568. 27. Renzetti LM, Wang MB, Ryan JP. Contribution of intracellular calcium to gallbladder smooth muscle contraction. Am J Physiol 1990; 259:G1–G5.
Page 55
28. Spedding M, Paoletti R. Classification of calcium channels and the sites of action of drugs modifying channel function. Pharmacol Rev 1992; 44:363–376. 29. Clas D, Hould FS, Rosenthall L, Arzoumanian A, Fried GM. Nifedipine inhibits cholecystokinininduced gallbladder contraction. J Surg Res 1989; 46:479–483. 30. Jonderko K, Nowak A, Kasicka Jonderko A, Sliwinski Z, Kucio C. Effect of nifedipine on interdigestive gallbladder volume and postprandial gallbladder emptying in man. Dig Dis Sci 1991; 36:1434–1440. 31. Portincasa P, Howard A, Murphy GM, Dowling RH. Cholecystokinin receptor binding by human gallbladder myocyte membranes (abstr). Gut 1986; 27:1261A. 32. Portincasa P, Murphy GM, Dowling RH, Palasciano G, Albano O. A method proposed for the preparation of plasma membranes from myocytes of human gallbladders. In: Gorrod JW, Albano O, Papa S, eds. Molecular Aspect of Human Disease. London: Ellis Horwood, 1988, pp 241–249. 33. Upp JR, Jr, Nealon WH, Singh P, Fagan CJ, Jonas AS, Greeley GH, Jr, Thompson JC. Correlation of cholecystokinin receptors with gallbladder contractility in patients with gallstones. Ann Surg 1987; 205:641–648. 34. Schjoldager B, Shaw MJ, Powers SP, Schmalz PF, Szurszewski J, Miller LJ. Bovine gallbladder muscularis: source of a myogenic receptor for cholecystokinin. Am J Physiol 1988; 254:G294–G299. 35. Oliver AS, Vigna SR. CCKA and CCKBlike receptors in the gallbladder and stomach of the alligator (Alligator mississippiensis). Gen Comp Endocrinol 1997; 105:91–101. 36. Schneider H, Sanger H, Hanisch E. In vitro effects of cholecystokinin fragments on human gallbladders. Evidence for an altered CCKreceptor structure in a subgroup of patients with gallstones. J Hepatol 1997; 26:1063–1068. 37. Matthews G. Neurotransmitter release. Annu Rev Neurosci 1996; 19:219–233. 38. Hess P. Calcium channels in vertebrate cells. Annu Rev Neurosci 1990; 13:337–356. 39. Yu P, De Petris G, Biancani P, Amaral J, Behar J. Cholecystokinincoupled intracellular signaling in human gallbladder muscle. Gastroenterology 1994; 106:763– 770. 40. Bitar KN, Bradford BG, Putney JW, Makhlouf GM. Stoichiometry of contraction and Ca2+ mobilization by inositol1,4,5trisphosphate in isolated gastric smooth muscle cells. J Biol Chem 1986; 261:16591–16596. 41. Pearson RK, Hadac EM, Miller LJ. Structural analysis of a distinct subtype of CCK receptor on human gastric smooth muscle tumors. Am J Physiol 1989; 256:G1005–G1010. 42. Masui H, Wakabayashi I. Preferential inhibition by ethanol of phorbol esterinduced smooth muscle contraction of guinea pig gallbladder. Life Sci 1998; 62:673– 678. 43. Chen Q, De Petris G, Yu P, Amaral J, Biancani P, Behar J. Different pathways mediate cholecystokinin actions in cholelithiasis. Am J Physiol 1997; 272:G838– G844. 44. Singletary BK, Van Thiel DH, Eagon PK. Estrogen and progesterone receptors in human gallbladder. Hepatology 1986; 6:574–578. 45. van der Werf SDJ, Van BergeHenegouwen GP, Ruben AT, Palsma DM. Biliary lipids, bile acid metabolism, gallbladder motor function and small intestinal transit during ingestion of a subfifty oral contraceptive. J Hepatol 1987; 4:318–326. 46. Eltze M, Konig H, Ullrich B, Grebe T. Contraction of guineapig gallbladder: muscarinic M3 or M4 receptors? Eur J Pharmacol 1997; 332:77–87. 47. Parkman HP, Pagano AP, Ryan JP. Subtypes of muscarinic receptors regulating gallbladder cholinergic contractions (abstr). Gastroenterology 1998; 114:A819. 48. Nelson DK, Glasbrenner B, Dahmen G, Riepl RL, Malfertheiner P, Adler G. M1 muscarinic mechanisms regulate intestinalphase gallbladder physiology in humans. Am J Physiol 1996; 271:824–830. 49. Liu F, Naruse S, Ozaki T, Sazi T, Kondo T, Toda Y. Effect of gastrinreleasing peptide (GRP) on guinea pig gallbladder contraction in vitro. J Gastroenterol 1995; 30:764–767.
Page 56
50. Yau WM. Mode of stimulation of gallbladder contraction by substance K. Am J Physiol 1990; 259:G838–G841. 51. Meldrum LA, Bojarski JC, Calam J. Effects of substance P and other neuropeptides on guineapig gallbladder muscle. Digestion 1987; 37:193–199. 52. Moummi C, Gullikson GW, Gaginella TS. Effect of endothelin1 on guinea pig gallbladder smooth muscle in vitro. J Pharmacol Exp Ther 1992; 260:549–553. 53. Parkman HP, Vozzelli MA, Pagano AP, Cowan A. Pharmacological analysis of receptors for bombesinrelated peptides on guinea pig gallbladder smooth muscle. Regul Pept 1994; 52:173–180. 54. Parkman HP, Pagano AP, Ryan JP. Dual effects of PACAP on guinea pig gallbladder muscle via PACAPpreferring and VIP/PACAPpreferring receptors. Am J Physiol 1997; 272:1433–1438. 55. Lluis F, Fujimura M, Lonovics J, Guo YS, Gomez G, Greeley GH, Jr, Townsend CM, Jr, Thompson JC. Peptide YY and gallbladder contraction. Studies in vivo and in vitro. Gastroenterology 1988; 94:1441–1446. 56. Lennon F, Feeley TM, Clanachan AS, Scott GW. Effects of histamine receptor stimulation on diseased gallbladder and cystic duct. Gastroenterology 1984; 87:257–262. 57. Andersson KE, Andersson R, Hedner P, Persson CG. Parallelism between mechanical and metabolic responses to cholecystokinin and prostaglandin E2 in extrahepatic biliary tract. Acta Physiol Scand 1973; 89:571–579. 58. Das A, Baijal SS, Saraswat VA. Effect of aspirin on gallbladder motility in patients with gallstone disease: a randomized, doubleblind, placebocontrolled trial of two dosage schedules. Dig Dis Sci 1995; 40:1782–1785. 59. Falcone RC, Hubbs SJ, Vanderloo JD, Prosser JC, Little J, Gomes B, Aharony D, Krell RD. Characterization of bradykinin receptors in guinea pig gall bladder. J Pharmacol Exp Ther 1993; 266:1291–1299. 60. Cabrini DA, Calixto JB. Characterization of desArg9bradykinininduced contraction in guineapig gallbladder in vitro. Eur J Pharmacol 1997; 331:31–38. 61. Freedman SM, Wallace JL, Shaffer EA. Characterization of leukotrieneinduced contraction of the guineapig gallbladder in vitro. Can J Physiol Pharmacol 1993; 71:145–150. 62. Boivin M, Raymond MC, Riberdy M, Trudel L, StPierre S, Poitras P. Plasma motilin variation during the interdigestive and digestive states in man. J Gastrointest Motil 1990; 2:240–246. 63. Vantrappen G, Janssens J, Peeters TL, Bloom SR, Christofides ND, Hellemans J. Motilin and the interdigestive migrating motor complex in man. Dig Dis Sci 1979; 24:497–500. 64. Stolk MFJ, van Erpecum KJ, Smout AJPM, Akkermans LMA, Jansen JB, Lamers CBHW, Peeters TL, vanBergeHenegouwen GP. Motor cycles with phase III in antrum are associated with high motilin levels and prolonged gallbladder emptying. Am J Physiol 1993; 264:G596–G600. 65. Luiking YC, Peeters TL, Stolk MFJ, Nieuwenhuijs VB, Portincasa P, Depoortere I, Van BergeHenegouwen GP, Akkermans LMA. Motilin induces gall bladder emptying and antral contractions in the fasted state in humans. Gut 1998; 42:830–835. 66. Catnach SM, Ballinger AB, Law AP, Nellans H, Fairclough PD. Dosedependent stimulation of gallbladder contraction by intravenous erythromycin in man. Aliment Pharmacol Ther 1993; 7:55–59. 67. Catnach SM, Fairclough PD, Trembath RC, O'Donnell LJ, McLean AM, Law PA, Wickham JE. Effect of oral erythromycin on gallbladder motility in normal subjects and subjects with gallstones. Gastroenterology 1992; 102:2071–2076. 68. Fiorucci S, Distrutti E, Quintieri A, Sarpi L, Spirchez Z, Gulla N, Morelli A. Larginine/nitric oxide pathway modulates gastric motility and gallbladder emptying induced by erythromycin and liquid meal in humans. Dig Dis Sci 1995; 40:1365–1371.
Page 57
69. Fiorucci S, Distrutti E, Bassotti G, Gerli R, Chiucchiu S, Betti C, Santucci L, Morelli A. Effect of erythromycin administration on upper gastrointestinal motility in scleroderma patients. Scand J Gastroenterol 1994; 29:807–813. 70. Fiorucci S, Bosso R, Morelli A. Erythromycin stimulates gallbladder emptying and motilin release by atropinesensitive pathways. Dig Dis Sci 1992; 37:1678– 1684. 71. Fiorucci S, Scionti L, Bosso R, Desando A, Bottini P, Marino C, Morelli A. Effect of erythromycin on gallbladder emptying in diabetic patients with and without autonomic neuropathy and high levels of motilin. Dig Dis Sci 1992; 37:1671–1677. 72. Jebbink MC, Masclee AAM, van der Kleij FG, Schipper J, Rovati LC, Jansen JB, Lamers CBHW. Effect of loxiglumide and atropine on erythromycininduced reduction in gallbladder volume in human subjects. Hepatology 1992; 16:937–942. 73. Masclee AAM, Ledeboer ML, Gielkens HAJ, van der Kleij FG, Jebbink MC, Lamers CBHW. Effect of erythromycin on gallbladder emptying in patients with antrectomy or truncal vagotomy. Am J Gastroenterol 1995; 90:973–977. 74. Arienti V, Magri F, Boriani L, Maconi G, Bassein L, Baraldini M, Marzio L, Gasbarrini G. Effect of a single dose of oral erythromycin on gastric and gallbladder emptying. Simultaneous assessment by ultrasound. Dig Dis Sci 1994; 39:1309–1312. 75. Masclee AA, Ledeboer ML, Gielkens HG, van der Kleij FG, Jebbink MC, Lamers CB. Effect of erythromycin on gallbladder emptying in patients with antrectomy or truncal vagotomy. Am J Gastroenterol 1995; 90:973–977. 76. Yamasaki T, Chijiiwa K, Chijiiwa Y. Direct contractile effect of motilin on smooth muscle cells from human gallbladder. J Surg Res 1994; 56:89–93. 77. Peeters TL. The potentials of erythromycin derivatives in the treatment of gastrointestinal motility disorders. Z Gesamte Inn Med 1991; 46:349–354. 78. KoutsovitiPapadopoulou M, Kounenis G, Batzias G, Elezoglou V. Effect of cisapride on the isolated guinea pig gall bladder and common bile duct. Gen Pharmacol 1997; 29:863–867. 79. Nakayama S, Neya T, Yamasato T, Takaki M, Itano N. Effects of cisapride on the motility of the digestive tract in dogs and guinea pigs. Nippon Heikatsukin Gakkai Zasshi 1985; 21:1–9. 80. Xu QW, Shaffer EA. Cisapride improves gallbladder contractility and bile lipid composition in an animal model of gallstone disease. Gastroenterology 1993; 105:1184–1191. 81. Schatzmann HJ. The calcium pump of the surface membrane and of the sarcoplasmic reticulum. Annu Rev Physiol 1989; 51:473–485. 82. Casteels R, Van Breemen C. Active and passive Ca2+ fluxes across cell membranes of the guineapig taenia coli. Pflugers Arch 1975; 359:197–207. 83. Kline LW, Zhang ML, Pang PKT. Cyclic AMP induces a relaxation response in the bullfrog Rana catesbeiana, but nitric oxide does not. J Exp Biol 1997; 200:2669–2674. 84. Archer SL, Huang JM, Hampl V, Nelson DP, Shultz PJ, Weir EK. Nitric oxide and cGMP cause vasorelaxation by activation of a charybdotoxinsensitive K channel by cGMPdependent protein kinase. Proc Natl Acad Sci USA 1994; 91:7583–7587. 85. Schultz KD, Bohme E, Kreye VA, Schultz G. Relaxation of hormonally stimulated smooth muscular tissues by the 8 bromo derivative of cyclic GMP. Naunyn Schmiedebergs Arch Pharmacol 1979; 306:1–9. 86. Rasmussen TN, Harling H, Rehfeld JF, Holst JJ. Calcitonin generelated peptide (CGRP), a potent regulator of biliary flow. Neurogastroenterol Motil 1997; 9:215–220. 87. Keaveny AP, Offner GD, Afdhal NH. Inducible nitric oxide synthase (iNOS) is the principal isoform expressed in human gallbladder (abstr). Gastroenterology 1998; 114: A526. 88. Luiking YC, Weusten BLAM, Portincasa P, Van der Meer R, Smouth AJPM, Akkermans LMA. Effect of longterm oral Larginine on esophageal motility and gallbladder dynamics in healthy humans. Am J Physiol 1998; 37:G984–G991.
Page 58
89. Tankurt E, Apaydin S, Ellidokuz E, Igcl E, Guven H, I Simsek, O Gonen. The prokinetic effect of domperidone in gallbladder—not upon dopaminergic receptors. Pharm Res 1996; 34:153–156. 90. Perrson CGA. Adrenoreceptors in the gallbladder. Acta Pharmacol Toxicol 1972; 31:177–185. 91. Greaves R, Miller J, O'Donnell L, McLean A, Farthing MJ. Effect of the nitric oxide donor, glyceryl trinitrate, on human gall bladder motility. Gut 1998; 42:410– 413. 92. Chen Q, Amaral J, Oh S, Biancani P, Behar J. Gallbladder relaxation in patients with pigment and cholesterol stones. Gastroenterology 1997; 113:930–937. 93. Sanger P, Schneider H, Hanisch E. Nonadrenergic noncholinergic regulation of gallstone containing and gallstone free human gallbladders. Zentralbl Chir 1997; 122:418–424. 94. Kline LW, Pang PKT. Cyclic AMP modulates part of the relaxant action of calcitonin generelated peptide in guinea pig gallbladder strips. Regul Pept 1997; 72:55–59. 95. Haruma K, Wiste JA, Camilleri M. Effect of octreotide on gastrointestinal pressure profiles in health, functional and organic gastrointestinal disorders. Gut 1994; 35:1064–1069. 96. Aldman G, Holmgren S. VIP inhibits CCKinduced gallbladder contraction involving a betaadrenoceptor mediated pathway in the rainbow trout, Oncorhynchus mykiss, in vivo. Gen Comp Endocrinol 1992; 88:287–291. 97. Guo Y, Singh P, Upp JR, Jr, Thompson J. Speciesspecific effects of neurotensin on gallbladder contraction in vitro. Dig Dis Sci 1989; 34:21–26. 98. Yiangou Y, Christofides ND, Gu J, Piper PJ, Polak JM, Bloom SR. PHIlike immunoreactivity in the gallbladder and in vitro effect of porcine PHI on smooth muscle of the gallbladder. FEBS Lett 1984; 175:307–312. 99. Hussaini SH, Pereira SP, Veysey MJ, Kennedy C, Jenkins P, Murphy GM, Wass JAH, Dowling RH. Role of gall bladder emptying and intestinal transit in the pathogenesis of octreotide induced gallbladder stones. Gut 1996; 38:775–783. 100. Lonovics J, Devitt P, Rayford PL, Thompson JC. Actions of VIP, somatostatin, and pancreatic polypeptide on gallbladder tension and CCKstimulated gallbladder contraction in vitro. Surg Forum 1979; 30:407–409. 101. Jennings LJ, Salido GM, Pozo MJ, Davison JS, Sharkey KA, Lea RW, Singh J. The source and action of histamine in the isolated guineapig gallbladder. Inflamm Res 1995; 44:447–453. 102. Schulz M, Hanisch E, Guldutuna S. In vitro contractility of the musculature of human gallbladders with and without gallstones—relevance of the prostaglandin system for CCK regulated motoricity. Z Gastroenterol 1993; 31:376–387. 103. Brotschi EA, LaMorte WW, Williams LFJ. Effect of dietary cholesterol and indomethacin on cholelithiasis and gallbladder motility in guinea pig. Dig Dis Sci 1984; 29: 1050–1056. 104. O'Donnell LJ, Wilson P, Guest P, Catnach SM, McLean A, Wickham JE, Fairclough PD. Indomethacin and postprandial gallbladder emptying. Lancet 1992; 339:269–271. 105. Murray FE, Stinchcombe SJ, Hawkey CJ. Effect of indomethacin and misoprostol on fasted gallbladder volume and mealinduced gallbladder contractility in humans. Dig Dis Sci 1992; 37:1228–1231. 106. Carey MC. Formation of cholesterol gallstones: the new paradigm. In: Paumgartner G, Stiehl A, Gerok W, eds. Trends in Bile Acid Research. Dordrecht, The Netherlands: Kluwer, 1989, pp 259–281. 107. Apstein MD, Carey MC. Pathogenesis of cholesterol gallstones: a parsimonious hypothesis. Eur J Clin Invest 1996; 26:343–352. 108. Konikoff FM, Chung DS, Donovan JM, Small DM, Carey MC. Filamentous, helical and tubular microstructures during cholesterol crystallization from bile: evidence that billiary cholesterol does not nucleate classic monohydrate plates. J Clin Invest 1992; 90:1156–1161.
Page 59
109. Portincasa P, van Erpecum KJ, Jansen A, Renooij W, Gadellaa M, vanBergeHenegouwen GP. Behavior of various cholesterol crystals in bile from gallstone patients. Hepatology 1996; 23:738–748. 110. Portincasa P, van Erpecum KJ, vanBergeHenegouwen GP. Cholesterol crystallisation in bile. Gut 1997; 41:138–141. 111. Carey MC. Formation and growth of cholesterol gallstones: the new synthesis. In: Fromm H, Leushner U, eds. Bile Acids—Cholestasis—Gallstones: Advances in Basic and Clinical Bile Acid Research. Dordrecht, The Netherlands: Kluwer, 1996, pp 147–175. 112. Portincasa P. Gallbladder and Bile in Health and Gallstone Disease: The Role of Motility, Gallstones and Bile Lipid Composition. Ph.D. thesis. Utrecht, The Netherlands: Utrecht University, 1995. 113. Neiderhiser DH, Roth HP. Absorption of cholesterol by the gallbladder. J Lipid Res 1976; 17:117–124. 114. Neiderhiser DH, Morningstar WA, Roth H. Absorption of lecithin and lysolecithin by the gallbladder. J Lab Clin Med 1973; 82:891–897. 115. Purdum PP, Shamburek RD, Hylemon PB, Moore EW. Rapid phospholipid transfer across cultured human gallbladder epithelia. Gastroenterology 1992; 102:A871. 116. Ostrow JD. Absorption by the gallbladder of bile acid salts, sulfobromophtalein and iodipamide. J Lab Clin Med 1969; 74:482–494. 117. Carey MC, Cahalane MJ. Whither biliary sludge? Gastroenterology 1988; 95:508–523. 118. Harvey PRC, Upadhya GA, Strasberg SM. Immunoglobulins as nucleating proteins in the gallbladder bile of patients with cholesterol gallstones. J Biol Chem 1991; 266: 13996–14003. 119. Upadhya GA, Harvey PR, Strasberg SM. Effect of human biliary immunoglobulins on the nucleation of cholesterol. J Biol Chem 1993; 268:5193–5200. 120. van Erpecum KJ, Portincasa P, Eckhardt E, Go PMNYH, vanBergeHenegouwen GP, Groen AK. Ursodeoxycholic acid reduces protein levels and their nucleationpromoting activity in gallbladder bile. Gastroenterology 1996; 110:1225–1237. 121. LaMorte WW. Biliary motility and abnormalities associated with cholesterol cholelithiasis. Curr Opin Gastroenterol 1993; 9:810–816. 122. Doty JE, Pitt HA, Kuchenbecker SL, DenBesten L. Impaired gallbladder emptying before gallstone formation in prairie dog. Gastroenterology 1983; 85:168– 174. 123. Fridhandler TM, Davison JS, Shaffer EA. Defective gallbladder contractility in the ground squirrel and prairie dog during the early stages of cholesterol gallstone formation. Gastroenterology 1983; 85:830–836. 124. Li YF, Moody FG, Weisbrodt NW, Zalewsky CA, Coelho JC, Senninger N, Gouma D. Gallbladder contractility and mucus secretion after cholesterol feeding in the prairie dog. Surgery 1986; 100:900–904. 125. Yu P, Chen Q, Harnett K, Biancani P, Behar J. Impaired IP3 and DAG production in human gallbladder muscle with cholesterol stones (abstr). Gastroenterology 1994; 107: A1220. 126. Shaffer EA, McOrmond P, Duggan H. Quantitative cholescintigraphy: assessment of gallbladder filling and emptying and duodenogastric reflux. Gastroenterology 1980; 79: 899–906. 127. Portincasa P, Mitra P, Maghsoudldoo M, Murphy GM, Dowling RH. An inegrated study of gallbladder (GB) motorfunction in gallstone patients (abstr). Gut 1987; 28:1395A. 128. DeCarvalho S. Atherosclerosis: I. A leiomyoproliferative disease of the arteries resulting from the breakdown of the endothelial barrier to potent blood growth factors. Angiology 1995; 36:497–710. 129. Festi D, Frabboni R, Bazzoli F, Sangermano A, Ronchi M, Rossi L, Parini P, Orsini M, Primerano AM, Mazzella G, Aldini R, Roda E. Gallbladder motility in cholesterol gallstone disease: effect of ursodeoxycholic acid administration and gallstone dissolution. Gastroenterology 1990; 99:1779–1785.
Page 60
130. van Erpecum KJ, vanBergeHenegouwen GP, Stolk MFJ, Hopman WPM, Jansen JBMJ, Lamers CBHW. Fasting gallbladder volume, postprandial emptying and cholecystokinin release in gallstone patients and normal subjects. J Hepatol 1992; 14:194–202. 131. Kishk SMA, Darweesh RMA, Dodds WJ, et al. Sonographic evaluation of resting gallbladder volume and postprandial emptying in patients with gallstones. AJR 1987; 148: 875–879. 132. Forgacs IC, Murphy GM, Dowling RH. Influence of gallstones and UDCA on gallbladder emptying. Gastroenterology 1984; 87:299–307. 133. Pauletzki JG, Cicala M, Holl J, Sauerbruch T, Schafmayer A, Paumgartner G. Correlation between gallbladder fasting volume and postprandial emptying in patients with gallstones and healthy controls. Gut 1993; 34:1443–1447. 134. Jazrawi RP, Pazzi P, Petroni ML, Prandini N, Paul C, Adam JA, Northfield TC. Postprandial gallbladder motor function: refilling and turnover of bile in health and cholelithiasis. Gastroenterology 1995; 109:582–591. 135. van der Weft SD, vanBergeHenegouwen GP, Palsma DM, Ruben AT. Motor function of the gallbladder and cholesterol saturation of duodenal bile. Neth J Med 1987; 30:160–171. 136. Pomeranz IS, Shaffer EA. Abnormal gallbladder emptying in a subgroup of patients with gallstones. Gastroenterology 1985; 88:787–791. 137. Putz P, Willems G. Effect of a lithogenic diet on cell proliferation in the murine gallbladder epithelium. Digestion 1981; 22:16–23. 138. Wahlin T. Effects of lithogenic diets on mouse gallbladder epithelium: a histochemical, cytochemical and morphometric study. Virchows Arch B Cell Pathol 1976; 22:273–286. 139. Wang DQH, Paigen B, Carey MC. Phenotipic characterization of Lith genes determining susceptibility to cholesterol (Ch) gallstone formation in inbred mice (abstr). Hepatology 1995; 22:289A. 140. Khanuja B, Cheah YC, Hunt M, Nishina PM, Wang DQ, Chen HW, Billheimer JT, Carey MC, Paigen B. Lith 1, a major gene affecting cholesterol gallstone formation among inbred strains of mice. Proc Natl Acad Sci USA 1995; 92:7729–7733. 141. Hayashi A, Lee SP, Savard C. Bidirectional transfer of cholesterol between gallbladder epithelial cells and bile. Am J Physiol 1996; 271:G410–G414. 142. Tilvis RS, Aro J, Stranberg TE, Lempinen M, Miettinen TA. Lipid composition of bile and gallbladder mucosa in patients with acalculous cholesterolosis. Gastroenterology 1982; 82:607–615. 143. Metzger AL, Adler R, Heymsfield S. Diurnal variations in biliary lipid composition. N Engl J Med 1973; 288:333–336. 144. Sahlin S, Ahlberg J, Reihner E, Stahlberg D, Einarsson K. Cholesterol metabolism in human gallbladder mucosa: relationship to cholesterol gallstone disease and effects of chenodeoxycholic acid and ursodeoxycholic acid treatment. Hepatology 1992; 16:320–326. 145. Xu QW, Shaffer EA. The potential site of impaired gallbladder contractility in an animal model of cholesterol gallstone disease. Gastroenterology 1996; 110:251–257. 146. Yu P, Chen Q, Biancani P, Behar J. Membrane cholesterol alters gallbladder muscle contractility in prairie dogs. Am J Physiol 1996; 271:G56–G61. 147. Yu P, Chen Q, Harnett KM, Amaral J, Biancani P, Behar J. Direct G protein activation reverses impaired CCK signaling in human gallbladders with cholesterol stones. Am J Physiol 1995; 269:G659–G665. 148. Brenner RR. Effect of unsaturated acids on membrane structure and enzyme kinetics. Prog Lipid Res 1984; 23:69–96. 149. Phillips MC. Cholesterolphospholipid interactions and the exchangeability of cholesterol between membranes. In: PJ Quin, RJ Cherry, eds. Structural and Dynamic Properties of Lipids and Membranes. Portland, OR: London, 1992, pp 103–118. 150. Portincasa P, van Erpecum KJ, Jansen A. Renooij W, Gadellaa M, vanBergeHenegou
Page 61
wen GP. Nucleation behavior of various cholesterol (xol) crystals in bile from gallstone patients (abstr). Gastroenterology 1995; 108:A1150. 151. Tint GS, Salen G, Shefer S. Effect of ursodeoxycholic acid and chenodeoxycholic acid on cholesterol and bile acid metabolism. Gastroenterology 1986; 91:1007–1018. 152. van de Heijning BJM, van de Meeberg P, Portincasa P, Doornewaard H, Hoebers FJP, van Erpecum KJ, vanBergeHenegouwen GP. Effects of ursodeoxycholic acid therapy on in vitro gallbladder contractility in patients with cholesterol gallstones. Dig Dis Sci 1999; 44:190–196. 153. Li YF, Weisbrodt NW, Moody FG, Coelho JU, Gouma DJ. Calciuminduced contraction and contractile protein content of gallbladder smooth muscle after highcholesterol feeding of prairie dogs. Gastroenterology 1987; 92:746–750. 154. Yu P, Harnett KM, Biancani P, De Petris G, Behar J. Interaction between signal transduction pathways contributing to gallbladder tonic contraction. Am J Physiol 1994; 265:1082–1089. 155. De Petris G, Yu P, Biancani P, Behar J. GTP and inositol triphosphate restores the gallbladder contraction after CCK stimulation in prairie dogs fed a high cholesterol diet (abstr). Gastroenterology 1993; 104:A359. 156. Xiao ZL, Chen Q, Amaral J, Biancani P, Behar J. Excess membrane cholesterol alters CCK and VIPinduced G protein activation in human gallbladders with cholesterol tones (abstr). Gastroenterology 1998; 114:A861. 157. Xiao ZL, Chen Q, Amaral J, Biancani P, Jensen RT, Behar J. Excess membrane cholesterol alters CCK receptor binding affinity and capacity of human gallbladder muscle (abstr). Gastroenterology 1998; 114:A86. 158. Yu P, De Petris G, Biancani P, Amaral J, Behar J. Abnormal calcium channel function in smooth muscle from human gallbladders with cholesterol stones. Gastroenterology 1993; 104:A606. 159. Jennings LJ, Wei Xu Q, Nelson MT, Mawe GM. Cholesterol modulates guineapig gallbladder smooth muscle action potential characteristics (abstr). Gastroenterology 1998; 114:A772. 160. Ryan JP. Motility of the gallbladder and biliary tree. In: Johnson LR, ed. Physiology of the Gastrointestinal Tract. New York: Raven Press, 1987, pp 695–722. 161. Itoh Z, Takahashi I. Periodic contractions of the canine gallbladder during the interdigestive state. Am J Physiol 1981; 240:G183–G189. 162. Itoh Z, Takeuki I, Aizawa I, Mori K, Taminato Y, Seino Y, Imura H, Yanaihara N. Changes in plasma motilin concentration and gastrointestinal contractile activity in conscious dogs. Am J Dig Dis 1978; 23:929–935. 163. Toouli J, Bushell M, Stevenson G, Dent J, Wycherly A, Iannos J. Gallbladder emptying in man related to fasting duodenal migrating motor contractions. Aust N Z J Surg 1986; 56:147–151. 164. Marzio L, Neri M, Capone F, Di Felice F, De Angelis C, Mezzetti A, Cuccurullo F. Gallbladder contraction and its relationship to interdigestive duodenal motor activity in normal human subjects. Dig Dis Sci 1988; 33:540–544. 165. Qvist N, Oster Jorgensen E, Rasmussen L, Kraglund K, Pedersen SA. The relationship between gallbladder dynamics and the migrating motor complex in fasting healthy subjects. Scand J Gastroenterol 1988; 23:562–566. 166. Nilsson BI, Svenberg T, Tollstrom T, Hellstrom PM, Samuelson K, Schnell PO. Relationship between interdigestive gallbladder emptying, plasma motilin and migrating motor complex in man. Acta Physiol Scand 1990; 139:55–61. 167. Scott RB, Strasberg SM, ElSharkawy TY, Diamant NE. Regulaton of the fasting enterohepatic circulation of bile acids by the migrating myoelectric complex in dogs. J Clin Invest 1983; 71:644–654. 168. Takahashi I, Dodds WJ, Hogan WJ, Itoh Z, Baker K. Effect of vagotomy on biliarytract motor activity in the opossum. Dig Dis Sci 1988; 33:481–489.
Page 62
169. Ao YF. Contractile function of the gallbladder after gastrectomy. Chung Hua Wai Ko Tsa Chih 1990; 28:386–389, 444. 170. Takahashi T, May DO. Cholinergic dependence of gallbladder response to cholecystokinin in the guinea pig in vivo. Am J Physiol 1991; 261:G565–G569. 171. Nino Murcia M, Burton D, Chang P, Stone J, Perkash I. Gallbladder contractility in patients with spinal cord injuries: a sonographic investigation. Am J Roentgenol 1990; 154:521–524. 172. Tandon RK, Jain RK, Garg PK. Increased incidence of biliary sludge and normal gallbladder contractility in patients with high spinal cord injury. Gut 1997; 41:682–687. 173. Conte VP. Normal neuronal features of the human gallbladder and structural changes in cholelithiasis patients. Rev Hosp Clin Fac Med Sao Paulo 1989; 44:211–213. 174. Plevris JN, Harrison DJ, Bell JE, Bouchier IAD. The immunocytochemical characteristics of human gall bladder innervation. Eur J Gastroenterol Hepatol 1994; 6:151–158. 175. McKirdy ML, Johnson CD, McKirdy HC. Inflammation impairs neurally mediated responses to electrical field stimulation in isolated strips of human gallbladder muscle. Dig Dis Sci 1994; 39:2229–2234. 176. Moody FG, HaleyRussell D, Li YF, Husband KJ, Weisbrodt NW, Dewey RB. The effects of lithogenic bile on gallbladder epithelium. Ann Surg 1989; 210:406–415, discussion 415–416. 177. Conter RL, Washington JL, Liao CC, Kauffman GLJ. Gallbladder mucosal blood flow increases during early cholesterol gallstone formation. Gastroenterology 1992; 102:1764–1770. 178. Sanabria JR, Upadhya A, Müllen B, Harvey PRC, Strasberg SM. Effect of deoxycholate on immunoglobulin G concentration in bile: studies in humans and pigs. Hepatology 1995; 21:215–222. 179. Stolk MFJ, van de Heijning BJM, van Erpecum KJ, Verheem A, Akkermans LMA, vanBergeHenegouwen GP. Effect of bile salts on in vitro gallbladder motility: preliminary study. Ital J Gastroenterol 1996; 28:105–110. 180. Shaffer EA, Bomzon A, Lax H, Davison JS. The source of calcium for CCKinduced contraction of the guineapig gallbladder. Regul Pept 1992; 37:15–26. 181. Xu QW, Shaffer EA. The influence of bile salts on small intestinal motility in the guinea pig in vitro. Gastroenterology 1992; 103:29–35. 182. Bomzon A, Ljubuncic P. Bile acids as endogenous vasodilators. Biochem Pharmacol 1995; 49:177–183. 183. Berr F, Pratschke E, Fischer S, Paumgartner G. Disorders of bile acid metabolism in cholesterol gallstone disease. J Clin Invest 1992; 90:859–868. 184. Shoda J, He BF, Tanaka N, Matsuzaki Y, Osuga T, Yamamori S, Miyazaki H, Sjovall S. Increased deoxycholate in supersaturated bile of patients with cholesterol gallstones disease and its correlation with de novo syntheses of cholesterol and bile acids in liver, gallbladder emptying, and small intestinal transit. Hepatology 1995; 21:1291–1302. 185. Hussaini SH, Maghsoudloo M, Murphy GM, Petit R, Wass JAH, Dowling RH. Octreotide (OT) increases the proportion of deoxycholic acid in gallbladder (GB) bilethe prime mover in the pathogenesis of octreotideinduced gallbladder stones (GBS)? (abstr). Gut 1992; 33:57S. 186. Xu QW, Scott RB, Tan DTM, Shaffer EA. Slow intestinal transit: a motor disorder contributing to cholesterol gallstone formation in the ground squirrel. Hepatology 1996; 23:1664–1672. 187. Portincasa P, Di Ciaula A, Palmieri V, Velardi A, Van BergeHenegouwen GP, Palasciano G. Tauroursodeoxycholic acid, ursodeoxycholic acid and gallbladder motility in gallstone patients and healthy subjects. Ital J Gastroenterol 1996; 28:111–113. 188. Sylwestrowicz TA, Shaffer EA. Gallbladder function during gallstone dissolution: effect of bile acid therapy in patients with gallstones. Gastroenterology 1988; 95:740–748. 189. van Erpecum KJ, vanBergeHenegouwen GP, Stolk MFJ, Hopman WP, Jansen JB, Lamers CBHW. Effects of ursodeoxycholic acid on gallbladder contraction and cholecys
Page 63
tokinin release in gallstone patients and normal subjects. Gastroenterology 1990; 99:836–842. 190. Williams CN. Bile acid metabolism and the liver. Clin Biochem 1976; 9:149–152. 191. Strom BL, West SL. The epidemiology of cholesterol gallstone disease. In: Cohen S, Soloway RD, eds. Gallstones. New York: Churchill Livingstone, 1985, pp 1–26. 192. Attili AF, Casale R, Di Lauro G, Festuccia V, Natali L, Pasqualetti P. Assessment of gallbladder motility in patients with alcoholic hepatic cirrhosis after a fatty meal: a realtime ultrasonography study. Minerva Gastroenterol Dietol 1992; 38:45–48. 193. Severi C, Grider JR, Makhlouf GM. Identification of separate bombesin and substance P receptors on isolated muscle cells from canine gallbladder. J Pharmacol Exp Ther 1988; 245:195–198. 194. Poston GJ, Singh P, Maclellan DG, Yao CZ, Uchida T, Townsend CM, Jr, Thompson JC. Agerelated changes in gallbladder contractility and gallbladder cholecystokinin receptor population in the guinea pig. Mech Ageing Dev 1988; 46:225–236. 195. Pomeranz IS, Davison JS, Shaffer EA. Direct determination of the contractility of the guinea pig gallbladder: a new in vivo model. Can J Physiol Pharmacol 1985; 63:1038–1042.
Page 65
4— Canalicular Lipid Secretion James M. Crawford University of Florida College of Medicine, Gainesville, Florida I— Introduction Bile is the primary excretory route for organic compounds whose water solubility is insufficient for elimination in urine. The primary organic constituents of bile are shown in Table 1. Hepatic secretion of bile salts, phospholipids, and cholesterol into bile forms the basis for elimination of excess cholesterol and a wide variety of endogenous and exogenous amphiphilic and hydrophobic compounds from the body. Bile salt secretion per se is necessary for promoting lipid digestion in the gut. The coordinated secretion of bile salts, phospholipids, and cholesterol occurs at the hepatocyte canalicular membrane. This process is the result of molecular gradients established by hepatocellular proteins, followed by the inexorable operation of biophysical principles governing the behavior of these three classes of compounds. In this chapter, the structural and physical principles governing biliary lipid secretion are reviewed. II— The Bile Canaliculus A— The Hepatocyte In the mammalian liver, hepatocytes make up approximately 85% of all cells and are organized into cribriform, anastomosing sheets or ''plates" of hepatocytes surrounded on two sides by the vascular sinusoids. Between abutting hepatocytes are bile canaliculi—channels 1 to 2 µm in diameter formed by grooves in the plasma membranes of adjoining hepatocytes. Canaliculi are delineated from the vascular space by tight junctions running parallel to the axis of the bile canaliculi between abutting hepatocytes. The plasma membranes of the bile canaliculus define the apical pole of hepatocytes; the entirety of the plasma membrane on the vascular side of the tight junctional network is termed basolateral. Each hepatocyte possesses multiple apical canalicular domains (typically three) reflecting the multiple interfaces between hepatocytes and their neighbors (1). Numerous hepatocellular microvilli protrude into the canalicular space (Fig. 1). Including the microvilli, the area of the canalicular plasma membrane makes up about 12% of total surface area of the hepatocellular plasma membrane (1). When microvillar surface area is included, this amounts to an astounding surface area of 760 cm2/cm3 of liver volume (2), equivalent to over 100 m2 in one human liver. (This is compared to 240 m2 of apical surface membrane area in the human small intestine.) There frequently is a portion of the apical plasma membrane that laps over the tight junctional region of each cellular interface; this appears as a flattened microvillus in crosssectional images of canaliculi, yet it actually appears to be an intermittently continuous
Page 66 Table 1 Organic Solutes in Human Bile Solute
Concentration (mg/dL)
Bile salts
150–3500
Comment Amphiphilic steroid detergents
Phospholipid
28–810
Membrane phospholipid
Cholesterol
60–320
Waterinsoluble steroid
Bilirubin conjugates
50–200
Watersoluble tetrapyrrole
Protein
80
Watersoluble protein
<5
Watersoluble proteins
Lysosomal proteins
<5
Watersoluble proteins
Canalicular membrane proteins
6
Intrinsic membrane proteins
Plasma proteinsalbumin, other
marginal ridge (3). Although not proven, it is tempting to speculate that this marginal ridge provides flapvalve resistance to backflow of fluid into the vascular space. Intracellular actin and myosin filaments arranged circumferentially around the canaliculus help propel secreted bile along the canaliculi. The canalicular channels constitute the outermost reaches of the biliary tree; in the rat, they represent 0.22% of liver volume (2). Canalicular channels between hepatocytes in the perivenular region of the hepatic parenchyma are the most upstream, farthest from inflowing blood (via the portal vein and hepatic artery radicles within portal tracts) and farthest from outflowing bile (via portal tract interlobular bile ducts). Each anastomosing canalicular network spans a linear distance of between 0.5 to 1.0 mm, which is the distance between terminal hepatic venules and portal tracts in the mammalian liver. The canalicular channels merge en route to the periportal region of the hepatic parenchyma, ultimately entering intermediate channels termed canals of Hering (Fig. 2). Canals of Hering are transitional channels, composed of abutting hepatocytes and small bile duct cholangiocytes. The canals of Hering extend out from interlobular bile ducts into the parenchyma, penetrating approximately onethird of the distance between portal tracts and effluent hepatic venules (4). Canals of Hering then coalesce, penetrate the mesenchyme of the portal tract, and link up with interlobular bile ducts. On the basis of threedimensional reconstruction of normal
Figure 1 Schematic of the apical surface of a hepatocyte. A small portion of the hepatocellular apical membrane is depicted, showing the tight junctions delimiting the canalicular surface from the lateral cellular surface and microvilli protruding into the canalicular lumen. An abutting hepatocyte forms the other half of the channel. The pericanalicular actin microfilament web is depicted; this plays a role in canalicular contractility.
Page 67
Figure 2 Schematic of canalicular microanatomy in the hepatic lobule. Bile canalicular channels between hepatocytes (far more than depicted) anastomose to enter into canals of Hering, which drain into interlobular bile ducts within portal tracts. Canals of Hering extend up to onethird of the distance between portal tracts and terminal hepatic venules.
human liver, canaliculi feed into canals of Hering over an average radial sector of about 70 to 90° (5). The canals, in turn, are arranged around the interlobular bile duct in roughly 10µm steps. Although it is not yet known whether there is a random or spiral arrangement of the canals of Hering, the repeat distance for each canalicular network is likely to be on the order of every 50 to 100 µm. Latex injection studies indicate that canals of Hering are accompanied by penetrating terminal venules of the portal vein system (6). It therefore may be posited that the blood supply to the hepatic parenchyma is linked on a microscopic scale to the watershed for bile formation. This concept is formalized in the termed cholehepaton (6), which denotes the smallest parenchymal unit responsible for hepatocellular metabolic function and bile formation. B— Structural Lipid Although there may be some contribution of epithelial cells lining the biliary tree to secretion and absorption of bile salts (7), hepatocytes are solely responsible for the entry of phospholipid and cholesterol into bile. As biliary lipid must therefore enter bile from the canalicular pole of the hepatocyte, it is important to consider the assembly of the canalicular plasma membrane. The physical structure of the hepatocyte canalicular membrane is unique in mammalian biology, in that it contains the highest mole per liter of cholesterol and sphingomyelin and is the least fluid of cellular membranes (8). This renders the canalicular membrane highly resistant to the detergent action of bile salts within the canalicular space. Phosphatidylcholine is the major glycerophospholipid in the canalicular membrane, along with lesser amounts of phosphatidylethanolamine, phosphatidylserine, and phosphatidylinositol (Table 2). The molar ratios of cholesterol/phospholipid (0.70) and sphingomyelin/phosphatidylcholine (0.75) are higher than in any other hepatocellular membrane. Cholesterol binds with strong affinity to sphingomyelin, imparting a high resistance to detergent dissolution. As sphingomyelin is present in both hemileaflets of the canalicular membrane, interdigitation of the disparate acyl moieties of
Page 68 Table 2 Lipid Composition of Canalicular Membrane and Bile
Species
% Molar composition Canalicular membrane
Trivial name
Bile
PC
Phosphatidylcholine
35.5
90.0
PE
Phosphatidylethanolamine
23.8
4.4
PS
Phosphatidylserine
11.2
1.2
PI
Phosphatidylinositol
4.4
1.4
SPH
Sphingomyelin
22.1
2.7
Lipid classes
a
Class
C16:0
Palmitic acid
16:0/16:0
2.3
Trb
C16:1
Palmitoleic acid
16:0/16:1
0.6
0.6
C18:0
Stearic acid
16:0/18:0
0.4
C18:1
Oleic acid
16:0/18:1
6.9
8.2
C18:2
Linoleic acid
16:0/18:2
19.7
53.7
C20:0
Arachidic acid
16:0/20:4
16.2
14.7
C22:0
Behinic acid
16:1/18:2
0.5
0.5
C24:0
Lignoceric acid
18:0/18:1
1.1
0.3
C20:4
Arachidonic acid
18:0/18:2
15.0
9.9
18:0/20:4
18.7
2.7
18:1/18:2
2.5
2.2
15.6
7.0
Phosphatidylcholine fatty acyl species
>C20:4 Polyunsaturated fatty acids
a
(polyenes)
polyenes
Tr
Refers to fatty acyl groups in positions sn1/sn2. Tr = trace. Source: Adapted from Refs. 76, 154, and 193. b
the sphingomyelin between the hemileaflets is possible. This, combined with the high cholesterol content, renders the canalicular membrane highly shearresistant as well. The localization of sphingomyelin is significant, in that sphingomyelin is located predominantly in the external hemileaflet of the bilayer (9). In most eukaryotic cells, plasma membrane cholesterol is located mostly on the inner hemileaflet of the plasma membrane. However, since cholesterol exhibits a stronger affinity for sphingomyelinrich plasma membranes than other cellular membranes (10), one might suspect that cholesterol and sphingomyelin directly interact in the external hemileaflet of the canalicular membrane. While we must state that the precise localization of cholesterol in the canalicular membrane is unknown, the canalicular membrane is poised to place high quantities of cholesterol in direct contact with the canalicular lumen. A further consideration is the potential existence of cholesterolrich and cholesterolpoor microdomains (10,11). Within a sphingomyelin:cholesterolrich membrane, the formation of microdomains of more fluid lipid within this plasma membrane bilayer is possible. Stable microdomains of fluid phosphatidylcholine can be formed at 25°C. As will be seen, the formation of transient but unstable microdomains at the physiological temperature of 37°C may be a critical step in enabling biliary lipid secretion. Any excess cholesterol not bound by sphingomyelin within the surrounding bilayer will be capable of diffusing into the cholesterolpoor fluid microdomains. This may have relevance to the efflux of cholesterol into bile and is discussed further on. There are four potential mechanisms for the delivery of structural membrane lipids to the canalicular membrane (12,13): (a) vesicular traffic, (b) monomeric exchange through the
Page 69
action of cytosolic proteins, (c) lateral diffusion from the basolateral plasma membrane, and (d) transbilayer movement of lipid molecules between hemileaflets of the canalicular membrane. Only the general features of hepatocellular membrane trafficking are known; the specifics of lipid delivery to the canalicular membrane of the hepatocyte have not been worked out. There is an extensive and growing literature describing the vesicular trafficking of membrane proteins to the canalicular pole (14); structural lipid is a necessary accompaniment of such traffic. Vesiclemediated transcytosis from plasma to bile of bulk fluid and of proteins, such as albumin and IgA, necessitates the movement of considerable amounts of membrane lipid from the basolateral to canalicular pole of the hepatocyte. Based on published quantitative data (15,16), one can calculate that vesicles insert membrane lipid equivalent to 9% of the surface area of the canalicular membrane every minute (17). Such rates of delivery necessitate a robust membrane retrieval mechanism, which is plausible given evidence for the presence of active canalicular membrane transporters in subapical vesicles (18). Membrane retrieval must be highly selective, however, since the membrane proteins dipeptidyl peptidase IV and HA4 (an ectoATPase) are effectively retained within the canalicular membrane, being retrieved for intracellular degradation at the rate of approximately 6 to 7% per day (19). The actual lipid composition of these intracellular vesicles is not precisely defined, but they do not appear to be enriched in biliary phosphatidylcholines or in cholesterol. However ill defined, to date vesicular delivery of structural lipid appears to be the only mechanism operative for maintaining bulk canalicular membrane composition. A case in point is the delivery of sphingomyelin. Studies with many cell types have provided extensive evidence that sphingomyelin is synthesized predominantly in the Golgi apparatus (20,21) and delivered to the plasma membrane in sphingomyelincontaining vesicles (reviewed in Refs. 22 to 24). Notably, sphingomyelin is delivered to the plasma membrane at rates comparable to protein delivery (t 1/2 = 20 to 30 min) (25), suggesting similar mechanisms of vesicular sorting and trafficking. In other polarized cell types, delivery of sphingomyelincontaining vesicles to the apical pole is partially microtubuledependent (26), implicating both vesiclemediated trafficking and other delivery mechanisms. Intriguing support for alternative pathways comes from the observation in CaCo2 intestinal cells that disruption of protein trafficking from the Golgi apparatus to the plasma membrane by either monensin or Brefeldin A did not abrogate sphingomyelin delivery but rather abolished specific sorting mechanisms, leading to increased apical content of sphingomyelin following Brefeldin A treatment (26). In rat hepatocytes, while monensin and Brefeldin A treatment completely disrupted protein transport from the Golgi apparatus, they did not disrupt sphingomyelin delivery (27). However, because isolated rate hepatocytes do not retain their polarity, these experiments could not distinguish between basolateral and canalicular delivery mechanisms. Thus, the specifics of sphingomyelin delivery to the canalicular membrane of hepatocytes remain unknown, although the use of new polarized cell systems (28) and techniques to identify relevant proteins (29) may permit progress in this area. Vesicular trafficking of lipid does not account for the delivery of biliary phosphatidylcholine. First, despite evidence for assembly of biliary phosphatidylcholines in the endoplasmic reticulum, no subcellular vesicle populations of these lipids have been identified (30). Second, biliary phosphatidylcholines are derived from the endoplasmic reticulum (31,32) rather than from transcytosis of basolateral plasma membrane lipids. Third, the transport of newly synthesized phosphatidylcholine from the endoplasmic reticulum to the plasma membrane is extremely rapid (t 1/2 < 1 min) and is ATPindependent and insensitive to inhibitors that disrupt protein traffic (22). In contrast, monomeric exchange of lipid between cytosolic proteins and the canalicular membrane appears to have no role in the maintenance of canalicular structure but may be critical for delivery of biliary phospholipid. As will be seen, transbilayer movement of such phospholipid then presents the appropriate molecules to the bile canalicular lumen. Lateral diffusion presents an interesting dilemma in that tight junctions present a barrier to diffusion within the external hemileaflet but not the internal hemileaflet (33). For structural phospholip
Page 70 Table 3 Protein Content of Hepatocellular Membranes and Bile Organelle Protein mass (%)
Molecules of protein: molecules of phospholipid
Endoplasmic reticulum
70%
1:23
Golgi apparatus
50%
1:40
Intracellular vesicles
25%
1:70
Canalicular membrane
45%
1:43
Bile
< 1%
1:>4000
ids, no evidence has been presented to suggest that lateral diffusion from basolateral to apical domains plays a role in maintaining canalicular membrane structure. As discussed below, lateral diffusion may be critical for the delivery of sterol to bile. Consideration must also be given to the protein content of the hepatocyte canalicular membrane. As shown in Table 3, lipid membranes of the hepatocyte contain high percentages of intrinsic protein. The endoplasmic reticulum, for example, is virtually saturated with protein at 70% protein (g protein/100 g). The canalicular membrane is 45% protein, including many of the critical transport proteins for the secretion of biliary solutes (see Table 4). Notable is the extremely low content of canalicular membrane proteins in bile. As discussed subsequently, the high rate of lipid secretion on the one hand and very low rate of membrane protein release on the other must be accomplished by a highly selective mechanism. III— Biliary Lipids A— Bile Salts Bile acids are amphiphilic, detergentlike molecules synthesized in the liver from cholesterol (34). They possess one, two, or three hydroxyl groups on the steroid moiety and a 5carbon side chain that terminates in a carboxyl group. Hepatic amidation of the carboxyl group with the amino acids taurine or glycine converts these molecules from free acids with a titratable carboxyl group to bile salts, which remain charged at physiological pH. The water solubility is thereby increased, enabling bile salts to be extremely effective solubilizing agents within the Table 4 Proteins in the Human Hepatocyte Canalicular Membrane Pertinent to Biliary Lipid Secretion Solute
Transport protein
Bile salts
BSEP: bile salt export pumpa
Phosphatidylcholine
MDR3: multidrugresistance P glycoprotein
Cholesterol
unknown
Chromosomal location 2q36 37q21
b
Organic anions
MRP2: multidrugresistance protein
10q24
Amphiphilic basic or cationic compounds
MDRI: multidrugresistance P glycoprotein
17q21
BSEP; also referred to as sister gene of Pglycoprotein, or SPGP. MRP2; also known as canalicular multispecific organic anion transporter, or CMOAT. Source: Based on Muller M, Jansen PLM. The secretory function of the liver: new aspects of hepatobiliary transport. J Hepatol 28:344–354, 1998. a
b
Page 71
biliary tree and gut lumen. Their effectiveness as detergents also presents a danger: excessive accumulation of bile salts may have severe local consequences, since these molecules are capable of disrupting lipid structure in cellular membranes as well (35). The synthesis of bile acids from cholesterol itself involves about 20 different enzymes distributed among the endoplasmic reticulum, cytosol, mitochondria, and peroxisomes, as addressed elsewhere in this volume. Physiological conservation of bile salts is accomplished by the enterohepatic circulation, whereby intestinal reabsorption of bile salts (primarily in the ileum) returns bile salts to the liver via the splanchnic circulation (36). Thus, the majority (>95%) of bile salts secreted into bile are taken up across the basolateral plasma membrane of the hepatocyte from the sinusoidal space; the small remainder is derived from de novo hepatic synthesis. Bile salt uptake across the basolateral plasma membrane of the hepatocyte is concentrative in that intracellular bile salt concentrations appear to be three to fourfold greater than in the 10 to 50 M concentrations found in plasma (37,38). Uptake into the mammalian liver occurs primarily via a 49kDa Na+/bile salt cotransporter [designated Ntcp (39); step la in Fig. 3], which utilizes the sodium gradient established by the basolateral Na+, K+ATPase. A phylogenetically older, Na+independent transporter for bile salts may also be operative to some extent in the mammalian liver, particularly in the immature individual (40). For the small portion of bile salts that is unconjugated in the gut by bacterial action and returned to the liver as free bile acids and for bile salts that are less hydrophilic than the trihydroxycholates, diffusion across the basolateral plasma membrane bilayer accounts for a portion of hepatocellular uptake (41). Following hepatocellular bile salt uptake from plasma, bile salts are avidly bound by cytosolic proteins (Fig. 3, step 1b) (42). The predominant binding proteins appear to be 3 hydroxysteroid dehydrogenase in the rat (43) and a related protein, dihydrodiol dehydrogenase in the human (44). These cytosolic proteins are members of a monomeric reductase gene family (44). Inhibition of bile salt binding to 3 hydroxysteroid dehydrogenase by indomethacin in the rat delays bile salt delivery to bile (45–47), suggesting that binding to cytosolic protein facilitates rather than retards delivery to the canalicular membrane. Bile salts are known to interact with intracellular organelles (48–53). They may even be transported into the lumina of the endoplasmic reticulum or trafficking vesicles through the action of a protein transporter (54), in keeping with observations in other cell systems (55). Although a role for vesicular trafficking of bile salts was proposed some years ago (56), the primary and perhaps only route of bile salt delivery to the canalicular pole of the hepatocyte appears to be cytosolic diffusion of proteinbound bile salts, with reversible but nonvectorial interactions of bile salts with intracellular membranes (17). Information is not available on whether diffusion of either free bile salt monomers or bile saltprotein complexes is potentially ratelimiting for transcellular transport, as has been implicated for other small molecules such as 3,5,3'triiodothyronine (57), dextrans (58), and fatty acids (59). B— Phospholipids More than 90% of biliary phospholipid is phosphatidylcholine, in contrast to the lipid composition of the canalicular membrane (Table 2). A contrast holds also for the fatty acyl species of the canalicular membrane and biliary phospholipid. Unlike the canalicular membrane, which contains a broad mixture of fatty acyl species (Table 2), biliary phosphatidylcholines are predominantly composed of palmitic, linoleic, and stearic acids, with palmitic (80%) or stearic (20%) acids esterified in the sn1 positions and linoleic, oleic, and arachidonic acids (in rank order) in the sn2 position (60). The biliary phospholipid species are more hydrophilic than the bulk lipids in the canalicular membrane (34) and are more susceptible to elution from the membrane by bile salts (61). Moreover, phosphatidylcholine and sphingomyelin are preferentially located in the exoplasmic hemileaflet of the canalicular membrane, phosphatidylethanolamine is evenly distributed, and phosphatidylserine and phosphatidylinositol are exclusively in the endoplasmic hemileaflet (62). Any proposed mechanisms for delivery of biliary phos
Page 72
Figure 3 Routes for bile salt (1), phosphatidylcholine (2), and cholesterol (3) delivery to bile. Bile salts are taken up across the basolateral membrane via the sodiumdependent bile salt transporter, ntcp (step 1a), and bind to intracellular proteins (step 1b). Delivery to the canalicular membrane is primarily via diffusion while bound to protein. Free bile salts are excreted into bile as monomers by specific transporters (step 1c). Biliarytype phosphatidylcholine appears to be delivered from the endoplasmic reticulum primarily via binding to phosphatidylcholine transfer protein (PCTP, step 2a); a role for vesicular trafficking through the Golgi apparatus (step 2b), followed by apical vesicle trafficking (step 2c), cannot be excluded. Biliary cholesterol is derived primarily from cholesterol ester in the lipid core and free cholesterol in the surface monolayer of circulating lipoproteins. Lipoproteins such as LDL enter the cell via receptormediated endocytosis (step 3a, see inset, which depicts the molecular structure of LDL). Lysosomal degradation of the lipoproteins releases free cholesterol, which may transfer to the canalicular plasma membrane by binding to cytosolic proteins such as sterol carrier protein 2 (SCP2, step 3b). Alternatively, intracellular distribution to the Golgi network (step 3c) may permit vesicular trafficking with sphingomyelinrich vesicles (step 2c). In the case of HDL, binding to the SRBI scavenger receptor (not shown) permits exchange of free cholesterol and cholesteryl esters to the basolateral plasma membrane (step 3d, see inset). Following the hydrolysis of cholesteryl esters by hepatic lipase, the free cholesterol may flip to the internal hemileaflet and diffuse laterally past the tight junctional region for entry into the canalicular plasma membrane domain. Intracellular structures and inset are drawn to scale. Bar = 2 m. Inset, bar = 2 nm.
pholipids to the canalicular membrane must be consistent with these striking differences in phospholipid classes and distribution between the canalicular membrane and bile. Hepatic phosphatidylcholines may be derived from several sources: uptake of exogenous phospholipids from circulating lipoproteins; de novo synthesis via acylation of glycero3phosphate to phosphatidic acid, dephosphorylation to form diglyceride, and reaction with CDPcholine to form phosphatidylcholine (the Kennedy pathway), headgroup remodeling of existing glycerophospholipids, and deacylation of triglycerides to provide diglycerides. With the exception of one report (63), exogenous phospholipids contained in circulating lipoproteins do not appear to contribute significantly to biliary phospholipids. [Liposomal phosphatidylcholines
Page 73
injected into the circulation as a pharmaceutical measure also are extensively degraded and altered by hepatocytes (64).] Synthesis of phosphatidylcholine de novo is quantitatively small and contributes only about 3% to biliary phosphatidylcholine output (65) unless phosphatidylcholine synthesis is stimulated by experimental infusion of biliarytype free fatty acids (66). Remodeling of other glycerophospholipids—such as Nmethylation of phosphatidylethanolamine, base exchange, or transesterification of lysolecithin—is not a significant contributor to biliary phosphatidylcholine synthesis (67). Rather, utilization of existing acylglycerides within the endoplasmic reticulum is the primary route for hepatic phosphatidylcholine synthesis (32). In particular, deacylation of triglycerides is the major source of biliary phosphatidylcholine molecules (67). Endogenous hepatocellular stores of triglycerides appear to be sufficient, since biliary phosphatidylcholine secretion is maintained in the absence of lipoprotein lipids. Compartmentation of biliary phosphatidylcholines within subcellular membranes does not appear to be operative (67). Instead, subselection of phosphatidylcholines for delivery to the canalicular membrane appears to occur at the level of intracellular trafficking. An answer to the enigma of how biliary phosphatidylcholines are delivered to the canalicular membrane in a highly specific bile saltstimulated fashion has been proposed by Cohen and colleagues (68). Phosphatidylcholinetransfer protein (PCTP) is an abundant cytosolic protein, and generally is thought to promote bidirectional molecular exchange between membranes (69). Cohen et al. demonstrated that PCTP is highly efficient in the unidirectional transfer of phosphatidylcholine molecules from model membranes of lipid composition matching that of the endoplasmic reticulum to model membranes mimicking the canalicular membrane (Fig. 3, step 2a) (68). The transfer process is relatively specific for biliarytype phosphatidylcholine molecules. Moreover, it is markedly stimulated by the presence of bile salts at the low micromolar concentrations thought to exist within the hepatocyte (step 1b). Realistic calculations indicate that PCTPmediated transfer can account for all the biliary phosphatidylcholine present in bile. An additional cytosolic lipidbinding protein, sterol carrier protein 2 (SCP2), may play a scavenger role in phosphatidylcholine delivery (70). While vesicular delivery of biliarytype phosphatidylcholine via the Golgi apparatus may yet be operative (Fig. 3, steps 2b and 2c), transfer via PCTP constitutes an attractive hypothesis for the highly efficient delivery of biliary type phosphatidylcholine molecules to the canalicular membrane. C— Cholesterol In humans, daily dietary intake of cholesterol is estimated to be 0.3 to 0.5 g/day, and daily biosynthesis (much of it in the liver) is between 0.6 to 0.9 g/day, for a total input of about 1.2 g/day (71). The predominant means by which the body eliminates excess cholesterol is via secretion into bile, either as free cholesterol or as bile acids. Fecal loss of bile acids alone accounts for 0.2 to 0.6 g/day in humans (36), and biliary cholesterol secretion accounts for an additional 0.6 g/day. Sloughing of the skin (0.085 g/day) and use for steroid hormone biosynthesis (0.05 g/day) round out the balance sheet (71). These numbers are given to make clear the importance of the liver in general and the hepatocyte canalicular membrane in particular for maintenance of cholesterol homeostasis. Without effective mechanisms to eliminate both bile salts and cholesterol in bile, our mammalian existence would crumble into atherosclerotic oblivion. The solubility of cholesterol in water is vanishingly small. As bile is an aqueous fluid, cholesterol must be solubilized by other agents. Bile salts alone increase the aqueous solubility of cholesterol severalfold. The combined action of bile salts and phosphatidylcholine increases cholesterol solubility over a millionfold (72). Thus, it is a biological tour de force that the canalicular membrane is capable of immense rates of secretion of monomeric bile salts (12 to 36 g/day in humans) (8); that bile salt secretion "pulls" vesicles of phospholipid, presumably composed of phosphatidylcholine, out of the explasmic face of the canalicular membrane (73)
Page 74
at rates of 4 to 12 g/day (see below); and that these two processes permit the solubilization of cholesterol in bile and its effective elimination from the body at rates of up to 0.6 g/day. It is no surprise that excess rates of cholesterol secretion predispose to its precipitation as cholesterol gallstones (74). Cholesterol is found almost exclusively in cell membranes. Despite the fact that intracellular membranes in the hepatocyte (particularly the endoplasmic reticulum and mitochondria) possess a surface area 20fold greater than that of the plasma membrane (1), 80% of cholesterol is in the plasma membrane (75). In the basolateral membrane, the molar ratio of cholesterol to phospholipid is 0.41. It is 0.76 in the canalicular membrane (76), giving this membrane the highest molar content of any hepatocyte membrane fraction. Cholesterol has a high affinity for membranes rich in sphingomyelin (10,77,78), and the sphingomyelin content of the canalicular membrane also is higher than that of any other heptaocellular membrane (molar ratio 0.62 in the canalicular membrane and 0.25 in the basolateral membrane) (8). As the canalicular domain of the hepatocyte occupies only 13% of the total plasma membrane surface area of the hepatocyte (1), most cholesterol still resides in the basolateral plasma membrane. Nevertheless, the unique lipid composition of the canalicular membrane is in keeping with its key role in the elimination of cholesterol into bile. As discussed below, most of the cholesterol secreted into bile is derived from circulating plasma lipoproteins (22,79), principally lowdensity lipoprotein (LDL), intermediatedensity lipoprotein (IDL), highdensity lipoprotein (HDL), and chylomicron remnants, in that order (34). Newly synthesized cholesterol does not contribute substantively to biliary cholesterol secretion (80,81). However, for that cholesterol which is newly synthesized in the liver, a greater proportion is secreted into bile than into plasma (82). Lipoprotein cholesterol exists either as free cholesterol in the outer phospholipid monolayer or as cholesterol esters within the triglyceride core (83). Receptor mediated endocytosis appears to be the primary mechanism for uptake of LDL lipid and apoprotein (Fig. 3, step 3a); this cholesterol is largely reesterified for secretion back into plasma as VLDL (84). Lysosomal hydrolysis of lipoprotein cholesteryl esters also generates free cholesterol (10,85,86). Rapid transit to the basolateral and apical plasma membranes (75) implicates cytosolic trafficking via binding proteins, similar to that seen for phosphatidylcholine. A strong candidate is sterol carrier protein2 (SCP2) (Fig. 3, step 3b) (87–89); this particularly may play a role for cholesterol delivery to bile (90). Trafficking via membranes (10), including an as yet illdefined sterolrich organelle (91), has also been proposed. In particular, the synthesis of sphingomyelin within the Golgi apparatus (20,21) and its delivery to the plasma membrane via vesicles (27,92,93) raises the possibility that cholesterol is preferentially delivered to the canalicular membrane via the Golgi apparatus (Fig. 3, step 3c, followed by step 2c) in sphingomyelinrich vesicles or membrane "rafts" (13,22–24,94,95). Cholesterol released from LDL degradation in lysosomes does not appear to pass through the endoplasmic reticulum (10). As elucidated further on, plasma HDL may play a critical role in contributing cholesterol for biliary secretion. Free cholesterol and cholesteryl esters in HDL may partition into the outer hemileaflet of the hepatocellular basolateral plasma membrane (Fig. 3, step 3d) (83,96–98). Cholesterol diffuses readily in the planar membrane and may freely flip across the plasma membrane to the inner hemileaflet. From there, distribution among intracellular membranes occurs rapidly. Endogenously synthesized cholesterol is not likely to travel via vesicles, since inhibitors of vesicle trafficking, particularly microtubule inhibitors, do not affect its movement (99). Endosomal recycling of sphingomyelinrich membranes from the canalicular domain (24,95) may also act as scavengers for intracellular cholesterol, in essence returning cholesterol that may have distributed to the cell interior to the plasma membrane (100). Considering that sphingomyelin synthesis can occur on the inner hemileaflet of recycling endosomes (101), cholesterol might also reside on the inner hemileaflet and thereby be delivered to the canalicular membrane via endosomal fusion in the correct topology for elution into bile. Despite the attractiveness of these various hypotheses for membrane trafficking of cholesterol, assiduous
Page 75
efforts to isolate cholesterolcarrying ''vesicles" from rat liver secreting excess amounts of cholesterol into bile have not been successful (102); a potential role for intracellular delivery vesicles in humans has not been excluded (103). IV— Lipid Secretion into Bile A— Bile Salt Secretion into Bile Bile salts are secreted into the canalicular lumen against a considerable concentration gradient, which has been estimated at about 100fold (38,104). The primary transport protein is the bile salt export protein (bsep), formerly referred to as the "sister of Pglycoprotein" (spgp) (105). This is an ATPindependent transport protein that is a member of the ABCtype transporter family. Physiological studies have demonstrated that canalicular membrane bile salt transport capacity mediated by this protein is modulated by transhepatic bile salt flux. In humans, a 12h fast followed by a meal leads to a two to sixfold increase in portal venous blood concentrations and hepatic secretion of bile salts (106). In rats, exposure of the liver to increased bile salt flux increases the maximal hepatic secretory rate (107–109). While local allosteric regulation may play some role (110), the major mechanism regulating canalicular membrane transport of bile salts is vesicular delivery of the relevant transport proteins (17), presumably bsep. Apical membrane transport proteins appear to reside in a subapical storage/retrieval endocytic compartment (18,111). Exposure of the liver to such stimuli as an osmotic load (112) or a bile salt load (113) stimulates the insertion of intracellular vesicles containing bile salt transport proteins into the canalicular membrane; a decrement in bile salt flux leads to a decrease in transport capacity and protein content (113). The mechanisms for upregulation appear to involve bile salt stimulation of cAMP and calciumdependent intracellular signaling systems (114), with a possible additional role for direct interaction of bile salts with hepatocellular membranes on the basis of hydrophobicity (115). B— Phosphatidylcholine Secretion into Bile A hepatocellular delivery system for phosphatidylcholine utilizing cytoplasmic phosphatidylcholine transfer protein (PCTP) will place phosphatidylcholine molecules within the endoplasmic hemileaflet of the canalicular membrane (Fig. 4, steps 1a and 1b) (68). Although spontaneous "flipping" of phospholipid across membranes is exceedingly slow (116), phospholipid translocase function has been identified both in microsomal (117) and canalicular membrane preparations (Fig. 4, step 2c) (118). A major finding was the discovery that mice lacking the mdr2 Pglycoprotein gene product are essentially incapable of secreting phospholipid into bile (119). Subsequent studies conclusively demonstrate that this Pglycoprotein acts as a phospholipid translocator (119–121). The mdr2 gene product may not be the only mechanism for phospholipid translocation, as preliminary evidence has been presented that other gene products may play a similar role (122,123). Collectively, these recent investigations implicate proteinmediated translocation of biliary phosphatidylcholine molecules from the endoplasmic to the exoplasmic hemileaflet of the canalicular membrane as a key event in biliary phospholipid secretion (124). The next event in phospholipid secretion is thought to be one of two possibilities (Fig. 4): (a) the formation of vesicles budding from the external hemileaflet of the canalicular membrane or (b) solubilization of phosphatidylcholine molecules from the external hemileaflet by bile salt micelles (125–127). The evidence for these two possibilities is examined below. 1— Vesicular Secretion Phospholipid vesicles are well documented in bile, both by ultrastructural and biophysical techniques (reviewed in Ref. 17). Reasoning that routine chemical fixation was too slow to
Page 76
Figure 4 Schematic of canalicular phospholipid secretion, depicting (A) vesicular and (B) micellar secretion. Intracellular bile salts interact with the endoplasmic reticulum membranes (step 1a), promoting the release of phosphatidylcholines and binding by cytosolic PCTP (step 1b). PCTP then deposits individual molecules within the internal hemileaflet of the canalicular membrane (step 1c). The possibility of vesicular traffic via the Golgi apparatus (step 1d) has not been excluded. Regardless of delivery mechanism, the mdr2 Pglycoprotein "flips" phosphatidylcholine molecules from the internal to the external hemileaflet (step 1e). Monomeric bile salts excreted into the lumen by specific transport proteins (step 2a) are capable of promoting vesiculation of biliarytype phosphatidylcholine microdomains within the external hemileaflet of the canalicular membrane (panel A, step 2b). The zone of vesicle budding is 50% thicker than the surrounding canalicular membrane bilayer, but the molecular structure of this region is unknown. Alternatively, biliarytype phosphatidylcholines may be extracted into mixed micelles (panel B, step 2c).
capture the vesiculation event, our laboratory prepared rat liver tissue for electron microscopy with an accelerated chemical fixation protocol and with ultrarapid in situ cryofixation (73). By both techniques, phospholipid vesicles were present on the exoplasmic face of the canalicular membrane, and their presence was dependent upon the cosecretion of physiological bile salts. A membrane barrier was always maintained, suggesting that bilayer exocytosis did not occur. Rather, a distinct zone of fusion was observed between adherent vesicles and the canalicular membrane. Our findings are consistent with the hypothesis that phospholipid vesicles form from microdomains of biliarytype phosphatidylcholine within the explasmic hemileaflet of the canalicular membrane (128). Quantitative estimates indicate that this vesiculation mechanism can account for all phospholipid secreted into bile (73) and that this process elutes approximately 8% of the external hemileaflet surface area into bile per minute (Fig. 5) (17). According to this hypothesis, three steps are required for canalicular phosphatidylcholine secretion: delivery to the canalicular membrane endoplasmic hemileaflet via PCTPmediated cytosolic diffusion, proteinmediated translocation from the endoplasmic to the exoplasmic hemileaflet, and bile saltdependent vesiculation of lipid microdomains in the exoplasmic hemileaflet. This proposal is supported by physiological studies of two types. First, excessive secretion of amphiphilic organic anions into the canalicular lumen along with bile salts "uncouples" phospholipid secretion from bile salt secretion [(129–132), see below]. This now appears to be due to the binding of the cosecreted solutes with bile salts within the canalicular lumen, preventing bile salt backextraction of phosphatidylcholine from the canalicular membrane (30). Second, bile salt—induced secretion of phosphatidylcholine is enhanced by an in
Page 77
Figure 5 Schematic of membrane dynamics at the canalicular pole of the hepatocyte. Transcytotic vesicular traffic, presumably passing through a subapical sorting compartment, delivers structural membrane lipid to the canalicular membrane at a rate of about 9% per minute (17). This vesicular traffic does not appear to contribute to biliary lipid secretion. Rather, deposition of phosphatidylcholine molecules via the action of PCTP is involved, followed by proposed vesiculation of the external hemileaflet of the canalicular membrane (see Fig. 4). Notably, the rate of phospholipid secretion into bile is computed as 8% of the external surface area of the canalicular membrane per minute (17).
creased intraluminal dwell time of bile salts (133) and by increased bile salt hydrophobicity (125). Considering that bile salts do not cross membrane bilayers (134), secreted bile salts will interact only with phospholipids in the external hemileaflet. Finally, ultrastructural vesiculation of the canalicular membrane is present in wildtype mice but is absent in mice lacking the mdr2 Pglycoprotein gene product (135). 2— Micellar Extraction of Phospholipids Micellar solubilization of lipids from the hepatocyte canalicular membrane has long been suggested as the mechanism for biliary lipid secretion (126,136). Specifically, luminal bile salts might induce the biliary secretion of lipids by direct solubilization of canalicular membrane lipids into mixed micelles (137). Evidence in favor of this mechanism has been provided by studies in the secretory vesicle mutants of yeast, in which the mdr2 Pglycoprotein has been expressed (138). Phospholipid translocation from the cytoplasmic to the luminal hemileaflet of isolated membrane vesicles is accomplished in an ATPdependent fashion, in keeping with the presumed function of the mdr2 Pglycoprotein. Importantly, the presence of the bile salt taurocholate within the lumen enhances mdr2 Pglycoproteindependent translocation of phospholipid (139). As these isolated vesicles are no larger than 100 nm in diameter (P. Gros, personal communication, 1996), it is highly unlikely that daughter vesicles could bud within the small interior space. Similar observations have been obtained with canalicular membrane vesicles isolated from rat liver (140); again, the formation of daughter vesicles is highly unlikely. Rather, these data are interpreted as consistent with bile salt solubilization of phospholipid from the luminal hemileaflet of the membrane vesicles in the form of mixed micelles. It is possible that both vesicular and micellar mechanisms of biliary lipid secretion may be operative simultaneously in vivo (120,124), as their operation ultimately will be dictated by the physical properties of the canalicular membrane and the component lipid molecules in the membrane and in the biliary space. It remains unclear whether micellization can achieve the specific extraction of biliarytype phospholipids and whether it is sufficiently rapid to explain the high rates of phospholipid secretion. The phase behavior of biliary lipids at the
Page 78
low concentrations of bile salts present within the yeast secretory vesicles (about 55 to 63 M) dictates that vesicles, not micelles, will form under the experimental conditions described (61,141). Moreover, the micellar secretion hypothesis is based on a single technique employing a fluorescent NBDphosphatidylcholine analogue (139,140). The vesicular secretion paradigm is supported not only by ultrastructural studies (73,135) but also by extensive biophysical studies of freshly secreted bile, involving not only study of phosphatidylcholine secretion (61,142–144) but also of cholesterol secretion (145–147). 3— Uncoupling of Phospholipid Secretion Regardless of whether a vesicular or micellar mechanism is operative for biliary phospholipid secretion (or whether both are involved), an interesting phenomenon of lipid "uncoupling" from bile salt secretion can occur. Specifically, certain organic anions are able to inhibit the secretion of phospholipids and cholesterol dose dependently without influencing the secretion of bile salts (130). Only relatively hydrophilic organic anions induce uncoupling, and this effect is exerted only after their secretion into the bile canaliculus (148). Thus, relatively hydrophilic organic anions—including ampicillin, bilirubin ditaurate, and sulfated taurolithocholate—inhibit biliary phospholipid and cholesterol secretion; more hydrophobic organic anions such as indocyanine green and rose bengal do not. Interestingly, these physiological effects correlate well with the high affinity of the hydrophilic organic anions for bile salt micelles (130,148). An attractive explanation for biliary lipid "uncoupling" is the proposal that organic anions secreted into the biliary space bind directly to luminal bile salts, thereby preventing their micellization of canalicular membrane lipids by impairing the detergent activity of bile salts (149). This hypothesis was examined by Verkade et al. (150) utilizing an in vitro system. Release of entrapped fluorescent carboxyfluorescein from small unilamellar vesicles composed of cholesterol/phosphatidylcholine was examined as a function of medium bile salt and organic anion composition. Bile saltinduced release of fluorescence was reduced by certain organic anions. However, the inhibitory effects did not correlate with the in vivo uncoupling effects of these organic anions, irrespective of the bile salt species used or the lipid composition of the vesicles. Ultracentrifugation and dynamic lightscattering studies indicated that the organic anions did interact with mixed bile salt/phosphatidylcholine/cholesterol micelles but did not directly inhibit micellization. Although these experiments do not elucidate the mechanism of the "uncoupling" phenomenon, they do not provide supporting evidence for a micellization mechanism of biliary lipid secretion. 4— Selectivity of Biliary Phospholipid Secretion A question present from the outset of biliary lipid studies is how selectivity for the phosphatidylcholine species present in bile is accomplished. The identification of the mdr2 Pglycoprotein in mice and MDR3 Pglycoprotein in humans has permitted demonstration that this protein shows a high preference for translocation of phosphatidylcholines from the inner to the outer hemileaflet of the canalicular membrane (151). The basis for subselection of fatty acyl species of phosphatidylcholines remains unclear (152). However, in vitro analysis of phosphatidylcholine transfer between vesicles (68) and statistical analysis of in vivo biliary lipid secretion strongly implicate cytoplasmic phosphatidylcholine transfer protein (PCTP) as the basis for this specificity. In particular, the binding affinity of more fluid phosphatidylcholine species for PCTP largely explains the molecular species of phosphatidylcholine secreted into bile (69,153). Although a "subpool" of biliarytype phospholipid has long been postulated (154), current evidence does not require that such a pool should exist. Rather, the action of PCTP and mdr2 Pglycoprotein may be sufficient to deliver the appropriate biliary phosphatidylcholines to the external hemileaflet of the canalicular membrane. The final basis for secretion of specific phosphatidylcholine species into bile remains unclear. If biliary phosphatidylcholines diffused widely within the external hemileaflet, then only a micellar basis of bile saltinduced extraction might impart specificity. One must then
Page 79
postulate that the hydrophobicity of luminal bile salts dictates which phospholipid species are extracted (155). However, if transient microdomains of biliarytype phosphatidylcholines developed in the external hemileaflet in the immediate vicinity of mdr2 Pglycoproteins, then either vesiculation or bile saltinduced micellar extraction would explain the specificity of the final step in phospholipid secretion. C— Cholesterol Secretion into Bile The literature on biliary cholesterol secretion is extensive and spans over three decades (156–158), with numerous contributions in recent years (82,159). Several fundamental concepts have emerged, which are covered in greater detail elsewhere in this volume. First, a more hydrophobic bile salt pool promotes cholesterol secretion into bile (158,160), presumably on the basis of enhanced solubilization of phospholipid/cholesterol mixed lipid structures within bile. Second, cholesterol secretion occurs most efficiently when cosecreted with phospholipid, but it may also occur in the absence of phospholipid secretion, based on experiments with mdr2 deficient mice (161). In the latter instance, the presence of very hydrophobic bile salts in the canalicular lumen appears to be sufficient for extraction of cholesterol from the canalicular membrane in the form of bile salt:cholesterol mixed micelles. Third, the predominant source of cholesterol secreted into bile is plasma; cholesterol newly synthesized within the hepatocyte does not contribute significantly to biliary secretion (80,162,163). Several key lines of investigation now provide insights into how biliary cholesterol secretion may occur. The first pertains to the basolateral plasma membrane HDL scavenger receptor. When HDL particles encounter the liver, there is marked dyssynchrony in the hepatocellular internalization of HDL constituents, with selective uptake of free cholesterol and cholesterol esters (9,164–166). This appears to occur without endocytosis or degradation of the lipoprotein particle. Recently, a cell surface receptor was identified that was capable of mediating hepatocellular HDL binding. The candidate is from the family of scavenger receptor proteins that show broad ligand specificities. Such proteins are capable of binding a wide variety of negatively charged molecules, such as anionic phospholipids, native and chemically modified LDL, and even whole cells expressing aberrant negatively charged ligands. To date, three classes of scavenger receptors have been identified (A, B, and C), which are widely distributed among tissues (167). Cloning of the murine homologue of the class B scavenger receptor type I (mSRBI) complementary cDNA has been achieved. Expression of mSRBI in Chinese hamster ovary cells (CHO cells) confers the ability to bind HDL with high affinity and saturability, with subsequent transfer of free cholesterol from HDL to the cells (168). Overexpression of mSRBI in transgenic mice results in an 85% reduction in plasma cholesterol levels and a doubling of biliary cholesterol output, without alteration in bile salt or phospholipid secretion rates (169). Protein expression is maximal in the liver. Notably, HDL particles are not endocytosed directly but rather appear to bind reversibly to the mSRBI scavenger receptor. These findings strongly suggest that the HDL scavenger receptor can play a major role in hepatic uptake of cholesterol from HDL (170,171). This interpretation is corroborated by the finding in humans that dramatic reductions in circulating plasma LDL and VLDL cholesterol levels do not affect secretion rates of biliary cholesterol (79). An unsolved issue is the actual mechanism for transfer of cholesterol from HDL to the hepatocyte. One hypothesis is that the SRBI receptor brings the HDL particle into close proximity to the hepatocellular plasma membrane, thus promoting exchange of free cholesterol over this short distance. Recent evidence suggests that proximity is not sufficient to effect transfer; SRBI directly facilitates transfer of lipid through interactions with its extracellular domain (172). This occurs not only for free cholesterol but also for the considerable amount of cholesteryl ester contained within the HDL particle (173). In the latter instance, hydrolysis of cholesteryl esters (and glycerolipids) is necessary to facilitate remodeling of the nascent HDL particle and release of free cholesterol (174). It is therefore notable that hepatic lipase is found on both the sinusoidal endothelium
Page 80
and the microvilli of the hepatocellular basement membrane (175–177), exactly where it would be required by this proposed mechanism. Once deposited in the basolateral plasma membrane (Fig. 3, step 3d), free cholesterol may diffuse laterally along the bilayer. Diffusion in the exoplasmic hemileaflet is restricted by the pericanalicular tight junctions (33), but diffusion in the endoplasmic hemileaflet is not. Thus, cholesterol may enter into the canalicular domain via the endoplasmic hemileaflet and distribute between both hemileaflets. The distribution of cholesterol between the inner and outer hemileaflets of the canalicular membrane is not known; it is possible (95) but not proven (10) that cholesterol will partition preferentially into the outer hemileaflet. Cholesterol entry into the canalicular membrane via aqueous exchange and lateral diffusion may then contribute substantively to cholesterol secretion into bile (163,178–180), particularly for HDL free cholesterol (181–183). These concepts have been supported by recent physiological evidence, using nonmetabolizable plant steroids as a model for biliary sterol secretion. Isolated rat livers were perfused with VLDL, LDL, and HDL obtained from patients with hereditary phytosterolemia, which leads to markedly elevated levels of circulating plant sterols. Biliary secretion of plant sterols was minimal during perfusion with VLDL or LDL, whereas with perfusion of HDL there was a marked increase in biliary sterol secretion (184). The increase in biliary secretion occurred over 5 to 10 min, which is faster than that expected for conventional receptormediated endocytosis, lysosomal degradation, and release of free cholesterol from within the cell. The mechanism for cholesterol entry into bile has not yet been elucidated. It has long been postulated that phospholipid and cholesterol enter bile simultaneously from the canalicular membrane (126), despite different routes of delivery to the canalicular membrane (reviewed in Ref. 34). The concept of microdomain formation is predicated on the existence of cholesterolpoor fluid patches of phosphatidylcholine in the canalicular membrane. Two mechanisms are possible for transfer of cholesterol from the surrounding canalicular membrane to newly formed phospholipid vesicles (Fig. 6): (a) lateral diffusion within the exoplasmic hemileaflet into the nascent vesicle while it is still attached and (b) transfer to free vesicles within the canalicular lumen via aqueous diffusion. The latter process is well documented in model systems and is extremely rapid when the donor membrane is supersaturated with cholesterol (185) and when bile salts are present (186). That lateral diffusion might be occurring is suggested by the intriguing observations in our ultrastructural work that (a) cryofixed vesicles were not circular in cross section but rather ellipsoidal and (b) the long axis of adherent vesicles was perpendicular to the canalicular membrane (73). It is possible that lateral diffusion of cholesterol into the external hemileaflet of a nascent vesicle from the surrounding canalicular membrane would "stiffen" the sides and decrease the lateral curvature; the apex of the forming vesicle and the base would remain relatively free of cholesterol. Upon release of the vesicle into the lumen, cholesterol would redistribute, producing the circular vesicular profiles observed by chemical fixation. The critical issue of cholesterol entry into bile remains unresolved (187) yet is central to the longstanding concept that hepatic secretion of cholesterolsupersaturated bile contributes to the pathogenesis of gallstones (126). The low biliary output of intrinsic canalicular membrane proteins that occurs under physiologic conditions (188,189) may also be due to lateral diffusion of proteins into nascent vesicles. V— Consequences of Impaired Biliary Lipid Secretion A— Cholestasis Cholestasis is defined as the systemic retention of biliary constituents as a result of failure of bile formation (reviewed in Ref. 35). Any substantive obstruction to bile flow through the biliary tree results in systemic retention of biliary constituents. Extrahepatic causes include gallstone obstruction of the biliary tree, pancreatic cancer, and bile duct stricture following cholecystectomy. Intrinsic bile duct diseases include primary biliary cirrhosis, primary scleros
Page 81
Figure 6 Schematic of possible mechanisms for cholesterol secretion into bile. Cholesterol may exchange freely with intraluminal phospholipid vesicles by binding to aqueous bile salts within the lumen (step 1a). Alternatively, cholesterol may diffuse laterally from cholesterolrich regions of the canalicular membrane into nascent vesicles during their formation (step 1b).
ing cholangitis, and biliary atresia. In all of these instances, it is logical to presume that hepatocellular excretion of biliary lipid is impaired on the basis of obstruction to bile flow. However, the inflammatory milieu induced by intra or extrahepatic obstruction has a profound effect on hepatocellular transport function, over and above simple fluid blockage. Specifically, conditions as mundane as shortterm obstruction of the common bile duct induce entry of potent proinflammatory substances, especially gutderived bacterial lipopolysaccharide, into the portal circulation. Lipopolysaccharide in particular, and the proinflammatory cytokines produced by an inflamed biliary tree, are potent downregulators of hepatocyte transport function (190). Thus, obstructive cholestasis features not only a reduction or cessation in bile flow but also intrinsic downregulation of hepatocyte bile secretion. The most prominent effect is an almost complete cessation of basolateral bile salt uptake by the hepatocyte, mediated by NTCP (191). The canalicular membrane transporters listed in Table 4 are affected variably in obstructive or inflammatory cholestasis (192). It is hypothesized that downregulation of NTCP function serves to protect hepatocytes from uptake and retention of potentially toxic bile salts while also substantially shutting down bile formation (191). Nonobstructive cholestasis, in contrast, appears to involve hepatocellular dysfunction, which cannot necessarily be viewed as protective. In most instances, socalled hepatocellular cholestasis is not the result of massive hepatocellular destruction. Rather, hepatocellular cholestasis is frequently the outcome of relatively innocuous insults in which hepatic mass is maintained. Hepatocellular cholestasis can arise from a host of hepatic insults, as reviewed elsewhere (35). Inflammationinduced cholestasis is exceedingly common, especially virus or druginduced hepatitis, and results from intrinsic failure of living hepatocytes to secrete bile (190). Inflammation emanating from inflamed sinusoids, inflamed portal tracts, or parenchymal foci of inflammation and hepatocyte destruction has a profound effect on adjacent living hepatocytes. Hepatocytes are essentially hijacked into producing acutephase reactants while losing the ability to secrete bile. The final common pathway of reduced NTCP function, reduced
Page 82
bile salt uptake and near cessation of bile salt secretion across the canalicular membrane, is much the same as in obstructive cholestasis. While it is difficult to argue that systemic retention of bile salts in the absence of obstruction is protective, reduced hepatocellular uptake of bile salts may nevertheless reduce total destruction of hepatocytes and thereby permit recovery once the cholestatic agent is removed. 1— Bile Salt Retention Experimentally, intravenous infusion of high doses of bile salts leads to hepatocellular cholestasis, presumably the result of breakdown in the integrity of the canalicular membrane (193,194). However, such pure bile salt "toxicity" is unusual clinically, as the systemic consequences of cholestasis are the logical outcome of the properties of the molecules retained. Within the peripheral blood, bile salt concentrations increase from their normal levels of 5 M or less to concentrations exceeding 300 M (195,196). Although bile salts are watersoluble, as amphiphilic compounds they nevertheless may partition into body tissues such as the subcutis. A prominent feature of severe bile salt retention is pruritus, an intense itching sensation of the skin. In some patients with chronic cholestasis, as from primary biliary cirrhosis, the pruritus may be so severe as to generate suicidal thoughts. However, it is not clear that the pruritus is simply the outcome of dermal irritation. There is extensive evidence that pruritus arises from central nervous system activation of the opioid signaling network (197). Bile salts do not cross the bloodbrain barrier, and transport proteins are in place to export into the cerebrospinal fluid those bile salts that do enter the brain substance (Peter MeierAbt, personal communication, 1998). Thus, the mechanisms causing the pruritus of cholestasis remain unclear. 2— Phospholipid Retention It is reasonable to posit that there are no detrimental consequences of systemic retention of phosphatidylcholine. This phospholipid is a normal constituent of hepatocytes, and mice defective in the canalicular mdr2 Pglycoprotein exhibit no ultrastructural abnormalities of their bile canaliculi (James Crawford, personal observation, 1997). 3— Cholesterol Retention In contrast, systemic retention of cholesterol cannot be considered innocuous, despite the fact that it, too, is a normal constituent of mammalian membranes. Marked elevations of circulating cholesterol bound to lipoproteins lead to sequestration of cholesterol in virtually every body tissue. A prominent physical sign of cholesterol retention is xanthomas—subcutaneous collections of macrophages filled with cholesteryl esters. Curiously, atherosclerosis is not a prominent feature of chronic cholestatic diseases, suggesting that as yet poorly understood mechanisms are in place for minimizing the detrimental effects of cholesterol retention on vascular structure. Shedding of keratinocytes and enterocytes from the skin and gut, respectively, are woefully inadequate as mechanisms for cholesterol loss. Fecal loss of secreted bile salts (about 2.5 g/day) and of free cholesterol (about 3 g/day) constitutes the only effective mechanism for maintaining cholesterol homeostasis. Attempts to downregulate cholesterol biosynthesis or enhance cholesterol bioconversion to bile salts do not satisfactorily prevent marked elevations in systemic cholesterol levels. These futile efforts underscore the central role of bile formation in cholesterol homeostasis. One may speculate that the development of other hepatic complications, such as the hepatic encephalopathy of endstage primary biliary cirrhosis, precludes full expression of the systemic consequences of longterm cholesterol retention. B— Inherited Diseases of Bile Secretory Function Elucidation of inherited defects in canalicular transporter function has been one of the most exciting areas of hepatic biology in recent years. Excellent reviews are available elsewhere
Page 83
(198,199) and are addressed elsewhere in this volume. Mention must be made of the inherited defect in MDR3, responsible for phosphatidylcholine secretion into bile. In the original report of the mdr2 (/) knockout mice, a histological cholangitis developed over several months of life, followed eventually by the development of hepatocellular carcinomas (119). Dietary feeding of the hydrophilic bile acid ursodeoxycholic acid greatly reduced the hydrophobicity of the ambient bile acid pool and led to marked improvement in liver histology and animal longevity (200). It is postulated that the inability of bile salts to extract phosphatidylcholine from the hepatocellular canalicular membrane produces excessive concentrations of free bile salts that are not bound up in mixed micelles with phospholipid. This, in turn, leads to detergentinduced toxicity of the downstream biliary tree and hence a chemical cholangitis (124). Human patients with an autosomal recessive inherited defect in MDR3 also develop severe cholestatic liver disease, with portal tractbased inflammation, fibrosis, and bile duct damage, which strikingly resembles the disease of the murine mdr2deficient mice (201,202). This disease, now designated progressive familial intrahepatic cholestasis type 3 (PFIC3), features elevated serum levels of gamma glutathione transpeptidase, an enzyme of the canalicular plasma membrane and apical membranes of the bile duct cholangiocytes. Thus, regardless of mechanism, in both mouse and human the continued secretion of bile salts without phospholipid into bile leads to a relentless cholangiodestructive disease. At the very least, this points out the fact that, outside of the hepatocyte canalicular membrane itself, there is no other plasma membrane in the body that can maintain its integrity in the face of mM concentrations of detergent bile salts. VI— Future Directions A— Unresolved Issues in Proposed Phospholipid Vesicle Formation Important questions remain in understanding biliary lipid secretion by hepatocytes. For the vesicular secretion paradigm, these are as follows: 1— Microdomains The formation of lipid microdomains within lamellar membranes is an important general phenomenon in biology (203–205). It is unknown whether transient cholesterolpoor microdomains of biliary phospholipid actually exist in the exoplasmic hemileaflet of the canalicular membrane. There is both thermodynamic and kinetic evidence that fluid microdomains of cholesterolpoor phospholipid can coexist with less fluid cholesterolrich membrane domains, particularly when the cholesterolrich region contains sphingomyelin, a lipid for which cholesterol exhibits a particular affinity (206–210). Importantly, the preferential interaction of cholesterol with sphingomyelin is retained even when phosphatidylcholine enters into the monolayer (211). Lateral diffusion of phospholipid into the cholesterolrich membrane regions is restricted and exceedingly slow (212), leading to relative immiscibility of cholesterolrich and cholesterolpoor microdomains (213). The action of a canalicular membrane phospholipid translocator (118) might promote the creation of transient focal domains of biliarytype phosphatidylcholine in the exoplasmic hemileaflet, the diameter of which is likely to fall between 50 and 100 nm on the basis of in vitro model studies (213). To date, conventional techniques such as freeze fracture (214) impart too long a fixation time to permit capture of transient microdomains. Ultrarapid cryofixation followed by freezefracture has not been successful in identifying microdomains (Lukas Landmann, personal communication). The ability to document such events will be an important test of the vesicular secretion hypothesis.
Page 84
2— Vesicle Formation How might vesicles form from the exoplasmic hemileaflet? Monomeric bile salts are readily capable of partitioning into membranes and interact preferentially with more fluid lipid domains, such as those composed of biliarytype phosphatidylcholines (61). Since bile salts are several hundredfold more concentrated within the canalicular lumen than within the cell, the exoplasmic hemileaflet of the canalicular membrane will be the primary site into which bile salts partition (215). Membrane "solubilizing" bile salts remain exclusively in the exoplasmic hemileaflet of the membrane, as transmembrane "flipflop" of fully ionized bile salts does not occur (134,216). Asymmetrical accumulation of phosphatidylcholine and of bile salt molecules will generate a localized increase of the lateral pressure gradient in the exoplasmic leaflet of the membrane. Focal ''bulging" may also be accentuated by interactions of sphingomyelin molecules across the lipid bilayer, imparting shear resistance to the surrounding sphingomyelinrich canalicular membrane (217). Bile saltmediated instability of the phospholipid bilayer structure (68,218) may promote restructuring of the hemileaflet microdomain, whereby phospholipid head groups reorient into a newly formed unilamellar vesicle, as occurs in the phosphatidylcholine surface monolayer of circulating lipoproteins (219,220). Such dramatic reorganization of membrane structure, and transient departure from a conventional bilayer structure, can occur without compromising membrane integrity (221). The small quantities of phosphatidylethanolamine present in the external hemileaflet are known to promote lamellartoinverted hexagonalphase transitions in monolayers (222). Inverted hexagonal phases of phosphatidylcholine are predicted by the phase diagram for bile formation (223). The possibility of inverted hexagonalphase microdomains within the external hemileaflet of the canalicular membrane is an intriguing component of the vesicular secretion paradigm (159). One part of the hypothesis for cholesterol entry into nascent vesicles described earlier is the lateral diffusion of cholesterol into fluid phosphatidylcholine domains. It is therefore interesting to note that small amounts of cholesterol in a phospholipid layer can actually "soften" the lipid layer rather than making it more rigid (224). Moreover, the presence of bile salts within the canalicular lumen will also reduce the affinity of cholesterol for sphingomyelin, further increasing the likelihood of cholesterol release into bile (225). How might water and solutes enter the interior of nascent vesicles? Water permeability through pure lipid bilayers is considerable and increases as membrane fluidity increases and cholesterol content decreases (226). In addition, water permeability increases at the boundaries between different lipid phases in membranes (227,228). Bile salts can induce marked increases (up to 500 Da) in both water and solute permeability through cholesterolpoor membranes without solubilization of the bilayers (229,230). We postulate that water enters nascent vesicles through the bile saltperturbed fluid microdomains of cholesterolpoor buds of biliary phosphatidylcholines. As newly released vesicles would undergo almost immediate osmotic collapse in the absence of interior solutes (231), luminal solutes (such as the inorganic salts normally present in bile) also may be swept into the interior of forming vesicles. Does canalicular motility contribute to vesicle formation? Regular contractions of bile canaliculi have been documented both in vitro and in vivo (232,233) and are mediated by the pericanalicular cytoskeleton of actin and myosin II. Membrane deformations destabilize membrane structure (234), and it is possible that contractile motions of the canalicular membrane further promote the formation of biliary vesicles. What are the alterations in vesicles following their secretion into bile? Vesicle dissolution appears to be a normal, dynamic process occurring during the brief residence time of lipid vesicles within the bile canalicular lumen. Bile saltinduced vesicle dissolution in vitro results in progressive diminution in the size of membrane vesicles as lipid is progressively dissolved (61,235). This phenomenon has also been observed in quasielastic lightscattering measurements of living hepatocyte couplets, whereby increasing intraluminal bile salt concentrations at first promote the elaboration of lipid vesicles but then induce dissolution of the same vesicles
Page 85
within the canalicular lumen (104). Dynamic alterations in vesicular size and shape have been noted in human biles (236). 3— Canalicular Phospholipid "Flippases" Low rates of phospholipid secretion may be observed transiently even in mice lacking the mdr2 gene (135), and gene products other than the mdr2 locus are capable of conferring phospholipid translocation across membranes (237–240). Identification of all the proteins involved is necessary to gain a full understanding of biliary phospholipid secretion (240). B— Lipoprotein X Obstructive cholestasis leads to the appearance of an abnormal lipoprotein in plasma, designated lipoprotein X, or LpX (241). In contrast to normal plasma lipoproteins, LpX is a unilamellar vesicle with an aqueous lumen (242). To test the hypothesis that LpX originates as a biliary vesicle that is regurgitated in bile, Oude Elferink and colleagues examined the elaboration of LpX in mice deficient for mdr2 (243). In wildtype mice, bile duct ligation led to a dramatic increase in plasma cholesterol and phospholipid concentrations, contained largely as LpX particles 40 to 100 nm in diameter. There was a complete absence of LpX in mdr2 (/) mice. Transgenic insertion of the MDR3 human homologue of mdr2 restored the presence of LpX during bile duct ligation. Besides demonstrating that the plasma concentration of this protein during obstructive cholestasis is determined by the activity of the canalicular phospholipid translocator, these findings raise the intriguing possibility that biliary lipid may be regurgitated into plasma in a vesicular form not unlike that elaborated in bile. It remains to be seen whether such regurgitation is transcellular (via reverse transcytosis of cargo vesicles) or paracellular (across disrupted tight junctions). It is also unclear whether LpX lipid vesicles might form de novo at the basolateral membrane. The extracellular concentrations of bile salts in plasma are 100fold less than those in the lumen of the bile canaliculus, and it is the presumed action of detergent bile salts that completes the canalicular vesiculation process. C— Cholesterol Delivery to Bile By far the most critical issue is elucidation of the exact mechanisms by which cholesterol is delivered to bile and how such delivery is regulated. In humans, the lack of correlation between circulating levels of plasma cholesterol and biliary cholesterol secretion rates has frustrated efforts to link atherosclerosis and cholesterol gallstone disease into a coherent concept of pathogenesis (244). Nevertheless, recent progress in elucidating the genetic underpinnings of cholesterol gallstone disease (see Chapter 6) are providing great promise for gaining an coherent understanding of the genetic basis for these two diseases (245,246). Moreover, characterization of hepatocellular uptake mechanisms, especially by the SRBI scavenger receptor, may provide a direct causal link between the pathobiology of atherosclerosis and the regulation of hepatic cholesterol secretion into bile (247). VII— Conclusion The rapid progress in the genetic, molecular, and structural basis of hepatocellular biliary lipid secretion has enabled a more comprehensive understanding of this complex event (248). While there are many details to verify or disprove, it is reasonable to state that "the source of the bile" has been identified.
Page 86
Acknowledgments This work was supported by National Institutes of Health grant R01DK53512. The graphic artwork of Aleta R. Crawford is gratefully acknowledged. References 1. ER Weibel, W Staubli, HR Gnagi, FA Hess. Correlated morphometric and biochemical studies on the liver cell: I. Morphometric model, stereological methods, and normal morphometric data for rat liver. J Cell Biol 42:68–91, 1969. 2. B Hornstein, L Stammler, L Bianchi, L Landmann. Ethinylestradiol increases volume and decreases sinusoidal membrane surface in the rat liver: a stereological analysis. Hepatology 16:217–223, 1992. 3. JD Vial, FR Simon, AM Mackinnon. Effect of bile duct ligation on the ultrastructural morphology of hepatocytes. Gastroenterology 70:85–92, 1976. 4. R Saxena, ND Theise, JM Crawford. Microanatomy of the liver: exploring the hidden interfaces. Hepatology 30:1339–1346, 1999. 5. ND Theise, R Saxena, BC Portmann, SN Thung, H Yee, L Chiriboga, A Kumar, JM Crawford. The canals of Hering and hepatic stem cells in humans. Hepatology 30:1425–1433, 1999. 6. W Ekataksin, K Wake. New concepts in biliary and vascular anatomy of the liver. Prog Liv Dis 15:1–30, 1997. 7. KN Lazaridis, L Pham, P Tietz, PA Marinelli, PC DeGroen, S Levine, PA Dawson, NF LaRusso. Rat cholangiocytes absorb bile acids at their apical domain via the ileal sodiumdependent bile acid transporter. J Clin Invest 100:2714–2721, 1997. 8. D Schachter. The hepatocyte plasma membrane: organization and differentiation. In: Arias IM, Boyer JL, Fausto N, Jakoby WB, Schachter DA, Shafritz DA, eds. The Liver: Biology and Pathobiology, 3rd ed. New York: Raven Press, 1994, pp 145–154. 9. CJ Fielding, PE Fielding. Molecular physiology of reverse cholesterol transport. J Lipid Res 36:211–228, 1995. 10. L Liscum, KW Underwood. Intracellular cholesterol transport and compartmentation. J Biol Chem 270:15443–15446, 1995. 11. F Schroeder, JR Jefferson, AB Kier, J Knittel, TJ Scallen, WG Wood, I Hapala. Membrane cholesterol dynamics: cholesterol domains and kinetic pools. Proc Soc Exp Biol Med 196:235–252, 1991. 12. A Van Helvoort, G Van Meer. Intracellular lipid heterogeneity caused by topology of synthesis and specificity in transport. Example: Sphingolipids. FEBS Lett 369:18–21, 1995. 13. P Verkade, K Simons. Lipid microdomains and membrane trafficking in mammalian cells. Histochem Cell Biol 108:211–220, 1997. 14. G Ihrke, GV Martin, MR Shanks, M Schrader, TA Schroer, AL Hubbard. Apical plasma membrane proteins and endolyn78 travel through a subapical compartment in polarized WIFB hepatocytes. J Cell Biol 141:115–133, 1998. 15. BF Scharschmidt, JR Lake, EL Renner, V Licko, RW Van Dyke. Fluid phase endocytosis by cultured rat hepatocytes and perfused rat liver: implications for plasma membrane turnover and vesicular trafficking of fluid phase markers. Proc Natl Acad Sci USA 83: 9488–9492, 1986. 16. JR Lake, V Licko, RW Van Dyke, BF Scharschmidt. Biliary secretion of fluidphase markers by the isolated perfused rat liver: role of transcellular vesicular transport. J Clin Invest 76:676–689, 1985. 17. JM Crawford. The role of vesiclemediated transport pathways in hepatocellular bile secretion. Semin Liver Dis 16:169–190, 1996.
Page 87
18. RPJ Oude Elferink, CTM Bakker, H Roelofsen, E Middelkoop, R Ottenhoff, M Helin, PLM Jansen. Accumulation of organic anion in intracellular vesicles of cultured rat hepatocytes is mediated by the canalicular multispecific organic anion transporter. Hepatology 17:434–444, 1993. 19. LJ Scott, AL Hubbard. Dynamics of four rat liver plasma membrane proteins and polymeric IgA receptor: rates of synthesis and selective loss into the bile. J Biol Chem 267: 6099–6106, 1992. 20. D Jeckel, A Karrenbauer, R Birk, RR Schmidt, F Wieland. Sphingomyelin is synthesized in the cis Golgi. FEBS Lett 261:155–157, 1990. 21. AH Futerman, B Stieger, AL Hubbard, RE Pagano. Sphingomyelin synthesis in rat liver occurs predominantly at the cis and medial cisternae of the Golgi apparatus. J Biol Chem 265:8650–8657, 1990. 22. PJ Trotter, DR Voelker. Lipid transport processes in eukaryotic cells. Biochim Biophys Acta 1213:241–262, 1994. 23. JC Mclntyre, RG Sleight. Mechanisms of intracellular membrane lipid transport. Curr Topics Mem 40:453–481, 1994. 24. M Koval, RE Pagano. Lipid recycling between the plasma membrane and intracellular compartments: transport and metabolism of fluorescent sphingomyelin analogues in cultured fibroblasts. J Cell Biol 108:2169–2181, 1989. 25. NG Lipsky, RE Pagano. Intracellular translocation of fluorescent sphingolipids in cultured fibroblasts: endogenously synthesized sphingomyelin and glucocerebroside analogues pass through the Golgi apparatus en route to the plasma membrane. J Cell Biol 100:27–34, 1985. 26. G Van Meer, W Van't Hof. Epithelial sphingolipid sorting is insensitive to reorganization of the Golgi by nocodazole, but is abolished by monensin in MDCK cells and by brefeldin A in Caco2 cells. J Cell Sci 104:833–842, 1993. 27. YJ Shiao, JE Vance. Sphingomyelin transport to the cell surface occurs independently of protein secretion in rat hepatocytes. J Biol Chem 268:26085–26092, 1993. 28. G Ihrke, EB Neufeld, T Meads, MR Shanks, D Cassio, M Laurent, T Schroer, RE Pagano, AL Hubbard. WIFB cells: an in vitro model for studies of hepatocyte polarity. J Cell Biol 123:1761–1775, 1993. 29. MMP Zegers, KJM Zaal, D Hoekstra. Functional involvement of proteins, interacting with sphingolipids, in sphingolipid transport to the canalicular membrane in the human hepatocytic cell line, HepG2. Hepatology 27:1089–1097, 1998. 30. HJ Verkade, RJ Vonk, F Kuipers. New insights into the mechanism of bile acidinduced biliary lipid secretion. Hepatology 21:1174–1189, 1995. 31. GM Patton, JM Fasulo, SJ Robins. Origin of bile lecithins: evidence for extensive remodeling and a novel pathway of synthesis. Gastroenterology 104:A970, 1993. 32. GM Patton, JM Fasulo, SJ Robins. Hepatic phosphatidylcholines: evidence for synthesis in the rat by extensive reutilization of endogenous acylglycerides. J Lipid Res 35:1211–1221, 1994. 33. G Van Meer, K Simons. The function of tight junctions in maintaining differences in lipid composition between the apical and the basolateral cell surface domains of MDCK cells. EMBO J 5:1455–1464, 1986. 34. MC Carey, JT LaMont. Cholesterol gallstone formation: 1. Physicalchemistry of bile and biliary lipid secretion. Prog Liv Dis 10:139–163, 1992. 35. RM Green, JM Crawford. Hepatocellular cholestasis: pathobiology and histological outcome. Semin Liver Dis 15:372–389, 1995. 36. MC Carey, WC Duane. Enterohepatic circulation. In: Arias IM, Boyer JL, Fausto N, Jakoby WB, Schachter D, Shafritz DA, eds. The Liver: Biology and Pathobiology, 3rd ed. New York: Raven Press, 1994, pp 719–767. 37. FJ Suchy. Hepatocellular transport of bile acids. Semin Liver Dis 13:235–247, 1993. 38. MH Nathanson, JL Boyer. Mechanisms and regulation of bile secretion. Hepatology 14:551–566, 1991.
Page 88
39. PJ Meier. The bile secretory polarity of hepatocytes. J Hepatol 9:124–129, 1989. 40. JL Boyer, B Hagenbuch, M Ananthanarayanan, FJ Suchy, B Stieger, PJ Meier. Phylogenic and ontogenic expression of hepatocellular bile acid transport. Proc Natl Acad Sci USA 90:435–438, 1993. 41. RW Van Dyke, JE Stephens, BF Scharschmidt. Bile acid transport in cultured hepatocytes. Am J Physiol 243:G484–G492, 1982. 42. A Stolz, H Takikawa, M Ookhtens, N Kaplowitz. The role of cytoplasmic proteins in hepatic bile acid transport. Annu Rev Physiol 51:161–176, 1989. 43. A Stolz, L Hammond, H Lou, H Takikawa, M Ronk, JE Shively. cDNA cloning and expression of the human hepatic bile acidbinding protein: a member of the monomeric reductase gene family. J Biol Chem 268:10448–10457, 1993. 44. H Lou, L Hammond, V Sharma, RS Sparkes, AJ Lusis, A Stolz. Genomic organization and chromosomal localization of a novel human hepatic dihydrodiol dehydrogenase with high affinity bile acid binding. J Biol Chem 269:8416–8422, 1994. 45. H Takikawa, A Stolz, N Kaplowitz. Cyclical oxidationreduction of the C3 position on bile acids catalyzed by rat hepatic 3ahydroxysteroid dehydrogenase: I. Studies with the purified enzyme, isolated rat hepatocytes, and inhibition by indomethacin. J Clin Invest 80:852–860, 1987. 46. H Takikawa, JC FernandezCheca, J Kuhlenkamp, A Stolz, M Ookhtens, N Kaplowitz. Effect of indomethacin on the uptake, metabolism and excretion of 3 oxocholic acid: studies in isolated hepatocytes and perfused rat liver. Biochim Biophys Acta 1084:247–250, 1991. 47. H Takikawa, Y Sugiyama, JC FernandezCheca, J Kuhlenkamp, M Ookhtens, N Kaplowitz. Evidence that interference with binding to hepatic cytosol binders can inhibit bile acid excretion in rats. Hepatology 23:1642–1649, 1996. 48. FA Simion, B Fleischer, S Fleischer. Subcellular distribution of bile acids, bile salts and taurocholate binding sites in rat liver. Biochemistry 23:6459–6466, 1984. 49. FA Simion, B Fleischer, S Fleischer. Two distinct mechanisms for taurocholate uptake in subcellular fractions from rat liver. J Biol Chem 250:10814–10822, 1984. 50. MA Goldsmith, S Huling, AL Jones. Hepatic handling of bile salts and protein in the rat during intrahepatic cholestasis. Gastroenterology 84:978–986, 1983. 51. FJ Suchy, WF Balistreri, J Hung, P Miller; SA Garfield. Intracellular bile acid transport in rat liver as visualized by electron microscope autoradiography using a bile acid analogue. Am J Physiol 245:G681–G689, 1983. 52. Y Lamri, A Roda, M Dumont, G Feldmann, S Erlinger. Immunoperoxidase localization of bile salts in rat liver cells: evidence for a role of the Golgi apparatus in bile salt transport. J Clin Invest 82:1173–1182, 1988. 53. JM Crawford, S Barnes, RC Stearns, CL Hastings, JJ Godleski. Ultastructural localization of a fluorinated bile salt in hepatocytes. Lab Invest 71:42–51, 1994. 54. C Alves, P Von Dippe, M Amoui, D Levy. Bile acid transport into hepatocyte smooth endoplasmic reticulum vesicles is mediated by microsomal epoxide hydrolase, a membrane protein exhibiting two distinct topological orientations. J Biol Chem 268:20148–20155, 1993. 55. S Ruetz, AE Lindsey, CL Ward, RR Kopito. Functional activation of plasma membrane anion exchangers occurs in a preGolgi compartment. J Cell Biol 121:37–48, 1993. 56. JM Crawford, CA Berken, JL Gollan. Role of the hepatocyte microtubular system in the excretion of bile salts and biliary lipid: implications for intracellular vesicular transport. J Lipid Res 29:144–156, 1988. 57. BA Luxon, RA Weisiger. A new method for quantitating intracellular transport: application to the thyroid hormone 3,5,3'triiodothyronine. Am J Physiol 263:G733–G741, 1992. 58. K LubyPhelps. Physical properties of cytoplasm. Curr Opin Cell Biol 6:3–9, 1994. 59. RA Weisiger. Cytoplasmic transport of lipids: role of binding proteins. Comp Biochem Physiol [B] 115B:319–331, 1996.
Page 89
60. DW Hay, MC Carey. Chemical species of lipids in bile. Hepatology 12(suppl):6S–16S, 1990. 61. DE Cohen, M Angelico, MC Carey. Structural alterations in lecithincholesterol vesicles following interactions with monomeric and micellar bile salts: physical chemical basis for subselection of biliary lecithin species and aggregative states of biliary lipids during bile formation. J Lipid Res 31:55–70, 1990. 62. JA Higgins, WH Evans. Transverse organization of phospholipids across the bilayer of plasmamembrane subfractions of rat hepatocytes. Biochem J 174:563– 567, 1978. 63. I Portal, T Clerc, V Sbarra, H Portugal, AM Pauli, H Lafont, B Tuchweber, I Yousef, F Chanussot. Importance of highdensity lipoproteinphosphatidylcholine in secretion of phospholipid and cholesterol in bile. Am J Physiol 264:G1052–G1056, 1993. 64. HJ Verkade, JTP Derksen, A Gerding, GL Scherphof, RJ Vonk, F Kuipers. Differential hepatic processing and biliary secretion of headgroup and acyl chains of liposomal phosphatidylcholines. Biochem J 275:139–144, 1991. 65. SJ Robins, H Brunengraber. Origin of biliary cholesterol and lecithin in the rat: contribution of new synthesis and preformed hepatic stores. J Lipid Res 23:604– 608, 1982. 66. SJ Robins, JM Fasulo, VF Robins, GM Patton. Utilization of different fatty acids for hepatic and biliary phosphatidylcholine formation and the effect of changes in phosphatidylcholine molecular species on biliary lipid secretion. J Lipid Res 32:985–992, 1991. 67. GM Patton, JM Fasulo, SJ Robins. Evidence that hepatic triglycerides provide acylglycerides for synthesis of bile phosphatidylcholines. Am J Physiol 267:G1028–G1034, 1994. 68. DE Cohen, MR Leonard, MC Carey. In vitro evidence that phospholipid secretion into bile may be coordinated intracellularly by the combined actions of bile salts and the specific phosphatidylcholine transfer protein of liver. Biochemistry 33:9975–9980, 1994. 69. KWA Wirtz. Phospholipid transfer proteins. Annu Rev Biochem 60:73–99, 1991. 70. AN Leonard, DE Cohen. In vitro evidence that sterol carrier protein 2 may provide a scavenger pathway for phosphatidylcholine secretion into bile. Hepatology 22:295A, 1995. 71. Vlahcevic ZR, Hylemon PB, Chiang JYL. Hepatic cholesterol metabolism. In: Arias IM, Boyer JL, Fausto N, Jakoby WB, Schachter DA, Shafritz DA, eds. The Liver: Biology and Pathobiology. New York: Raven Press, 1994, pp 379–389. 72. MC Bourgès, DM Small, DG Dervichian. Biophysics of lipid associations: III. The quaternary systems lecithinbile saltcholesterolwater. Biochim Biophys Acta 144:189–201, 1967. 73. JM Crawford, GM Möckel, AR Crawford, SJ Hagen, VC Hatch, S Barnes, JJ Godleski, MC Carey. Imaging biliary lipid secretion in the rat: ultrastructural evidence for vesiculation of the hepatocyte canalicular membrane. J Lipid Res 36:2147–2163, 1995. 74. JT LaMont, MC Carey. Cholesterol gallstone formation: 2. Pathobiology and pathomechanics. Prog Liv Dis 10:165–191, 1992. 75. RG Sleight. Intracellular lipid transport in eukaryotes. Annu Rev Physiol 49:193–208, 1987. 76. J Rosario, E Sutherland, L Zaccaro, FR Simon. Ethinylestradiol administration selectively alters liver sinusoidal membrane lipid fluidity and protein composition. Biochemistry 27:3939–3946, 1988. 77. PD Thomas, MJ Poznansky. Cholesterol transfer between lipid vesicles: effect of phospholipids and gangliosides. Biochem J 251:55–61, 1988. 78. MB Sankaram, TE Thompson. Interaction of cholesterol with various glycerophospholipids and sphingomyelin. Biochemistry 29:10670–10675, 1990. 79. CG Hillebrant, B Nyberg, K Einarsson, M Eriksson. The effect of plasma low density lipoprotein apheresis on the hepatic secretion of biliary lipids in humans. Gut 41:700–704, 1997.
Page 90
80. J Scheibner, M Fuchs, E Hörmann, G Tauber, EF Stange. Biliary cholesterol secretion and bile acid formation in the hamster: the role of newly synthesized cholesterol. J Lipid Res 35:690–697, 1994. 81. K Empen, K Lange, EF Stange, J Scheibner. Newly synthesized cholesterol in human bile and plasma: quantitation by mass isotopomer distribution analysis. Am J Physiol 272:G367–G373, 1997. 82. RHJ Bandsma, F Stellaard, RJ Vonk, GT Nagel, RA Neese, MK Hellerstein, F Kuipers. Contribution of newly synthesized cholesterol to rat plasma and bile determined by mass isotopomer distribution analysis: bilesalt flux promotes secretion of newly synthesized cholesterol into bile. Biochem J 329:699–703, 1998. 83. E Schnitzer, D Lichtenberg. Reevaluation of the structure of low density lipoproteins. Chem Phys Lipids 70:63–74, 1994. 84. Glickman RM, Sabesin SM. Lipoprotein metabolism. In: Arias IM, Boyer JL, Fausto N, Jakoby WB, Schachter DA, Shafritz DA, eds. The Liver: Biology and Pathobiology. New York: Raven Press, 1994, pp 391–414. 85. DR Voelker. Lipid transport pathways in mammalian cells. Experientia 46:569–579, 1990. 86. MP Reinhart. Intracellular sterol trafficking. Experientia 46:599–611, 1990. 87. L Puglielli, A Rigotti, AV Greco, MJ Santos, F Nervi. Sterol carrier protein2 is involved in cholesterol transfer from the endoplasmic reticulum to the plasma membrane in human fibroblasts. J Biol Chem 270:18723–18726, 1995. 88. L Puglielli, A Rigotti, L Amigo, L Nunez, M Santos, AV Greco, F Nervi. Evidence for the involvement of sterol carrier protein (SCP2) in a rapid, microtubule, and Golgiindependent transport of newlysynthesized cholesterol to bile. Gastroenterology 108: A1150, 1995. 89. MT Vanier, P Pentchev, C RodriguezLafrasse, R Rousson. NiemannPick disease type C: an update. J Inherit Metab Dis 14:580–595, 1991. 90. L Puglielli, A Rigotti, L Amigo, L Nufiez, AV Greco, MJ Santos, F Nervi. Modulation of intrahepatic cholesterol trafficking: Evidence by in vivo antisense treatment for the involvement of sterol carrier protein2 in newly synthesized cholesterol transport into rat bile. Biochem J 317:681–687, 1996. 91. Y Lange, TL Steck. Cholesterolrich intracellular membranes: a precursor to the plasma membrane. J Biol Chem 260:15592–15597, 1985. 92. T Babia, JW Kok, M Van der Haar, R Kalicharan, D Hoekstra. Transport of biosynthetic sphingolipids from Golgi to plasma membrane in HT29 cells: involvement of different carrier vesicle populations. Eur J Cell Biol 63:172–181, 1994. 93. W Van't Hof, G Van Meer. Lipid polarity and sorting in epithelial cells. Curr Topics Mem 40:539–563, 1994. 94. R Bittman, CR Kasireddy, P Mattjus, JP Slotte. Interaction of cholesterol with sphingomyelin in monolayers and vesicles. Biochemistry 33:11776–11781, 1994. 95. D Allan, K Kallen. Transport of lipids to the plasma membrane in animal cells. Prog Lipid Res 32:195–219, 1993. 96. VN Schumaker, ML Phillips, JE Chatterton. Apolipoprotein B and lowdensity lipoprotein structure: implications for biosynthesis of triglyceriderich lipoproteins. Adv Prot Chem 45:205–248, 1994. 97. J Sines, R Rothnagel, M Van Heel, JW Gaubatz, JD Morrisett, W Chiu. Electron cryomicroscopy and digital image processing of lipoprotein(a). Chem Phys Lipids 67–68: 81–89, 1994. 98. R Van Antwerpen, JC Gilkey. Cryoelectron microscopy reveals human low density lipoprotein substructure. J Lipid Res 35:2223–2231, 1994. 99. L Urbani, RD Simoni. Cholesterol and vesicular stomatitis virus G protein take separate routes from the endoplasmic reticulum to the plasma membrane. J Biol Chem 265:1919–1923, 1990.
Page 91
100. Y Lange, F Strebel, TL Steck. Role of the plasma membrane in cholesterol esterification in rat hepatoma cells. J Biol Chem 268:13838–13843, 1993. 101. M Koval, AH Futerman, RE Pagano. Sphingomyelin synthesis in endosomal compartments. Trends Cell Biol 5:148–149, 1995. 102. ID Roman, A Thewles, R Coleman. Fractionation of livers following diosgenin treatment to elevate biliary cholesterol. Biochim Biophys Acta Lipids Lipid Metab 1255:77–81, 1995. 103. HA Ahmed, RP Jazrawi, PM Goggin, J Dormandy, TC Northfield. Intrahepatic biliary cholesterol and phospholipid transport in humans: effect of obesity and cholesterol cholelithiasis. J Lipid Res 36:2562–2573, 1995. 104. G Möckel, S Gorti, RK Tandon, R Tanaka, MC Carey. Microscope laser light scattering spectroscopy of vesicles within canaliculi of rat hepatocyte couplets. Am J Physiol 269:G73–G84, 1995. 105. T Gerloff, B Stieger, B Hagenbuch, J Madon, L Landmann, J Roth, AF Hofmann, PJ Meier. The sister of Pglycoprotein represents the canalicular bile salt export pump of mammalian liven. J Biol Chem 273:10046–10050, 1998. 106. B Angelin, I Bjorkhem, E Einarsson, S Ewerth. Hepatic uptake of bile acids in man: fasting and postprandial concentrations of individual bile acids in portal venous and systemic blood serum. J Clin Invest 70:724–731, 1982. 107. RD Adler, FJ Wannagat, RK Ockner. Bile secretion in selective biliary obstruction: adaptation of taurocholate transport maximum to increased secretory load in the rat. Gastroenterology 73:129–136, 1977. 108. JB Watkins, CD Klaassen. Effect of repeated oral administration of taurocholate on hepatic excretory function in rat. J Pharmacol Exp Ther 218:182–187, 1981. 109. FR Simon, EM Sutherland, M Gonzalez. Regulation of bile salt transport in rat liver: evidence that increased maximum bile salt secretory capacity is due to increased cholic acid receptors. J Clin Invest 70:401–411, 1982. 110. J Graf, M Rupnik, G Zupancic, R Zorec. Osmotic swelling of hepatocytes increases membrane conductance but not membrane capacitance. Biophys J 68:1359–1363, 1995. 111. H Roelofsen, CTM Bakker, B Schoemaker, M Heijn, PLM Jansen, RPJ Oude Elferink. Redistribution of canalicular organic anion transport activity in isolated and cultured rat hepatocytes. Hepatology 21:1649–1657, 1995. 112. D Häussinger, F Schliess. Cell volume and hepatocellular function. J Hepatol 22:94–100, 1995. 113. M Arrese, M Pizarro, N Solis, L Accatino. Adaptive regulation of hepatic bile salt transport: role of bile salt hydrophobicity and microtubuledependent vesicular pathway. J Hepatol 26:694–702, 1997. 114. H Roelofsen, CJ Soroka, D Keppler, JL Boyer. Cyclic AMP stimulates sorting of the canalicular organic anion transporter (MRP2/cMoat) to the apical domain in hepatocyte couplets. J Cell Sci 111:1137–1145, 1998. 115. JM Crawford, DCJ Strahs, AR Crawford, S Barnes. The role of bile salt hydrophobicity in hepatic microtubuledependent bile salt secretion. J Lipid Res 35:1738–1748, 1994. 116. JD Jones, TE Thompson. Mechanism of spontaneous, concentrationdependent phospholipid transfer between bilayers. Biochemistry 29:1593–1600, 1990. 117. JM Backer, EA Dawidowicz. Reconstitution of a phospholipid flippase from rat liver microsomes. Nature 327:341–343, 1987. 118. F Berr, PJ Meier, B Stieger. Evidence for the presence of a phosphatidylcholine translocator in isolated rat liver canalicular plasma membrane vesicles. J Biol Chem 268: 3976–3979, 1993. 119. JJM Smit, AH Schinkel, RPJ Oude Elferink, AK Groen, E Wagenaar, L Van Deemter, CAAM Mol, R Ottenhoff, NMT Van der Lugt, MA Van Roon, et al. Homozygous disruption of the murine mdr2 pglycoprotein gene leads to a complete absence of phospholipid from bile and to liver disease. Cell 75:451–462, 1993.
Page 92
120. RPJ Oude Elferink, R Ottenhoff, M van Wijland, JJM Smit, AH Schinkel. Regulation of biliary lipid secretion by mdr2 Pglycoprotein in the mouse. J Clin Invest 95:31–38, 1995. 121. RPJ Oude Elferink, R Ottenhoff, M van Wijland, CMG Frijters, C van Nieuwkerk, AK Groen. Uncoupling of biliary phospholipid and cholesterol secretion in mice with reduced expression of mdr2 Pglycoprotein. J Lipid Res 37:1065–1075, 1996. 122. M Fuchs, MC Carey, DE Cohen. Functional reconstitution of a liver specific longchain phosphatidylcholine transmembrane translocator. Gastroenterology 108:A1067, 1995. 123. GA KullakUblick, PJ Meier, B Hagenbuch, F Berr, B Steiger. Expression einer kanalikulären phosphatidylcholinflippase der rattenleber in Xenopus laevis oozyten (abstr). German Association for the Study of the Liver, 2:52, 1995. 124. RPJ Oude Elferink, GNJ Tytgat, AK Groen. The role of mdr2 Pglycoprotein in hepatobiliary lipid transport. FASEB J 11:19–28, 1997. 125. R Coleman, K Rahman. Lipid flow in bile formation. Biochim Biophys Acta 1125:113–133, 1992. 126. DM Small. The formation of gallstones. Adv Intern Med 16:243–264, 1970. 127. CF Higgins. To flip or not to flip? Curr Biol 4:259–260, 1994. 128. DJ Cabral, DM Small. Physical chemistry of bile. In: Schultz SG, Forte JG, Rauner BB, eds. Handbook of Physiology, Section 6: The Gastrointestinal System. Vol. 3: Salivary, Gastric, Pancreatic, and Hepatobiliary System. Bethesda, MD: American Physiological Society, 1989, pp 621–622. 129. MD Apstein, AR Russo. Ampicillin inhibits biliary cholesterol secretion. Dig Dis Sci 30:253–256, 1985. 130. HJ Verkade, MJ Wolbers, R Havinga, DRA Uges, RJ Vonk, F Kuipers. The uncoupling of biliary lipid from bile acid secretion by organic anions in the rat. Gastroenterology 99:1485–1492, 1990. 131. F Kuipers, JPT Derksen, A Gerding, GL Scherphof, RJ Vonk. Biliary lipid secretion in the rat: the uncoupling of biliary cholesterol and phospholipid secretion from bile acid secretion by sulfated glycolithocholic acid. Biochim Biophys Acta 922:136–144, 1987. 132. MJ Monte, RA Parslow, R Coleman. Inhibitory action of cyclobutyrol on the secretion of biliary cholesterol and phospholipids. Biochem J 266:165–171, 1990. 133. HJ Verkade, H Wolters, A Gerding, R Havinga, V Fidler, RJ Vonk, F Kuipers. Mechanism of biliary lipid secretion in the rat: a role for bile acidindependent bile flow. Hepatology 17:1074–1080, 1993. 134. F Kamp, JA Hamilton. Movement of fatty acids, fatty acid analogues, and bile acids across phospholipid bilayers. Biochemistry 32:11074–11086, 1993. 135. AR Crawford, AJ Smith, VC Hatch, RPJ Oude Elferink, P Borst, JM Crawford. Hepatic secretion of phospholipid vesicles in the mouse critically depends on mdr2 or MDR3 Pglycoprotein expression. J Clin Invest 100:2562–2567, 1997. 136. R Coleman. Biochemistry of bile secretion. Biochem J 244:249–261, 1987. 137. IM Yousef, MM Fisher. Can J Biochem 54:1040–1056, 1976. 138. S Ruetz, P Gros. Phosphatidylcholine translocase: A physiological role for the mdr2 gene. Cell 77:1071–1081, 1994. 139. S Ruetz, P Gros. Enhancement of mdr2mediated phosphatidylcholine translocation by the bile salt taurocholate: implications for hepatic bile formation. J Biol Chem 270:25388–25395, 1995. 140. AT Nies, Z Gatmaitan, IM Arias. ATPdependent phosphatidylcholine translocation in rat liver canalicular plasma membrane vesicles. J Lipid Res 37:1125– 1136, 1996. 141. SU Egelhaaf, P Schurtenberger. Shape transformations in the lecithinbile salt system: from cylinders to vesicles. J Phys Chem 98:8560–8573, 1994. 142. DE Cohen, M Angelico, MC Carey. Quasielastic light scattering evidence for vesicular secretion of biliary lipids. Am J Physiol 257:G1–G8, 1989.
Page 93
143. DE Cohen, MR Fisch, MC Carey. Principles of laser lightscattering spectroscopy: applications to the physiochemical study of model and native biles. Hepatology 12(suppl): 113S–122S, 1990. 144. DE Cohen, MC Carey. Physical chemistry of biliary lipids during bile formation. Hepatology 12(suppl):1435–1485, 1990. 145. GJ Somjen, Y Merikovsky, P Lelkes, T Gilat. Cholesterolphospholipid vesicles in human bile: an ultrastructural study. Biochim Biophys Acta 879:14–21, 1986. 146. GJ Somjen, T. Gilat. A nonmicellar mode of cholesterol transport in human bile. FEBS Lett 156:265–268, 1983. 147. GJ Somjen, T Gilat. Contribution of vesicular and micellar carriers to cholesterol transport in human bile. J Lipid Res 26:699–704, 1985. 148. HJ Verkade, R Havinga, A Gerding, RJ Vonk, F Kuipers. Mechanism of bile acidinduced biliary lipid secretion in the rat: effect of conjugated bilirubin. Am J Physiol 264:G462–G469, 1993. 149. G Yamashita, S Tazuma, G Kajiyama. Effects of organic anions on biliary lipid secretion in rats. Importance of association with biliary lipid structures. Biochem J 286:193–196, 1992. 150. HJ Verkade, MAC De Bruijn, MA Brink, H Talsma, RJ Vonk, F Kuipers, AK Groen. Interactions between organic anions, micelles and vesicles in model bile systems. Biochem J 320:917–923, 1996. 151. AJ Smith, JLPM TimmermansHereijgers, B Roelofsen, KWA Wirtz, WJ van Blitterswijk, JJM Smit, AH Schinkel, P Borst. The human MDR3 Pglycoprotein promotes translocation of phosphatidylcholine through the plasma membrane of fibroblasts from transgenic mice. FEBS Lett 354:263–266, 1994. 152. RD Shamburek, CC Schwartz. Selective composition of biliary phosphatidylcholines is affected by secretion rate but not by bile acid hydrophobicity. J Lipid Res 34:1833–1842, 1993. 153. WW LaMorte, ML Mooker, S Kay. Determinants of the selection of phosphatidylcholine molecular species for secretion into bile in the rat. Hepatology 28:631–637, 1998. 154. T Kawamoto, T Akino, M Nakamura, M Mori. Metabolism of individual molecular species of phosphatidylcholine in the liver subcellular membranes and bile. Biochim Biophys Acta 619:35–47, 1980. 155. H Miyake, S Tazuma, G Kajiyama. Bile salt hydrophobicity modulates subselection of biliary lecithin species in rats depleted of bile salt pool. Dig Dis Sci 43:921–926, 1998. 156. HO Wheeler, KK King. Biliary excretion of lecithin and cholesterol in the dog. J Clin Invest 51:1337–1350, 1972. 157. NF LaRusso, NE Hoffman, AF Hofmann, TC Northfield, JL Thistle. Effect of primary bile acid ingestion on bile acid metabolism and biliary lipid secretion in gallstone patients. Gastroenterology 69:1301–1314, 1975. 158. K Einarsson, SM Grundy. Effects of feeding cholic acid and chenodeoxycholic acid on cholesterol absorption and hepatic secretion of biliary lipids in man. J Lipid Res 21:23–34, 1980. 159. CMG Frijters, AK Groen, RPJO Elferink. MDR2 Pglycoproteinmediated lipid secretion and its relevance to biliary drug transport. Adv Drug Deliv Rev 25:201–215, 1997. 160. LE Bilhartz, JM Dietschy. Bile salt hydrophobicity influences cholesterol recruitment from rat liver in vivo when cholesterol synthesis and uptake are constant. Gastroenterology 95:771–779, 1988. 161. RPJ Oude Elferink, R Ottenhoff, M van Wijland, CMG Frijters, C van Nieuwkerk, AK Groen. Uncoupling of biliary phospholipid and cholesterol secretion in mice with reduced expression of mdr2 Pglycoprotein. J Lipid Res 37:1065–1075, 1996. 162. SJ Robins, JM Fasulo, PD Lessard, GM Patton. Hepatic cholesterol synthesis and the secretion of newly synthesized cholesterol in bile. Biochem J 289:41–44, 1993.
Page 94
163. SJ Robins, JM Fasulo, MA Collins, GM Patton. Evidence for separate pathways of transport of newly synthesized and preformed cholesterol into bile. J Biol Chem 260:6511–6513, 1985. 164. E Bravo, KM Botham, MA Mindham, PA Mayes, T Marinelli, A Cantafora. Evaluation in vivo of the differential uptake and processing of highdensity lipoprotein unesterified cholesterol and cholesteryl ester in the rat. Biochim Biophys Acta Lipids Lipid Metab 1215:93–102, 1994. 165. S Jäckle, F Rinninger, T Lorenzen, H Greten, E Windler. Dissection of compartments in rat hepatocytes involved in the intracellular trafficking of highdensity lipoprotein particles or their selectively internalized cholesteryl esters. Hepatology 17:455–465, 1993. 166. MW Scobey, FL Johnson, LL Rudel. Delivery of highdensity lipoprotein free and esterified cholesterol to bile by the perfused monkey liver. Am J Physiol 257:G644–G652, 1989. 167. AK Hatzopoulos, A Rigotti, RD Rosenberg, M Krieger. Temporal and spatial pattern of expression of the HDL receptor SRBI during murine embryogenesis. J Lipid Res 39:495–508, 1998. 168. S Acton, A Rigotti, KT Landschulz, S Xu, HH Hobbs, M Krieger. Identification of scavenger receptor SRBI as a High Density Lipoprotein receptor. Science 271:518–520, 1996. 169. KF Kozarsky, MH Donahee, A Rigotti, SN Iqbal, ER Edelman, M Krieger. Overexpression of the HDL receptor SRB1 alters plasma HDL and bile cholesterol levels. Nature 387:414–417, 1997. 170. M Arrese, JM Crawford. Of plaques and stones: the SRBI scavenger receptor. Hepatology 26:1072–1074, 1997. 171. M Krieger. The ''best" of cholesterols, the "worst" of cholesterols: a tale of two receptors. Proc Natl Acad Sci USA 95:4077–4080, 1998. 172. X Gu, B Trigatti, S Xu, S Acton, J Babitt, M Krieger. The efficient cellular uptake of high density lipoprotein lipids via scavenger receptor class B type I requires not only receptormediated surface binding but also receptorspecific lipid transfer mediated by its extracellular domain. J Biol Chem 273:26338–26348, 1998. 173. K Fluiter, DR Van der Westhuijzen, TJC Van Berkel. In vivo regulation of scavenger receptor BI and the selective uptake of high density lipoprotein cholesteryl esters in rat liver parenchymal and Kupffer cells. J Biol Chem 273:8434–8438, 1998. 174. P MarquesVidal, C Azéma, X Collet, H Chap, BP Perret. Hepatic lipasemediated hydrolysis versus liver uptake of HDL phospholipids and triacylglycerols by the perfused rat liver. Biochim Biophys Acta 1082:185–194, 1991. 175. F Burgaya, J Peinado, M Llobera, I Ramirez. Hepatic endothelial lipase activity in neonatal rat liver. Biosci Rep 9:559–564, 1989. 176. B Breedveld, K Schoonderwoerd, AJM Verhoeven, R Willemsen, H Jansen. Hepatic lipase is localized at the parenchymal cell microvilli in rat liver. Biochem J 321:425–430, 1997. 177. DA Sanan, J Fan, A Bensadoun, JM Taylor. Hepatic lipase is abundant on both hepatocyte and endothelial cell surfaces in the liver. J Lipid Res 38:1002–1013, 1997. 178. SJ Robins, JM Fasulo, CR Pritzker, GM Patton. Hepatic transport of unesterified cholesterol in the rat is traced by the plant sterol, sitostanol. Hepatology 22:240A, 1995. 179. CC Schwartz, LG Halloran, ZR Vlahcevic, DH Gregory, L Swell. High and low density lipoprotein metabolism in man: preferential utilization of HDL free cholesterol by the liver for biliary cholesterol secretion. Science 200:62–64, 1978. 180. CC Schwartz, M Berman, ZR Vlahcevic, LG Halloran, DH Gregory, L Swell. Multicompartment analysis of cholesterol metabolism in man: characterization of the hepatic bile acid and biliary cholesterol precursor sites. J Clin Invest 61:408–423, 1978. 181. SJ Robins, JM Fasulo, G Salen, GM Patton. Preferential sterol transport from HDL to bile. Hepatology 22:240A, 1995.
Page 95
182. E Bravo, A Cantafora, L Argiolas. Hepatic uptake and metabolism of free cholesterol from different lipoprotein classes: an in vivo study in the rat. Biochim Biophys Acta 1003:315–320, 1989. 183. E Brava, A Cantafora. Hepatic uptake and processing of free cholesterol from different lipoproteins with and without sodium taurocholate administration: an in vivo study in the rat. Biochim Biophys Acta 1045:74–80, 1990. 184. SJ Robins, JM Fasulo. High density lipoproteins, but not other lipoproteins, provide a vehicle for sterol transport to bile. J Clin Invest 99:380–384, 1997. 185. TL Steck, FJ Kezdy, Y Lange. An activationcollision mechanism for cholesterol transfer between membranes. J Biol Chem 263:13023–13031, 1988. 186. ZR Vlahcevic, EC Gurley, DM Heuman, PB Hylemon. Bile salts in submicellar concentrations promote bidirectional cholesterol transfer (exchange) as a function of their hydrophobicity. J Lipid Res 31:1063–1071, 1990. 187. F Berr, HC Jaeger, T Bitterle, J Mössner. Evidence for secretory coupling of phosphatidylcholine molecular species to cholesterol in rat bile. J Hepatol 26:1069–1078, 1997. 188. SG Barnwell, PJ Lowe, R Coleman. The effects of colchicine on secretion into bile of bile salts, phospholipids, cholesterol and plasma membrane enzymes: bile salts are secreted unaccompanied by phospholipids and cholesterol. Biochem J 220:723–731, 1984. 189. A Rigotti, L Núñez, L Amigo, L Puglielli, J Garrido, M Santos, S González, G Mingrone, A Greco, F Nervi. Biliary lipid secretion: immunolocalization and identification of a protein associated with lamellar cholesterol carriers in supersaturated rat and human bile. J Lipid Res 34:1883–1894, 1993. 190. JM Crawford. Cellular and molecular biology of the inflamed liver. Curr Opin Gastroenterol 13:175–185, 1997. 191. M Trauner, M Arrese, H Lee, JL Boyer, SJ Karpen. Endotoxin downregulates rat hepatic ntcp gene expression via decreased activity of critical transcription factors. J Clin Invest 101:2092–2100, 1998. 192. M Trauner, M Arrese, CJ Soroka, M Ananthanarayanan, TA Koeppel, SF Schlosser, FJ Suchy, D Keppler, JL Boyer. The rat canalicular conjugate export pump (Mrp2) is downregulated in intrahepatic and obstructive cholestasis. Gastroenterology 113:255–264, 1997. 193. IM Yousef, SG Barnwell, F Gratton, B Tuchweber, A Weber, CC Roy. Liver cell membrane solubilization may control maximum secretory rate of cholic acid in the rat. Am J Physiol 252:G84–G91, 1987. 194. WGM Hardison, DE Hatoff, D Miyai, RG Weiner. Nature of the bile acid maximum secretory rate in the rat. Am J Physiol 241:G337–G343, 1981. 195. C Gartung, M Ananthanarayanan, MA Rahman, S Schuele, S Nundy, CJ Soroka, A Stolz, FJ Suchy, JL Boyer. Downregulation of expression and function of the rat liver Na+/bile acid cotransporter in extrahepatic cholestasis. Gastroenterology 110:199–209, 1996. 196. C Gartung, S Schuele, SF Schlosser, JL Boyer. Expression of the rat liver Na+/taurocholate cotransporter is regulated in vivo by retention of biliary constituents but not their depletion. Hepatology 25:284–290, 1997. 197. NV Bergasa, EA Jones. The pruritus of cholestasis: potential pathogenic and therapeutic implications of opioids. Gastroenterology 108:1582–1588, 1995. 198. M Muller, PLM Jansen. The secretory function of the liver: new aspects of hepatobiliary transport. J Hepatol 28:344–354, 1998. 199. AS Knisely, NB Freimer. Insight into bile duct differentiation takes (notched) wings. Hepatology 27:298–299, 1998. 200. CMJ Van Nieuwkerk, RPJO Elferink, AK Groen, R Ottenhoff, GNJ Tytgat, KP Dingemans, MAVB Weerman, GJA Offerhaus. Effects of ursodeoxycholate and cholate feeding on liver disease in FVB mice with a disrupted mdr2 Pglycoprotein gene. Gastroenterology 111:165–171, 1996.
Page 96
201. JML De Vree, E Jacquemin, E Sturm, D Cresteil, PJ Bosma, J Aten, JF Deleuze, M Desrochers, M Burdelski, O Bernard, et al. Mutations in the MDR3 gene cause progressive familial intrahepatic cholestasis. Proc Natl Acad Sci USA 95:282–287, 1998. 202. JF Deleuze, E Jacquemin, C Dubuisson, D Cresteil, M Dumont, S Erlinger, O Bernard, M Hadchouel. Defect of multidrugresistance 3 gene expression in a subtype of progressive familial intrahepatic cholestasis. Hepatology 23:904–908, 1996. 203. R Welti, M Glaser. Lipid domains in model and biological membranes. Chem Phys Lipids 73:121–137, 1994. 204. OG Mouritsen, K Jorgensen. Dynamical order and disorder in lipid bilayers. Chem Phys Lipids 73:3–25, 1994. 205. NM Hooper. Membrane biology: do glycolipid microdomains really exist? Curr Biol 8: R114–R116, 1998. 206. F Schroeder. Lipid domains in plasma membranes from rat liver. Eur J Biochem 132: 509–516, 1983. 207. PFF Almeida, WLC Vaz, TE Thompson. Percolation and diffusion in threecomponent lipid bilayers: effect of cholesterol on an equimolar mixture of two phosphatidylcholines. Biophys J 64:399–412, 1993. 208. YK Shin, DE Budil, JH Freed. Thermodynamics and dynamics of phosphatidylcholinecholesterol mixed model membranes in the liquid crystalline state: effects of water. Biophys J 65:1283–1294, 1993. 209. PLG Chong, F Liu, MM Wang, K Truong, IP Sugar, RE Brown. Fluorescence evidence for cholesterol regular distribution in phosphatidylcholine and in sphingomyelin lipid bilayers. J Fluoresc 6:221–230, 1996. 210. F Liu, IP Sugar, PLG Chong. Cholesterol and ergosterol superlattices in threecomponent liquid crystalline lipid bilayers as revealed by dehydroergosterol fluorescence. Biophys J 72:2243–2254, 1997. 211. P Mattjus, JP Slotte. Does cholesterol discriminate between sphingomyelin and phosphatidylcholine in mixed monolayers containing both phospholipids. Chem Phys Lipids 81:69–80, 1996. 212. HM McConnell. Equilibration rates in lipid monolayers. Proc Natl Acad Sci USA 93: 15001–15003, 1996. 213. MB Sankaram, TE Thompson. Cholesterolinduced fluidphase immiscibility in membranes. Proc Natl Acad Sci USA 88:8686–8690, 1991. 214. C Rahner, B Stieger, L Landmann. Structurefunction correlation of tight junctional impairment after intrahepatic and extrahepatic cholestasis in rat liver. Gastroenterology 110:1564–1578, 1996. 215. R Schubert, K Beyer, H Wolburg, K Schmidt. Structural changes in membranes of large unilamellar vesicles after binding of sodium cholate. Biochemistry 25:5263–5269, 1986. 216. DJ Cabral, DM Small, HS Lilly, JA Hamilton. Transbilayer movement of bile acids in model membranes. Biochemistry 26:1801–1804, 1987. 217. PFF Almeida, WLC Vaz, TE Thompson. Lateral diffusion and percolation in twophase, twocomponent lipid bilayers: topology of the solidphase domains in plane and across the lipid bilayer. Biochemistry 31:7198–7210, 1992. 218. JW Nichols. Low concentrations of bile salts increase the rate of spontaneous phospholipid transfer between vesicles. Biochemistry 25:4596–4601, 1986. 219. AR Tall, DM Small. Body cholesterol removal: role of plasma highdensity lipoproteins. Adv Lipid Res 17:1–51, 1980. 220. T Handa, H Saito, K Miyajima. Phospholipid monolayers at the trioleinsaline interface: production of microemulsion particles and conversion of monolayers to bilayers. Biochemistry 29:2884–2890, 1990. 221. P Ostapchuk, P Hearing, D Ganem. A dramatic shift in the transmembrane topology of a viral envelope glycoprotein accompanies hepatitis B viral morphogenesis. EMBO J 13:1048–1057, 1994.
Page 97
222. DP Siegel, RM Epand. The mechanism of lamellartoinverted hexagonal phase transitions in phosphatidylethanolamine: Implications for membrane fusion mechanisms. Biophys J 73:3089–3111, 1997. 223. DM Small, MC Bourgès, DG Dervichian. The biophysics of lipidic association: I. The ternary systems lecithinbile saltwater. Biochim Biophys Acta 125:563– 580, 1966. 224. J Lemmich, K Mortensen, JH Ipsen, T Honger, R Bauer, OG Mouritsen. The effect of cholesterol in small amounts on lipid bilayer softness in the region of the main phase transition. Eur Biophys J Biophys Lett 25:293–304, 1997. 225. KJ Van Erpecum, MC Carey. Influence of bile salts on molecular interactions between sphingomyelin and cholesterol: relevance to bile formation and stability. Biochim Biophys Acta Lipids Lipid Metab 1345:269–282, 1997. 226. PL Yeagle. Cholesterol and the cell membrane. Biochim Biophys Acta 822:267–287, 1985. 227. JH Ipsen, OG Mouritsen, M Bloom. Relationships between lipid membrane area, hydrophobic thickness, and acylchain orientational order: the effects of cholesterol. Biophys J 57:405–412, 1990. 228. TX Xiang, BD Anderson. Phase structures of binary lipid bilayers as revealed by permeability of small molecules. Biochim Biophys Acta Bio Membr 1370:64– 76, 1998. 229. P Walde, J Sunamoto, CJ O'Connor. The mechanism of liposomal damage by taurocholate. Biochim Biophys Acta 905:30–38, 1987. 230. CJ O'Connor, RG Wallace, K Iwamoto, T Taguchi, J Sunamoto. Bile salt damage of egg phosphatidylcholine liposomes. Biochim Biophys Acta 817:95–102, 1985. 231. A Carruthers, DL Melchior. Studies of the relationship bertween bilayer water permeability and bilayer physical state. Biochemistry 22:5797–5807, 1983. 232. C Oshio, MJ Phillips. Contractility of bile canaliculi: implications for liver function. Science 212:1041, 1981. 233. N Watanabe, N Tsukada, CR Smith, MJ Phillips. Motility of bile canaliculi in the living animal: implications for bile flow. J Cell Biol 113:1069–1080, 1991. 234. D Needham. Measurement of interbilayer adhesion energies. Methods Enzymol 220: 111–129, 1993. 235. A De la Maza, AM Manich, JL Parra. Intermediate aggregates resulting in the interaction of bile salt with liposomes studied by transmission electron microscopy and light scattering techniques. J Microsc 186:75–83, 1997. 236. A Kaplun, Y Talmon, FM Konikoff, M Rubin, A Eitan, M Tadmor, D Lichtenberg. Direct visualization of lipid aggregates in native human bile by light and cryotransmission electronmicroscopy. FEBS Lett 340:78–82, 1994. 237. M Fuchs, MC Carey, DE Cohen. Evidence for an ATPindependent longchain phosphatidylcholine translocator in hepatocyte membranes. Am J Physiol 273:G1312–1319, 1997. 238. GA KullakUblick, T Gerloff, B Hagenbuch, F Berr, PJ Meier, B Stieger. Expression of a rat liver phosphatidylcholine translocator in Xenopus laevis oocytes. Hepatology 23: 1254–1259, 1996. 239. L Cornacchia, H Domdey, J Mössner, F Berr. Expression of a nonmdr2coded liver phosphatidylcholine membrane transport protein in Xenopus laevis oocytes. Biochem Biophys Res Commun 231:277–282, 1997. 240. DE Cohen, M Fuchs. Transmembrane translocation of phosphatidylcholines and biliary lipid secretion: evidence for "flippases" in the canalicular plasma membrane. Hepatology 24:1546–1548, 1996. 241. D Seidel, P Alaupovic, RH Furman. A lipoprotein characterizing obstructive jaundice: I. Method for quantitative separation and identification of lipoproteins in jaundiced subjects. J Clin Invest 48:1211–1223, 1969. 242. P Laggner, O Glatter, K Muller, O Kratky, G Kostner, A Holasek. The lipid bilayer structure of the abnormal human plasma lipoprotein X: an xray small anglescattering study. Eur J Biochem 77:165–171, 1977.
Page 98
243. RPJ Oude Elferink, R Ottenhoff, J Van Marle, CMG Frijters, AJ Smith, AK Groen. Class III Pglycoproteins mediate the formation of lipoprotein X in the mouse. J Clin Invest 102:1749–1757, 1998. 244. ECR Quintao. Is reverse cholesterol transport a misnomer for suggesting its role in the prevention of atheroma formation. Atherosclerosis 116:1–14, 1995. 245. B Khanuja, YC Cheah, M Hunt, PM Nishina, DQH Wang, HW Chen, JT Billheimer, MC Carey, B Paigen. Lith1, a major gene affecting cholesterol gallstone formation among inbred strains of mice. Proc Natl Acad Sci USA 92:7729–7733, 1995. 246. D Machleder, B Ivandic, C Welch, L Castellani, K Reue, AJ Lusis. Complex genetic control of HDL levels in mice in response to an atherogenic diet— coordinate regulation of HDL levels and bile acid metabolism. J Clin Invest 99:1406–1419, 1997. 247. A Rigotti, BL Trigatti, M Penman, H Rayburn, J Herz, M Krieger. A targeted mutation in the murine gene encoding the high density lipoprotein (HDL) scavenger receptor class B type I reveals its key role in HDL metabolism. Proc Natl Acad Sci USA 94: 12620–12615, 1997. 248. DM Heuman. The source of the bile: The legacy of Mathias Möckel. Hepatology 24: 274–276, 1996.
Page 99
5— Bile Ductal Secretion and Its Regulation Won Kyoo Cho Indiana University School of Medicine and The Richard L. Roudebush VA Medical Center, Indianapolis, Indiana I— Introduction As ancient Babylonian and Greeks thought, bile secretion serves a very important role in our wellbeing. Bile secretion assists fat digestion by forming micelles with bile acids, neutralizes gastric acid with bicarbonate secretion, and facilitates removal of toxins or waste materials. The role of bile duct epithelium in bile formation has been recognized since 1920s (1). Early physiological studies suggested the importance of the bile duct epithelium in the modification of bile via secretion and absorption of ions, water, bile acids, carbohydrates, and proteins. In fact, cholangiocytes contribute about 10% of daily bile output in the rat and about 40% in the human, although they represent only 2 to 5% of total liver cells (2,3). In spite of rigorous attempts to isolate and study the cholangiocytes in the past, the relatively small numbers of cholangiocytes in the liver and their tight associations with hepatic parenchymal cells had made their isolation a difficult task. However, recent advances in isolation methods have rejuvenated the research efforts to understand the physiology and biology of bile ducts. This chapter focuses on these recent developments in this exciting research area. II— Anatomy Greater than 2 km of bile ducts and ductules in the human liver deliver bile from hepatocyte to the second portion of the duodenum at the papilla of Vater (4). Bile ducts are lined with cholangiocytes resting on a basement membrane and sealed with tight junctions. Bile ducts receive their blood supply from the peribiliary vascular plexus arising from the hepatic artery as well as from portal blood and lymph vessels for gas and nutrients (5). The biliary system is categorized into extrahepatic and intrahepatic bile ducts. The extrahepatic bile ducts consist of the right and left hepatic ducts, which join to form the common hepatic duct. The intrahepatic bile ducts are divided into 12 generations of bile duct branches with decreasing diameter from the hilum. of the liver toward canals of Hering in the periphery. Commonly, these bile duct branches are named according to the size of their diameters (Table 1). About six definable segments with 12 branches are recognized based on the diameters of the segments. Recent studies indicate that larger bile ducts are lined by larger, columnar cholangiocytes with basally displaced nuclei and a lower nucleustocytoplasm ratio, while smaller ducts are lined by less tall columnar or cuboidal cells with a higher nucleustocytoplasm ratio (6,7). The cholangioles or ductules are the smallest intrahepatic ducts, measuring less than 15 m in diameter, and are lined by 3 to 4 cuboidal cholangiocytes (6). The canals of Hering,
Page 100 Table 1 Terminology of Intrahepatic Bile Ducts Type of ducts
Generation of branching Approximate diameter ( m)
Left and right hepatic duct
First generation
Segmental duct
Second generation
Area duct
>800 400–800
Third generation
300–400
Septal duct
Interlobular duct
15–100
Mediumsized
40–100
Smallsized
15–40
Ductules (cholangioles)
>100
Twelfth generation
<15
the smallest extensions of the intrahepatic biliary tree, are lined by both hepatic parenchymal cells and cholangiocytes with an intact basement membrane. They establish the connections between the hepatic parenchyma and the biliary tree. III— Functions of the Biliary System Until recently, bile ducts were considered merely passive conduits delivering bile produced by hepatocytes to the duodenum. However, it has become more apparent that bile ducts play many other active roles in hepatic biology and pathophysiology. In addition to their active involvement in bile secretion and absorption of various ions and biochemicals, as further discussed in this chapter, they also participate actively in immune functions as well as proliferation/differentiation during growth and liver regeneration (8,9). They secrete peptides and mediators such as IgA immunoglobulins (2), interleukins (10), transforming growth factor beta (TGF b ) (11), monocyte chemotactic protein1 (MCP1) (12), and endothelin1 (13), which may play important roles in intercellular communication, proliferation, inflammation, and immunomodulation. Furthermore, bile ducts are increasingly being recognized as major sites of diverse pathological processes in inflammatory, immunological, infectious, toxic, ischemic, and neoplastic liver diseases as well as in congenital and acquired "vanishing bile duct" disorders (9). Covering such diverse areas of rapidly expanding research in biliary physiology is beyond the scope of this chapter. Thus, this chapter focuses on the functions of the biliary system in bile secretion and modification. IV— Experimental Models of Biliary Secretion Physiological in vivo animal models using dog, pig, and rat have been widely available to study bile secretion. Although these study models have contributed greatly to our understanding of bile secretion and hepatic physiology, the complexities of bile secretion involving both hepatocytes and bile ducts and possible influences from other organs or neurohumoral effects have limited their usefulness. Important ex vivo models to study bile ductal secretion are isolated perfused rat liver (IPRL) with and without bile duct ligation. By using these experimental models, studies in the 1960s showed that secretin stimulated bicarbonaterich choleresis from the bile ducts, and that the increase in was associated with a reciprocal decrease in Cl in bile. More recently, the development of in vitro models—such as isolated bile duct cells, isolated bile duct units, and bile duct cell monolayers as well as longterm cultures—have provided better tools to study biliary secretion.
Page 101
A— Isolated Perfused Rat Liver Although many in vivo and ex vivo animal models have been used to study bile secretion, IPRL models have been the best characterized and most widely utilized since their use was first reported by Claude Bernard over 100 years ago (14). Other animals—such as cat, dog, guinea pig, pig, and monkey—have also been used for isolated perfused liver studies, but the rat is most commonly used, not only because of well established techniques but also because of its easy availability, convenient size, and low cost (14). Moreover, IPRL permits repeated sampling of the perfusate and bile, easy administration of drugs or hormones, and alteration of temperature or perfusate. It also allows experiments to be conducted independent of neurohumoral effects or influence from other organ systems. Therefore, this model has been used extensively to study hepatic metabolism, synthesis, and extraction as well as bile secretion, drug toxicity, and lobular heterogeneity (14). In bile secretion studies, IPRL has been an invaluable tool to examine various aspects of the bile secretory process and its regulation, but IPRL has some limitations for studying bile duct secretion. The bile collected from the IPRL represents the final products after hepatic secretions and bile ductular modifications; thus it is not possible to examine each component of bile secretion selectively. In rat livers, since cholangiocytes contribute only ~10% of bile output, alterations in secretion from bile duct level are not readily detectable. To overcome this limitation, bile duct ligation or naphthylisothiocyanate feeding has been employed to induce selective bile duct proliferations to augment the bile duct secretory component of bile secretion. This bile ductproliferated rat liver model has been used successfully to demonstrate that secretin, bombesin, and VIP stimulate bile duct secretion (15–17). B— Isolated Bile Duct Cells Isolation of cholangiocytes has been a difficult task, since they represent only 2 to 5% of total liver cells and their size and density are similar to those of other nonparenchymal and inflammatory cells in the liver (2,3). Detailed discussions of the recent advances in cholangiocyte isolation are presented elsewhere (18). Initially, a large number of relatively pure cholangiocytes were isolated from animals with bile duct proliferation resulting from bile duct ligation or naphthylisothiocyanate treatment. These isolation techniques involved differential gradient centrifugation and counterflow elutriation techniques (3,19). Although these cholangiocytes were isolated from rat livers with bile duct proliferation, subsequent studies showed that they had cellular and functional characteristics similar to those from normal livers (3,19). Further improvements in isolation techniques—such as isopycnic and Percoll gradients, immunoabsorption (20), and immunomagnetic isolation using bile duct—specific antibodies (21)—allowed the isolation of pure cholangiocytes from normal rat livers with purity as high as >95% and cell yields up to 1 million cholangiocytes per normal rat liver (21). These cell preparations were extensively characterized as cholangiocytes by morphology, using light and electron microscopy, GT histochemistry, or immunocytochemical detection of intermediate filament proteins such as cytokeratins 7 and 19 or other specific antigenic determinants such as HEA 125 for human cholangiocytes (3,20,22). Currently, primary cholangiocytes have been isolated from rat, mouse, pig, and human livers. Furthermore, these isolation techniques have also been used to isolate cholangiocytes not only from normal human livers (22) but also from diseased livers of patients with primary biliary cirrhosis (23). Although most of these isolation and characterization methods can be applied to other animal species, some important species differences need to be considered. For example, mouse cholangiocytes do not constitutively express GT (24), while in guinea pig liver, GT is expressed in cholangiocytes as well as in sinusoidal endothelial cells (25). The isolation of pure cholangiocytes has contributed greatly to the study of biliary physiology and secretion. Intracellular pH (pHi) studies using fluorescent dyes such as BCECF, radioisotope efflux studies, and electrophysiological studies using patchclamping techniques
Page 102
have been used to characterize ion transport mechanisms and their regulation (19,26–28). Measurements of secondary messenger levels have permitted the examination of signal transduction pathways involved in the regulation of bile duct secretion by various neuroendocrine peptides such as secretin or somatostatin (29,30). The expression and regulation of various proteins such as transporters and receptors have been studied using molecular and cellular techniques (7,23,31). In addition, the development of methods to isolate apical and basolateral plasma membrane fractions from isolated cholangiocytes using sucrose gradients has helped to study the subcellular distribution of proteins such as ion transporters (32). Although these primary cholangiocyte preparations were invaluable to study biliary physiology, they have certain inherent limitations. Their relatively low cell yield and diminishing viability after isolation as well as their high demand for labor and cost in isolation procedures have restricted their use. Furthermore, these isolated cell preparations lose cell polarity and local cellular contacts, which are important for various aspects of epithelial cell physiology such as ion transport and the expression of cellular proteins. In addition, although the size of the cell and the nucleus/cytoplasm ratio may help to determine the approximate origins of the isolated cells with respect to the anatomical location (6,7), the lack of specific markers makes it impossible to ascertain from which biliary segment they are derived. The localization of the origins of these isolated cholangiocytes may be important, since recent studies indicate functional heterogeneities among cholangiocytes from different segments of bile ducts (6,7). C— Bile Duct Cell Culture/Monolayer To overcome some of the problems associated with primary cell cultures, longterm biliary cell cultures and cell lines have been developed to study biliary physiology. These include the cholangiocarcinoma cell lines MzChA1 and SkChA1, which were isolated from human adenocarcinomas derived from the gallbladder and extrahepatic biliary tree, respectively (33). They have been used to study ion channels (27,28), intracellular pH (34), and signal transduction pathways (27). More recently, by using retroviral transduction of SV40 large T antigen, immortalized bile duct cell lines from mouse (35) and human (36) livers were developed. Furthermore, by using the same technique, immortalized cholangiocytes were also isolated from patients with cystic fibrosis (37). The development of these techniques to prepare immortalized cholangiocytes from diseased livers should help to study various liver diseases affecting bile ducts. However, these transformed or tumor cell lines have intrinsic limitations related to their origin and the nature of transformed cells, so that they may not exhibit normal cholangiocyte phenotypes. In addition, longterm cholangiocyte cultures also have been established from both rat and human livers (38–40). Rat cholangiocytes can form confluent polarized monolayers with tight junctions, providing an invaluable tool to study ion transport and secretion (39,41). Although these cell cultures are thought to be derived from "normal" cholangiocytes, they also have certain problems, such as clonal selection and dedifferentiation, which can modify their phenotypes and gene expression with multiple passages. Therefore, they may not represent normal cholangiocytes, raising questions on the validity of applying the results to normal biliary physiology. Furthermore, as with the isolated cholangiocyte model, functional heterogeneities among cholangiocytes cannot be studied with these cell lines, since they represent only the phenotypes of certain cholangiocyte populations from which these cell lines derived. D— Isolated Bile Duct Unit To overcome some of the problems associated with the isolated cholangiocytes, recent efforts have focused on the isolation of intact, polarized bile duct units. Initially, microdissection techniques were used to isolate intrahepatic bile ducts from pig livers (42). This cell preparation could be used within 6 h of isolation to study the regulation of intracellular pHi during secretin stimulation. Subsequently, similar bile duct units were microdissected from rat livers following
Page 103
collagenase perfusion and injection of a liquid trypan blueagar solution into the portal vein and hepatic artery to better distinguish bile ducts from blood vessels (43). These isolated bile ducts were cultured for 36 to 48 h to allow the open ends to seal and form bile duct units. These IBDUs were then microinjected with fluorescent pH and Cl markers to measure luminal pH and [Cl] with secretin stimulation (43). This preparation method is very laborintensive and timeconsuming, yielding only a small number of IBDUS. It also cannot be used to isolate smaller interlobular IBDUS, ranging from 15 to 100 m in diameter, which make up the major portion of the Bile Duct Epithelium (BDE) and are thought to represent the main "active" secretory site. More recently, we have developed a novel intact polarized isolated bile duct unit (IBDU) from rat liver, consisting of an enclosed lumen lined by polarized cholangiocytes (44) (Fig. 1). In contrast to the earlier isolation methods from pig (42) and rat livers (43), our newer isolation method eliminates the tedious and time consuming microdissection step and can produce many functional and polarized IBDUs from small to mediumsized (30 to 100 m) bile ducts from rat (44). The isolation method involves collagenase perfusion, teasing out parenchymal cells, mincing, and serial enzymatic digestion of the remaining stroma with pronase, collagenase, DNase, and hyaluronidase (44). After selecting digested bile duct fragments using sized meshes, segments of bile ducts averaging 25 m in diameter can be identified by glutamyl transferase and cytokeratin (7,19) immunostaining (44). When placed on matrigel and cultured for 24 to 48 h, these segments seal off their luminal ends and develop spheroids with an enclosed lumen lined by cholangiocytes, as demonstrated by Nomarski, confocal, and electron microscopy (Fig. 1) (44). Functionally, we have demonstrated these IBDUs to be a powerful functional secretory model to study the effects of agonists and inhibitors on biliary secretion (16,44). The IBDUs respond to neuroendocrine peptides such as secretin, bombesin, and VIP by increasing secretion into their enclosed luminal spaces, and these changes in volume can be measured by quantitative videomicroscopy (16,44). Using BCECF microfluorometric pH and micropuncture techniques, we have shown that these peptides stimulate fluid and bicarbonate secretion at the level
Figure 1 Videomicroscopy of isolated bile duct unit (IBDU) from the rat. Nomarski images of the IBDU show that IBDU is consisted of a central lumen enclosed by cholangiocytes. Videomicroscopic images before (left panel) and after (right panel) stimulation with VIP (100 nM) for 30 min demonstrate that the enclosed lumen expands with an increased secretion into the lumen.
Page 104
Figure 2 Ion transport mechanisms in the cholangiocyte. Many ion transport mechanisms have been characterized recently, but the precise localizations for some of these transporters are not determined; thus they are tentatively shown in the figure. At the apical membrane, a exchanger (AE2), which is presumably coupled to Cl channels, mainly mediates secretion into the bile following administration of choleretic agents such as secretin. A number of different Cl channels have been characterized, including cystic fibrosis transmembrane conductance regulator (CFTR), Ca2+activated Clchannel (CDCC), as well as highconductance anion channel (HCAC). The CDCC is thought to be activated by increased intracellular Ca2+ levels from luminal purinergic nucleotides or UDCA. Apical Na+/H+ exchanger (NHE2) is thought to mediate Na+ reabsorption. At the basolateral membrane, in addition to Na+/H+ exchanger (NHE1), acid extrusion is mainly mediated by symport in the rat, Na+dependent exchanger in the human, and H+ATPase in pig cholangiocytes to counteract intracellular acidification from apical secretion. Basolateral Na+/K+ATP ase in conjunction with K+ channels establish and regulate Na+ gradient and membrane potentials, which are vital for various ion transport activities. High intracellular Cl level is maintained by a basolateral Na+/K+/2Cl cotransport. Water transport across bile duct epithelium is facilitated by water channels present in both basolateral and apical membranes. In addition, a carbonic anhydrase present in cholangiocytes helps intracellular pH regulation.
Page 105
of the bile duct by increasing exchange (16,17,45). Furthermore, studies using various specific inhibitors have enabled us to further characterize the underlying ion transport mechanisms involved in these neuroendocrine peptidestimulated biliary secretory responses (46,47). These novel IBDUs represent the only intact functional polarized in vitro bile duct model that permits direct measurements of net fluid secretion to study the biliary secretion in small to mediumsized interlobular segments of the biliary tree where CFTR resides (7,44). Moreover, the lumen is large enough to be microinjected with cellimpermeable pHsensitive BCECF dextran to study changes in luminal pH (16,44). In addition, this isolation method can be of help in the study of various populations of IBDUs from different segments of biliary tree with apparent functional and structural heterogeneities (6,7) by selecting different sizes of bile duct fragments during enzymatic digestion steps. Although each experimental cell model has its shortcomings, they have provided invaluable clues to the complex puzzles in biliary physiology required to elucidate the mechanism and regulation of biliary secretion and modification at the cellular and molecular levels, as discussed below. V— Fluid and Solute Transport Biliary fluid and electrolyte transport have been extensively reviewed elsewhere (48–50). In this review, current information on the mechanism and regulation of biliary secretion and modification of bile by cholangiocytes is summarized. A— Electrolyte Transport Since secretin was shown to stimulate bicarbonaterich choleresis from bile ducts in the 1960s, ion transport mechanisms in cholangiocytes have been extensively studied and characterized (Fig. 2). These ion transporters can be broadly classified as carriers, such as exchangers and cotransporters, or ion channels. 1— Ion Transporters A number of ion transporters have been characterized in cholangiocytes using ion transport and pH studies (Table 2). Some of these transporters are active transporters such as Na+/K+ ATPase and H+ATPase, which require energy generated from ATP hydrolysis, while others are facilitated transporters, such as Na+/H+ exchanger and Na+/K+/2Cl cotransporter, which are driven by ion electrochemical gradients. These transporters play important roles in cellular homeostasis, ion transport, and pH regulation.
Page 106 Table 2 Ion Transporters Present in Bile Duct Epithelium Ion transporter
Animals
+
Localization
Stoichiometry
Activator
Inhibitor
Rat, human
Apical
1:1
Secretin, forskolin
DIDS
Human
Basolateral
1:1
DIDS
H ATPase
Pig
Basolateral
ATP, ?secretin
Bafiolmycin A1
Na+/H+ exchanger: NHE1
Rat, human
Basolateral
1:1
Amiloride
NHE2
Rat, human
Apical
1:1
Amiloride
Rat
?Basolateral
1:3
Depolarization
DIDS
All
Basolateral
3:2
ATP
Ouabain
Rat
?Basolateral
Bumetanide
Na independent
exchanger
Na+dependent
exchanger
+
cotransporter Na+/K+ ATPase +
+
Na /K /2Cl cotransporter
Key: DIDS, 4,4'diisothiocyano2,2'stilbene disulfonic acid.
Page 107 +
a. Na Independent in response to their changes in chemical gradient (19,26,45,51). This transporter is thought to be closely coupled to Cl channel for its function and activation during secretinstimulated biliary secretion (26). This exchanger has been localized to small and mediumsized bile ducts in humans (51) but to medium and largesized bile ducts in rats (7). The physiological significance of the heterogeneous distribution of this transporter is unknown. b. Na+Dependent c.
symporter of rat cholangiocytes, into the cell (52).
in response to transmembrane electrical potential changes (19,26,45).
d. Na+/H+ Exchanger. Driven by the Na+ electrochemical gradient, this transporter exchanges a Na+ with H+ and is inhibited by amiloride (19,26). Functional pH studies in rat and human cholangiocytes indicate that they may function as an acid extruder following an acid load to maintain phi (19,26,36,52). However, its activity is inversely proportional to pHi due to its known allosteric inhibition by H+ and has a ''set point" of pH 7.15 in rat cholangiocytes (19,26). Thus, Na+/H+ exchanger (NHE) is thought to be relatively inactive under basal conditions in media containing and may play a minor role in recovery from acid load under physiological conditions. Recent molecular study has confirmed the presence of NHE isoforms 1 and 2 in cholangiocytes, while hepatocytes appears to express only isoform 1 (55). The NHE1 isoform is thought to be localized in the basolateral domain, while NHE2 isoform may be present in the apical domain as in other Na+ absorbing epithelia such as gallbladder (8) and is thought to be involved in the Na+ reabsorption. e. H+ATPase. Like Na+/K+ ATPase, H+ATPase is an active transporter requiring ATP as an energy source to transport H+ against its electrochemical gradient. Some functional pH studies suggest the presence of bafilomycininhibitable, vacuolartype H+ATPase in the basolateral domain of pig cholangiocytes, which may mediate a counterregulatory pH adjustment during secretinstimulated bicarbonate secretion (53). However, pH studies in rat and human cholangiocytes (45,52,54) and immunocytochemical studies of rat bile ducts (45,54) failed to show the presence of H+ATPase in these epithelia. f. Na+/K+ATPase. In many epithelial cells, this active electrogenic transporter provides electrochemical gradients necessary for diverse physiological processes by pumping out three Na+ ions in exchange for two K+ ions by using the energy from ATP hydrolysis. Immunocytochemical study of the human biliary tree showed that this transporter is localized in the basolateral domain (57). This active transporter maintains the Na+ gradient required for the functioning of various Na+dependent transporters discussed in this section. g. Na+/K+/2Cl Cotransporter. Radioisotope uptake and efflux data suggest the presence of Na+/K+/2C cotransporter inhibitable by bumetanide in transformed bile duct cell
Page 108
lines (27). This ion transporter may play an important role in basolateral Cl uptake to maintain the high Cl gradient in cholangiocytes necessary to secrete Cl into the bile when Cl channels are activated (27). 2— Ion Channels Ion channels are selective transmembranous proteins that provide an aqueous pore for the movement of ions driven by their electrochemical gradients. In bile duct epithelia, a number of different ion channels have been identified and characterized, as shown Table 3. a. Chloride Channels. Recent electrophysiological and radioisotope efflux studies have demonstrated various Cl channels in cholangiocytes. Separate Ca2+ and cAMPdependent Cl currents have been identified and are thought to participate actively in the regulation of fluid and electrolyte secretion in bile duct epithelial cells. 1. cAMPDependent Chloride Channels or CFTR. The cAMPdependent Cl channel is the best studied ion channel in bile duct epithelium. Both structurally and functionally it appears to be the cystic fibrosis transmembrane conductance regulator (CFTR), which mediates secretinstimulated biliary secretion (26,28,58). It is expressed only on the luminal membranes of cholangiocytes but not on hepatocytes (58). This Cl channel has a linear currentvoltage relationship with no time dependence and is stimulated by forskolin or CAMP analogues, but it is not inhibited by DIDS (28,58). This Cl channel is also thought to be closely coupled to the secretion during secretinstimulated choleresis (26,28). However, studies in other cells suggest that CFTR has multiple other functions, which include regulation of other Cl channels (59) or Na+ channels (60), ATP transport across membranes (59), regulation of endocytotic recycling (61), and protein glycosylation and sulfation (62). Therefore, CFTR is a complex multifunctional protein that is responsible for the myriad heterogeneous pathologies in cystic fibrosis (CF) patients and the defects in biliary secretion in these patients may contribute to the pathogenesis of cholestatic liver disease in CF. 2. CalciumDependent Chloride Channels. In bile duct epithelium, intracellular Ca2+ levels increase with extracellular nucleotide triphosphates such as ATP or UTP, muscarinic agonists such as acetylcholine and carbacol, as well as the bile acid ursodeoxycholic acid (UDCA) (34,63–65). Increase in intracellular Ca2+ level stimulates the Ca2+dependent Cl channels (CDCCS) via activation of Ca2+/calmodulindependent protein kinase II (50,64). These channels are outwardly rectified, show timedependent activation at depolarizing potentials, and reverse near the equilibrium potential for Cl (64). They can be stimulated by ionomycin and inhibited by the Cl channel blocker DIDS. Although it is plausible that increased intracellular Ca2+ levels may induce biliary secretion, there is no direct evidence that CDCCs have any significant role in biliary secretion in cholangiocytes. In fact, one secretion study in rat bile duct units has shown that increases in intracellular Ca2+ have minimal direct effects on bile duct secretion (65). Therefore it remains to be determined whether CDCCs can selectively modulate biliary secretion and to determine their physiological role in cholangiocytes. 3. HighConductance Anion Channels. Electrophysiological studies have identified highconductance anion channels (HCACS) in cholangiocytes (66,67), which are generally closed in basal conditions and have significant permeability to both Cl and (2:1). They are shown to be insensitive to CAMP or Ca2+ but are regulated cooperatively by membrane potential and pertussis toxin (PTX)sensitive G proteins in a membranedelimited manner (66,67). They can be activated by GDP and inhibited by GTP and PTX (67). Although HCACs are thought to be involved in the regulation of pH, cell volume, and secretion of fluid and electrolytes, their role in biliary secretion and physiology remains to be further elucidated. b. Potassium Channels. Potassium channels are involved in the regulation of electrogenic solute transport in various epithelia including bile duct epithelia (27,68,69). The biliary secretion induced by various secretagogues such as secretin, bombesin, and VIP is dependent on K+ channels, presumably related to the role of the cell membrane potential as described for
Page 109 Table 3 Ion Channels Present in Bile Duct Epithelium Channels
Localization
Ion selectivity
I/V relation
Time dependence
Activator
Apical
Cl > I
Linear
No
Secretin, forskolin
NPPB
Calciumdependent (CDCC)
?Apical
I > Cl
Outward
Yes
ATP, UDCA, ionoycin
DIDS
Highconductance anion channel
?
Cl > gluconate
Potassium channels
?Basolateral
Water channels (aquaporin)
Both
Linear
Inhibitor
cAMPdependent (CFTR)
Chloride channels
PTX
ATP
BaCl2
Secretin
HgCl2
Key: NPPB, 5nitro2'(3phenylpropylamino)benzoate; DIDS, 4,4'diisothiocyano2,2'stilbene disulfonic acid; PTX, pertussis toxin.
Page 110 +
secretin (26,46,47). These secretagogues may facilitate secretion by activating K channels and hyperpolarizing cholangiocytes, which provides an electrogenic driving force for Cl conductance via Cl channels or balances the membrane depolarization resulting from exit at the luminal surface. Among many types of K+ channels known in different cell types, maxiK+ channels, which are inhibited both by BaCl2 and TEA, appear to be involved in the secretin and bombesinstimulated fluid secretion (47). While the main effects of various secretogogues such as secretin and DBcAMP are thought to be induced by an increase in apical Cl conductance that results in membrane depolarization, it does not exclude the possibility that a primary increase in basolateral K+ conductance may also occur. Whether this K+ channel involvement represents a primary increase in basolateral K+ conductance, as occurs in some Cl secreting epithelia (68), or a secondary increase driven by membrane depolarization resulting from Cl efflux remains to be determined. In fact, patchclamp experiments in pancreatic duct cells have shown that following exposure of these cells to secretin, the activity of maxiK+ channels increases more than 2000 times above control values mainly by a substantial reduction in the time that the channel is in the closed state (69). Electrophysiological studies will be needed to further clarify this question in regard to cholangiocytes. 3— Carbonic Anhydrase A histochemical study has demonstrated that carbonic anhydrase activity is preferentially localized to bile ductules, with weaker reactions in peripheral zones of the liver lobules (70). Previous in vivo studies in bile fistula rats (71,72) showed that acetazolamide significantly decreased UDCAinduced bile flow and bicarbonate secretion. In pigs (70), acetazolamide also inhibited UDCAdependent as well as secretinstimulated biliary secretion but had no effect on basal secretion in sodium taurocholateinfused pigs. These studies indicate the involvement of carbonic anhydrase in bicarbonate secretion into bile. Recent studies using isolated rat bile duct units confirmed that carbonic anhydrase has a minimal function during basal secretion but may be necessary to generate additional when secretion is stimulated by various secretagogues such as secretin, bombesin, and VIP (46,47). Thus, these findings suggest that carbonic anhydrase in cholangiocytes may provide another mechanism, in addition to symport or H+ATPase, to increase the supply when the demand for increases during NHCO.GIF secretagoguestimulated secretion. B— Water Transport Water movement through transcellular and paracellular pathways passively follows osmotic driving forces produced by active solute transport in epithelial cells. Tight junctions play important roles in establishing cell polarity and barrier. In bile duct epithelium, in vitro data indicate that transepithelial water transport occurs primarily through a transcellular pathway and is mainly mediated by water channel or aquaporin (73). Recent molecular and immunohistochemical studies have shown that aquaporin 1 (AQP 1) is present in apical as well as basolateral domains of cholangiocytes but not in hepatocytes (73,74). Furthermore, this water channel is thought to be regulated by membrane recycling and involved in the secretininduced biliary secretion (75). C— Bile Acid Transport/Cholehepatic Circulation In vivo studies of the effect of administration of bile acids on bile secretion in anesthetized biliary fistula hamsters, rats, and guinea pigs as well as the isolated perfused hamster liver showed that UDCA induced a marked bicarbonaterich hypercholeresis, which was far greater than that induced by any other bile acid administered (76). As an explanation for this UDCA
Page 111
induced bicarbonaterich hypercholeresis, the cholehepatic shunt hypothesis was proposed (76). This hypothesis postulates that a fraction of UDCA taken up by hepatocytes is secreted into canalicular bile in the unconjugated form and is protonated by a H+ ion derived from carbonic acid that was generated by the hydration of luminal CO2 by carbonic anhydrase present in biliary ductular cells. The protonated UDCA is reabsorbed, thus leaving a bicarbonate anion in the lumen. The bile acid passes through the cholangiocyte and is transported across the basolateral membrane; it returns to the sinusoids via the peribilliary capillary plexus and is taken up by the hepatocytes to then be resecreted into bile. Thus, this recycling of the UDCA by cholehepatic shunt pathway can provide a plausible explanation for the observed UDCAinduced bicarbonaterich hypercholeresis. Although bile acid transport was thought to occur in cholangiocytes, the details of this process were not known until recently. Molecular and radioisotope uptake studies in confluent polarized monolayers of normal rat cholangiocytes have demonstrated a Na+dependent, unidirectional, apical to basolateral transport of [3H] taurocholate, indicating the presence of a bile acid transporter in bile duct epithelium (31,41). Kinetic studies in purified apical membrane vesicles and molecular characterizations in rat cholangiocytes have shown that a bile acid transporter is present in the cholangiocytes and is identical to the rat ileal apical Na+dependent bile acid transporter (31,41). Immunoblots and immunohistochemistry have demonstrated that this 48kDa protein is present only in apical membranes of large but not small cholangiocytes (31,41). In addition, the 14kDa ileal cytosolic binding protein is also shown to be present in large but not small cholangiocytes (31) and may help to transport absorbed bile acids across the cholangiocyte. Therefore, these studies indicate that conjugated bile acids can be taken up by this apical bile acid transporter and may modify canalicular bile secretion and modulate ductal bile secretion. However, the real physiological significance of this bile acid uptake system in cholangiocytes and the transport mechanism across the basolateral membrane is poorly understood. Moreover, the UDCAinduced hypercholeresis may not use this transport mechanism, since the protonated UDCA is thought to be reabsorbed mainly by permeation across the apical membrane (76). D— Glucose Transport For a long time, physiological studies have shown that biliary epithelia absorb simple sugars such as glucose and xylose from the bile system by Na+dependent and independent transport systems (77,78). This ductular absorption of monosaccharides also causes decreased bile flow and increased taurocholate concentration in bile, suggesting that this process also induces water reabsorption (78). Although the physiological roles of this ductular absorption of sugar are not clear, it may prevent bacterial growth in gallbladder by decreasing the nutrients in bile and may serve as a mechanism for water absorption from bile duct epithelium (78). Recent studies have demonstrated the presence of two glucose transporters in bile duct epithelium. Radiolabeled nonmetabolizable monosaccharide uptake studies using polarized rat cholangiocyte monolayer revealed a vectorial, saturable, apical glucose uptake which was Na+dependent and inhibitable by phlorizin, a competitive inhibitor of the Na+/glucose cotransporter (41). Further molecular studies using RTPCR also confirmed the expression of the Na+/glucose transporter SGLT1 in cholangiocytes (41). In addition, the facilitated glucose transporter GLUT1 has also been shown to be present in cholangiocytes (41,79). However, in contrast to the previous immunofluorescent study in rat liver sections showing its apical and lateral plasma membrane localization (79), a more recent molecular study using polarized rat cholangiocyte monolayers showed that GLUT1 was detected in vesicles enriched in basolateral plasma membrane fractions (41). The reason for this discrepancy is not clear. These studies suggest that cholangiocytes take up glucose from bile via SGLT1 (and GLUT1) present on the apical domain and transport glucose across the basolateral membrane via GLUT1, thus accounting for glucose absorption from bile.
Page 112
E— Amino Acid Transport Measurements of biliary amino acids in the rat, guinea pig, rabbit, and dog reveal that glutamate (0.06 to 0.5 mM), cystine (0.2 to 1.1 mM), and glycine (1.7 to 2.8 mM) account for most of the total amino acids in bile (80,81). In these animals, glutathione contributes to the major portion of bile acidindependent bile flow. It is secreted into bile but is broken down by GT present on the apical membranes of bile duct epithelium (80,81). Subsequently, the glutamate and cysteine moieties derived from the hydrolysis of glutathione are partially reabsorbed either as peptides, free amino acids, or conjugates (81). These data indicate that cholangiocytes have transport systems for amino acids involved in the intrahepatic reabsorption of amino acids from bile. One recent study in primary cultures of rat cholangiocytes demonstrated that cholangiocytes have highcapacity Na+dependent as well as a Na+independent uptake systems for glutamate (82) that are different from those in hepatocytes. These transport systems may play an important role in reclaiming the glutathione from the bile (80,81). VI— Regulation of Biliary Secretion Physiological in vivo studies in dogs in 1960s demonstrated that the feeding of food stimulated bicarbonaterich bile secretion (83). Secretin, identified by Bayliss and Starling in 1902, has been considered as the main regulator of biliary bicarbonate secretion (84). However, as with the Pavlovian response in gastric secretion, bicarbonaterich bile secretion increased also with sham feeding (85), indicating the importance of neural regulation in postprandial choleresis. Although there are many unanswered questions in the regulation of bile secretion, the unavailability of adequate experimental models and complexities of bile secretion involving hepatocytes and bile ducts has limited our understanding of this process. In this section, recent understandings of this fascinating topic are reviewed with special focus on endocrine, neural, and paracrine regulation of biliary secretion. A— Endocrine The first experiment showing that hepatic bile secretion is under hormonal control was performed in 1825 by Leuret and Lassaigne (86). They found that bile flow increased when vinegar was applied to duodenal mucosa. Subsequently Bayliss and Starling in 1902 described secretin as the responsible secretagogue (84). As shown in Table 4, many hormones have been shown to influence bile secretion in in vivo models but only recently, with the developments of various in vitro and ex vivo models of bile secretion, have these processes been examined systematically. Interestingly, endocrine control has a quite limited role in bile aciddependent bile secretion, but it has variable influences in canalicular bile acidindependent and bile ductular secretions. Some hormones—such as secretin, CCK, and glucagon— have stimulatory effects on bile secretion, while somatostatin and substance P are known to be inhibitory. Furthermore, these hormones also interact among themselves to modulate bile secretion. 1— Secretin Discovered in 1902 as the first hormone, secretin has been shown to stimulate bicarbonaterich bile secretion (84), but it was not until the 1960s that its choleretic effect was thought to occur by acting at the bile ducts (87). This secretinstimulated biliary exchange mechanism (87). Studies in the bile ductligated rat model with bile duct
Page 113 Table 4 Effects of Neuroendocrine Peptides on Bile Secretion Neuroendocrine peptide
Bile acid dependent
Bile acid independent
Bombesin
0
0
Bile ductal
CCK
0
+
Gastrin
0
0
Glucagon
0
+
Secretin
0
0
+
Somatostatin
/0
+
+/
Substance P
0
0
VIP
+
0
+
proliferation have also demonstrated that secretin acts on the bile duct epithelium to stimulate biliary bicarbonate secretion (3). Autoradiographic studies using 125I labeled secretin confirmed the presence of secretin binding sites on bile ducts but not on hepatocytes (88). Our recent pH studies in isolated rat cholangiocytes indicate that secretin stimulates biliary bicarbonate secretion by stimulating pH (19,26). In contrast, in human cholangiocytes, stimulation of Na+independent
entry to maintain the intracellular
secretion in human cholangiocytes.
In addition, secretin is also thought to stimulate secretion by membrane recycling of vesicles containing ion transporters (53,90) and water channels (75). In pig bile duct epithelium, secretin induces a colchicineinhibitable decrease in the number of cytoplasmic vesicles, which is associated with an increase in the basolateral membrane surface area (91,92). Moreover, functional pH data suggest that these vesicles may contain vacuolartype H+ATPase (53). These findings indicate that in pig cholangiocytes, secretin appear to recruit more H+ATPase to the basolateral membrane by vesicular exocytosis to counteract the increased intracellular acid load during the secretinstimulated secretion (53). In contrast, rat and human cholangiocytes lack H+ATPase (45,52). In rat cholangiocytes, some evidence exists for secretininduced microtubuledependent vesicular translocation of vesicles and AQP1 water channels to the plasma membrane (75,90), which may serve as a mechanism for secretinstimulated biliary fluid secretion.
Page 114
2— Gastrin Gastrin is a hormone produced by G cells in the antrum of the stomach. Gastrin actually comprises a family of closely related hormones that vary in peptide length. Although best known as a major regulator of gastric acid secretion, gastrin can stimulate pancreatic secretion and inhibit the reabsorption of water and electrolytes in the intestine, but it has no effect on gallbladder secretion (93–95). As for bile secretion, gastrin has been shown to induce choleresis in in vivo and ex vivo studies in dogs (93,96), but nonsulfated gastrin I had no effect on bile flow (96). One recent study in the rat demonstrates that gastrin receptors are expressed in cholangiocytes (97). However, that particular study instead shows that gastrin I has no effect on basal biliary secretion from the bile ducts but has inhibitory effects on secretin induced increases in cAMP levels in cholangiocytes as well as secretinstimulated choleresis in anesthetized rats (97). The reasons for the observed discrepancy in biliary secretory responses to gastrin in these studies are not clear but may be partly related to the use of different experimental models and the types and concentrations of gastrin used. 3— Somatostatin Since its initial isolation from sheep hypothalamus in 1973, somatostatin, a cyclic peptide with 14 or 28 amino acid residues, has been found throughout the body (98,99). It is produced by specialized endocrine cells such as D cells in the gastrointestinal tract and neurons and can act as a hormone and neuropeptide (98). Five subtypes (SSTRs 1 to 5) of somatostatin receptors have been identified in various tissues and are coupled to G proteins to inhibit adenyl cyclase or K+ channels (99). Somatostatin has been shown to inhibit exocrine secretion, release of neuroendocrine peptides and substances, gastrointestinal motility, splanchnic blood flow, and certain types of tissue growth and proliferation (98). In various animal models, somatostatin also has been shown to inhibit bile secretion both directly as well as indirectly by inhibiting the release of other secretagogues such as secretin (95). Rat cholangiocytes express SSTR2 mRNA exclusively, whose level increases with bile duct ligation (30). While many in vivo studies on dogs and in vitro studies in rat IBDU (16,17,100) suggested that somatostatin had no effect on secretininduced choleresis or biliary secretion, one study in bile ductligated rats showed that it inhibited both the secretinstimulated choleresis in IPRL and increase in cAMP levels in cholangiocytes (30). The reason for this discrepancy is not clear but may be related to the known upregulation of somatostatin receptors with bile duct ligation. In addition, somatostatin inhibits the bombesinstimulated biliary secretion (16) but not the choleretic effect of VIP (17,101). B. Neural Since the late nineteenth century, it has been recognized that the liver, gallbladder, and bile ducts are richly innervated by sympathetic and parasympathetic systems as well as the right phrenic nerve via anterior and posterior plexuses (102–104). Sympathetic innervation comes mainly from the T710 and reaches celiac ganglia via greater splanchnic nerves, while parasympathetic fibers are derived mainly from vagus nerves (105). Afferent nerves are poorly understood and thought to be localized in the hepatic branch of the vagus as well as in the anterior plexus of the hepatic nerve (103). In addition, some neuropeptidergic nerves are also present in portal triads and along the sinusoids (106,107). Although there are considerable variations in the hepatic nerve supply among different species, the intrinsic innervation by nerve fibers mainly follows the vascular and biliary structures. Some fibers may enter the hepatic lobule, where they form a network around hepatocytes and extend into the sinusoids (106,107). The terminal plexus in the liver parenchyma may be homologous to the Auerbach and Meissner plexuses in the gastrointestinal tract or the cardiac and pulmonary plexuses (106,107). These hepatic nerves have been shown to be important for both efferent and sensory
Page 115
functions in carbohydrate and lipid metabolism, regulation of the hepatic microcirculation, and osmo and chemoreception. Although nerve fibers are abundant within the portal tract, blood vessels, and bile ducts, their roles in bile secretion are poorly understood, partly due to the inherent complexities of bile secretion involving both hepatocytes and bile duct cells, inadequate study models, and interspecies variations in bile secretion. It is also difficult to determine whether these effects occur directly from neurotransmitters or indirectly from neurally mediated alterations in perfusion, released hormones, and/or metabolic changes. 1— Autonomic Nerve System A direct effect of vagal tone on bile flow has been suggested, since truncal vagotomy decreases spontaneous bicarbonate secretion and reduces insulininduced choleresis (108). However, vagal stimulation in dogs increases bile flow but has no effect in rabbits and cats (109,110). Recently, acetylcholine receptor subtype M3 but not M1 or M2 was identified in rat bile ducts (111). Interestingly, acetylcholine alone does not have any stimulatory effect on secretion from bile duct cells but augments the secretinstimulated activity of the exchanger by secretin (cAMP) and acetylcholine (Ca2+) may have an important role in the regulation of biliary bicarbonate secretion to neutralize the gastric acid delivered to the small intestines. However, given the pharmacological doses used for acetylcholine (10 M) in the study, the real physiological significance of this finding is unknown and needs further study. Adrenergic control mechanisms are even less understood. Splanchnic nerve section or dopamine administration increases bile secretion (110,112), while electrical stimulation of sympathetic nerves or norepinephrine administration inhibits bile secretion (113). Therefore the role of sympathetic nerves in the regulation of bile secretion appears to be inhibitory but remains unclear. 2— Neuropeptidergic Nerve System There are abundant neuropeptidergic nerves present in portal triads and along the sinusoids (106,107). Immunoreactivity to various neuropeptides within ganglion cells and nerves in the hepatic plexus and in the intrahepatic periarterial spaces suggests that these peptides are intrinsic to these structures (106). Thus, these neuropeptides likely regulate the function of the bile duct epithelium and/or the hepatic vasculature (107). Nevertheless, the functional significance of these hepatic peptidergic nerves in biliary physiology and bile secretion has not been clearly established until recently, partly because of the lack of adequate in vitro models and partly due to complex interactions with other secretagogues. a. Bombesin. Bombesin is a neuropeptide of 14 amino acid residues initially isolated from the skin of the European frogs Bombina bombina and Bombina variegata variegata (114). Subsequently, its mammalian homologue, gastrinreleasing peptide (GRP), was isolated from mammalian gut extracts and found to have identical biological actions and similar potency as bombesin (115,116). Bombesin/GRP is almost exclusively confined to nerve fibers (117) and GRPlike immunoreactivity was identified within ganglion cells and nerves in the hepatic plexus and in the intrahepatic periarterial spaces, suggesting an intrinsic origin for this peptide (106). Moreover, nerve cell bodies with bombesinlike immunoreactivity are also localized in hypothalamic and medullary nuclei in the central nervous system involved in the regulation of autonomic functions (118). Immunoreactivity to these peptides and their receptors also has been detected in the gastrointestinal tracts of various mammals (106). They have been shown to increase gastric secretion (119) and gut motility (120), stimulate gallbladder contraction (120), and release numerous peptides such as neurotensin, motilin, insulin, CCK, secretin, and glucagon (121–124).
Page 116
In the dog and pig, bombesin/GRP increase bicarbonaterich bile secretion (123–125). Our recent studies in the rat demonstrate that like secretin, bombesin can directly stimulate biliary bicarbonate and fluid secretion from bile ducts (16). The ion transport mechanisms (Fig. 3) involved in this bombesinstimulated biliary secretion appear to be similar to those of secretin (45). Bombesin stimulates the symporter by altering the Cl concentration gradient and membrane potential (45,47) (Fig. 3). However, unlike secretin, the response of the bombesin is not dependent on microtubules nor is it associated with increased cAMP levels in isolated cholangiocytes (127). These findings indicate that bombesin has a distinctly different underlying mechanism from that of secretin to induce secretion from cholangiocytes. Currently, the signal transduction pathway mediating the bombesinstimulated secretion in cholangiocytes is unknown but does not appear to involve cAMP, cGMP, or Ca2+dependent pathways (127). Study of modulating roles of other neuroendocrine peptides on the bombesin response has revealed that somatostatin but not substance P had a direct inhibitory effect on bombesinstimulated biliary secretion from bile ducts (16). However, in the in vivo setting, these inhibitory neuropeptides may also have additional indirect modulatory effects by affecting the release or metabolism of bombesin from nerve terminals. These studies suggest that central and/or hepatic nerves may regulate secretion by releasing neuropeptides that can activate specific receptors on cholangiocytes, thereby modulating biliary transport and secretion. Therefore, autonomic and/or intrinsic hepatic neural pathways might regulate biliary secretion through neuropeptides such as bombesin by producing a bicarbonaterich choleresis to counteract the increase in acid loads during food digestion or a Pavlovian response. b. Vasoactive Intestinal Peptide. A major regulatory enteric neuropeptide of 28 amino acid residues was initially identified by Said and Mutt in 1970 from porcine intestine (128) and named as vasoactive intestinal peptide (VIP) due to its potent vasodilatory properties. In addition to its vasoactive effects, VIP plays important roles in the regulation of blood flow (129); gastrointestinal motility (130); and salivary, gastric, pancreatic, intestinal, and gallbladder secretions (131,132). VIP is found exclusively in nervous tissue in enteric, peripheral, and central neurons in various mammals (106,133) and is considered a principal nonadrenergic, noncholinergic neurotransmitter (133,134). Although most VIP nerve fibers in the gastrointestinal tract are considered to be of intrinsic origin, some are supplied by extrinsic nerves, such as vagal nerve branches for human subdiaphragmatic enteric nerves (135) and the pelvic nerve in the cat (136). These findings suggest that VIP nerves may interact with other peripheral and central neurons to produce complex neuroregulation of gastrointestinal function and physiology. VIP has been known to increase bicarbonaterich biliary secretion in the human (137), dog (138), and rat (139). In addition, it increases bile acid secretion in rats with bile acid supplementation (139) but not in humans with partial bile acid depletion for 6 to 8 days after cholecystectomy and choledocholithotomy with common bile duct cannulation (137). Our recent studies in the rat have demonstrated that VIP stimulates biliary fluid and from bile duct cells by stimulating the exchanger, but more potently than secretin or bombesin (17,46) (Fig. 3). In addition, VIP also has a stimulatory effect on bile acid secretion, presumably from hepatocytes (17). Although the ion transport mechanisms responsible for the VIPstimulated choleresis from rat bile duct appear to be similar to that of secretin (46), the underlying signal transduction system is not known but appears to be independent of cAMP, cGMP, or Ca2+ signal transduction pathways, as with bombesin (140). However, unlike bombesin, VIPstimulated biliary secretion is inhibited by colchicine and is not inhibited by somatostatin or substance P (17). These studies indicate that, as with bombesin, VIP may also serve an important regulatory role in biliary secretion by stimulating a bicarbonaterich choleresis during food digestion or Pavlovian response to counteract acid loads from the stomach.
Page 117
Figure 3 Regulation of bile ductal secretion. A number of neuroendocrine peptides and mediators regulate bile ductal secretion via specific receptors and signal transduction pathways. Secretin stimulates and somatostatin (and possibly gastrin) inhibits biliary secretion by their effects on cAMP/PKAdependent pathways which appear to regulate the Cl channel activity by phosphorylation, but the signal transduction pathways of bombesin and VIP are not known. Increases in intracellular Ca2+ by ATP, UDCA, or others have no direct effect on biliary secretion, but acetylcholine augments the secretin response, presumably via Ca2+dependent pathways. Although all the specific details of processes involved are not understood, the end result appears to be stimulatory or inhibitory effects on the activity of exchanger as well as on the membrane recycling of vesicles containing various transporter(s), resulting increased or decreased fluid and secretions into bile.
Page 118
c. Substance P. In 1931 von Euler and Gaddum discovered substance P, the first gut neuropeptide to be found. Substance P has 11amino acid residues. It is found in nerve fibers in the myenteric and submucosal plexus of the gastrointestinal tract and has been shown to play a role in the regulation of intestinal secretion (141). In bile secretion, substance P has anticholeretic effects in in vivo studies in the dog, but it failed to produce a significant inhibitory effect on secretin, bombesin, or VIP stimulated fluid secretion in the dog or IBDU from rat (16,17,100). These findings indicate that substance P may inhibit hepatocyte canalicular bile secretion. Furthermore, the inhibitory neuroendocrine peptides like substance P or somatostatin may also act in vivo by inhibiting the release of bombesin from local nerve terminals. C— Paracrine Paracrine regulation of bile secretion is poorly understood. Recent studies in rat bile duct cells and human biliary cell lines indicate that biliary secretion may be modulated by extracellular ATP (64). In bile from the human, rat, and pig, adenosine nucleotides are present in micromolar concentrations sufficient to activate purinergic receptors (142). Extacellular ATP stimulates P2u receptors on apical membrane of bile duct cell and increases intracellular Ca2+ levels, thus resulting in activation of Cl conductances (65,143) (Fig. 3). Therefore, this purinergic receptor activation by the nucleotides released presumably from hepatocytes might constitute a paracririe mechanism for modulation and coordination of biliary secretion from hepatocytes and cholangiocytes. In addition, various bile acids may also have some modulatory effect on bile secretion. Bile acids are taken up by cholangiocytes by the Na+dependent bile acid transporter in the apical membrane as discussed previously (31,41). Furthermore, UDCA increases the intracellular Ca2+ levels in cholangiocytes (63), but UDCA has no significant effect on ion transporters (144). Although it is speculated that bile acids may have some modulatory effects on biliary secretion, the physiological effects or significance remain unknown. VII— Conclusion The advances in our understanding in biliary physiology and secretion in recent years have been spectacular, but this area of research is still young. Although many human liver diseases—such as CF liver disease, primary biliary cirrhosis, primary sclerosing cholangitis, and vanishing bile duct syndrome—are thought to arise mainly from the defects or injury in the bile ducts, with impaired biliary secretion, our knowledge in the pathophysiology and pathogenesis of these liver diseases is still very limited. However, as we begin to learn more of the physiology and biology of bile ducts, we will be better able to understand the underlying disease process and to develop treatments for these liver diseases. For example, initially, the CF liver disease was thought to be due to defects in hepatocytes. However, recent demonstration that CFTR is only expressed in cholangiocytes but not hepatocytes drastically changed the research focus to bile ducts. In fact, the CF liver diseases are currently thought to arise from the impaired bile duct secretion from defects in the cAMPdependent Cl channel, resulting obstruction of bile ductules from this thick biliary secretion. As discussed in this review, Cl channels other than CFTR are expressed in bile duct epithelium (Fig. 2); thus one possible therapeutic approach is to develop methods to activate noncAMPdependent biliary secretory pathways to compensate for the CFTR defect. Therefore, further understanding in biliary physiology and biology will eventually allow us to develop methods to treat and prevent such cholangiopathy. References 1. P Rous, PD McMaster. Physiologic causes of stasis bile. J Exp Med 34(21):75–95, 1921.
Page 119
2. N Tavoloni. The intrahepatic biliary epithelium: an area of growing interest in hepatology. Semin Liver Dis 7(4):280–292, 1987. 3. G Alpini, R Lenzi, L Sarkozi, N Tavoloni. Biliary physiology in rats with bile ductular cell hyperplasia—Evidence for a secretory function of proliferated bile ductules. J Clin Invest 81:569–578, 1988. 4. WHH Andrews. Excretory function of the liver: a reassessment. Lancet 2:166–169, 1955. 5. WE Burkel. The fine structure of the terminal branches of the hepatic arterial system in the rat. Anat Res 167:329–349, 1970. 6. A Benedetti, C Bassotti, K Rapino, L Marucci, AM Jezequel. A morphometric study of the epithelium lining the rat intrahepatic biliary tree. J Hepatol 24(3):335– 342, 1996. 7. G Alpini, S Roberts, SM Knutz, Y Ueno, S Gubba, PV Podila, G Lesage, NF LaRusso. Morphological, molecular, and functional heterogeneity of cholangiocytes from normal rat liver. Gastroenterology 110:1636–1643, 1996. 8. M Strazzabosco. New insights into cholangiocyte physiology. J Hepatol 27(5):945–952, 1997. 9. DH Adams. Biliary epithelial cells: innocent victims or active participants in immunemediated liver disease? J Lab Clin Med 128:528–530, 1996. 10. K Matsumoto, H Fujii, G Michalopoulos, JJ Fung, AJ Demetris. Human biliary epithelial cells secrete and respond to cytokines and hepatocyte growth factors in vitro: interleukin6, hepatocyte growth factor and epidermal growth factor promote DNA synthesis in vitro. Hepatology 20(2):376–382, 1994. 11. S Milani, H Herbst, D Schuppan, H Stein, C Surrenti. Transforming growth factors 1 and 2 are differentially expressed in fibrotic liver disease. Am J Pathol 139:1221–1229, 1991. 12. F Marra, R De Franco, C Grappone, M Pinzani, S Milani, S Pastacaldi, et al. Expression of monocyte chemotactic proteina during active hepatic fibrogenesis in humans. Ital J Gastroenterol 28:300, 1996. 13. C Housset, A Carayon, B Housset, C Legendre, L Hannoun, R Poupon. Endothelin1 secretion by human gallbladder epithelial cells in primary culture. Lab Invest 69(6): 750–755, 1993. 14. GJ Gores, LJ Kost, NF LaRusso. The isolated perfused rat liver: conceptual and practical considerations. Hepatology 6(3):511–517, 1986. 15. G Alpini, R Lenzi, W Zhai, PA Slott, MH Liu, L Sarkozi, N Tavolini. Bile secretory function of intrahepatic biliary epithelium in the rat. Am J Physiol 20:G124– 133, 1989. 16. WK Cho, A Mennone, SA Rydberg, JL Boyer. Bombesin stimulates bicarbonate secretion from rat cholangiocytes: implications for neural regulation of bile secretion. Gastroenterology 13:311–321, 1997. 17. WK Cho, JL Boyer. Vasoactive intestinal polypeptide is a potent regulator of bile secretion from rat cholangiocytes. Gastroenterology 117(2):420–428, 1999. 18. G Alpini, JO Phillips, B Vroman, NF LaRusso. Recent advances in the isolation of liver cells. Hepatology 20(2):494–514, 1994. 19. M Strazzabosco, A Mennone, J Boyer. Intracellular pH regulation in isolated rat bile duct epithelial cells. J Clin Invest 87:1503–1512, 1991. 20. DC Hixon, JP Allison. Monoclonal antibodies recognizing oval cells induced in liver of rats by N2fluorenylacetamide or ethionine and a choline deficient diet. Cancer Res 45: 3750–3760, 1985. 21. M Ishii, B Vroman, NF LaRusso. Isolation and morphologic characterization of bile duct epithelial cells from normal rat liver. Gastroenterology 97(5):1236–1247, 1989. 22. R Joplin, AJ Strain, JM Neuberger. Immunoisolation and culture of biliary epithelial cells from normal human liver. In Vitro Cell Dev Biol 25(12):1189–1192, 1989. 23. R Joplin, AJ Strain, JM Neuberger. Biliary epithelial cells from the liver of patients with primary biliary cirrhosis: isolation, characterization, and shortterm culture. J Pathol 162(3):255–260, 1990.
Page 120
24. K Paradis, HL Sharp. In vitro ductlike structure formation after isolation of bile ductular cells from a murine model. J Lab Clin Med 113(6):689–694, 1989. 25. AJ Lanca, Y Israel. Histochemical demonstration of sinusoidal gglutamyltransferase activity by substrate protection fixation: comparative studies in rat and guinea pig liver. Hepatology 14(1):857–863, 1991. 26. D Alvaro, WK Cho, A Mennone, JL Boyer. Effect of secretin on intracellular pH regulation in isolated rat bile duct epithelial cells. J Clin Invest 92:1314–1325, 1993. 27. S Basavappa, J Middleton, AW Mangel, JM McGill, JA Cohn, JG Fitz. Cl and K+ transport in human biliary cell lines. Gastroenterology 104:1796–1805, 1993. 28. JM McGill, TW Gettys, S Basavappa, JG Fitz. Secretin activates Clchannels in bile duct epithelial cells through a cAMPdependent mechanism. Am J Physiol 266:G731–G736, 1994. 29. R Lenzen, G Alpini, N Tavoloni. Secretin stimulates bile ductular secretory activity through the cAMP system. Am J Physiol 263(4 pt 1):G527–G532, 1992. 30. PS Tietz, G Alpini, LD Pham, NF Larusso. Somatostatin inhibits secretininduced ductal hypercholeresis and exocytosis by cholangiocytes. Am J Physiol 269(1 pt 1):G110–G118, 1995. 31. G Alpini, SS Glaser, R Rodgers, JL Phinizy, WE Robertson, J Lasater, A Caligiuri, Z Tretjak, GD LeSage. Functional expression of the apical Na+dependent bile acid transporter in large but not small rat cholangiocytes. Gastroenterology 113(5):1734–1740, 1997. 32. P Tietz, S Levine, R Holman, C Fretham, NF LaRusso. Characterization of apical and basolateral plasma membrane domains derived from cultured rat cholangiocytes. Analytical Biochemistry 254(2): 192–199, 1997. 33. A Knuth, H Gabbert, W Dippold, et al. Biliary adenocarcinoma: characterization of three new human tumor cell lines. J Hepatol 1:579–596, 1985. 34. M Strazzabosco, C Poci, C Spirli, L Sartori, A Knuth, G Crepaldi. Effect of ursodeoxycholic acid on intracellular pH in a bile duct epitheliumlike cell line. Hepatology 19(1): 145–154, 1994. 35. K Paradis, ON Le, P Russo, M StCyr, H Fournier, D Bu. Characterization and response to interleukin 1 and rumor necrosis factor of immortalized murine biliary epithelial cells. Gastroenterology 109(4): 1308–1315, 1995. 36. SA Grubman, RD Perrone, DW Lee, SL Murray, LC Rogers, Ll Wolkoff, AE Mulberg, V Cherington, DM Jefferson. Regulation of intracellular pH by immortalized human intrahepatic biliary epithelial cell lines. Am J Physiol 266(6 pt 1):G1060–G1070, 1994. 37. SA Grubman, SL Fang, AE Mulberg, RD Perrone, LC Rogers, DW Lee, D Armentano, SL Murray, HL Dorkin, SH Cheng, et al. Correction of the cystic fibrosis defect by gene complementation in human intrahepatic biliary epithelial cell lines. Gastroenterology 108(2):584–592, 1995. 38. Y Yang, SE Raper, JA Cohn, JF Engelhardt, JM Wilson. An approach for treating the hepatobiliary disease of cystic fibrosis by somatic gene transfer. Proc Natl Acad Sci USA 90:4601–4605, 1993. 39. B Vroman, NF LaRusso. Development and characterization of polarized primary cultures of rat intrahepatic bile duct epithelial cells. Lab Invest 74:303–313, 1996. 40. R Joplin, T Hishida, H Tsubouchi, Y Daikuhara, R Ayres, JM Neuberger, AJ Strain. Human intrahepatic biliary epithelial cells proliferate in vitro in response to human hepatocyte growth factor. J Clin Invest 90(4):1284–1289, 1992. 41. KN Lazaridis, L Pham, B Vroman, PC de Groen, NF LaRusso. Kinetic and molecular identification of sodiumdependent glucose transporter in normal rat cholangiocytes. Am J Physiol 272(5 pt 1):G1168–G1174, 1997. 42. O Villanger, T Veel, MR Holthe, EJ Cragoe Jr, MG Raeder. Secretin stimulates bile ductules to secrete both H+ and 376, 1992. 43. S Roberts, S Kuntz, G Gores, NF LaRusso. Regulation of bicarbonatedependent duc
ions. Acta Physiol Scand 146:369–
Page 121
tular bile secretion assessed by luminal micropuncture of isolated rodent intrahepatic bile ducts. Proc Natl Acad Sci USA 90:9080–9084, 1993. 44. A Mennone, D Alvaro, WK Cho, JL Boyer. Isolation of small polarized bile duct units. Proc Natl Acad Sci USA 92:6527–6531, 1995. 45. WK Cho, A Mennone, JL Boyer. Intracellular pH regulation in bombesinstimulated secretion in isolated bile duct units from rat liver. Am J Physiol 275:G1028– G1036, 1998. 46. WK Cho, SA Rydberg, JL Boyer. Characterization of VIPstimulated bile secretion in isolated perfused rat liver (IPRL) and cholangiocytes (abstr). Hepatology 4:147A, 1996. 47. WK Cho, JL Boyer. Characterization of ion transport mechanisms involved in bombesinstimulated biliary secretion in rat cholangiocytes. J Hepatology 30(6): 1045–1051, 1999. 48. JL Boyer. Bile duct epithelium: frontiers in transport physiology. Am J Physiol 33:G1–G5, 1996. 49. RA Marinelli, NF Larusso. Solute and water transport pathways in cholangiocytes. Semin Liver Dis 16(2):221–229, 1996. 50. AP Kwiatkowski, JM McGill. Electrolyte transport in biliary epithelia. J Lab Clin Med 130(1):8–13, 1997. 51. E MartinezAnso, JE Castillo, J Diez, JF Medina, J Prieto. Immunohistochemical detection of chloride/bicarbonate anion exchangers in human liver. Hepatology 19(6): 1400–1406, 1994. 52. M Strazzabosco, R Joplin, A Zsembery, L Wallace, C Spirli, L Fabris, A Granato, A Rossanese, C Poci, JM Neuberger, L Okolicsanyi, G Crepaldi. Na(+) dependent and independent Cl/HCO3 exchange mediate cellular HCO3 transport in cultured human intrahepatic bile duct cells. Hepatology 25(4):976–985, 1997. 53. O Villanger, T Veel, MG Raeder. Secretin causes H+ secretion from intrahepatic bile ductules by vacuolartype H+ATPase. Am J Physiol 265:G719–G724, 1993. 54. WK Cho, A Mennone, JL Boyer. Forskolin stimulates secretion in isolated polarized bile ductular units by mechanisms lacking a H+ATPase response—evidence for heterogeneity of the biliary epithelium (abstr). Hepatology 8:296A, 1993. 55. U Marti, C Elsing, EL Renner, S LiechtiGallati, J Reichen. Differential expression of Na+,H(+)antiporter mRNA in biliary epithelial cells and in hepatocytes. J Hepatol 24(4):498–502, 1996. 56. K Yoshitomi, BC Burckhardt, E Froemter. Rheogenic sodiumbicarbonate cotransport in the peritubular cell membrane of rat renal proximal tubule. Pflugers Arch 405:360–366, 1985. 57. JY Scoazec, AF Bringuier, JF Medina, E MartinezAnso, D Veissiere, G Feldmann, C Housset. The plasma membrane polarity of human biliary epithelial cells: in situ immunohistochemical analysis and functional implications. J Hepatol 26(3):543–553, 1997. 58. JA Cohn, TV Strong, MR Picciotto, AC Nairn, FS Collins, JG Fitz. Localization of the cystic fibrosis transmembrane conductance regulator in human bile duct epithelial cells. Gastroenterology 105:1857–1864, 1993. 59. EM Schwiebert, ME Egan, TH Hwang, SB Fulmer, SS Allen, GR Cutting, WB Guggino. CFTR regulates outwardly rectifying chloride channels through an autocrine mechanism involving ATP. Cell 81(7):1063–1073, 1995. 60. MJ Stutts, CM Canessa, JC Olsen, M Hamrick, JA Cohn, BC Rossier, RC Boucher. CFTR as a cAMPdependent regulator of sodium channels (see comments). Science 269(5225):847–850, 1995. 61. NA Bradbury, T Jilling, G Berta, EJ Sorscher, RJ Bridges, KL Kirk. Regulation of plasma membrane recycling by CFTR. Science 91:319–328, 1992. 62. A Dosanjh, W Lencer, D Brown, DA Ausiello, JL Stow. Heterologous expression of delta F508 CFTR results in decreased sialylation of membrane glycoconjugates. Am J Physiol 266(2 pt 1):C360–C366, 1994.
Page 122
63. GH Shimokura, JM McGill, T Schlenker, JG Fitz. Ursodeoxycholate increases cytosolic calcium concentration and activates Cl currents in a biliary cell line. Gastroenterology 109(3):965–972, 1995. 64. JM McGill, S Basavappa, AW Mangel, GH Shimokura, JP Middleton, JG Fitz. Adenosine triphosphate activates ion permeabilities in biliary epithelial cells. Gastroenterology 107:236–243, 1994. 65. MH Nathanson, AD Burgstahler, A Mennone, JL Boyer. Characterization of cytosolic Ca2+ signaling in rat bile duct epithelia. Am J Physiol 271(1 pt 1):G86– G96, 1996. 66. JM McGill, S Basavappa, JG Fitz. Characterization of highconductance anion channels in rat bile duct epithelial cells. Am J Physiol 262(4 pt 1):G703–G710, 1992. 67. JM McGill, TW Gettys, S Basavappa, JG Fitz. GTPbinding proteins regulate high conductance anion channels in rat bile duct epithelial cells. J Membr Biol 133 (3):253–261, 1993. 68. OH Petersen. Calciumactivated potassium channels and fluid secretion by exocrine glands. Am J Physiol 251:G1–G13, 1986. 69. MA Gray, JR Greenwell, AJ Garton, BE Argent. Regulation of maxiK+ channels on pancreatic duct cells by cyclic AMPdependent phosphorylation. J Membr Biol 115: 203–215, 1990. 70. T Buanes, T Grotmol, T Veel, T Landsverk, Y Ridderstrale, MG Raeder. Importance of carbonic anhydrase for canalicular and ductular choleresis in the pig. Acta Physiol Scand 133:535–544, 1988. 71. M Corbic, C Munoz, M Dumont, G de Couet, S Erlinger. Effect of systemic pH, PCO2 and bicarbonate concentration on biliary bicarbonate secretion in the rat. Hepatology 5: 594–599, 1985. 72. JJ GarciaMarin, M Dumont, M Corbic, G de Couet, S Erlinger. Effect of acidbase balance and acetazolamide on ursodeoxycholateinduced biliary bicarbonate secretion. Am J Physiol 248:G20–G27, 1985. 73. SK Roberts, M Yano, Y Ueno, L Pham, G Alpini, P Agre, NF LaRusso. Cholangiocytes express the aquaporin CHIP and transport water via a channel mediated mechanism. Proc Natl Acad Sci USA 91(26):13009–13013, 1994. 74. S Nielsen, BL Smith, El Christensen, P Agre. Distribution of aquaporin CHIP in secretory and resorptive epithelia and capillary endothelia. Proc Natl Acad Sci USA 90:7275–7279, 1993. 75. RA Marinelli, L Pham, P Agre, NF LaRusso. Secretin promotes osmotic water transport in rat cholangiocytes by increasing aquaporin1 water channels in plasma membrane: evidence for a secretininduced vesicular translocation of aquaporin1. J Biol Chem 272(20): 12984–12988, 1997. 76. YB Yoon, LR Hagey, AF Hofmann, D Gurantz, EL Michelotti, JH Steinbach. Effect of sidechain shortening on the physiologic properties of bile acids: hepatic transport and effect on biliary secretion of 23norursodeoxycholate in rodents. Gastroenterology 90(4):837–852, 1986. 77. P Guzelian, JL Boyer. Glucose reabsorption from bile: evidence for a biliohepatic circulation. J Clin Invest 53:526–535, 1974. 78. M Lira, CD Schteingart, JH Steinbach, K Lambert, JA McRoberts, AF Hofmann. Sugar absorption by the biliary ductular epithelium of the rat: evidence for two transport systems (see comments). Gastroenterology 102(2):563–571, 1992. 79. HJ Hacker, B Thorens, R Grobholz. Expression of facilitative glucose transporter in rat liver and choroid plexus: a histochemical study in native cryostat sections. Histochemistry 96(5):435–439, 1991. 80. N Ballatori, R Jacob, JL Boyer. Intrabiliary glutathione hydrolysis: a source of glutamate in bile. J Biol Chem 261(17):7860–7865, 1986. 81. N Ballatori, R Jacob, C Barrett, JL Boyer. Biliary catabolism of glutathione and differential reabsorption of its amino acid constituents. Am J Physiol 254(1 pt 1):G1–G7, 1988.
Page 123
82. I EisenmannTappe, S Wizigmann, R Gebhardt. Glutamate uptake in primary cultures of biliary epithelial cells from normal rat liver. Cell Biol Toxicol 7(4):315– 325, 1991. 83. DL Nahrwold, MI Grossman. Secretion of bile in response to food with and without bile in the intestine. Gastroenterology 53:11–17, 1967. 84. WM Bayliss, EH Starling. The mechanism of pancreatic secretion. J Physiol 28:325–353, 1902. 85. K Powell, L Miller, F Brooks. Effect of sham feeding on bile flow in cholecystectomized dogs. Proc Soc Exp Biol Med 118:481–483, 1965. 86. DL Kaminski, DL Nahrwold. Neurohormonal control of biliary secretion and gallbladder function. World J Surg 3(4):449–456, 1979. 87. WGM Hardison, JC Norman. Electrolyte composition of the secretin fraction of bile from the perfused pig liver. Am J Physiol 214:758–763, 1968. 88. M Farouk, SR Vigna, DC McVey, WC Meyers. Localization and characterization of secretin binding sites expressed by rat bile duct epithelium. Gastroenterology 102:963–968, 1992. 89. D Alvaro, A Mennone, JL Boyer. Role of kinases and phosphatases in the regulation of fluid secretion and 273:G303–G313, 1997.
exchange in cholangiocytes. Am J Physiol
90. A Kato, GJ Gores, NF LaRusso. Secretin stimulates exocytosis in isolated bile duct epithelial cells by a cyclic AMPmediated mechanism. J Biol Chem 267:15523–15529, 1992. 91. T Buanes, T Grotomol, T Landsverk, M Raeder. Secretin empties bile duct cell cytoplasm of vesicles when it initiates ductular Gastroenterology 95:417–424, 1988.
secretion in the pig.
92. T Veel, T Buanes, T Grotmol, E Engeland, MG Raeder. Colchicine blocks the effects of secretin on bile duct cell tubulovesicles and plasma membrane geometry and impairs ductular secretion in the pig. Acta Physiol Scand 139:603–607, 1990. 93. S Konturek. The influence of gastrin analogues on gastric, pancreatic and bile secretion. Acta Med Polona 8(2):201–212, 1967. 94. DL Nahrwold, AN Shariatzedeh. Role of the common bile duct in formation of bile and in gastrininduced choleresis. Surgery 70(1): 147–153, 1971. 95. L Thulin, C Johansson. Gastrointestinal hormones. Acta Chir Scand Suppl 482:69–72, 1978. 96. DL Kaminski, YG Deshpande. Effect of gastrin I and gastrin II on canine bile flow. Am J Physiol 236(5):E584–E588, 1979. 97. SS Glaser, RE Rodgers, JL Phinizy, WE Robertson, J Lasater, A Caligiuri, Z Tretjak, GD LeSage, G Alpini. Gastrin inhibits secretininduced ductal secretion by interaction with specific receptors on rat cholangiocytes. Am J Physiol 273(5 pt 1):G1061–G1070, 1997. 98. S Reichlin. Somatostatin. N Engl J Med 309(24):1495–1501, 1983. 99. F Raulf, J Perez, D Hoyer, C Bruns. Differential expression of five somatostatin receptor subtypes, SSTR15, in the CNS and peripheral tissue. Digestion 55 (suppl 3):46–53, 1994. 100. I Magnusson, L Thulin, K Einarsson, K Bergstrom. Effects of substance P and somatostatin on taurocholatestabilized and CCK or secretininduced choleresis in the anesthetized dog. Scand J Gastroenterol 19:1007–1014, 1984. 101. B Nyberg, B Angelin, K Einarsson. Somatostatin does not block the effect of vasoactive intestinal peptide on bile secretion in man. Eur J Clin Invest 22:60–66, 1992. 102. WF Alexander. The innervation of the biliary system. J Comp Neurol 73:357–370, 1940. 103. F Magni, C Carobi. The afferent and preganglionic parasympathetic innervation of the rat liver, demonstrated by the retrograde transport of horseradish peroxidase. J Auton Nerv Syst 8(3):237–260, 1983. 104. T Terada, Y Nakanuma. Innervation of intrahepatic bile ducts and peribiliary glands in
Page 124
normal human livers, extrahepatic biliary obstruction and hepatolithiasis: an immunohistochemical study. J Hepatol 9(2):141–148, 1989. 105. D Sasse, UM Spornitz, IP Maly. Liver architecture. Enzyme 46(1–3):8–32, 1992. 106. F Carlei, NJ Lygidakis, V Speranza, WH Brummelkamp, JF McGurrin, R Pietroletti, E Lezoche, DG Bostwick. Neuroendocrine innervation of the hepatic vessels in the rat and in man. J Surg Res 45:417–426, 1988. 107. M elSalhy, R Stenling, L Grimelius. Peptinergic innervation and endocrine cells in the human liver. Scand J Gastroenterol 28(9):809–815, 1993. 108. RE Geist, RS Jones. Effect of selective and truncal vagotomy on insulinstimulated bile secretion in dogs. Gastroenterology 60(4):566–571, 1971. 109. DL Kaminski, J Dorighi, M Jellinek. Effect of electrical vagal stimulation on canine hepatic bile flow. Am J Physiol 227:487–493, 1974. 110. CA Tanturi, AC Ivy. On the existence of secretory nerves in the vagi for and the reflex excitation and inhibition of bile secretion. Am J Physiol 121:270–283, 1938. 111. D Alvaro, G Alpini, AM Jezequel, C Bassotti, C Francia, F Fraioli, R Romeo, L Marucci, G Le Sage, SS Glaser, A Benedetti. Role and mechanisms of action of acetylcholine in the regulation of rat cholangiocyte Secretory functions [published erratum appears in J Clin Invest 1997 Oct 15;100(8):following 2139]. J Clin Invest 100(6):1349–1362, 1997. 112. RF Harty, RC Rose, DL Nahrwold. Stimulation of hepatic bile secretion by dopamine. J Surg Res 17(5):359–363, 1974. 113. K Beckh, R Arnold, Regulation of bile secretion by sympathetic nerves in perfused rat liver. Am J Physiol 261(5 pt 1):G775–G780, 1991. 114. V Erspamer, G Falconieri Erspamer, M Inselvini. Some pharmacological actions of alytesin and bombesin (letter). J Pharm Pharmacol 22:875, 1970. 115. TJ McDonald, G Nilsson, M Vagne, M Ghatei, SR Bloom, V Mutt. A gastrin releasing peptide from the porcine nonantral gastric tissue. Gut 1978(19):767– 774, 1978. 116. JR Reeve Jr, JH Walsh, P Chew, B Clark, D Hawke, JE Shively, Amino acid sequences of three bombesinlike peptides from canine intestine extracts. J Biol Chem 258:5582–5588, 1983. 117. GJ Dockray, C Vaillant, JH Walsh. The neuronal origin of bombesinlike immunoreactivity in the rat gastrointestinal tract. Neuroscience 4:1561–1568, 1979. 118. M Panula, HYT Yang, E Costa. Neuronal localization of the bombesinlike immunoreactivity in the central nervous system of the rat. Reg Pept 4:275–283, 1982. 119. G Bertaccini, V Erspamer, M Impicciatore. The actions of bombesin on gastric secretion of the dog and the rat. Br J Pharmacol 49:437–444, 1973. 120. AGE Pearse, JM Polak, SR Bloom. The new gut hormones. Gastroenterology 80:746–761, 1977. 121. HR Fender, PJ Curtis, PL Rayford, JC Thompson. Effect of bombesin on serum gastrin and cholecystokinin in dogs. Surg Forum 27:414–416, 1976. 122. V Schusdziara, D Rouiller, V Harris, EF Pfeiffer, RH Unger. Effect of bombesin upon plasma somatostatinlike immunoreactivity, insulin, and glucagon in normal and chemically sympathectomized dogs. Regul Pept 1:89–96, 1980. 123. DL Kaminski, YG Deshpande. Effect of somatostatin and bombesin on secretinstimulated ductular bile flow in dogs. Gastroenterology 85(6):1239–1247, 1983. 124. H Glad, P Svendsen, MA Ainsworth, O Olsen, JF Rehfeld, OB Schaffalitzky De Muckadell. The effect of gastrinreleasing peptide on porcine pancreaticobiliary bicarbonate secretion is mediated by secretin. Scand J Gastroenterol 29:195–202, 1994. 125. WJ Kortz, JRB Nashold, E Delong, WC Meyers, Effects of bombesin on fasting bile formation. Ann Surg 203(1):1–7, 1986. 126. WK Cho, A Mennone, JL Boyer. Intracellular pH regulation in bombesinstimulated secretion in isolated bile duct units from rat liver. Am J Physiol 275 (5):G1028–G1036, 1998. 127. WK Cho, A Mennone, JL Boyer. Bombesin stimulates bile secretion in calcium and PKCindependent pathways. Gastroenterology 114(4):L0112, 1998.
Page 125
128. SI Said, V Mutt. Potent peripheral and splanchnic vasodilator peptide from normal gut. Nature 225(235):863–864, 1970. 129. S Eklund, J Fahrenkrug, M Jodal, O Lundgren, OB Schaffalitzky de Muckadell, A Sjoqvist. Vasoactive intestinal polypeptide, 5hydroxytryptamine and reflex hyperaemia in the small intestine of the cat. J Physiol 302:549–557, 1980. 130. KN Bitar, GM Makhlouf. Relaxation of isolated gastric smooth muscle cells by vasoactive intestinal peptide. Science 216:531–533, 1982. 131. N Ashton, BE Argent, R Green. Effect of vasoactive intestinal peptide, bombesin and substance P on fluid secretion by isolated rat pancreatic ducts. J Physiol 427:471–482, 1990. 132. IK Morton, SJ Phillips, SH Saverymuttu, JR Wood. Secretin and vasoactive intestinal peptide inhibit fluid absorption and induce secretion in the isolated gall bladder of the guineapig (proceedings). J Physiol 266(1):65P–66P, 1977. 133. MG Bryant, MM Polak, I Modlin, SR Bloom, RH Albuquerque, AG Pearse. Possible dual role for vasoactive intestinal peptide as gastrointestinal hormone and neurotransmitter substance. Lancet 1 (7967):991–993, 1976. 134. RK Goyal, S Rattan, SI Said. VIP as a possible neurotransmitter of noncholinergic nonadrenergic inhibitory neurones. Nature 288(5789):378–380, 1980. 135. JM Lundberg, T Hokfelt, J Kewenter, G Pettersson, H Ahlman, R Edin, A Dahlstrom, G Nilsson, L Terenius, K UvnasWallensten, S Said. Substance P, VIP , and enkephalinlike immunoreactivity in the human vagus nerve. Gastroenterology 77(3):468–471, 1979. 136. M Kawatani, IP Lowe, I Nadelhaft, C Morgan, WC De Groat. Vasoactive intestinal polypeptide in visceral afferent pathways to the sacral spinal cord of the cat. Neurosci Lett 42(3):311–316, 1983. 137. B Nyberg, K Einarsson, T Sonnenfeld. Evidence that vasoactive intestinal peptide induces ductular secretion of bile in humans. Gastroenterology 96:920–924, 1989. 138. L Thulin, M Hellgren. Choleretic effect of vasoactive intestinal peptide. Acta Chir Scand 142:235–237, 1976. 139. G Ricci, J Fevery. The action of VIP on bile secretion and bile acid output in the nonanesthetized rat. Biochem Pharmacol 34(20):3765–3767, 1985. 140. WK Cho, JL Boyer. VIP stimulates bile secretion in cholangiocytes by cAMP, PKAindependent mechanisms (abstr). Hepatology 26(4):397A, 1997. 141. KA Hubel. Intestinal nerves and ion transport: stimuli, reflexes, and responses. Am J Physiol 248(3 pt 1):G261–G271, 1985. 142. RS Chari, SM Schutz, JE Haebig, GH Shimokura, PB Cotton, JG Fitz, WC Meyers. Adenosine nucleotides in bile. Am J Physiol 270(2 pt 1):G246–G252, 1996. 143. LI Wolkoff, RD Perrone, SA Grubman, DW Lee, SP Soltoff, LC Rogers, M Beinborn, SL Fang, SH Cheng, DM Jefferson. Purinoceptor P2U identification and function in human intrahepatic biliary epithelial cell lines. Cell Calcium 17(5):375–383, 1995. 144. D Alvaro, A Mennone, JL Boyer. Effect of ursodeoxycholic acid on intracellular pH regulation in isolated rat bile duct epithelial cells. Am J Physiol 265:G783– G791, 1993.
Page 127
6— Epidemiology, Risk Factors, and Pathogenesis of Gallstones Nezam H. Afdhal Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts Gallstone disease is the most common and costly of all digestive diseases resulting in 700,000 cholecystectomies annually and as many as 1 million hospitalizations. The cost of treating gallstonerelated disease in the United States has been estimated at approximately $5 billion dollars each year. In recent years, the development of laparoscopic cholecystectomy has significantly reduced hospital inpatient costs and recovery time, but this cost saving has been offset by a 28% increase in the number of cholecystectomies (1). Over the past two decades, a great deal has been learned about the epidemiology of and risk factors for gallstones (see Table 1). Ultrasonography has played a major role in this process, providing a rapid, riskfree method of screening large populations. Prior to the availability of ultrasound, most studies relied on highly selective autopsy data and limited oral cholecystography. I— Epidemiology Epidemiological data are now available from a number of large European and American populations (see Table 2). These studies have revealed a marked variation in overall gallstone prevalence between different ethnic populations. In general, there appear to be higher rates of cholelithiasis in western Caucasians, Hispanic, and Native American populations and lower rates in eastern European and African populations (2–6). In North America, the most detailed prevalence data are available for Hispanics and Native Americans. One report evaluated three distinct HispanicAmerican populations using ultrasonography: Puerto Ricans, Mexican Americans, and Cuban Americans (7,8). Even among Hispanic patients, there is a marked difference in gallstone prevalence dependent on ethnicity. Hispanic women between the ages of 20 and 40 years were found to have an overall prevalence rate of gallstones of 11.2%. The prevalence rose to 22.2% between the ages 40 and 60 years. The highest incidence was in those of MexicanAmerican origin, where a 1.5 to 1.8 times greater overall prevalence rate was seen compared to Cuban Americans and Puerto Rican females. The higher rate in Mexican Americans may represent genetic admixture with Native Americans where the prevalence rate is the highest in the world (see below). The prevalence rate was much lower in Hispanic men: 1.5 and 6% in the same respective age groups. Native Americans have the highest prevalence of cholelithiasis in North America. As an example, 73% of female Pima Indians over the age of 25 years have gallstones (9). Similar
Page 128 Table 1 Risk Factors for Cholelithiasis Age Female sex Parity Obesity Rapid weight loss Hypertriglyceridemia Genetic: Pima Indians, Chileans Medications: estrogen, clofibrate, ceftriaxone, sandostatin Terminal ileal resection Gallbladder hypomotility: pregnancy, diabetes, postvagotomy Somatostatinoma Total parenteral nutrition Spinal cord injury
high rates have been found in other Native American populations, such as Chippewa and Micmac Indians (10,11). In CaucasianAmerican populations in North America, the prevalence of cholelithiasis is less well established because there have not been any large ultrasoundbased studies. In a Michigan study using oral cholecystography, men between the ages 35 to 55 years were found to have an overall prevalence of 11%, while a small ultrasound survey of Canadian women between the ages of 15 and 50 years found an overall prevalence of 16.7% (12). Large ultrasoundbased studies from Europe have characterized both gallstone prevalence and incidence. As an example, the Multicenter Italian Study of Cholelithiasis (MICOL) examined nearly 33,000 subjects aged 30 to 69 years in 18 cohorts in 10 Italian regions (13,14). The overall prevalence of gallstone disease was 18.8% in women and 9.5% in men (14). Similar results were noted in the Sirmione study which found an overall prevalence rate of 11% in 1930 Italian subjects between the ages of 18 and 65 years (15). Ultrasounds were repeated on the same patients at 5year intervals. The 10year cumulative incidence of new gallstones was 4.6%. A similar study from Denmark examining the incidence of gallstones showed a cumulative incidence of 2.3%, with the incidence initially appearing higher in women with a female male ratio of 4.7 at age 35, which was reduced to 1.2 at age 65 years (16). Compared to the above ethnic groups, African Americans appear to have the lowest prevalence of cholelithiasis. Autopsybased studies performed in the 1950s found that African Americans had onehalf to onequarter the risk of cholelithiasis compared to Caucasian Americans; later reports have shown a 40% lower risk of hospitalization for gallstonerelated disease (17). II— Risk Factors In addition to the variability of gallstones in different ethnic populations, a number of other risk factors for this condition have been identified. A— Age Age is a major risk factor for the gallstones. Gallstones are exceedingly rare in children except in the presence of hemolytic states; in addition, less than 5% of all cholecystectomies are performed in children. In a study from Bari, where 1400 children and adolescents underwent
Page 129 Table 2 The Prevalence of Gallstone Disease in Selected Populations Population Mexican American 1982–84
Test
Female N
Percent GSD
Male N
Percent GSD
416
13.8
360
2.6
259
26.4
202
9.7
79
44.4
72
15.5
52
10.8
Age
US
20–39 40–59 60–74
Cuban Americans 1982–84
US
94
19.2
60–74
43
21.7
22
14.3
Puerto Ricans 1982–84
US
20–39
184
9
95
2.0
Rome, Italy 1981–82
Bristol, England 1987–89
Copenhagen, Denmark 1982–84
Okinawa, Japan 1984
Pima Indians 1967–68
20–39
US
US
US
US
OCG
40–49
157
60–74
41
20–29
39
0
73
5.1
21.2
81
3.3
12.1
24
11.1
158
2.5
44
2.3
30–39
404
5.9
312
2.0
40–49
311
10.9
430
6.7
17.8
203
14.7
250
14.4
50–59
168
>65
40
20–29
25 —
—
305
3.9
30–39
328
6.4
40–49
199
6.5
430
7.5 11.5
—
—
50–59
141
14.2
226
60–69
85
22.4
182
30
454
4.7
457
4.8
40
460
6.1
473
1.5
50
450
14.5
465
6.6
451
12.9
60
398
22.3
0–19
381
0
20–29
126
30–39
163
40–49
122
396
1.7
1.0
3
132
1.0
3.5
145
2.5
3
135
2.0 1.5
50–59
191
4
272
60–69
167
9
202
4.5
188
15.0
>70
107
9.5
15–25
45
12.7
25–34
47
73.2
45
4.4
35–44
53
70.8
51
11.1
45–54
47
75.8
51
31.9
47
66.3
57
67.8
55–64
50
>65
58
62 89.5
45
0
Key: US, ultrasound; OCG, oral cholecystogram. Source: Adapted from Ref. 128.
ultrasound, gallstones were only found in 2. However in the United States, with obesity becoming more of a problem in young adolescents and adults, we are seeing more clinical gallstone disease (18). Age 40 appears to represent the cutoff between relatively low and high rates of cholecystectomies. This observation was validated in the Sirmione study, in which the incidence between the ages of 40 and 69 years was four times higher than that in younger subjects (15). Among the 135 patients with gallstones, only 1 was between the ages of 18 and 21 years.
Page 130
B— Gender As noted above, a higher prevalence of gallstones has been observed in women in all age groups. The difference between women and men is particularly striking in young adults and this gap narrows with age. The GREPCO study found that a femaletomale ratio of 2.9 between the ages of 30 to 39 years; the ratio narrowed to 1.6 between the ages of 40 to 49 years and 1.2 between the ages of 50 to 59 years (19,20). Interestingly, women with gallstone disease are more likely to have had a cholecystectomy than males, and this was confirmed in the HHANES study, where 49% of women but only 28% of men with gallstones had a cholecystectomy. The higher rates in young women is probably hormonal, since estrogens have been shown to increase biliary cholesterol secretion and progesterone reduces gallbladder contractility. In countries such as Italy where the pregnancy rate is falling dramatically, sex differences in gallstone disease are less apparent even for the younger age groups. C— Pregnancy Pregnancy is a major risk factor for the development of cholesterol gallstones. The risk is related to both the frequency and number of pregnancies. In one report, for example, the prevalence of gallstones increased from 1.3% in nulliparous females to 12.2% in multiparous females (21). Another study recruited 272 women in the first trimester of pregnancy (22,23). The incidence of new biliary sludge and gallstones was 31 and 2%, respectively. Sex hormones induce a variety of physiological changes in the biliary system, which ultimately cause bile to become supersaturated with cholesterol, thereby promoting gallstone formation. These changes include the following: 1. Cholesterol supersaturation occurs as a result of an estrogeninduced increase in cholesterol secretion and a progesteroneinduced reduction in bile acid secretion (24). 2. Pregnancy induces a qualitative change in bile acid synthesis characterized by relative overproduction of hydrophobic bile acids such as chenodeoxycholate, thereby reducing the ability of bile to solubilize cholesterol (25). 3. Progesteroneinduced slowing of gallbladder emptying further promotes the formation of stones by causing bile stasis. These changes normalize 1 to 2 months following delivery. In the postpartum period, gallbladder sludge resolves in 61% of cases (23), and approximately 30% of stones smaller than 10 mm disappear due at least in part to desaturation of bile. Thus although there is a higher risk of gallstone and sludge development in the third trimester and early puerperium, there is also spontaneous resolution of gallbladder disease during these time periods as the physiology of bile secretion and gallbladder motility return to the normal state. D— Oral Contraceptives and Estrogen Replacement Therapy Studies evaluating the risk associated with estrogen use have been conflicting, although the majority of studies have shown that estrogen therapy is associated with higher rates of gallstones. Postmenopausal women given estrogen replacement had 3.7 times the relative risk of developing symptomatic gallstones compared to nonusers (8,26–28), and those women undergoing cholecystectomy were 2.5 times more likely to have used estrogen in the past than those not requiring cholecystectomy. In the Nurses' Health Study of almost 55,000 postmenopausal women, those currently using postmenopausal hormones were at an increased risk of cholecystectomy (relative risk 2.1) compared to neverusers (29). For current users, the risk of cholecystectomy increased with increasing duration of hormone use and higher doses of estrogen. The risk for past hormone users decreased substantially in women who had discontinued use within the preceding
Page 131
1 to 3 years (relative risk 1.6, a small but significant risk persisted for women who had stopped taking estrogen replacement therapy 5 or more years previously (relative risk 1.3). There is also an increased risk in men receiving estrogen therapy. In one study of men who had had a myocardial infarction, treatment with estrogen or clofibrate was associated with more than a twofold increase in risk of gallbladder disease compared to those receiving placebo (30). In another report of men with prostate cancer, new gallstones detected by ultrasonography developed at 1 year in 5 of 28 men treated with estrogen compared to none of 26 who underwent orchiectomy (31). Estrogentreated men had a 40% increase in biliary cholesterol excretion compared to agematched controls. Oral contraceptive use also appears to cause a slight increase risk of gallstone formation. Women under the age of 40 and those taking highdose estrogen (>50 g) preparations have the greatest added risk (28). It has been suggested that oral contraceptives have only a transient effect on gallstone formation. In support of this hypothesis, a casecontrol study found a slightly higher incidence of gallstones shortly after starting oral contraceptives, an effect that disappeared after 10 years (32). A similar relationship was noted in a metanalysis of controlled epidemiological studies (33). E— Family History and Genetics Family history studies suggest that genetics has a significant role in the development of gallstones. One report performed oral cholecystography in 171 firstdegree relatives of patients with gallstones and 200 agematched controls (34). Gallstones occurred more than twice as often in the family group: 20.5 versus 9%. A more recent study evaluated 330 firstdegree relatives of 105 patients with gallstones using ultrasonography; cholelithiasis was found in 15.5% of firstdegree relatives compared to only 3.6% of matched controls (35). The risk was greater in female relatives in both of these studies. The great variation in gallstone rates amongst different ethnic groups described above could be due to genetic as well as dietary and cultural habits. A dramatic example occurs in Pima Indians, who have exceptionally high rates of cholesterol gallstones: 73% in women over the age of 25 years (9). Although no gallstone genes have been described for humans, there are gallstonesusceptible mice that have a complex polygenic basis for developing gallstones (36). F— Obesity Obesity (defined as weight greater than 120% of ideal body weight) is a wellestablished risk factor for the development of cholesterol gallstones, presumably due to enhanced cholesterol absorption, synthesis, and secretion (37–41). The risk is particularly high in women and in those with morbid obesity as well as in younger age groups, among whom a threefold increase in risk has been reported (15). It has also been suggested that the incidence of gallbladder disease in morbidly obese subjects may be higher than expected from ultrasonography or oral cholecystography. In one report, 62 morbidly obese patients underwent prophylactic cholecystectomy at the time of a gastric exclusion procedure (42). Among the 47 who had normal imaging studies, 40 had abnormal histological findings in the gallbladder. G— Rapid Weight Loss Rapid weight loss is also a risk factor for gallstone formation, occurring in approximately 35% of patients after proximal gastric bypass (40,43,44). High rates of gallstone formation have also been associated with very low calorie diets (45). Gallstones that form in association with rapid weight loss appear to more common in Caucasians and women. The mechanism by which this occurs is incompletely understood. One report evaluated changes in gallbladder bile during periods of weight loss (44,46–48). Bile mucin content
Page 132
increased 18fold and bile calcium concentration rose 40%. These factors may promote cholesterol nucleation and stone formation. Motility is also affected by dieting and a reduction in effective gallbladder emptying in patients on very low calorie diet who receive less than 10 g fat daily has been described (49,50). In contrast to the general population, in which the great majority of gallstones are asymptomatic, persons with weight lossrelated cholelithiasis are more likely to be symptomatic. In one series, for example, 28% of patients required urgent cholecystectomy within 3 months after a gastric exclusion procedure (42). Prophylaxis with ursodeoxycholic acid (UDCA) has been shown to be effective at reducing the risk of stone formation during rapid weight loss. In one trial of 1004 patients treated with a very low calorie diet, ultrasonography was performed at baseline and at 8 and 16 weeks (45). The incidence of gallstones was 28% in the placebo group, compared to 8, 3, and 2% of those treated with 300, 600, and 1200 mg/day of UDCA, respectively. In another controlled trial, 68 obese subjects were randomized to UDCA (1200 mg/day), aspirin, or placebo at the time of entry into a 520 kcal/day diet program (50a). The patients were treated for up to 16 weeks and were reevaluated at 4 weeks and at 3 weeks after treatment. None of the patients treated with UDCA developed gallstones or cholesterol crystals in the bile compared to five and six patients, respectively, in the placebo group (p < 0.001). The bile saturation index fell by about 18% with UDCA compared to a 20% elevation in the patients receiving placebo. These findings have led to the recommendation that obese patients should be started on UDCA 600 mg at night during very low caloric diets or after weight reduction surgery. H— Diabetes Mellitus Diabetes mellitus appears to be associated with an increased risk of gallstones (51). A casecontrol study compared 336 patients who had gallstones or had undergone cholecystectomy to 336 controls (44,52). Diabetes was more prevalent in the patients with gallbladder disease (11.6 versus 4.8%). In another casecontrol study, an increased prevalence of gallstones in diabetes could only be demonstrated in women (42 versus 26% in nondiabetic women) (53). Why diabetes predisposes to gallstones is not well understood. Gallbladder hypomotility is common in diabetics and is seen as a response of smooth muscle to hyperglycemia; it can also be caused by autonomic neuropathy (54). I— Serum Lipids The precise role of serum lipids on gallstone formation is not known. Gallstones appear to be positively associated with apolipoprotein E4 phenotype and elevated serum triglycerides (55,56). In contrast, a negative association exists between gallstones and HDL. There is no conclusive evidence linking elevated serum cholesterol and gallstones. J— Gallbladder Stasis Conditions that result in bile stasis are associated with a higher prevalence of gallstones. In the normal state, the gallbladder avidly absorbs water from bile. If bile remains within the gallbladder for a prolonged period of time, it can become overly concentrated with cholesterol, which in turn promotes stone formation. Common examples of this mechanism include spinal cord injuries, prolonged fasting, and the use of total parenteral nutrition (57–59). A much rarer cause of stasisinduced gallstones are somatostatinomas. A somatostatinoma is a neuroendocrine tumor that secretes somatostatin, a potent inhibitor of gallbladder emptying. By the same mechanism, the use of octreotide, a somatostatin analog, has also been associated with gallstone formation (60).
Page 133
K— Other Drugs A number of widely used drugs can promote gallstone and biliary sludge formation. Clofibrate, a cholesterollowering agent, is strongly associated with gallstones. Clofibrate reduces bile acid secretion by inhibiting the ratelimiting enzyme in bile acid synthesis, cholesterol 7 hydroxylase. This results in cholesterol supersaturated bile and stone precipitation (61). Ceftriaxone, a the thirdgeneration cephalosporin, is a major cause of biliary sludge formation in hospitalized patients. Bile excretion accounts for up to 40% of ceftriaxone elimination and in bile; drug concentrations can reach 200 times that of the serum (62). When supersaturated, ceftriaxone complexes with calcium and precipitates out of bile. This process is probably potentiated in intensive care unit patients who are not fed enterally and have bile stasis. III— Cholesterol Gallstone Pathogenesis—An Overview There are three predominant types of gallstones: cholesterol, brown pigment, and black pigment stones. Pigment stones are addressed later in the chapter by Solloway; in this chapter the focus is on cholesterol gallstones. Cholesterol gallstones can contain from 50 to 99% cholesterol and are the commonest type of stones in countries where a westernstyle diet, rich in fat, is ingested. In the United States, they account for 80% of gallstones, and the risk factors for their development are given in the first section of this chapter. In the last few decades, critical advances have occurred in our understanding of how gallstones form; we will examine an overview of gallstone formation in this chapter. Many of the following chapters focus on the specific defects associated with gallstone formation, and this chapter will serve more as an overview of the problem, with more specific details found in the following chapters. Cholesterol gallstone formation can most simply be described as a failure of the constituents of bile to maintain cholesterol in the solubilized form, leading to a precipitation of cholesterol crystals and then growth to form gallstones. However, multiple pathophysiological mechanisms are involved in gallstone formation, including supersaturation of bile with cholesterol, as well as alteration of mucosal and motor function. The prolithogenic state is best characterized by excess cholesterol in bile, a more rapid nucleation of bile, an alteration of biliary mucin and proteins, and finally a loss of normal gallbladder contractility (63–67). These events lead to the formation initially of cholesterol crystals and then cholesterol gallstones, a process depicted in Fig. 1. Each of these events appears critical for gallstone formation; in animal models and humans, manipulation of one of these components can lead to the prevention of gallstones (50,68–71). Thus gallstone formation is the culmination of a number of distinct but closely related pathophysiological pathways, which are described in this and subsequent chapters. A— Cholesterol Metabolism Cholesterol homeostasis is closely regulated by the liver in humans and biliary cholesterol secretion remains as one of the critical pathways by which one is able to excrete excess cholesterol. The three major sources of hepatic cholesterol are dietary intake, plasmaderived cholesterol transported from the tissues as lowdensity lipoprotein (LDL), highdensity lipoprotein (HDL), and very low density lipoprotein (VLDL) as well as newly synthesized cholesterol within the hepatocyte (Fig. 2). The hepatocyte must process approximately 1800 mg of cholesterol daily, and it does this through three pathways: (1) esterification of cholesterol into lipoproteins and secretion back into plasma; (2) synthesis of bile salts; and (3) excretion of free cholesterol into bile. Biliary excretion of cholesterol accounts for more than 50% of the metabolism of free cholesterol in the hepatocyte. In situations where there is excess hepatocyte cholesterol, this excretory pathway becomes even more important but results in the formation of a cholesterol supersaturated bile. Robins has shown that most biliary cholesterol
Page 134
Figure 1 Venn diagram showing triple defect necessary for gallstone formation. Gallstones form in the mucin gel layer in the gallbladder.
Figure 2 Schematic of cholesterol metabolism in the hepatocyte. (BSEP, bile salt export pump; MDR2, phospholipid flipase.)
Page 135
is derived from HDL uptake (72), which is mediated by scavenger receptors such as SRBI (73). Cholesterol synthesis accounts for little of that excreted into bile, and cholesterol derived from LDL is predominantly metabolized into bile salts (Fig. 2). Since cholesterol is insoluble in water, the cosecretion of the other lipid components of bile, such as bile salts and phospholipids, are critical for maintaining cholesterol in a soluble state. B— Biliary Lipids and the Physical Chemistry of Bile Bile is formed at the hepatocytecanalicular membrane in the liver, modified as it passes through the larger bile ducts, and stored in the gallbladder, where it is further modified by the gallbladder mucosa. Bile contains water, electrolytes, hepatocyte and biliary canalicular proteins, and lipids (Table 3). The three major lipid moieties in bile are the primary and secondary bile acids, phosphatidylcholine (lecithin), and cholesterol. These molecules are depicted graphically in Fig. 3 according to their hydrophobic and hydrophilic characteristics. The relative concentration of the three lipid components of bile can be plotted as a ternaryphase diagram and predicts the physicochemical characteristics of the lipid carriers in bile (Fig. 4) (74,75). In unsaturated bile, the cholesterol saturation index (CSI) < 1.0. Cholesterol is carried as mixed micelles; it is solubilized by lecithin and bile salts in the gallbladder. As the CSI rises above 1.0 and bile is supersaturated by cholesterol, the threshold for the solubilization of cholesterol as mixed micelles is reached and cholesterol is now present in vesicles (76–79). Vesicles are bilayer structures composed of cholesterol and phospholipids, with small concentrations of bile salts; they are thermodynamically unstable (80). Fusion of vesicles can result in giant multilamellar vesicles or liquid crystals, which appear as Maltese crosses on polarized light microscopy and from which cholesterol monohydrate crystals can nucleate (81). In humans, bile is frequently supersaturated with cholesterol; but not all persons Table 3 Constituents of Human Canalicular Bile Bile salts, 12 g/L
Cholates, 35%
Glycine conjugates, 75%
Chenodeoxycholates, 35%
Taurine conjugates, 24.8%
Deoxycholates, 25%
Free bile acids, 0.2%
Lithocholates, 1% Miscellaneous, 4%
Phospholipids, 5 g/L
Phosphatidylcholine, 96% Phosphatidyl ethanolamine, 3%
Cholesterol, 1 g/L
Free, unesterified, 99%
Bilirubin, 0.2 g/L
Diglucuronide, 80% Monoglucuronide, 18% Unconjugated, 2%
Proteins, 2 g/L
Albumin, 50% Immunoglobulins, 23% CBP/APF Serum proteins, 9% Canalicular proteins, 1%
Electrolytes
Sodium, 150 meq/L Magnesium, 2 meq/L Calcium, 3 meq/L Potassium, 5 meq/L Chloride, 110 meq/L Bicarbonate, 30 meq/L
Key: CBP, calcium binding protein; APF, anionic polypeptide fraction.
Page 136
Figure 3 Cartoon depicting the hydrophobic and hydrophilic structure of the lipid constituents of bile.
Figure 4 Phase diagram for a physiological mixture of mixed bile salts, lecithin, and cholesterol. The zones representing cholesterol in 1, 2 and 3 phases are demarcated by solid lines. Distinct pathways of nucleation from liquid crystals through intermediate transition forms are shown by the zones marked A to E. (Adapted from Refs. 74, 75, and 129.)
Page 137
with supersaturated bile develop gallstones. The degree of cholesterol supersaturation of bile has been shown by several investigators to be the most important determinant of gallstone formation (82). Thus, cholesterol supersaturation is a sine qua non for gallstone formation, but other pathophysiological defects in nucleation and motility are also required. C— Biliary Lipid Secretion and Cholesterol Supersaturation Cholesterol metabolism is closely regulated by the liver, and excess dietary cholesterol is predominantly excreted as free cholesterol in bile. Therefore, western diets high in cholesterol lead to biliary supersaturation. In certain populations, such as Pima Indians and in Chileans, an unexplained genetic defect results in very high cholesterol supersaturation and a predilection for gallstone development. However, since the saturation of cholesterol is also dependent on the relative concentrations of bile salts and lecithin, secretion of these lipids into bile is critical. The major pathways for both lecithin and bile acid secretion have been elucidated. Bile salts are synthesized in the hepatocyte and undergo an enterohepatic circulation, with reabsorbtion through ileal bile acid receptors in the terminal ileum. Approximately 300 mg of cholesterol is utilized for bile acid synthesis daily via two pathways involving the enzymes 7 hydroxylase and sterol27hydroxylase (83). Bile salt secretion across the canalicular membrane into the canalicular space is an active process involving the newly recognized ATPdependent bile salt export pump (BSEP) (84). BSEP is a canalicular protein and was previously known as the sister of pglycoprotein. Lecithin is transported to the inner leaflet of the canalicular membrane by a specific phosphatidylcholine (PC) transport protein that is highly selective for PC species of lecithin (85). Once at the inner leaflet, PC is transferred into bile by a protein flippase, multidrug resistance 2 protein (MDR2) (86). In knockout mice, the absence of this protein leads to complete loss of both cholesterol and phospholipids in bile (87). The secretion of both bile salt and PC appears to be closely linked to cholesterol secretion into bile. Elegant experiments using rapid cryofixation electron microscopy have demonstrated vesicular budding of cholesterol from the outer leaflet of the canalicular membrane into bile in association with bile acid secretion (88). Rapid solubilization of these cholesterol vesicles by the high bile salt concentration in the canalicular space results in the formation of mixed micelles. To date no specific cholesterol transporter has been described; cholesterol secretion may be predominantly due to bile saltinduced rapid fluxes across the canalicular membrane. Thus, the physical chemistry of biliary lipids and their secretion into bile act as the background upon which gallstone formation occurs. Gallstones will not occur without cholesterol supersaturation, and this is probably the most important criterion for stone development. However, in the rest of this chapter, we explain why other processes are so important for stone formation. D— Role of the Gallbladder in Gallstone Formation The gallbladder is critical for cholesterol gallstone formation, since nearly all stones are formed in this organ. The gallbladder mucosa has certain critical functions that are necessary to protect the mucosa from injury due to the high bile salt concentrations (up to 300 mM) seen in humans. The gallbladder mucosa is a simple epithelial cell lining and is able to secrete mucins and hydrogen ions and to absorb water (89). The secretion of mucin is critical for the prevention of autodigestion of the gallbladder, since these mucins are highly negatively charged and exert important GibbsDonnan forces to protect the mucosa from bile salts. Acidification of bile is through an active Na+/H+ transporter and the acidification is necessary to prevent calcium salt precipitation in bile (90,91). In gallstone formation, both of these processes are impaired, resulting in excess mucin secretion, which is prolithogenic and increased free calcium with calcium salt precipitation onto gallstones. These issues are dealt with in detail in the sections on gallbladder mucin and biomineralization.
Page 138
Water absorption by the gallbladder is important for the concentration of bile and effective storage of bile salts in the gallbladder in the interprandial state. Concentration of bile salts can rise to as high as 300 mM in bile and there is an associated concentration of total lipids from 3 g/dL in hepatic bile to as high as 10 g/dL in gallbladder bile. The high lipid concentration is critical for the nucleation of cholesterol monohydrate crystals and explains why crystallization occurs in the gallbladder rather than in hepatic bile. Lipid absorption by the gallbladder mucosa results in preferential absorption of PC versus cholesterol, especially from vesicles. This leads to a very high ratio of cholesterol to PC in vesicles (2:1) and results in vesicles becoming thermodynamically more unstable. Unilamellar vesicles fuse to become multilamellar vesicles enriched in cholesterol, and these vesicles contain domains from which nucleation of solid cholesterol monohydrate crystals can occur. Thus the gallbladder is critical for modification of biliary lipids, leading to a cholesterolsupersaturated unstable bile that is susceptible to nucleation. E— Nucleation of Cholesterol Monohydrate Crystals We have reviewed the events that lead up to the development of a bile that is able to nucleate cholesterol crystals. However, many normal persons who never develop crystals or gallstones have a similar lipid profile in the gallbladder with strongly cholesterolsupersaturated bile. Holzbach demonstrated, in a critical study examining biles of similar cholesterol saturation from patients with and without gallstones, that biles from gallstoneforming patients had a more rapid nucleation (usually <3 days) of cholesterol crystals in vitro (92–95). This time to the appearance of crystals is known as the nucleation time or crystal appearance time and is an important cofactor in gallstone formation. Cholesterol nucleates from bile by a process of heterogeneous nucleation, where some nidus promotes initial crystallization. Our current understanding of nucleation is that bile contains a balance of both pro and antinucleating factors and that an imbalance in these factors results in the more rapid nucleation seen in gallstone biles. Despite intensive investigation, there has been little consensus on the exact nature of pronucleating agents (96,97). Certainly both lipid and protein nucleating agents have been described; in every case their exact role is unclear. This issue is discussed further in Chapter 11. Several proteins have been well characterized as nucleating agents; these are listed in Table 4 (98–109). The mechanism by which proteins act as nucleating agents is unclear. Some authors have suggested that the total protein content and the hydrophilichydrophobic content of proteins (110) are the critical determinants of nucleation, whereas others have shown differences in proteins from patients with multiple gallstones versus those with solitary stones (111,112). Gallbladder mucin is a potent nucleating agent and appears to work via the promotion of vesicular aggregation and fusion to form multilamellae and specific domains of mucin have been shown to be critical for its pronucleating properties (81,103). Further studies on other proteins to examine how they interact with lipids in bile should add to our knowledge of the mechanism of nucleation. In addition, antinucleating factors have been described; these act to stabilize cholesterolcontaining vesicles and to retard vesicle fusion and nucleation (93,113,114). The bestdescribed include apolipoprotein AI and AII, and these have been shown to be effective in vivo and in vitro (115–117). A potent and effective orally delivered antinucleating agent would have an important role in prevention of gallstones. Alteration of the lipid content of bile can also affect the nucleation of cholesterol. Nucleation is more rapid in the presence of higher concentrations of deoxycholate, which affect vesicle membrane stability. The more hydrophobic bile acids destabilize membranes when compared to hydrophilic bile salts such as taurodeoxycholate or ursodeoxycholate. In the case of deoxycholate, these findings are important, since levels of deoxycholate in gallbladder bile are increased during cholelithiasis (118). Deoxycholate is formed by bacterial degradation of bile acids and reuptake from the bowel (119). In situations of cholesterol excess, there may also be abnormal motility of the intestine, with a prolongation of transit time. This may lead
Page 139 Table 4 Nucleating Factors Present in Bile That May Inhibit or Promote Nucleation of Cholesterol Monohydrate Crystals Pronucleators
Antinucleators Proteins
Gallbladder mucin
Apolipoproteins AI and AII
Immunoglobulins M and G
128kDa Helix pomatia binding dimer
Aminopeptidase N
28kDa IgA fragment
Alpha1acid glycoprotein
Phospholipase c
Haptoglobin
Fibronectin
Nonprotein Factors Total lipid conc. >5 g/dL
Total lipid conc. <3 g/dL
High vesicular cholesterol: phospholipid Low vesicular cholesterol: phospholipid ratio ratio High calcium
Low calcium
Increased secondary hydrophobic bile acids
Increased hydrophilic bile acids
Polyunsaturated PL species
Saturated PL species
High CSI
Low CSI
Key: PL, phospholipid; CSI, cholesterol saturation index. Source: Adapted from Ref. 129.
to increased levels of deoxycholate in bile and these findings have been demonstrated by Dowling (see Chap. 4) in patients with sandostatininduced gallstones and those with constipation (60). Alterations to the length (increasing chain length to C16–C20) and hydrophobicity of the fatty acids in the Sn1 and Sn2 position of lecithin can also effect nucleation in vitro but have little relevance in vivo, where PC is overwhelmingly the dominant form of phospholipid. However, if one were able to derive a bile acid conjugated to longchain fatty acid, as has been shown by Gilat and Konikoff (personal communication), nucleation could be retarded and the lipids could enter bile through the enterohepatic circulation. F— Gallbladder Motility Several chapters in this book are devoted to the normal and abnormal motility of the gallbladder, particularly with reference to gallstone formation. In the preceding sections, we have shown that cholesterol crystals can be formed in the gallbladder, but these are of a size that would easily be ejected through the cystic duct by normal gallbladder ejection. However, simultaneously with the defects in lipid secretion and gallbladder mucosal function, there is also a defect in motility. Current theories suggest that the smooth muscle is also saturated with cholesterol, leading to an uncoupling of the contractile mechanism and decreased muscle contraction (120–124). There may also be a defect in the intrinsic nervous control of gallbladder motility. The resulting effect is the retention by the gallbladder of a gel phase— consisting of mucus, cholesterol crystals and microliths, calcium bilirubinate granules—which is described as biliary sludge (125–127) (see Chap. 21). Biliary sludge is not only a precursor of gallstones but is
Page 140
also able to provoke clinical symptoms such as pancreatitis, as this particulate matter can escape into the common bile duct and lead to inflammation of the pancreas. G— Gallstone Prevention As our understanding of gallstone formation has progressed, the ability to start considering prevention of gallstones is now a very real consideration. Primary prevention with exercise, avoidance of highcholesterol foods, and constipation are simple lifestyle measures that can be undertaken. Therapies to increase BSEP delivery of bile acids or to stimulate bile acid synthesis are becoming a reality. Alteration of nucleation with bile salt therapy or mucolytic agents may be possible in the future. Finally, drugs to stimulate contraction of the gallbladder such as CCK analogs or motilin agonists, can be used to prevent sludge formation. These exciting opportunities in basic and clinical research may make gallstone disease a medically treatable disorder and obviate the need for so many cholecystectomies. IV— Summary In this chapter we have discussed the epidemiology and multifactorial pathogenesis of gallstone formation and laid the groundwork for the following chapters, which discuss the specific mechanisms in much greater detail. The future for gallstone research looks exciting and the prevention of gallstone disease may soon become a reality in clinical medicine. References 1. Steiner CA, Bass EB, Talamini MA, Pitt HA, Steinberg EP. Surgical rates and operative mortality for open and laparoscopic cholecystectomy in Maryland (see comments). N Engl J Med 1994;330(6):403–408. 2. Zahor A, Sternby NH, Kagan A, Uemura K, Vanecek R, Vichert AM. Frequency of cholelithiasis in Prague and Malmo: an autopsy study. Scand J Gastroenterol 1974;9(1):3–7. 3. Lindstrom CG. Frequency of gallstone disease in a welldefined Swedish population: a prospective necropsy study in Malmo. Scand J Gastroenterol 1977;12 (3):341–346. 4. Brett M, Barker DJ. The world distribution of gallstones. Int J Epidemiol 1976;5(4):335–341. 5. Diehl AK, Schwesinger WH, Holleman DR, Jr, Chapman JB, Kurtin WE. Gallstone characteristics in Mexican Americans and nonHispanic whites (see comments). Dig Dis Sci 1994;39(10):2223–2228. 6. Heaton KW, Braddon FE, Mountford RA, Hughes AO, Emmett PM. Symptomatic and silent gall stones in the community. Gut 1991;32(3):316–320. 7. Maurer KR, Everhart JE, Ezzati TM, et al. Prevalence of gallstone disease in Hispanic populations in the United States [published erratum appears in Gastroenterology 1989 Jun;96(6):1630]. Gastroenterology 1989;96(2 Pt 1):487–492. 8. Maurer KR, Everhart JE, Knowler WC, Shawker TH, Roth HP. Risk factors for gallstone disease in the Hispanic populations of the United States. Am J Epidemiol 1990;131(5):836–844. 9. Sampliner RE, Bennett PH, Comess LJ, Rose FA, Burch TA. Gallbladder disease in Pima Indians: demonstration of high prevalence and early onset by cholecystography. N Engl J Med 1970;283(25):1358–1364. 10. Williams CN, Johnston JL, Weldon KL. Prevalence of gallstones and gallbladder disease in Canadian Micmac Indian women. Can Med Assoc J 1977;117 (7):758–760.
Page 141
11. Thistle JL, Schoenfield LJ. Lithogenic bile among young Indian women. N Engl J Med 1971;284(4):177–181. 12. Williams CN, Johnston JL. Prevalence of gallstones and risk factors in Caucasian women in a rural Canadian community. Can Med Assoc J 1980;122(6):664– 668. 13. Attili AF, Pazzi P, Galeazzi R. Prevalence of previously undiagnosed gallstones in a population with multiple risk factors. Dig Dis Sci 1995;40(8):1770–1774. 14. Attili AF CN, Roda N et al. Epidemiology of gallstone disease in Italy; prevalence data of the multicenter Italian study on cholelithiasis (MICOL). Am J Epidemiol 1995;141: 158. 15. Barbara L SC, MorselliLabate AM, et al. A ten year incidence of gallstone disease: the Sirmione study. J Hepatol. 1993;18:43. 16. Jensen KH, Jorgensen T. Incidence of gallstones in a Danish population. Gastroenterology 1991;100(3):790–794. 17. Sichieri R, Everhart JE, Roth H. A prospective study of hospitalization with gallstone disease among women: role of dietary factors, fasting period, and dieting. Am J Public Health 1991;81(7):880–884. 18. Waldhausen JH, Benjamin DR. Cholecystectomy is becoming an increasingly common operation in children. Am J Surg 1999;177(5):364–367. 19. Angelico F, Del Ben M, Barbato A, Conti R, Urbinati G. Tenyear incidence and natural history of gallstone disease in a rural population of women in central Italy: The Rome Group for the Epidemiology and Prevention of Cholelithiasis (GREPCO). Ital J Gastroenterol Hepatol 1997;29(3):249–254. 20. Attili AF, De Santis A, Capri R, Repice AM, Maselli S. The natural history of gallstones: the GREPCO experience. The GREPCO Group. Hepatology 1995;21 (3):655–660. 21. Valdiviseo V CC, Siegel F, et al. Pregnancy and cholelithiasis: pathogenesis and natural course of gallstones diagnosed in early puerperium. Hepatology 1993;17:1. 22. Maringhini A, Marceno MP, Lanzarone F, et al. Sludge and stones in gallbladder after pregnancy: prevalence and risk factors. J Hepatol 1987;5(2):218–223. 23. Maringhini A, Ciambra M, Baccelliere P, et al. Biliary sludge and gallstones in pregnancy: incidence, risk factors, and natural history. Ann Intern Med 1993;119 (2):116–120. 24. Everson GT, McKinley C, Kern F Jr. Mechanisms of gallstone formation in women: effects of exogenous estrogen (Premarin) and dietary cholesterol on hepatic lipid metabolism. J Clin Invest 1991;87(1):237–246. 25. Kern F Jr., Everson GT, DeMark B, et al. Biliary lipids, bile acids, and gallbladder function in the human female: effects of pregnancy and the ovulatory cycle. J Clin Invest 1981;68(5):1229–1242. 26. Grodstein F, Colditz GA, Hunter DJ, Manson JE, Willett WC, Stampfer MJ. A prospective study of symptomatic gallstones in women: relation with oral contraceptives and other risk factors. Obstet Gynecol 1994;84(2):207–214. 27. Misciagna G, Leoci C, Guerra V, et al. Epidemiology of cholelithiasis in southern Italy: Part II: Risk factors. Eur J Gastroenterol Hepatol 1996;8(6):585–593. 28. Scragg RK, McMichael AJ, Seamark RF. Oral contraceptives, pregnancy, and endogenous oestrogen in gall stone disease—a casecontrol study. Br Med J (Clin Res Ed) 1984;288(6433):1795–1799. 29. Everhart JE, Khare M, Hill M, Maurer KR. Prevalence and ethnic differences in gallbladder disease in the United States. Gastroenterology 1999;117(3):632– 639. 30. Group CDPR. Gallbladder disease as a side effect of drugs influencing lipid metabolism: experience in the coronary drug project. N Engl J Med 1977;296:1185. 31. Hendricksson P EK, Eriksson A, et al. strogen induced gallstone formation in males. relation to changes in serum and biliary lipids during hormonal treatment of prostatic carcinoma. J Clin Invest 1989;84:811. 32. Thijs C, Leffers P, Knipschild P. Oral contraceptive use and the occurrence of gallstone disease—a casecontrol study. Prev Med 1993;22(1):122–131.
Page 142
33. Thijs C KP. Oral contraceptives and the risk of gallbladder disease: a metaanalysis. Am J Public Health 1993;22:122. 34. Gilat T, Feldman C, Halpern Z, Dan M, BarMeir S. An increased familial frequency of gallstones. Gastroenterology 1983;84(2):242–246. 35. Sarin SK, Negi VS, Dewan R, Sasan S, Saraya A. High familial prevalence of gallstones in the firstdegree relatives of gallstone patients (see comments). Hepatology 1995;22(1): 138–141. 36. Khanuja B, Cheah YC, Hunt M, et al. Lithl, a major gene affecting cholesterol gallstone formation among inbred strains of mice. Proc Natl Acad Sci USA 1995;92(17):7729–7733. 37. Acalovschi MV, Blendea D, Pascu M, Georoceanu A, Badea RI, Prelipceanu M. Risk of asymptomatic and symptomatic gallstones in moderately obese women: a longitudinal followup study. Am J Gastroenterol 1997;92(1):127–131. 38. Everhart JE. Contributions of obesity and weight loss to gallstone disease. Ann Intern Med 1993;119(10):1029–1035. 39. Friedman GD, Kannel WB, Dawber TR. The epidemiology of gallbladder disease: observations in the Framingham Study. J Chronic Dis 1966;19(3):273–292. 40. Shiffman ML, Sugerman HJ, Kellum JH, Brewer WH, Moore EW. Gallstones in patients with morbid obesity: relationship to body weight, weight loss and gallbladder bile cholesterol solubility. Int J Obesity Rel Metab Disord 1993;17(3):153–158. 41. Stampfer MJ, Maclure KM, Colditz GA, Manson JE, Willett WC. Risk of symptomatic gallstones in women with severe obesity. Am J Clin Nutr 1992;55 (3):652–658. 42. Amaral JF TW. Gallbladder disease in the morbidly obese. Am J Surg 1985;149:551. 43. Shiffman ML, Sugerman HJ, Kellum JM, Brewer WH, Moore EW. Gallstone formation after rapid weight loss: a prospective study in patients undergoing gastric bypass surgery for treatment of morbid obesity. Am J Gastroenterol 1991;86(8):1000–1005. 44. Shiffman ML, Shamburek RD, Schwartz CC, Sugerman HJ, Kellum JM, Moore EW. Gallbladder mucin, arachidonic acid, and bile lipids in patients who develop gallstones during weight reduction. Gastroenterology 1993;105(4):1200–1208. 45. Shiffman ML, Kaplan GD, BrinkmanKaplan V, Vickers FF. Prophylaxis against gallstone formation with ursodeoxycholic acid in patients participating in a very lowcalorie diet program [see comments]. Ann Intern Med 1995;122(12):899–905. 46. Andersen T. Liver and gallbladder disease before and after verylowcalorie diets. Am J Clin Nutr 1992;56(1 Suppl):235S–239S. 47. Marks JW, Bonorris GG, Albers G, Schoenfield LJ. The sequence of biliary events preceding the formation of gallstones in humans. Gastroenterology 1992;103 (2):566–570. 48. Yang H, Petersen GM, Roth MP, Schoenfield LJ, Marks JW. Risk factors for gallstone formation during rapid loss of weight. Dig Dis Sci 1992;37(6):912–918. 49. Gebhard RL, Prigge WF, Ansel HJ, et al. The role of gallbladder emptying in gallstone formation during dietinduced rapid weight loss. Hepatology 1996;24 (3):544–548. 50. Marks JW, Bonorris GG, Schoenfield LJ. Effects of ursodiol or ibuprofen on contraction of gallbladder and bile among obese patients during weight loss. Dig Dis Sci 1996; 41(2):242–249. 50a. Broomfield PH, Chopra R, Sheinbaum RC, Bonorris GG, Silverman A, Schoenfield LJ, Marks JW. Effects of ursodexycholic acid and aspirin on the formation of lithogenic bile and gallstones during loss of weight. N Engl J Med 1988;319(24):1567–1572. 51. Sasazuki S, Kono S, Todoroki I, et al. Impaired glucose tolerance, diabetes mellitus, and gallstone disease: an extended study of male selfdefense officials in Japan. Eur J Epidemiol 1999;15(3):245–251. 52. De Santis A, Attili AF, Ginanni Corradini S, et al. Gallstones and diabetes: a casecontrol study in a freeliving population sample. Hepatology 1997;25(4):787– 790. 53. Chapman BA, Wilson IR, Frampton CM, et al. Prevalence of gallbladder disease in diabetes mellitus. Dig Dis Sci 1996;41(11):2222–2228.
Page 143
54. Hahm JS, Park JY, Park KG, Ahn YH, Lee MH, Park KN. Gallbladder motility in diabetes mellitus using real time ultrasonography. Am J Gastroenterol 1996;91 (11): 2391–2394. 55. Scragg RK, Calvert GD, Oliver JR. Plasma lipids and insulin in gallstone disease: a casecontrol study. Br Med J (Clin Res Ed) 1984;289(6444):521–525. 56. Kern F, Jr. Effects of dietary cholesterol on cholesterol and bile acid homeostasis in patients with cholesterol gallstones. J Clin Invest 1994;93(3):1186–1194. 57. Apstein MD, DaleckiChipperfield K. Spinal cord injury is a risk factor for gallstone disease. Gastroenterology 1987;92(4):966–968. 58. Moonka R, Stiens SA, Eubank WB, Stelzner M. The presentation of gallstones and results of biliary surgery in a spinal cord injured population. Am J Surg 1999;178(3): 246–250. 59. Moonka R, Stiens SA, Resnick WJ, et al. The prevalence and natural history of gallstones in spinal cord injured patients. J Am Coll Surg 1999;189(3):274–281. 60. Hussaini SH, Pereira SP, Veysey MJ, et al. Roles of gall bladder emptying and intestinal transit in the pathogenesis of octreotide induced gall bladder stones. Gut 1996;38(5): 775–783. 61. Stahlberg D RE, Rudling M et al. Influence of benzfibrate on hepatic cholesterol metabolism in gallstone patients: reduced activity of cholesterol 7 hydroxylase. Hepatology 1995;21:1025. 62. Shiffman ML, Keith FB, Moore EW. Pathogenesis of ceftriaxoneassociated biliary sludge. In vitro studies of calciumceftriaxone binding and solubility. Gastroenterology 1990;99(6):1772–1778. 63. Afdhal N, Smith B. Current concepts on the pathogenesis of cholesterol gallstones. Views Dig Dis 1990;22:13–18. 64. Apstein MD, Carey MC. Pathogenesis of cholesterol gallstones: a parsimonious hypothesis. Eur J Clin Invest 1996;26(5):343–352. 65. Carey MC, Lamont JT. Cholesterol gallstone formation. 1. Physicalchemistry of bile and biliary lipid secretion. Progr Liver Dis 1992;10:139–163. 66. Donovan JM. Physical and metabolic factors in gallstone pathogenesis. Gastroenterol Clin North Am 1999;28(1):75–97. 67. Lamont JT, Carey MC. Cholesterol gallstone formation: 2. Pathobiology and pathomechanics. Progr Liver Dis 1992;10:165–191. 68. Lee SP, Carey MC, LaMont JT. Aspirin prevention of cholesterol gallstone formation in prairie dogs. Science 1981;211(4489):1429–1431. 69. Lee SP. Lessons from experimental cholelithiasis: gallbladder and mucosa, nonsteroidal antiinflammatory drugs, and gallstones (editorial; comment). Gastroenterology 1991; 101(3):857–860. 70. Levenson DE, Fromm H. 10 g of fat per day may keep gallstones away. Gastroenterology 1993;104(6):1880–1881. 71. Xu QW, Shaffer EA. Cisapride improves gallbladder contractility and bile lipid composition in an animal model of gallstone disease. Gastroenterology 1993;105 (4):1184–1191. 72. Robins SJ FJ. High density lipoproteins, but not other lipoproteins, provide a vehicle for sterol transport to bile. J Clin Invest 1997;99:380–384. 73. Acton S RA, Landschulz KT, Xu S, Hobbs HH, Krieger M. Identification of a scavenger receptor SRB1 as a high density lipoprotein receptor. Science 1996;271:518–520. 74. Wang DQ, Carey MC. Characterization of crystallization pathways during cholesterol precipitation from human gallbladder biles: identical pathways to corresponding model biles with three predominating sequences. J Lipid Res 1996;37(12):2539–2549. 75. Wang DQ, Carey MC. Complete mapping of crystallization pathways during cholesterol precipitation from model bile: influence of physicalchemical variables of pathophysiologic relevance and identification of a stable liquid crystalline state in cold, dilute and hydrophilic bile saltcontaining systems. J Lipid Res 1996;37(3):606– 630.
Page 144
76. Cohen DE, Angelico M, Carey MC. Quasielastic light scattering evidence for vesicular secretion of biliary lipids. Am J Physiol 1989;257(1 Pt 1):G1–G8. 77. Donovan JM, Carey MC. Separation and quantitation of cholesterol ''carriers" in bile. Hepatology 1990;12(3 Pt 2):94S–104S; discussion 104S–105S. 78. Eckhardt ER, van de Heijning BJ, van Erpecum KJ, Renooij W, VanBergeHenegouwen GP. Quantitation of cholesterolcarrying particles in human gallbladder bile. J Lipid Res 1998;39(3):594–603. 79. Gantz DL, Wang DQ, Carey MC, Small DM. Cryoelectron microscopy of a nucleating model bile in vitreous ice: formation of primordial vesicles. Biophys J 1999;76(3):1436–1451. 80. Somjen GJ, Marikovsky Y, Lelkes P, Gilat T. Cholesterolphospholipid vesicles in human bile: an ultrastructural study. Biochim Biophys Acta 1986;879(1):14– 21. 81. Afdhal NH, Niu N, Nunes DP, et al. Mucinvesicle interactions in model bile: evidence for vesicle aggregation and fusion before cholesterol crystal formation. Hepatology 1995;22(3):856–865. 82. Miquel JF, Nunez L, Amigo L, et al. Cholesterol saturation, not proteins or cholecystitis, is critical for crystal formation in human gallbladder bile. Gastroenterology 1998;114(5): 1016–1023. 83. Javitt NB. Bile acid synthesis from cholesterol: regulation and auxiliary pathways. FASEB J 1994;8:1308–1311. 84. Gerloff T, Steiger B, Hagenbuch B, et al. The sister of pglycoprotein represents the canalicular bile salt export pump of mammalian liver. J Biol Chem 1998;173:10046–10050. 85. Cohen DE, Leonard MR, Carey MC. In vitro evidence that phospholipid secretion into bile may be coordinated intracellularly by the combined actions of bile salts and the specific phosphatidylcholine transfer protein of liver. Biochemistry 1994;33(33):9975–9980. 86. Smit JJ, Schinkel AH, Oude Elferink RP, et al. Homozygous disruption of the murine mdr2 Pglycoprotein gene leads to a complete absence of phospholipid from bile and to liver disease. Cell 1993;75(3):451–462. 87. Elferink R, Ottenhof R, Vanwijland M, Smit JJM, Schinkel AH, Groen AK. Regulation of biliary lipid secretion by the mdr2pglycoprotein in the mouse. J Clin Invest 1995; 95:31–38. 88. Crawford JM, Mockel GM, Crawford AR, et al. Imaging biliary lipid secretion in the rat: ultrastructural evidence for vesiculation of the hepatocyte canalicular membrane. J Lipid Res 1995;36(10):2147–2163. 89. Conter RL, Washington JL, Liao CC, Kauffman GL Jr. Gallbladder mucosal blood flow increases during early cholesterol gallstone formation (see comments). Gastroenterology 1992;102(5):1764–1770. 90. Rege RV, Moore EW. Evidence for H+ secretion by the in vivo canine gallbladder. Gastroenterology 1987;92(2):281–289. 91. Shiffman ML, Sugerman HJ, Moore EW. Human gallbladder mucosal function: effect of concentration and acidification of bile on cholesterol and calcium solubility. Gastroenterology 1990;99(5):1452–1459. 92. Holzbach RT. Factors influencing cholesterol nucleation in bile. Hepatology 1984;4(5 Suppl):173S–176S. 93. Holzbach RT. Metastability behavior of supersaturated bile. Hepatology 1984;4(5 Suppl):155S–158S. 94. Holzbach RT. Recent progress in understanding cholesterol crystal nucleation as a precursor to human gallstone formation. Hepatology 1986;6(6):1403–1406. 95. Holzbach RT, Kibe A, Thiel E, Howell JH, Marsh M, Hermann RE. Biliary proteins: unique inhibitors of cholesterol crystal nucleation in human gallbladder bile. J Clin Invest 1984;73(1):35–45.
Page 145
96. Afdhal NH, Smith BF. Cholesterol crystal nucleation: a decadelong search for the missing link in gallstone pathogenesis. Hepatology 1990;11(4):699–702. 97. Harvey PR, Strasberg SM. Will the real cholesterolnucleating and antinucleating proteins please stand up? (editorial; comment). Gastroenterology 1993;104 (2):646–650. 98. Abei M, Kawczak P, Nuutinen H, Langnas A, Svanvik J, Holzbach RT. Isolation and characterization of a cholesterol crystallization promoter from human bile (see comments). Gastroenterology 1993;104(2):539–548. 99. Harvey PR, Upadhya GA, Strasberg SM. Immunoglobulins as nucleating proteins in the gallbladder bile of patients with cholesterol gallstones. J Biol Chem 1991;266(21):3996–4003. 100. Lee TJ, Smith BF. Bovine gallbladder mucin promotes cholesterol crystal nucleation from cholesteroltransporting vesicles in supersaturated model bile. J Lipid Res 1989;30(4):491–498. 101. Luk AS, Kaler EW, Lee SP. Protein lipid interaction in bile: effects of biliary proteins on the stability of cholesterollecithin vesicles. Biochim Biophys Acta 1998;1390(3):282–292. 102. Miquel JF, Von Ritter C, Del Pozo R, Lange V, Jungst D, Paumgartner G. Fibronectin in human gallbladder bile: cholesterol pronucleating and/or mucin "link" protein? Am J Physiol 1994;267(3 Pt 1):G393–G400. 103. Nunes DP, Afdhal NH, Offner GD. A recombinant bovine gallbladder mucin polypeptide binds biliary lipids and accelerates cholesterol crystal appearance time. Gastroenterology 1999;116(4):936–942. 104. Nuutinen H, Corradini SG, Jungst D, et al. Correlation between biliary alpha 1acid glycoprotein concentration and cholesterol crystal nucleation time in gallstone disease. Dig Dis Sci 1995;40(6):1174–1178. 105. Nuutinen H, Abei M, Schwarzendrube J, et al. Biliary alpha 1acid glycoprotein concentrations in gallstonefree controls and in patients with multiple or solitary cholesterol gallstones. Dig Dis Sci 1995;40(8):1786–1791. 106. Offner GD, Gong D, Afdhal NH. Identification of a 130kilodalton human biliary concanavalin A binding protein as aminopeptidase N. Gastroenterology 1994;106(3):755–762. 107. Small DM. Cholesterol nucleation and growth in gallstone formation. N Engl J Med 1980;302:1305–1307. 108. Smith BF. Human gallbladder mucin binds biliary lipids and promotes cholesterol crystal nucleation in model bile. J Lipid Res 1987;28(9):1088–1097. 109. Smith BF. Gallbladder mucin as a pronucleating agent for cholesterol monohydrate crystals in bile. Hepatology 1990;12(3 Pt 2):183S–186S; discussion 186S– 188S. 110. Ahmed HA, Petroni ML, AbuHamdiyyah M, Jazrawi RP, Northfield TC. Hydrophobic/hydrophilic balance of proteins: a major determinant of cholesterol crystal formation in model bile. J Lipid Res 1994;35(2):211–219. 111. Van Erpecum KJ, Van BergeHenegouwen GP, Eckhardt ER, et al. Cholesterol crystallization in human gallbladder bile: relation to gallstone number, bile composition, and apolipoprotein E4 isoform. Hepatology 1998;27(6):1508–1516. 112. Jungst D, Lang T, von Ritter C, Pratschke E, Paumgartner G. Cholesterol nucleationtime in gallbladder bile of patients with solitary or multiple cholesterol gallstones. Hepatology 1992;15(5):804–808. 113. Busch N, Holzbach RT. Crystal growthinhibiting proteins in bile. Hepatology 1990;12(3 Pt 2):195S–198S; discussion 199S. 114. Busch N, Matiuck N, Sahlin S, Pillay SP, Holzbach RT. Inhibition and promotion of cholesterol crystallization by protein fractions from normal human gallbladder bile. J Lipid Res 1991;32(4):695–702. 115. Kibe A, Holzbach RT, LaRusso NF, Mao SJ. Inhibition of cholesterol crystal formationby apolipoproteins in supersaturated model bile. Science 1984;225 (4661):514–516.
Page 146
116. Secknus R, Yamashita G, Ginanni Corradini S, et al. Purification and characterization of a novel human 15 kd cholesterol crystallization inhibitor protein in bile. J Lab Clin Med 1996;127(2):169–178. 117. Tao S, Tazuma S, Kajiyama G. Apolipoprotein AI stabilizes phospholipid lamellae and thus prolongs nucleation time in model bile systems: an ultrastructural study. Biochim Biophys Acta 1993;1166(1):25–30. 118. Hay DW, Carey MC. Chemical species of lipids in bile. Hepatology 1990;12(3 Pt 2):6S–14S; discussion 14S–16S. 119. Marcus SN, Heaton KW. Deoxycholic acid and the pathogenesis of gallstones. Gut 1988;29(4):522–533. 120. Chen Q, Amaral J, Oh S, Biancani P, Behar J. Gallbladder relaxation in patients with pigment and cholesterol stones. Gastroenterology 1997;113(3):930–937. 121. Chen Q, Amaral J, Biancani P, Behar J. Excess membrane cholesterol alters human gallbladder muscle contractility and membrane fluidity. Gastroenterology 1999;116(3):678–685. 122. van de Heijning BJ, van de Meeberg PC, Portincasa P, et al. Effects of ursodeoxycholic acid therapy on in vitro gallbladder contractility in patients with cholesterol gallstones. Dig Dis Sci 1999;44(1):190–196. 123. Yu P, Harnett KM, Biancani P, De Petris G, Behar J. Interaction between signal transduction pathways contributing to gallbladder tonic contraction. Am J Physiol 1993;265(6 Pt 1):G1082–G1089. 124. Yu P, De Petris G, Biancani P, Amaral J, Behar J. Cholecystokinincoupled intracellular signaling in human gallbladder muscle. Gastroenterology 1994;106 (3):763–770. 125. Carey MC, Cahalane MJ. Whither biliary sludge? Gastroenterology 1988;95(2):508–523. 126. Ko CW, Sekijima JH, Lee SP. Biliary sludge. Ann Intern Med 1999;130(4 Pt 1):301–311. 127. Lee SP, Maher K, Nicholls JF. Origin and fate of biliary sludge. Gastroenterology 1988;94(1):170–176. 128. Everhart JE. Gallstones. In: Digestive Diseases in the United States: Epidemiology and Impact, 1994. NIH Publication No. 941447. Bethesda, MD: National Institutes of Health, 1994. 129. Afdhal NH, Carey MC. New Perspectives on gallstone pathogenesis and prevention. J Ir Coll Phys Surg 1995;24:267–278.
Page 147
7— Pigment Gallstones Roger D. Soloway and Nyingi M. Kemmer University of Texas Medical Branch, Galveston, Texas Jinguang Wu Peking University, Beijing, People's Republic of China I— Introduction Although the pathogenesis of cholesterol gallstones is increasingly well known, the pathogenesis of noncholesterol components in stones is varied and much less completely understood. Noncholesterol components in stones occur in three compositional and clinical settings: (a) as periodic, ringshaped crystallization of calcium salts on a protein scaffolding situated between the cholesterol crystal matrix of cholesterol gallstones; (b) as the crystallization of a variety of calcium salts in a protein matrix, forming black pigment stones in the gallbladder; and (c) as the precipitate of a supersaturated solution of the calcium salts of bilirubin and fatty acids forming brown pigment stones anywhere within the biliary tract (1–3). The epidemiology and pathogenesis of these three types of stones are mutually exclusive, meaning that only one type of stone will form at a given point in time. This exclusivity is reflected in their differing structures and compositions and is the subject of this chapter. Sludge, or microlithiasis, may be a stage in gallstone formation or may be a separate condition and is treated independently. II— Epidemiology A— General An examination of the geography of gallstone composition demonstrates striking differences in the operative frequency of gallstone types around the world. Although genetics plays an important role, infection, motility, and diet are also independent and important factors governing regional differences. Cholesterol stones account for 70 to 90% of gallstones in various areas in the United States (4) and usually contain a series of rings of calcium salts laid down on a protein scaffolding when examined in cross section (5). Black pigment stones make up the remainder of gallbladder stones in western countries and are composed of calcium salts and proteins with almost no cholesterol content. In the West, brown pigment stones occur predominantly as common duct stones developing more than 2 years after cholecystectomy (6), while in Asia, stones occur within the gallbladder but predominantly as pyogenic cholangitis and hepatolithiasis due to the formation of brown stones in the extrahepatic or intrahepatic bile ducts (7–13). The relationship of gallbladder sludge or microlithiasis to these stones is
Page 148
unclear, since less than 20% of patients with sludge later develop traditionally sized gallstones (14–17). B— Noncholesterol Content in Cholesterol Gallstones Studies of stone composition have revealed that the pigmented centers and rings of "mixed" cholesterol stones, the most common type of gallstone, contain primarily cholesterol (18). Cholesterol stones contain 70 to 100% cholesterol, and the crosssectional distribution of noncholesterol content on examination of individual stone sets appears to be unique for each patient (unpublished results). Studies comparing cholesterol content between cholesterol stones in different countries did not demonstrate striking average differences between series, although the range of proportions of cholesterol in stones in different operative series was quite wide (unpublished results). The mechanisms governing the formation and composition of the various components of cholesterol stones seem to be similar throughout the world. Although we did not recognize different crops of gallstones in a series of gallstones from Philadelphia, we did not consider the presence of sludge or microlithiasis in that series (4). In a more recent series from Germany, where this phenomenon was specifically examined, 29% of 310 sets of stones contained two distinct generations of stones (19). In Philadelphia (4), only 2 of more than 100 stone sets contained both a cholesterol and a distinct black pigment stone layer, while in Germany, 47% of gallstones contained a black or brown outer layer (19). It is unclear if this marked difference was due to the lack of differentiation by investigators between secondary deposition of noncholesterol components in a field of loosely packed cholesterol crystals or to the primary deposition of a noncholesterol layer of calcium salts and proteins (characteristic of black pigment stones) on the surface of a cholesterol stone. We and others have recently identified patients with several crops of stones in the same gallbladder (19–21) and others have identified patients with gallstones and sludge (19). Solitary and multiple gallstones demonstrate differences in response to bile acid dissolution treatment and in recurrence after dissolution (19,22). Pure cholesterol stones (socalled solitaires) represent about 20% of cholesterol stones in the United States (unpublished results) and 22.5% in Germany (70 of 310) (19). These stones contain no visible noncholesterol components (the most sensitive way to detect calcium bilirubinate). Mixed stones are typical of most patients with crops of stones (1). The various calcium carbonate polymorphs in cholesterol stones (23,24) differ in frequency between the United States, Bolivia, Mexico, and Italy (unpublished results), confirming the clinical impression from gallstone dissolution trials that gallstones from Europe more frequently become calcified or become calcified in the course of treatment monitoring. Calcite, aragonite, and vaterite were each identified in stones, but in different layers (23). Two studies found that carbonate was more prevalent in the periphery than in the center of cholesterol stones, suggesting that its deposition was more related to intermittent obstruction (23–25) than to stone nidation (26,27). Biliary obstruction is most frequently painful but may be tolerated to different extents in different cultures. Calcium hydroxyapatite, the usual form of calcium phosphate in stones, occurs less frequently than calcium carbonate. There may be regional environmental factors that influence stone composition. This could be due to differences in mineral content of the drinking water or in the predisposition of changes in the diet to increase the biliary secretion of calcium or carbonate and hydroxyapatite. Alternatively, if these salts are precipitated as a result of the activity of commensal biliary tract bacteria, as occurs for urinary stones or dental calculus (28), the average indigenous bacterial population may differ significantly between regions, and some bacteria in Europe may be more predisposed to calcium salt formation. New prospective and extensive regional studies are needed to provide additional information about the causation of differing regional patterns of cholesterol gallstone composition. Although there are virtually no black pigment stones formed in La Paz, Bolivia, or Mexico City, Mexico (29–30), the non
Page 149
cholesterol content of these stones was not significantly different from that of the stones studied in Philadelphia (4). C— Black Pigment Gallstones These stones are heterogeneous in composition but grouped as black because of the appearance of the metal salts of bilirubinate anionic copolymers after exposure to singlet oxygen (31,32). Calcium bilirubinate may vary from 10 to 90% of stone weight (4,33), and a variety of other calcium and metal salts of carbonate and hydroxyapatite are also present. These stones are associated primarily with old age (34,35) and to a much smaller extent with hemolysis (36,37), hyperalimentation (38), Crohn's disease (39,40) and cirrhosis (41–45). The incidence of these stones varies less strikingly by country of origin than for cholesterol stones when compared in operative series (4,34,46). In Galveston Texas, with a large Hispanic population, and in Rochester, Minnesota, with a large proportion of Scandinavian peoples, as well as in Scandinavia, the incidence of black pigment stones is 5% (3). In Japan, the incidence is 9%, with a significant incidence of brown pigment stones (35), a phenomenon not noted in western series. In the eastern United States the incidence is 30% (34); in India, the incidence is 40% (47); among Amerinds, the incidence is very low or nonexistent (29,30). Although the average age of these various populations differs, this is not enough to explain the differences. In the presence of hemolysis, as in sickle cell disease, all patients, in whom stones have been examined have had black pigment stones (32). This is also true for the majority of the small number of stones formed in patients receiving total parenteral nutrition (TPN) (38,48). However, since the large majority of stones and sludge forming in late pregnancy dissolve following delivery, with the return of the gallbladder and other muscles to normal levels of motility, the hypomotility of pregnancy probably predisposes to cholesterol rather than pigment gallstones. Those who have examined stone formation during hyperalimentation have demonstrated that hypomotility is crucial, since administration of a meal or of cholecystokinin octipeptide several times a week, resulting in gallbladder contraction, prevented stone formation even though TPN was continued (48). The mechanism by which advancing age causes an increase in the operative incidence of black pigment stones and a decrease in the incidence of cholesterol stones is unknown, but this has been observed repeatedly throughout the world when it was looked for (34,35). The most striking study to date, has been the study by Hikasa and associates, in which they fed 5weekold (adolescent) hamsters a cholesterol stoneproducing diet and 80% developed cholesterol stones. However, when they fed the same diet to 5monthold (middleaged) hamsters, only 60% developed cholesterol stones, while 20% formed black pigment stones (49). A number of questions have been left unanswered by this provocative study: (a) What is the effect of aging on causing the formation of black pigment stones? (b) What is special about the 20% of a homogenous strain that produce black pigment rather than cholesterol gallstones? (c) Why do 20% of this same strain not form stones at all at either age point selected? D— Brown Pigment Gallstones The frequency of primary brown pigment gallbladder stones varies from 0% in Bolivia (29,30), to 30% in Japan (35), to greater than 90% of operative series in rural regions in China (3) and it is thought to be related to the classic lowanimalprotein, lowcalorie, highvegetable diet of Asian peoples. In Asia, stones occurred primarily in the biliary tract and liver (50), associated with recurrent pyogenic cholangitis (51) or cholangiohepatitis (52). The operative frequency varies throughout Asia (53). Asian surgeons have become skillful in hepatic resection and duct reconstruction to reduce stone recurrence in areas of the liver damaged by infection. The strongest support for this hypothesis comes from Japan, where studies of gallstone incidence have been performed repeatedly over decades (7,8,43). With the advent of endoscopy, large clinics have been established to handle frequent removal of recurrent stones from patients with advanced disease. Prior to 1945, brown pigment stones accounted for more than 70% of stones
Page 150
removed at operation in Japan (7,8). Many of these stones were associated with stones concomitantly formed in the common bile duct or within the intrahepatic biliary system, sometimes in the absence of gallbladder stones. This pattern was typical throughout Asia and remains the pattern in rural China. By 1978, in a large Japanese study (35), brown pigment stones accounted for only 30% of operative series and were replaced by cholesterol stones as the predominant stone type found in Japan. This was thought to be due to the change in diet to a highercalorie, highprotein, highcarbohydrate, and more westernized diet. Additional support for this hypothesis came from the finding that during the 3 years of this large multicenter Japanese study (35), there was a trend toward a decreasing frequency of brown pigment stones. Patients in this study (35) being treated in rural hospitals were more likely to have brown pigment stones, reflecting their more traditional diet. The reason that brown pigment stones rarely form in the gallbladder in the West but are common or even predominant in Asia is unknown. However, eating a diet that does not stimulate bile flow as actively as a highprotein, highcalorie western diet may be a contributing factor. This dietary difference may cause alterations in the population of biliary bacteria, changes in biliary or duodenal motility, or differences in mucosal injury, which have variously been proposed as etiological factors. When Japanese or Chinese or other East Asian peoples emigrated to Australia, Hawaii, or the continental United States, the brown pigment stone disease they brought with them was instructive for surgeons in these regions. However, the first generation of their descendants born in the new country did not demonstrate this type of stone disease but instead reflected the disease of their locality. Our experience in communication with surgeons in these regions is that they were unable to identify U.S. or Australianborn patients of East Asian origin who had primary brown pigment stone disease. This type of primary disease is very uncommon in the west. In more than 30 years of searching for this type of patient and during collection of more than 2000 sets of stones from various western countries, we have identified only two Caucasian patients with primary brown pigment stone disease. The reasons that brown pigment stone rarely form in the gallbladder in the West but are common or even predominant in the East is unknown. Again, diet, differences in biliary bacterial populations, changes in motility, or differences in mucosal injury have been proposed. The primary incidence of brown pigment stones in the West (54) is secondary to choledocholithiasis; they are formed more than 2 years after cholecystectomy (6), most frequently for cholesterol cholelithiasis. In contrast to Asia, most of these patients do not have clinical episodes of bacterial cholangitis but bacteria can be cultured from the stones and they closely resemble, chemically and visually, primary stones found in Asian countries. Abnormalities of the biliary tract resulting in altered motility—including choledochal cysts, Caroli's disease, and paravaterian duodenal diverticula—have been associated with the formation of primary common bile duct stones (55–58). Similar material causes occlusion of biliary stents; its composition depends on the character of the stent material, which appears to act as a template, influencing the character of the precipitate (59–60). Studies have demonstrated that the components of this type of stone are characteristic of deposits in the various endoprostheses and are responsible for stent occlusion (59). The stent surface provides a template for precipitation, since the rate of precipitation varies with the composition of the stent (60). E— Sludge Sludge is more common than found by detection methods, since only 50% of cases are identified by ultrasound. It is usually thought to consist of either calcium bilirubinate or cholesterol microliths (14,15), although some believe that it is essentially calcium bilirubinate (61). Although it is known to develop during conditions inducing stasis—such as fasting (48), surgery (62), pregnancy, and weight loss—and its natural history in these conditions is now described, there is no information available about the frequency of sludge formation in response to these
Page 151
stimuli in various countries. Only in the minority of cases does sludge evolve into gallstones (63,64). III— Pathogenesis A— General The initiating factor(s) in the pathogenesis of pigment stones remains unknown (2,3,66). Some investigators have considered this to be a problem of supersaturation of components in the bulk phase of bile (65). Others, among whom we are included, favor the development of areas of supersaturation within localized microenvironments in glands, in mucous gel layers, or within developing stones (1,3,66). The question of whether proteins within stones provide the scaffolding on which crystallization occurs, whether they provide the limits within their gel meshwork for a myriad of microenvironments, or whether they precipitate within a crystalline field, providing the cement to hold the crystals together, or whether all of these mechanisms play a role remains unanswered. B— Noncholesterol Components in Cholesterol Stones Recent studies have suggested, that instead of being the nidus upon which cholesterol stones form, noncholesterol components [protein (67), bilirubin (68)] frequently precipitate following migration within an already nidated and growing cholesterol stone. This facilitates the formation of the rings of concentric pigmented precipitates present in many cholesterol stones when stones are cut in cross section (69). These are "mixed" or "combination" cholesterol stones. These pigmented Liesegang rings of precipitation (69) occur due to the diffusion of the noncholesterol components along concentration gradients (69) toward the center of the stone. The ratios of the components and the gradients are such that, at periodic intervals, precipitation begins (70). This attracts ions composing the precipitated salt from the surrounding gradient, adding to the precipitate in the initial ring and reducing the concentrations of these ions above or below the ring in the gradient. This phenomenon recurs repeatedly, forming a ringlike pattern. Cholesterol stones make up from 80 to 95% of most operative series in western countries and mixed stones constitute 67 to 80% of most cholesterol stone series. These stones usually occur in crops of stones with an identical crosssectional pattern, indicating that the gradients governing ring formation occurred within each stone but were widely distributed throughout the gallbladder. While the features characteristic of these stone crops are very similar within each stone group, each group was unique when compared with any other cholesterol stone crop with regard to the number of rings, intensity of pigment deposition, band width, distance between bands, and number of bands in a Liesegang system. In crops of larger stones, in cross section, there may be two or more Liesegang systems that have developed during stone growth with the coprecipitation of a variety of carbonate and phosphate calcium salts. Each system of rings may differ from the next. In addition, areas of chaotic precipitation may occur as well between ring systems of periodic precipitation. Such ring systems may prevent further exchange with the environment, since the pores present in the loosely packed cholesterol layers may be absent due to filling with protein and calcium salts. The clinical correlate of this phenomenon is the partial dissolution of cholesterol stones by chenodeoxycholic acid with the arrest of precipitation at the surface of one of these rings (71,72). These rings can be breached by lithotripsy, a focused ultrasound technique by which stones may be pulverized (73). This leads to a greater proportion of stones undergoing dissolution and a more rapid rate of stone dissolution (73,74). The remaining stones are socalled solitary cholesterol stones, or pure cholesterol stones, containing virtually no noncholesterol components. In contrast to mixed cholesterol stones, such stones do not recur after dissolution (19), implying a separate mechanism of nidation and/or growth.
Page 152
C— Black Pigment Stones These stones may present with bands containing many different shades of black, brown, or gray, dependent on highly individual composition from patient to patient. However, this succinct color classification makes certain that clinicians and researchers do not confuse these stones with brown pigment stones, simply because calcium bilirubinate is the predominant calcium salt (75). Calcium bilirubinate in these stones occurs as microcrystals or linear polymers (75) in a sea of protein macromolecules. By analogy from synthetic polymers, some areas may be flexible threads, while other areas may, through local compositional differences, be partially or completely crystalline (3). Their small crystal size may help to explain why xray diffraction of these stones has not yielded a molecular crystalline structure but rather an xray powder pattern (24,76,77). Calcium hydroxyapatite also occurs as a ropelike polymeric thread (31,78), but calcium carbonate has not been identified in this form. Crystalline material has been identified in separate rings in black stones, indicating changing conditions for precipitation during stone growth (79). In a series of studies using a variety of techniques, we have produced a spectrum of bilirubinates containing from 2 to 12% calcium (80,81). It is clear that bilirubin was not coordinating with calcium in a fixed stoichiometric fashion. Our infrared spectroscopic studies (80,81) demonstrated a series of differing relative intensity for the bilirubin propionic acid carbonyl group at 1700 cm1 and for the B and C ring pyrrole groups at 3400 cm1. The infrared spectral peak intensities of these two groups varied in a tight linear fashion, indicating the coordination of halves of the bilirubin molecule with calcium; there observations support the heterogeneity of calcium bilirubinate, perhaps due to a random ordering of hemiportions of neighboring molecules through metal coordination. We have constructed a number of models of calcium bilirubinate (3) but have not been able to confirm the molecular identity of any model due to the heterogeneous metal binding from molecule to molecule and the presence of multiple cations with similar binding capability but forming differing coordination complexes. Contrary to past theory (82,83), our infrared studies of calcium bilirubinate indicate that bilirubin, a linear tetrapyrrole, can coordinate with calcium by donating up to four hydrogens per molecule, from either of the pyrrole groups on the central B or C rings of the tetrapyrrole molecule or from either of the B and C ring propionic acid side chains (3). The A and B rings and the C and D rings appear to act independently in metal binding, so that one half of the molecule may retain its three hydrogen bonds in the configuration of protonated bilirubin while the other half may donate both a carboxyl and pyrrole hydrogen, forming a heterogenous compound termed an acid salt (84). Each bilirubin molecule forms onehalf of the calcium coordination structure for one calcium ion, with a neighboring bilirubin molecule providing the other half. Thus, calcium bilirubinate may form long metal ion polymeric chains that may also branch and form a network. When such stones were dissolved within seconds using DMSO (85) and the samples were examined by electrophoresis, the only pigment found to be present in human stones was unconjugated bilirubin (1). When stones from mice with hemolytic anemia were examined, it was found that they were made up of 37% bilirubin monoglucuronide, 6% diglucuronide, and 56% unconjugated bilirubinate (86). Monoglucuronide conjugates of bilirubin and conjugates with other sugars have been proposed to predispose to the formation of pigment precipitates (87,88). Others have suggested that bilirubin was present as a polybilirubinate (82,83) but there has been no evidence to support this in dissolution studies. Similarly, the suggestion that bilirubin was complexed via vinyl groups (88,89) has not been supported, since black stones can be completely dissolved using several methods (1), none of which identify products other than bilirubin. Vinyl bridging would not be expected to be reconstituted following stone dissolution. The equilibrium swelling attributed to bilirubin polymers (89,90) may instead be due to the associated glycoprotein network (91). Black stones have been noted to develop within the lumina of gallbladder glands in mice (92) and in humans (93,94) with chronic hemolytic anemia (92–94), where they likely nidate in a secreted protein matrix and in the surface mucous layer. Most human black pigment stones are found in the elderly (34,35), but they also occur in patients with chronic hemolysis and
Page 153
cirrhosis (36). Cirrhosis and hemolysis can be linked by intermittent or chronic hemolysis, respectively, resulting in hyperbilirubinbilia. This concentration of bilirubin exceeds the ion product for calcium bilirubinate and myriad microcrystals of bilirubin form on protein templates and, at times, in layers. Black stones have been noted to occur in crops and occasionally with similar crosssectional structure, as with cholesterol stones, suggesting that conditions for stone formation occurred throughout the gallbladder at a discrete point in time. However, these small stones are less uniform in size and shape and do not reach the size of many crops of cholesterol stones, suggesting that conditions for their formation are intermittent and less frequent or that they have not been present for as long a time. It is clear that gallbladder mucosal secretion, protein, and its associated calcium are necessary for black pigment stone formation, since cholecystectomy prevents recurrent stone formation in patients with hemolytic anemia, where the hyperbilirubinbilia continues unabated. The reason that stones may take years to develop in humans with hemolytic anemia and take a proportionally equivalent length of time to develop in mice with hemolytic anemia (92) remains unclear. If bilirubin supersaturation were solely responsible, precipitation should occur in utero or shortly after birth, but this is not the case in either mice or men. One hypothesis is that chronic hyperbilirubinbilia may take time to alter mucosal function and stimulate the production of altered proteins that serve as templates for calcium bilirubinate crystallization. A second hypothesis might invoke the development of gallbladder glands of increasing complexity and depth, creating a different precipitation microenvironment. Alternatively, conditions may favor the growth of commensal or other bacteria, which, in turn, secrete a biofilm that predisposes to calcium salt precipitation, including calcium bilirubinate. Oral calcium administration increases pigment stone formation in animals (95) through effects on mucin and/or bilirubin secretion. Intestinal resection also increases bilirubinbilia (39). The mean bilirubin content of stones from these patients with hemolysis exceeds that found in patients without hemolysis but the range of concentrations largely overlaps (36,38), suggesting that the processes by which these stones formed were similar. However, the increased concentration of bilirubin in the bile of patients with hemolysis appears to have influenced the increased proportion of calcium bilirubinate in stones. Black pigment stones also form behind biliary strictures, with or without clinical bacterial infection (36). A prime example is in patients with primary sclerosing cholangitis. Although calcium is the predominant metal in stones, small amounts of other divalent metals, primarily copper, occur in stones (96). We have undertaken in vitro studies of calcium and copper bilirubinate formation and have found that copper bilirubinate complexes have a different infrared spectrum from calcium bilirubinate, implying a different salt structure. Further studies indicate that it was present as a Cu2+ N chelate (80,81). Calculations (97) indicate that protonated bilirubin is flexible at the "ridgetile" angle between the two halves of the tetapyrrole, varying from 60 to 133°. Lightner et al. first postulated this lepidopterous effect on the basis of their studies of tetrapyrroles (98). Such flexibility is critical for two molecules of bilirubin to coordinate with Cu2+ (97). Copper salts are more soluble than calcium salts and in mixture may help keep calcium salts in solution (99) and therefore in a form that can be excreted. 1— Bilirubinate In our experience, the black pigment stone set containing the highest proportion of calcium bilirubinate contained 91% calcium bilirubinate (31), no other calcium salts and presumably 9% protein. Other sets of black stones contain less than 10% calcium bilirubinate and, despite the presence of predominantly calcium hydroxyapatite or calcium carbonate, appear black. The black color of these stones is from the outer layer and is due to the incorporation of singlet oxygen from the atmosphere or surrounding bile after the precipitate is left standing for a period of time (99–102). When such stones are fractured, inner layers are redbrown to brown in color, indicating that this interaction has not occurred.
Page 154
2— Carbonate This salt may form as part of the pigmented bands in cholesterol stones described above and as the predominant salt in some black pigment stones. It is stimulated by obstruction of the gallbladder and is responsible for the formation of ''milk of calcium" bile and the "porcelain" gallbladder. Three types of carbonate can be formed— vaterite, aragonite, and calcite (24)—indicating that changes in the conditions governing precipitation occurred during stone growth (79), perhaps due to changes in ionic concentrations and/or pH. We have identified, by infrared spectroscopy, differences in these polymorphs and have identified these in different layers of the same stone, as close as 10 to 50 m from each other, using infrared microspectroscopy of particles from neighboring stone layers (23). Alternatively, diffusion gradients within the stone may have provided the conditions for the formation of different polymorphs at the same time. Vaterite is very unstable in air or in a fluid environment and should have changed to calcite in our stored stones. However it is detected, even after long periods of storage (23). This suggests that it is coated with protein or other protection that prevents change. We unsuccessfully searched for altered infrared carbonate spectra suggesting stabilization, as in crustaceans (103), by intercalation of proteins within the calcite or vaterite crystal. Vaterite is intrinsically unstable and rapidly converts to calcite despite storage at 100°C (23). Despite this in vitro observation, this carbonate polymorph is identified in stones (23,24) after years of storage, suggesting that vaterite was somehow protected. Possibilities include a thin but effective protein coating that we have observed, using the scanning electron microscope (SEM), to be covering some crystal surfaces or due to intercalated protein in the vaterite crystal, as occurs in crustaceans (103). This intercalation should cause a marked change in the infrared spectrum of vaterite and should be easily identified. However, we have not identified such changes in areas of vaterite (unpublished results) despite a systematic search. Thus, protein crystal coating seems the most likely explanation. 3— Hydroxyapatite Glycine conjugated bile salts, through submicellar binding (104) to amorphous calcium phosphate nidi, can poison these microaggregates and prevent transformation to microcrystalline calcium hydroxyapatite and thus prevent growth to a sufficient size to cause precipitation (105). Phospholipid modifies this inhibitory effect (106). Human gallbladder mucin (107) and polyamines (108) also modify bile salt inhibition. Thus, like cholesterol, bile contains a series of accelerators and inhibitors, the balance of which determines whether calcium hydroxyapatite will precipitate in a given gallbladder. Bile salts have been proposed to regulate calcium and carbonate ion concentrations in bile (109). Calcium hydroxyapatite is also present in urinary stones and dental calculus (28). In these situations, bacteria, which produce biofilm, are capable of forming the scaffolding for this type of calcification and have been identified. It seems probable that a similar process is operative in the gallbladder. Studies by Swidzinski and colleagues (110), using bacterial DNA identification techniques, indicated that bacteria had been present in the gallbladder (110), which had previously been thought to be sterile (111). The benchmark of 105 colonies per milliliter used as the index of clinical infection of bile, urine and other fluids may not be appropriate in considering the role of bacteria in gallstone formation, since bacteria may exist in much smaller concentrations in the mucous layer along the gallbladder and biliary tract mucosa and may interact with the underlying mucosa to stimulate the production of calciumbinding proteins. 4— Metals The proportion of calcium salts in stones greatly exceeds its concentration in bile, although it is the most common cation (96). The activity of Ca2+ in bile is much less than its total concentration due to complexing with bile salts (112–114) phospholipids (114–116) and pro
Page 155
teins (117), either in precipitates or in soluble complexes. Calcium is highly concentrated within intracellular mucous globules, serving to hold mucous in a tightly packed form (117). After secretion, the stores of calcium are released into the mucous layer at concentrations greatly exceeding the concentrations of calcium in the bulk phase. Thus, a calcium gradient is likely to be established across the mucous layer, from mucosa to bile, facilitating the formation of Liesegang rings in precipitates developing in this layer. Since bile is also a major excretory pathway for calcium, the forces determining biliary calcium secretion and concentration are multiple and complex (118–120). Calcium salts are less soluble than magnesium salts and, even though magnesium is the second most common cation in bile (96), copper is the second most common cation in stones (96). As stated above, copper salts are more soluble than calcium salts and serve to keep calcium salts in solution when coppercalcium bilirubinates are formed (121). A provocative study in the prairie dog indicated that feeding calcium significantly increased biliary bilirubin, bilirubin monoglucuronide, and glycoprotein but not ionized calcium (95). These animals all demonstrated pigment stones and calcium bilirubinate sludge. These findings deserve examination in humans, since calcium supplementation is recommended for elderly females to prevent osteoporosis. This is just the group at risk for the development of pigment gallstones. A variety of other metals are present in stones, probably reflecting the wide range of divalent cations secreted in bile, the major excretion route for all divalent cations except calcium, where most is secreted in the urine. 5— Bile Acids In a series of studies, we have demonstrated that bile acids and calcium as well as other cations form periodic and fractal precipitates in agar gel systems, a model designed to resemble the mucin layer in the gallbladder. In the gallbladder, bile acids could diffuse across the mucous layer in a gradient opposite to that of calcium (see above), creating an opportunity for periodic precipitation at the optimal concentration of these two components. Because the concentration of calcium at the mucosal surface, following mucous granule release, is much greater than in bile, calcium bile salts are more likely to precipitate in the mucous layer than in the bulk phase of bile. In vitro, we have found that bilirubinate, carbonate, and hydroxyapatite precipitate in areas where bile salts have initially precipitated (122). Although only small amounts of bile acids have been identified in stones (122), with larger amounts in pigment than in cholesterol stones (122), calcium salts of bile acids may serve the critical role of acting as a nidation focus for other calcium salts. It is also reasonable to hypothesize that bile salts may redissolve to some extent and release calcium locally to then form bilirubinates, carbonates, or hydroxyapatites. However, experiments in agar gels over several months indicate that the calcium bile salt precipitates do not change in shape, as rings of calcium bilirubinate form superimposed on the original sites of precipitation (123). 6— Protein A mucin network was identified in stones by histochemical staining (124,125), and it was noted that protein and bilirubin were codistributed (33,126–128). On extraction and protein purification of black pigment gallstones, we found that as much as 12% could be identified as mucin glycoprotein, but even more was present as protein using other techniques (129). A series of calciumbinding proteins have facilitated the formation of both cholesterol and pigment gallstones (130–135). Precipitation may occur through sequestration of these smaller proteins (132–135) within the meshwork in the mucin layer formed by mucin glycoproteins. Studies of a variety of minerals to examine protein crystallization indicated that there were specific mineral components that could induce protein crystallization at lower critical levels of supersaturation than required for spontaneous growth (136). It might be postulated that the reverse is also true: proteins might act to facilitate proteinspecific patterns of precipitation at component concentrations below that found in the bulk phase of bile. Clearly, this concept needs testing.
Page 156
D— Brown Pigment Gallstones These stones, the product of stasis and infection (2,36,137–140), are formed through the action of bacterial enzymes to deconjugate bilirubin from glucuronic acid (137) and to hydrolyze phospholipids (141), providing large amounts of bilirubin and free fatty acids to precipitate with calcium. Crosssectioning these stones demonstrates that they are formed in layers. Unlike the regular spacing of Liesegang rings, these layers are of random thickness and composition. They vary from predominantly calcium bilirubinate to predominantly calcium salts of fatty acids, with layers containing all combinations of these components. Since calcium bilirubinate is reddishbrown and calcium salts of fatty acids are white, the periodicity of precipitation is clearly visible. The reason for this alternation between deconjugation of bilirubin and lecithin hydrolysis is unknown but may involve periodic changes in the species of bacteria present in bile. Alternatively, layers of bacteria in biofilm have been identified in these stones (138) and may provide stop and start signals for different types of precipitation. Bacteria have been associated with the calcium bilirubinate layers but less so with the calcium fatty acid layers (138). Characteristics of bacteria in stones and in bile were compared (139). Bacteria from stones contain the P1specific fimbriae, a minority of strains had the Forssman antigenspecific fimbriae, and none demonstrated mannosespecific fimbriae. All strains bound antiGal, a ubiquitous naturally occurring IgG specific for galactosyl residues. All of these characteristics distinguish these bacteria from gut flora. The authors postulate that these properties may enhance the ability of these bacteria to colonize the biliary tract and/or to initiate pigment gallstone formation (139). In contrast to black pigment stones, these stones contain calcium carbonate or hydroxyapatite very rarely. Protein is present, but it is not known if it has a central role, as in the case of cholesterol and black pigment stones, or whether it is coprecipitant, trapped in stone by the speed of precipitation or because mucin and other proteins bind to all precipitates. Possibly much of the protein found in these stones might be present as biofilm secreted by bacteria for their protection (138). When these films completely encase and protect all of the local bacteria, they may also act to prevent bacterial action in the surround medium. Based on SEM and spectroscopic techniques, we have concluded that black pigment stones form much more slowly, through microcrystallization in local microenvironments within a protein gel, while brown pigment stones precipitate much more rapidly through supersaturation of the bulk phase of bile. Teleologically, it seems important for brown pigment stones to form through rapid precipitation within the more actively flowing bile in the bile ducts compared to the relative stasis in the gallbladder. 1— Bilirubinate In brown pigment stones, calcium bilirubinate is present as a loosely packed amorphous precipitate (33). When examined by SEM before and after exposure to an oxygen plasma environment which dissolved away the protein network, no rodlike microcrystals or polymers of calcium bilirubinate were demonstrated, as in black stones (33). When dissolved in dimethyl sulfoxide (DMSO), only unconjugated bilirubin was identified (85). 2— Fatty Acid Salts When freshly obtained stones were examined using highperformance liquid chromatography (HPLC) separation, a variety of fatty acids salts were identified, mirroring the fatty acid composition of the biliary lecithin isolated from the same patient (141), strongly supporting their hydrolysis from biliary lecithin by bacterial phospholipases. These stones contained about 15% cholesterol, which is believed to be coprecipitated through reduction in the concentrations of lecithin and bile acids, occurring through hydrolysis and deconjugation, respectively. Alternatively, cholesterol might have coprecipitated passively
Page 157
due to the rapidity of the deconjugation and precipitation of stone components, lecithin, and bile acids, which hold cholesterol in solution. E— Sludge Although sludge has been shown to cause a number of clinical syndromes—such as recurrent pancreatitis, cholangitis, and biliary colic—questions concerning its pathogenesis remain. Does it form by mechanisms separate from those of the three major types of gallstones? Do the microliths, which rest between the bulk phase of bile and the mucous layer, grow to larger stones or must stones remain within the mucous layer to grow to larger size? Clearly, sludge can shift—as monitored by ultrasound, with changes in gallbladder position—as do larger stones. Why is it that stones large enough to cause ultrasound shadowing (142) (greater than 4 mm) infrequently follow the development of sludge? Whether the sludge represents microliths of cholesterol or calcium bilirubinate forming during total parenteral nutrition or weight loss, when the gallbladder is hypomotile, periodic gallbladder contraction and bile acid administration (143,144) have been shown to remove and/or dissolve sludge and prevent its consequences. IV— Conclusion This review has concentrated on organizing the observations of many investigators on the components of black and brown gallstones and has attempted to weave them together with a number of hypotheses. The conclusions drawn are open to investigation and are stated to provoke interest in this area by future investigators using different techniques. Acknowledgments Supported by Institutional Funds, Sealy and Smith Foundation, Marie B. Gale Professorship, past NIH support (RDS). Grants from the National Science Foundation of China, Institutional Funds (JG W). References 1. Soloway RD, Wu JG. Analysis of gallstones, In: Muraca M, ed. Methods in Biliary Research. Boca Raton, FL: CRC Press, 1995, pp. 167–190. 2. Trotman BW, Soloway RD. Pigment gallstone disease: summary of the National Institutes of HealthInternational workshop. Hepatology 1982; 2:879–884. 3. Soloway RD, Wu JG, Xu DF. Pigment gallstones and secondary calcification of gallstones. In: Swobodnik W, Ditschuneit H, Soloway RD, eds. Gallstone Disease. Pathophysiology and Therapeutic Approaches. Munich: SpringerVerlag, 1990, pp. 35–46. 4. Trotman BW, Ostrow JD, Soloway RD. Pigment vs. cholesterol cholelithiasis: comparison of stone and bile composition. Am J Dig Dis 1974; 19:585–590. 5. Malet PF, Weston NE, Trotman BW, Soloway RD. Cyclic deposition of calcium salts during growth of cholesterol gallstones. Scanning Electron Microsc 1985; 2:775–779. 6. Malet PF, Dabezies MA, Huang G, Long WB, Gadacz TR, Soloway RD. Quantitative infrared spectroscopy of common bile duct gallstones. Gastroenterology 1988; 94:1217–1221. 7. Miyake Jr H. Statistische, klinische und chemische Studien zur Aetiology der Gallensteine mit besonderer Beruchsichtigung der japanischen und deutschen Verhaltnisse. Arch Klin Chir 1913; 101:54–117.
Page 158
8. Miyake H, Kuwatsuka T, Tamesue N, Uzumi K. Comparative studies on the chemical composition of gallstones in Japan, China and the United States. Fukuoka Acta Med 1963; 54:781–826. 9. Diehl AK, Schweisinger WH, Holleman DR Jr, Chapman JB, Kurtin WE, et al. Gallstone characteristics in Mexican Americans and nonHispanic whites. Dig Dis Sci 1994; 60: 2223–2228. 10. Nakayama F. Quantitative microanalysis of gallstones. J Lab Clin Med 1969; 72:602–611. 11. Agarwal DK et al. Utility of biliary microcrystal analysis in predicting composition of common bile duct stones. Scand J Gastroenterol 1994; 29:352–354. 12. Mukaihara S. Classification and composition of human gallstones. Arch Jpn Chir 1981; 50:476–500. 13. Cetta F. The classification of biliary calculi and the clinicotherapeutic implications. Ann Ital Chir 1998; 69:701–708. 14. Lee SP, Nichols JF. Nature and composition of biliary sludge. Gastroenterology 1986; 90:677–686. 15. Lee, SP, Maher K, Nichols JF. Origin and fate of biliary sludge. Gastroenterology 1988; 88:170–176. 16. Chen CY, Shiesh SC, Lin XZ. Biliary sludge and pigment stone formation in bile ductligated guinea pigs. Dig Dis Sci 1999; 44:203–209. 17. Duchmann JC, Joly JP, Decrombecque C, Delcenserie R, Levy S, Capron D, Capron JP. Cirrhosis: a new but expected cause of biliary sludge. Alcohol Clin Exp Res 1997; 21:119–121. 18. Malet PF, Williamson CE, Trotman BW, Soloway RD. Composition of the pigmented center of cholesterol gallstones. Hepatology 1986; 6:477–481. 19. Wolpers C, Hofmann AF. Solitary versus multiple cholesterol gallbladder stones. Clin Invest 1993; 71:423–434. 20. Strasberg SM, Harvey PRC, Taylor DR, Soloway RD. Multiple crops of cholesterol gallstones in the same gallbladder indicate that cholesterol precipitates first and pigmented centers and layers develop during remodeling (abstr). Gastroenterology 1996; 110:A477. 21. Donovan JM. Physical and metabolic factors in gallstone pathogenesis. Gastroenterol Clin North Am 1999; 28:75–97. 22. Jungst D, Lang T, von Ritter C, Pratschke E, Paumgartner G. Cholesterol nucleation time in gallbladder bile of patients with solitary or multiple cholesterol gallstones. Hepatology 1992; 15:804–808. 23. Taylor DR, Crowther RS, Cozart JC, Sharrock P, Wu JG, Soloway RD. Calcium carbonate in cholesterol gallstones. Polymorphism, distribution and hypotheses about pathogenesis. Hepatology 1995; 22:488–496. 24. Bogren HG. The polymorphs of calcium carbonate in human gallstones. Scand J Clin Lab Invest 1983; 43:371–375. 25. Ruiz de Aguiar A, Medina Nunez JA, Lopez Domingo MI, Villacorta Patino J, Leguey Jimenez S. Calcium carbonate in cholesterol gallstones. J Hepatol 1988; 6:71–79. 26. Kaufman HS, Magnuson TH, Pitt HA, Frasca P, Lillemoe KD. The distribution of calcium salt precipitates in the core, periphery and shell of cholesterol, black pigment and brown pigment gallstones. Hepatology 1994; 19:1124–1132. 27. Agarwal DK, Choudhuri G, Kumar J. Chemical nature and distribution of calcium compounds in radiolucent gallstones. Scand J Gastroenterol 1993; 28:613– 616. 28. Hou RZ, Wu JG, Soloway RD, Zhang YF, Du YC, Liu F, Xu GX. Fourier transform infrared photoacoustic spectroscopy of dental calculus. Mikrochim Acta 1988; 2:133–136. 29. RiosDalenz J, Takabayashi A, Henson DE, Strom BL, Soloway RD. Cancer of the gallbladder in Bolivia: suggestions concerning etiology. Am J Gastroenterol 1985; 80:371–375.
Page 159
30. Strom BL, Soloway RD, RiosDalenz J, RodriguezMartinez HA, West SL, Kinman JL, Crowther RS, Taylor D, Polansky M, Berlin JA. Biochemical epidemiology of gallbladder cancer. Hepatology 1996; 23:1402–1411. 31. Li WH, Tong XB, Soloway RD, Zhou WJ, Weng SF, Shen GR, Xu DF, Taylor D, Hong NK, Hirakawa N, Crowther RD, Wu JG. Copper forms black bilirubinate related to the formation of free radicals and causes alteration of the pyrrole rings and tetrapyrrole structure. 2nd Bilirubin Workshop, Trieste Italy, 1992. 32. Yang ZL, Weng SF, Soloway RD, Taylor D, Wu JG, Xu GX. Free radicals are required for solidstate formation of calcium and barium bilirubinates (abstr). Gastroenterology 1991; 100:A814. 33. Soloway RD, Fayusal EB, Trotman BW, Weston NE, Ficca Jr JF. Water content of gallstones: location and contribution to a hypothesis concerning stone structure. Hepatology 1982; 2:223–229. 34. Trotman BW, Soloway RD. Pigment vs cholesterol cholelithiasis: clinical and epidemiologic aspects. Am J Dig Dis 1986; 20:735–740. 35. Hikasa Y, Nagase M, Soloway RD, Tanimura H, Setoyama M, Kato H. Gallstones in western Japan: 1. Epidemiologic factor affecting the type and location of gallstones. Arch J Surg 1981; 50:272–288. 36. Soloway RD, Trotman BW, Maddrey WC, Nakayama F. The influence of hemolysis, infection and stasis on the calcium salts in pigment gallstones. Dig Dis Sci 1986; 31: 454–460. 37. Fevery J, Verwilghen R, Tan TG, DeGroote J. Glucuronidation of bilirubin and the occurrence of pigment gallstones in patient with chronic hemolytic disease. Eur J Clin Invest 1980; 10:219–226. 38. O'Brien CB, Berman JM, Fleming CR, Malet PF, Soloway RD. Total parenteral nutrition gallstones contain more calcium bilirubinate than sickle cell gallstones (abstr). Gastroenterology 1986; 90:1752. 39. Brink MA, Slors JFM, Keulemans YCA, Mok KS, de Waart DR, Carey MC, Groen AK, Tytgat GNJ. Enterohepatic cycling of bilirubin: a putative mechanism for pigment gallstone formation in ileal Crohn's disease. Gastroenterology 1999; 116:1420–1427. 40. Lapidus A, Baugstad M, Astron M, Muhrbeck O. The prevalence of gallstone disease in a defined cohort of patients with Crohn's disease. Am J Gastroenterol 1999; 94:1261–1266. 41. Trotman BW, Morris III TA, Cheney HM, Ostrow JD, Sanchez HM, Soloway RD, and Conn HO. Pigment gallstone composition in cirrhotic and non–cirrhotic subjects. Am J Dig Dis 1978; 23:872–876. 42. Nicholas P, Rinaudo PA, Conn HO. Increased incidence of cholelithiasis in Laennec's cirrhosis. Gastroenterology 1972; 63:112–121. 43. Schwesinger WH, Kurtin WE, Levine BA, Page CP. Cirrhosis and alcoholism as pathogenetic factors in pigment gallstone formation. Ann Surg 1985; 201:319– 322. 44. Maggi A, Solenghi D, Pauzeri A, Borroni G, Cazzaniga M, Sangiovanni A, de Fazio C, Salerno F. Prevalence and incidence of cholelithiasis in patients with liver cirrhosis. Ital J Gastroenterol Hepatol 1997; 29:330–335. 45. Sakata R et al. formation of black pigment gallstone in a hamster model of experimental cirrhosis. Eur J Clin Invest 1997; 27:840–845. 46. Diehl AK, Schwesinger WH, Holleman DR Jr, Chapman JB, Kurtin WE. Clinical correlates of gallstone composition: distinguishing pigment from cholesterol stones. Am J Gastroenterol 1995; 90:967–972. 47. Soloway RD, Takabayashi A, RiosDalenz J, Nakayama R, Tandon RK, Trotman BW, Henson DE. Geographic differences in the operative incidence and type of pigment gallstone and in the noncholesterol components of cholesterol gallstones. Hepatol Rapid Lit Rev 1981; 11:1637–1638. 48. Messing B, Bories C, Kunstlinger F, Bernier JJ. Does total parental nutrition induce gallbladder sludge formation and lithiasis? Gastroenterology 1983; 84:1012– 1019.
Page 160
49. Hikasa Y, Nagase M, Tanimura H, Shioda R, Setoyama M, Kobashi N, Mukaihara S, Kamata T, Maruyama K, Kato H, Mori K, Soloway RD. Epidemiology and etiology of gallstones. Arch Jpn Chir 1980; 49:555–575. 50. Nagase M, Hikasa Y, Soloway RD, Tanimura H, Setoyama M, Kato H. Gallstones in western Japan: factors affecting the prevalence of intrahepatic stones. Gastroenterology 1980; 78:684–690. 51. Lam SK, Chan CW. Pyogenic cholangitis: recent etiology and management. Surg Dig Dis 1985; 3:12–24. 52. Leung JWC, Sung JY, Costerton JW. Bacteriological and electron microscopy examination of brown pigment stones. J Clin Microbiol 1989; 27:915–921. 53. Nakayama F, Soloway RD, Nakama T, Miyazaki K, Ichimiya H, Sheen PC, Ker CG, Ong GB, Choi TK, Boey J, Foong WC, Tan EC, Tung KH, Lee CN. Hepatolithiasis in East Asia: retrospective study. Dig Dis Sci 1986; 31:21–26. 54. Cetta F. The role of bacteria in pigment gallstone disease. Ann Surg 1991; 213:315–326. 55. Loveit T, Osnes M. Duodenal diverticula. Scand J Gastroenterol 1984; 19:579–581. 56. Kennedy RH, Thompson MH. Are duodenal diverticula associated with choledocholithiasis? Gut 1988; 29:1003–1006. 57. Lobo DN, Balfour TW, Iftikhar SY, Rowlands BJ. Periampullary diverticula and pancreaticobiliary disease. Br J Surg 1999; 86:588–597. 58. Kim MH, Myung SJ, Seo DW, Lee SK, Kim YS, Lee MH, Yoo BM, Min MI. Association of periampullary diverticula with primary choledocholithiasis but not with secondary choledocholithiasis. Endoscopy 1998; 30:601–604. 59. Moesch C, Sautereau D, Cessot F, Berry P, Mounier M, Gainant A, Pillegand. Physicochemical and bacteriological analysis of the contents of occluded biliary endoprostheses. Hepatology 1991; 14:1142–1146. 60. Hirakawa N, Burke D, Soloway RD, Schnall M, Wen G, Crowther RS, Wu JG. Stent composition determines the components precipitated in biliary sludge on the stent surface: an infrared study (abstr). Hepatology 1990; 11:1310. 61. Allen B, Bernhoft R, Blanchaert N, Svanvik J, Filly R; Gooding G, Way L. Sludge is calcium bilirubinate associated with bile stasis. Am J Surg 1981; 141:51. 62. Bolondi L, Gaiani S, Testa S, Labo G. Gall bladder sludge formation during prolonged fasting after gastrointestinal tract surgery. Gut 1985; 26:734–738. 63. Jungst D, Del Pozo R, Christon S, Miguel JF, Eder MI, Lange V, Frimberger E, Von Ritter C, Paumgarmer G. Sedimentation of biliary sludge: effect on composition of gallbladder bile from patients with cholesterol, mixed or pigment stones. Scand J Gastroenterol 1996; 31(930):273–278. 64. Janowitz P, Kratzer W, Zemmler T, Tudyka J. Wechsler JG. Gallbladder sludge: spontaneous course and incidence of complications in patients without stones. Hepatology 1994; 20:291–294. 65. Cahalane MJ, Neubrand MW, Carey MC. Physicalchemical pathogenesis of pigment gallstones. Semin Liver Dis 1988; 8:317–328. 66. Crowther RS, Soloway RD. Pigment gallstone pathogenesis: from man to molecules. Semin Liver Dis 1990; 10:171–180. 67. Sanabria JR, Upadhya AR, Harvey PRC, Strasberg SM. Diffusion of substances into human cholesterol gallstones. Gastroenterology 1994; 106:749–754. 68. Sanabria JR, Gordon ER, Harvey PRC, Goresky CA, Strasberg SM. Accumulation of unconjugated bilirubin in cholesterol pellets implanted in swine gallbladders. Gastroenterology 1996; 110:607–613. 69. Peng Q, Wu JG, Soloway RD, Hu TD, Huang WD, Xu YZ, Wang LB, Li XF, Li WH, Xu DF, Xu GX. Periodic and chaotic precipitation phenomena in a bile salt system related to gallstone formation. Biospectroscopy 1997; 3:195–206. 70. Xie DT, Qi OY, Soloway RD, Xu YZ, Wu JG, Xu GX. The kinetic process of
Page 161
Liesegang ring formation is described using computer analysis of videotaped recordings of a metal ioncholate diffusion system (abstr). Gastroenterology 1999; 116:G0139. 71. Whiting MJ, Jarvinen V, Watts JM. Chemical composition of gallstones resistant to dissolution therapy with chenodeoxycholic acid. Gut 1980; 21:1077–1081. 72. Freilich HS, Malet PF, Schwartz JS, Soloway RD and the National Cooperative Gallstone Study Group. Chemical and morphologic characteristics of cholesterol gallstones that failed to dissolve on chenodiol: the National Cooperative Gallstone Study. Gastroenterology 1986; 91:713–718. 73. Sackmann M, Pauletzki J, Sauerbruch T, Paumgartner G. The Munich Gallbladder Lithotripsy Study: results of the first 5 years with 711 patients. Ann Intern Med 1991; 114:290–296. 74. Schoenfield LJ, Berci G, Carnovale RL, et al. The effect of ursodiol on the efficacy and safety of extracorporeal shockwave lithotripsy of gallstones: the Dornier National Biliary Lithotripsy Study. N Engl J Med 1990; 323:1239–1245. 75. Malet PF, Takabayashi A, Trotman BW, Soloway RD, Weston NE. Black and brown pigment gallstones differ in microstructure and microcomposition. Hepatology 1984; 4:227–234. 76. Bogren H, Larsson K. Crystalline components of biliary calculi. Scand J Clin Lab Invest 1963; 15:457–462. 77. Bogren H, Larsson K. An xraydiffraction study of crystalline cholesterol in some pathological deposits in man. Biochem Biophys Acta 1963; 756569. 78. Qiu SM, Wen G, Wen J, Soloway RD, Crowther RS. Interaction of human gallbladder mucin with calcium hydroxyapatite: binding studies and the effect on hydroxyapatite formation. Hepatology 1995; 21:1618–1624. 79. Sutor DJ, Wooley SE. The sequential deposition of crystalline material in gallstones: evidence for changing gallbladder bile composition during the growth of some stones. Gut 1974; 15:130–131. 80. Shen GR, Li WH, Soloway RD, Wen J, Wu JG. Cu++ increases solubility of black Cabilirubinate complexes in vitro: possible protection against gallstones (abstr). Clin Res 1993; 41:A159. 81. Li WH, Tong XB, Soloway RD, Zhou WJ, Weng SF, Shen GR, Xu DF, Taylor D, Hong NK, Hirakawa N, Crowther RS and Wu JG. Copper facilitates formation of black bilirubinate salts when coprecipitated in in vitro formation of nonstoichiometric salts (abstr). Gastroenterology 1991; 100:A813. 82. Wosiewitz U, Schroebler S. On the chemistry of "black" pigment stones from the gallbladder. Clin Chim Acta 1981; 89:1–12. 83. Burnett W, Dwyer KR, Kennard CHL. Black pigment or polybilirubinate gallstones: composition and formation. Ann Surg 1981; 193:331–333. 84. Berman JM, Dabezies MA, Malet PF, Soloway RD. Fourier transform infrared spectroscopy (FTIR) can identify and quantitate the acid and neutral salts of calcium bilirubinate (abstr). Gastroenterology 1985; 88:1711. 85. Trotman BW, RoyChowdhury J, Wirt GD, Bernstein SE. Ethyl anthranilate diazotization of unconjugated and conjugated bilirubin using dimethyl sulfoxide: application to bile and serum. Anal Biochem 1982; 121:175–181. 86. Trotman BW, Nair CR, Bernstein SE. Monoconjugated bilirubin is a major component of hemolysisinduced gallstones in mice. Hepatology 1988; 8:919–924. 87. Keida Y, Nakano T, Tabata M, Shimuzu S, Nakayama. Significance of different conjugate forms of bilirubin in the formation of pigment gallstones. J Gastroenterol Hepatol 1991; 6:595–598. 88. Vitetta L, Sali A, Little P, Nayman J, Elzarka A. Primary "brown pigment" bile duct stones. HPB Surg 1991; 4:209–20. 89. Black, BE, Carr SH, Ohkubo H, Ostrow JD. Equilibrium swelling of pigment gallstones: evidence for network polymer structure. Biopolymers 1982; 21:601– 610.
Page 162
90. Ohkubo H, Ostrow JD, Carr SH, Rege RV. Polymer networks in pigment and cholesterol gallstones assessed by equilibrium swelling and infrared spectroscopy. Gastroenterology 1984; 87:805–814. 91. Guo H, Soloway RD, Xu DF, Crowther RS, Wu JG. Dependence of equilibrium swelling on an interaction between calcium bilirubinate and proteins (abstr). Gastroenterology 1990; 98:A254. 92. Trotman BW, Bernstein SE, Bove KE, Wirt GD. Studies on the pathogenesis of pigment gallstones in hemolytic anemia: description and characteristics of a mouse model. J Clin Invest 1980; 65:1301–1308. 93. Haider MZ, Ashebu S, Aduh P, Adekile AD. Influence of alphathalassemia on cholelithiasis in SS patients with elevated Hb F. Acta Haematol 1998; 100:147– 150. 94. Kalayaci AG, Albayrak D, Gunes M, Incesu L, Agac R. The incidence of gallbladder stones and gallbladder function in betathalassemic children. Acta Radiol 1999; 40:440–443. 95. Magnuson TH, Lillemoe KD, Peoples GE, Pitt HA. Oral calcium promotes pigment gallstone formation. J Surg Res 1989; 46:286–291. 96. Trotman BW, Petrella EJ, Soloway RD, Sanchez HM, Morris III TA, Miller WT. Evaluation of radiographic lucency or opaqueness of gallstones as a means of identifying cholesterol or pigment stones: correlation of lucency or opaqueness with calcium and minerals. Gastroenterology 1975; 68:1563–1566. 97. Ferraro JR, Wu JG, Soloway RD, Li WH, Xu YZ, Xu DF, Shen GR. Copper binding to bilirubin as determined by FTIR and EPR spectroscopy. Appl Spectrosc 1996; 50:922–927. 98. Lightner DA, Nogales DF, Holmes DL, Anstine DT. Bilirubin structure—an overview. International Bilirubin Workshop, Trieste, Italy, 1995. 99. Li WH, Shen GR, Soloway RD, Xu ZH, Yang ZL, Guo H, Wu JG. Characterization of black calcium bilirubinate by Fourier transform infrared spectroscopy (abstr). Gastroenterology 1991; 100:A766. 100. Yang ZL, Weng SF, Soloway RD, Taylor D, Wu JG, Xu GX. Free radicals are required for the solidstate formation of calcium and barium bilirubinates (abstr). Gastroenterology 1991; 100:A814. 101. Li WH, Shen GR, Soloway RD, Xu ZH, Yang ZL, Guo H, Wu JG. Characterization of black calcium bilirubinate by Fourier transform infrared spectroscopy (abstr). Gastroenterology 1991; 100:A766. 102. Li WH, Tong XB, Soloway RD, Zhou WJ, Weng SF, Shen GR, Xu DF, Taylor D, Hong NK, Hirakawa N, Crowther RS, Wu JG. Copper facilitates formation of black bilirubinate salt when coprecipitated with calcium: implications for black pigment gallstone formation (abstr). Gastroenterology 1992; 102:A842. 103. Berman A, Addadi L, Kvick A, Leiserowitz L, Nelson M, Weiner S. Intercalation of sea urchin proteins in calcite: study of a crystalline composite material. Science 1990; 250:664–666. 104. Qiu SM, Soloway RD, Crowther RS. Interaction of bile salts with calcium hydroxyapatite: inhibitors of apatite formation exhibit high affinity premicellar binding. Hepatology 1992; 16:1280–1289. 105. Qiu SM, Wen G, Hirakawa N, Soloway RD, Hong NK, Crowther RS. Glycochenodeoxycholic acid inhibits calcium phosphate precipitation in vitro by preventing the transformation of amorphous calcium phosphate to calcium hydroxyapatite. J Clin Invest 1991; 88:1265–1271. 106. Okido M, Soloway RD, Crowther RS. Influence of phospholipid on bile salt binding to calcium hydroxyapatite and on the poisoning of nascent hydroxyapatite crystals. Liver 1996; 16:321–325. 107. Qiu SM, Wen G, Wen J, Soloway RD, Crowther RS. Interaction of human gallbladder mucin with calcium hydroxyapatite: binding studies and the effect on hydroxyapatite formation. Hepatology 1995; 21:1618–1624.
Page 163
108. Crowther RC, Pritchard CM, Qiu SM, Soloway RD. Inhibition of calcium hydroxyapatite formation by polyamines. Liver 1993; 13:141–145. 109. Gleeson D, Qureshi Y, Murphy GM, Dowling RH. The role of bile acids in regulating biliary calcium and carbonate ions concentrations in man. In: Paumgartner G, Stiehl A, Gerok W, eds, Bile Acids and the Liver. Lancaster PA: MTP Press, 1987; pp 199–212. 110. Swidzinski A, Ludwig W, Pahlig H, Priem E Molecular genetic evidence of bacterial colonization of cholesterol gallstones. 1995; 108:860–864. 111. Goodhart GL, Levison ME, Trotman BW, Soloway RD. Pigment vs cholesterol cholelithiasis: bacteriology of gallbladder stone, bile, and tissue correlated with biliary lipid analysis. Am J Dig Dis 1978; 23:877–882. 112. Moore EW, Celic L, Ostrow JD. Interactions between ionized calcium and sodium taurocholate: bile salts are important buffers for prevention of calcium containing gallstones. Gastroenterology 1982; 83:1079–1089. 113. Sutor DJ, Wilkie LI, Jackson MJ. Ionized calcium in pathological human bile. J Clin Pathol 1980; 33:86–88. 114. PercyRobb IW. Contribution of biliary lipids to calcium binding in bile. Gastroenterology 1980; 78:696–702. 115. Rajogopalan N, Lindenbaum S. The binding of Ca2+ to taurine and glycineconjugated bile salt micelles. Biochim Biophys Acta 1982; 711:66–74. 116. Altenbach C, Sellig J. Ca2+ binding to phosphatidylcholine bilayers as studied by deuterium magnetic resonance: evidence for the formation of a Ca2+ complex with two phospholipid molecules. Biochemistry 1984; 23:3913–3920. 117. Verdugo P, DeyrupOlsen I, Aitken M, et al. Molecular mechanism of mucin secretion: the role of intragranular charge shielding. J Dent Res 1987; 66:506–508. 118. Cummings SA, Hofmann AF. Physiologic determinants of biliary calcium secretion in the dog. Gastroenterology 1984; 87:664–673. 119. Moore EW. Biliary calcium and gallstone formation. Hepatology 1990; 12:206S–214S. 120. Gallinger S, Harvey PRC, Petrunka CN, Strasberg SM. Effect of binding of ionize calcium on the in vitro nucleation of cholesterol and calcium bilirubinate in human gallbladder bile. Gut 1986; 27:1382–1386. 121. Shen GR, Li WH, Soloway RD, Wen J, Wu JG. Cu2+ increases solubility of black Cabilirubinate complexes in vitro: possible protection against gallstones (abstr). Clin Res 1993; 41:A159. 122. Schoenfield LJ, Sjovall J, Sjovall K. Bile acid composition of gallstones from man. J Lab Clin Med 1966; 68:186–194. 123. Yang Z, Soloway RD, Weng S, Zhang X, Taylor DR, Wu JG. The growth of periodic calcium bilirubinate rings following and superimposed upon the fractal (chaotic) precipitations of calcium deoxycholate in an agar gel system. Gastroenterology 1998; 114:A550. 124. Womack NA, Zeppa R, Irvin GL III. The anatomy of gallstones. Ann Surg 1963; 133: 937–945. 125. Sheen PC, Lee KT, Liu YE. Mucin content in gallbladders with brown pigment stones or combination stones with a brown periphery. Digestion 1998; 599:660– 664. 126. Been JM, Bills PM, Lewis D. Electron probe microanalysis in the study of gallstones. Gut 1977; 18:836–842. 127. Been JM, Bills PM, Lewis D. Microstructure of gallstones. Gastroenterology 1979; 76:548–555. 128. Maki T, Matsushiro T, Suzuki N, Nakamura N. Role of sulfated glycoprotein in gallstone formation. Surg Gyn Obstet 1974; 132:846–853. 129. Lamont JT, Ventola AS, Trotman BW, Soloway RD. Mucin glycoprotein content of human pigment gallstones. Hepatology 1983; 3:377–382. 130. Sutor DJ, Wooley SE. The organic matrix of gallstones. Gut 1974; 15:487–489. 131. Hay DW, Carey MC. Pathophysiology and pathogenesis of cholesterol gallstone formation. Semin Liver Dis 1990; 10:159–170.
Page 164
132. Nalbone G, Lafont HJ, Vigne JL, Domingo N, Hauton JC. Purification of the human anion polypeptide fraction of the apobile lipoprotein complex by zonal ultracentrifugation. Lipids 1985; 20:884–889. 133. Shimuzu S, Sabsay B, Veis A, Ostrow JD, Rege RV, Dawes LG. Isolation of an acid protein from cholesterol stones which inhibits the precipitation of calcium carbonate in vitro. J Clin Invest 1989; 84:1990–1996. 134. Okido M, Shimuzu S, Ostrow JD, Nakayama F. Isolation of a calciumregulatory protein from black pigment gallstones: similarity with a protein from cholesterol gallstones. Hepatology 1992; 15:1079–1085. 135. Kestell MF, Sekijima J, Lee SP, Park HZ, Long M, Kaler EW. A calcium binding protein in bile and gallstones. Hepatology 1992; 16:1315–1321. 136. McPherson A, Shlichta P. Heterogeneous and epitaxial nucleation of protein crystals on mineral surfaces. Science 1988; 239:385–387. 137. Maki T. Pathogenesis of calcium bilirubinate gallstone: role of E. coli b glucuronidase and coagulation by inorganic ions, polyelectrolytes and agitation. Ann Surg 1966; 164: 90–100. 138. Costerton JW, Irvin RT. The bacterial glycocalyx in nature and disease. Annu Rev Microbiol 1981; 35:299–324. 139. Tabata M, Nakayama F. Bacteria and gallstones: etiological significance. Dig Dis Sci 1981; 26:218–224. 140. Stewart L, Smith AL, Pellegrini CA, et al. Pigment gallstones form as a composite of bacterial microcolonies and pigment solids. Ann Surg 1987; 206:242–250. 141. Robins SJ, Fasulo JM, Patton GM. Lipids of pigment gallstones. Biochim Biophys Acta 1982; 712:21–25. 142. Good LI, Edell SL, Soloway RD, Trotman BW, Mulhern C, Arger PA. Ultrasonic properties of gallstones. Effect of stone size and composition. Gastroenterology 1979; 77: 258–263. 143. Broomfield PH, Chopra R, Sheinbaum RC, Bonorris GG, Silverman A, Schoenfield LJ, Marks JW. Effects of ursodeoxycholic acid and aspirin on the formation of lithogenic bile and gallstones during loss of weight. New Engl J Med 1988; 319:1567–1572. 144. Ros E, Navarro S, Bru C, GarciaPuges A, Valderrama R. Occult microlithiasis in idiopathic acute pancreatitis: prevention of relapses by cholecystectomy or ursodeoxycholic acid therapy. Gastroenterology 1991; 101:1701–1709.
Page 165
8— Hepatic Metabolism of Cholesterol, Bile Salts, and Phospholipids Douglas M. Heuman and Z. Reno Vlahcevic Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia I— Introduction The major lipids of bile are cholesterol, bile salts, and phosphatidylcholine. The liver plays a key role in synthesis and metabolism of each. In this chapter we summarize briefly the metabolic pathways and regulatory mechanisms involved in synthesis of these lipids as well as general aspects of their uptake and elimination. We also discuss the factors responsible for maintenance of hepatic homeostasis of cholesterol and bile salts, perturbations of which may contribute to pathogenesis of cholesterol gallstone disease, cholestasis, and hypercholesterolemia. II— Cholesterol A— Biology of Cholesterol Cholesterol is of critical importance in mammalian cell function. In humans all nucleated cells have the capacity to synthesize cholesterol. Because of the liver's size and its need for cholesterol for lipoprotein synthesis, bile salt synthesis, and bile secretion, the liver is quantitatively the most important site of cholesterol synthesis in most species (1). The majority of free cholesterol in cells is contained in membranes, especially the plasma membrane. Although cholesterol is very poorly watersoluble, its single hydroxy group constitutes a polar moiety. Cholesterol partitions into phospholipid bilayers, where it intercalates between phospholipid acyl chains, its hydroxy group interacting with phosphate and water at the lipidwater interface. Cholesterol stabilizes membranes, causing phospholipids to pack together more tightly (condensation) and limiting mobility of the phospholipid acyl chains (reduced fluidity). At the same time, the presence of cholesterol prevents phospholipids from forming immobile crystalline aggregates at cool temperatures (inhibition of liquidgel transition) and thereby permits cells to function over a broad range of temperatures. Cholesterol also dramatically reduces the susceptibility of phospholipid bilayers to disruption by surfactants, such as bile salts, and reduces bilayer permeability to small polar molecules. An appropriate membrane cholesterol concentration is necessary for the normal function of many membranespanning proteins, including receptors and transporters (2).
Page 166
B— Biosynthesis Cholesterol synthesis is a threepart process (Fig. 1). Initially, three acetate molecules derived from oxidation of fatty acids or carbohydrate are combined to form mevalonate, which then is decarboxylated to yield a branched fivecarbon isoprene, the building block from which sterols are constructed (Fig. IA). Next, six isoprene units are joined together in a series of reactions to produce squalene, a longchain unsaturated hydrocarbon (Fig. 1B). Squalene condenses internally at four points in a chain reaction that forms the 30carbon parent sterol, lanosterol, and lanosterol then undergoes a series of decarboxylations, isomerizations, and reductions to yield cholesterol (Fig. 1C). The principal ratelimiting step in the cholesterol biosynthetic pathway is synthesis of 3hydroxy3methylglutaryl coenzyme A (HMGCoA) from acetyl coenzyme A and its subsequent reduction to form mevalonate. The two enzymes involved in this process, cytosolic HMGCoA synthase (HMGCoAS) and microsomal HMG CoA reductase (HMGCoAR), constitute major sites of regulation of cholesterol synthesis. These enzymes are subject to complex regulation by a variety of effectors at many levels, including gene transcription, translational efficiency, mRNA turnover, protein turnover, and posttranslational modification such as phosphorylation/dephosphorylation (3–5). Distal enzymes in the cholesterol biosynthetic pathway that may also contribute to regulation of cholesterol biosynthesis include farnesyl diphosphate synthase and squalene synthase. All four of these enzymes are downregulated at the level of gene transcription in a negative feedback manner by cholesterol and various cholesterol oxidation products (oxysterols). This regulation is mediated via a short sequence in the 5'flanking region of the gene termed the sterol regulatory element (SRE) (6). An identical SRE in the 5'flanking region of the lowdensitylipoprotein (LDL) receptor gene permits uptake of preformed lipoprotein cholesterol and de novo cholesterol synthesis to be regulated coordinately. The SRE is activated by binding of a basic helixloophelix leucinezipper transcription factor. This factor is a fragment derived from cleavage of a larger peptide called the SREbinding protein (SREBP), a constitutive component of the smooth endoplasmic reticulum. Initial proteolytic cleavage of SREBP, which is ratedetermining for release of the transcription regulatory fragment, is catalyzed by a sterolsensitive enzyme, SREBP cleavage activation protein (SCAP). Depletion of cholesterol in the smooth endoplasmic reticulum produces a physical alteration in this membrane that causes SCAP to become activated and to cleave SREBP. Following a second, nonregulated cleavage, the Nterminal fragment of SRE is released. This regulatory fragment travels to the nucleus, binds to the SRE, and increases transcription of the associated genes, as illustrated in Fig. 2 (6). C— Esterification, Storage, and Lipoprotein Secretion Cholesterol esters are synthesized by attaching a fatty acid to the 3betahydroxy moiety of cholesterol. This modification eliminates the slight polarity of cholesterol and changes its physical chemical behavior. Cholesterol esters, unlike free cholesterol, have little affinity for phospholipid bilayers and instead tend to form oil drops along with other neutral lipids such as triglycerides. In lipoproteins, free cholesterol resides in the surface amphiphilic monolayer of phospholipid and proteins, while cholesterol esters are present along with triglycerides almost entirely in the hydrophobic core (7). Cholesterol esterification is a means whereby excess hepatic cholesterol can be diverted either to storage in cytosolic oil droplets or to export in very low density lipoproteins (VLDL). Cholesterol esterification is catalyzed by acylcoenzyme A:cholesterol acyltransferase (ACAT), a microsomal enzyme. At least two forms of ACAT exist with different expression in different tissues. ACAT1, the first form identified, is important in adrenals and macrophages but was found to be expressed poorly in liver (8); knockout of this gene in mice had no effect on hepatic cholesterol esterification (9). Recently a second form of ACAT, termed ACAT2, has
Page 167
Figure 1 Pathway and major intermediates of cholesterol biosynthesis. See text for details. A. Synthesis of isopentenyl pyrophosphate, the basic building block of sterols. Synthesis of mevalonate by HMGCoA reductase is the major ratelimiting step in the cholesterol biosynthetic pathway. B. Condensation of six isopentenyl pyrophospates results in the longchain hydrocarbon squalene. C. Internal condensation of squalene yields lanosterol and ultimately cholesterol.
Page 168
Figure 2 Transcriptional regulation of major genes regulating cholesterol uptake and biosynthesis. SRE, sterol regulatory element (gene promoter); SREBP, sterol regulatory element binding protein; SCAP, SREBP cleavage activating protein; HMGCoA, 3hydroxy3methylglutarylcoenzyme A. See text for details.
been identified, cloned and sequenced by several groups (10–12). ACAT2 is strongly expressed in liver and intestine and is a likely candidate for the major hepatic cholesterol esterifying enzyme. With this discovery, understanding of the regulation of ACAT on a molecular level is expected to progress rapidly. When demand for cholesterol increases within the hepatocyte, cholesterol can be retrieved from oil droplets via hydrolysis of cholesterol esters. The enzyme responsible for mobilization of this stored cholesterol, neutral cytosolic cholesterol ester hydrolase (CEH), has recently been cloned and characterized (13). CEH has been shown to be downregulated in response to feeding of cholesterol or increased cholesterol synthesis and is upregulated by maneuvers that inhibit cholesterol synthesis or enhance cholesterol degradation (14). The liver exports cholesterol to the tissues via synthesis and secretion of VLDL. These large particles have a low surfacetovolume ratio and contain a high proportion of cholesterol esters and triglycerides. The regulation of VLDL production is not well understood, but cholesterol ester availability appears to be an essential element, since manipulations that inhibit cholesterol synthesis or esterification clearly reduce VLDL secretion (15). D— Lipoprotein Cholesterol Uptake The liver takes up circulating lipoprotein cholesterol (predominantly cholesterol esters) via two major pathways employing receptormediated endocytosis. LDLs are recognized by a receptor specific for apoproteins B and E, more commonly termed the LDL receptor. Chylomicron
Page 169
remnants are taken up via a selective apoE receptor. The ingested particles are imported via clathrincoated pits to endocytotic vesicles (16). Ultimately the cholesterol esters are transferred to liposomes, where they are acted on by a cholesterol ester hydrolase whose activity is optimal at an acidic pH. This acidic CEH differs from the neutral cytosolic cholesterol ester hydrolase and is not thought to play a regulatory role in cholesterol homeostasis. Subsequent trafficking of lipoprotein cholesterol to various sites, including the cell plasma membrane, appears to involve both vesicular transport and cytosolic carrier proteins (17). The liver also takes up cholesterol esters from high density lipoproteins (HDL). Much of HDL cholesterol originates in peripheral tissues, and the delivery of this cholesterol to the liver for catabolism to bile acids or excretion into bile constitutes a pathway of ''reverse cholesterol transport" that plays an essential role in prevention of atherosclerosis. A recently identified receptor, termed the scavenger receptor B1 (SRB1), binds circulating HDL at the hepatocyte sinusoidal membrane and mediates transfer of cholesterol esters between HDL particles and hepatocytes (18). The HDL particles do not appear to be ingested. Rather, the transfer of cholesterol esters to the cell occurs selectively, without uptake of the apoproteins, by a mechanism that is currently unknown (19). Both expression of SR B1mRNA and rate of uptake of HDL cholesterol esters in hepatocytes are downregulated following treatment with ethinyl estradiol or cholesterol feeding; interestingly, an opposite pattern of regulation is noted in Kupffer cells (20). E— Elimination The liver is the only organ that plays a significant role in elimination of cholesterol from the body. Two principal pathways are responsible. First, cholesterol can be oxidized to form bile salts. Second, free cholesterol can be secreted directly into bile along with phosphatidylcholine in the form of large unilamellar vesicles. Bile salt synthesis and its regulation are discussed in some detail below. The processes by which cholesterol is transported to the canalicular membrane, packaged into vesicles, and secreted into bile are of critical importance in pathogenesis of gallstones and are discussed in a later chapter. III— Bile Salts A— Biology of Bile Salts Bile salts (also termed bile acids) are anionic detergents synthesized from cholesterol. They are produced only in the liver. Synthesis of bile acids represents a major pathway for elimination of cholesterol from the body. About half of daily cholesterol turnover in humans is attributable to synthesis of bile acids. Bile salts serve a number of biological functions. Secretion of bile salts drives bile secretion, creating an excretory route for elimination of many metabolic wastes. Bile salts also stimulate canalicular secretion of cholesterol and lecithin into bile. In the digestive tract, bile salts contribute to emulsification of lipids, activation of digestive lipases, and solubilization of products of lipid digestion such as monoglycerides. Bile salts are actively and efficiently reabsorbed from the distal small intestine, taken up by the liver, and resecreted into bile. The flux of bile salts in this enterohepatic circulation regulates key ratelimiting enzymes of bile salt synthesis and thereby controls the rate of cholesterol degradation. Thus perturbations of the enterohepatic circulation of bile salts can have profound effects on cholesterol homeostasis. B— Bile Salt Synthesis Structures of the primary bile salts of humans, cholic acid and chenodeoxycholic acid, are illustrated in Fig. 3. Studies of Bjorkhem (21) in the 1970s indicated that the first steps in bile
Page 170
Figure 3 Pathways of bile acid synthesis, showing key ratelimiting enzymes. Cyp7A, cholesterol 7 hydroxylase; Cyp27, sterol 27hydroxylase; Cyp12, sterol 12 hydroxylase; Cyp7B, oxysterol 7 hydroxylase.
Page 171
acid synthesis involve ring modifications. The initial step in this classical pathway of bile acid biosynthesis is hydroxylation in the 7 position. Following conversion to the 3 ketodelta 4 derivative, an additional hydroxylation may occur in the 12 position. Next, modification of the side chain begins with 27hydroxylation, followed by 24 hydroxylation and oxidative cleavage at C24. Finally, reduction of the keto group and double bond yields a saturated ring with a 3 hydroxy group and a 5 hydrogen. The ratedetermining enzyme of this classic "neutral" pathway of bile acid synthesis, cholesterol 7 hydroxylase, has been extensively characterized during the past 10 years. Cholesterol 7 hydroxylase is a unique microsomal cytochrome P450 isoenzyme (Cyp7A) (22). It is highly regulated. Expression of Cyp7A is downregulated by bile acids in the enterohepatic circulation at the level of gene transcription in a negative feedback inhibitory manner (23). The feedback inhibitory potency of different bile acids varies with their relative hydrophobicity (24). Recent studies in isolated cultured hepatocytes indicate that bile acids may exert their effects indirectly through activation of protein kinase C isoenzymes (25), leading in turn to a cascade of activation of a series of regulatory kinases. The precise molecular basis for bile acid regulation of cholesterol 7 hydroxylase remains unclear at this time. In some species such as rat, Cyp7A expression also is upregulated at the level of gene transcription by cholesterol (26). In other species such as hamster and rabbit, transcriptional regulation of Cyp7A by cholesterol is not observed, and under normal circumstances this does not appear to be of major importance in most humans. However upregulation of C7 H by cholesterol may account in part for the ability of some individuals to tolerate diets containing very high levels of cholesterol (27). Other regulatory factors in animal studies include insulin, glucocorticoids, and thyroid hormone (28). In rats, infusion of taurocholate intraduodenally produced marked downregulation of Cyp7A, whereas an equimolar infusion delivered intravenously was ineffective (29). These studies suggested that an additional intestinal factor may be involved in mediating or modulating bile acid biofeedback. Recent evidence indicates that an alternate pathway may, under some circumstances, account for a substantial fraction of bile acid biosynthesis. In this alternate pathway, sidechain oxidation precedes nuclear modifications. The initial step in this pathway is hydroxylation of the side chain at position 27 (30). The 7 hydroxylase activity in this pathway is not attributable to Cyp7A; rather, a second hydroxylase, termed oxysterol 7 hydroxylase or Cyp7B, is responsible (31). Cyp7B is also a microsomal cytochrome P450; unpublished preliminary data from our laboratories suggest that Cyp7B, like Cyp7A, may be regulated by bile salts in a negative feedback inhibitory manner. Sterol 27 hydroxylase, the initial enzyme in the alternate pathway of bile acid biosynthesis, is a mitochondrial cytochrome P450 (Cyp27). Like cholesterol 7 hydroxylase, it is inducible at the level of gene transcription following bile salt depletion and is suppressed by bile salt supplementation in the diet, though quantitatively the changes in activity are less pronounced for Cyp27 than for Cyp7A (32). Unlike Cyp7A, which is expressed only in liver, Cyp27 is expressed in many extrahepatic tissues as well. It has been suggested that oxidation of cholesterol by Cyp27 in these extraheptic sites may be a key step in the pathway of "reverse cholesterol transport" by which cholesterol is mobilized in the periphery, delivered to the liver, and eliminated. Cyp27 also is responsible for initiating sidechain oxidation in the classic pathway of bile acid synthesis and thus is required for both major pathways of bile acid synthesis. Genetic deficiency of Cyp27 results in cerebrotendinous xanthomatosis, and accelerated atherosclerosis (33). Patients with this disorder lack bile acids; instead, large amounts of polyhydroxylated C27 sterols (bile alcohols) are formed. The strongest evidence supporting the existence of an alternate pathway of bile acid synthesis comes from transgenic "knockout" mice in which the Cyp7A gene has been deleted. Although they require bile acid supplements in early life, these mice at maturity synthesize primary bile salts in sufficient quantities to survive and digest normally (34). Similarly in bile fistula rats, administration of the squalene synthase inhibitor squalestatin was found to suppress
Page 172
Cyp7A gene expression by more than 95% while preserving Cyp27; squalestatin reduced bile acid synthesis by less than 50% (35). Curiously, in the latter studies, bile acid synthesis was observed to gradually return to normal despite persistent suppression of Cyp7A and stable activity of Cyp27, suggesting the possibility of a third potentially ratelimiting step in bile acid biosynthesis. C— Bile Salt Pool Composition The proportions of cholic and chenodeoxycholic acids in the bile salt pool vary widely. The introduction of a 12 hydroxy moiety is catalyzed by a microsomal cytochrome P450, sterol 12 hydroxylase (Cyp12), whose activity appears to be a major determinant of the cholic/chenodeoxycholic ratio. The cDNA for this enzyme in rats recently has been cloned and sequenced (36). Preliminary studies of its regulation in rats (37) indicate that, like cholesterol 7 hydroxylase, its expression was downregulated by hydrophobic bile salts at the level of gene transcription. However, in contrast to cholesterol 7 hydroxylase, sterol 12 hydroxylase activity was induced by inhibitors of cholesterol synthesis and suppressed by cholesterol feeding. Most bile salts are efficiently amidated in human liver with glycine or taurine. This amidation (commonly termed conjugation) lowers the pKa from about 5 for the unconjugated bile salt to 3.9 for glycine conjugates or less than 1 for taurine conjugates (38). Amidated bile salts are secreted into bile and, after a period of storage and concentration in the gallbladder, are expelled into the proximal intestine. Between 95 and 99% of secreted bile salt normally is reabsorbed from the intestine, predominantly via sodiumdependent active transport in the distal ileum. A small fraction of bile salt escapes ileal absorption and reaches the colon, where bile salts are deconjugated by anaerobic bacteria. Several species of grampositive anaerobes, including some species of Clostridium and Eubacterium, further metabolize bile acids to remove the 7hydroxy group, as shown in Fig. 4. This apparently simple metabolic step requires a series of enzymecatalyzed reactions. The responsible bacterial operon has been extensively characterized (39). The resulting secondary bile acids, deoxycholic acid (from 7 dehydroxylation of cholate) and lithocholic acid (from 7 dehydroxylation of chenodeoxycholate) are absorbed by passive diffusion, return to the liver, and are reamidated. Deoxycholate conjugates become part of the circulating bile salt pool. Lithocholic acid is further modified by sulfation and glucuronidation; although secreted into bile, these metabolites are poorly reabsorbed from the intestine and are largely lost in the feces. The metabolic effects of bile salts are determined to a large extent by their relative hydrophobicity. Secondary bile salts are more hydrophobic than the corresponding primary bile salts. Enrichment of the bile salt pool with deoxycholate leads to increased biliary cholesterol secretion (40) and more potent suppression of cholesterol 7 hydroxylase (24). In some individuals, enrichment of the intestinal flora with bacteria capable of bile salt 7dehydroxylation is associated with a high proportion of biliary deoxycholate and increased secretion of supersaturated bile; suppression of these bacteria with antibiotics was found to reduce deoxycholate and normalize biliary cholesterol saturation (41). IV— Inborn Errors of Cholesterol and Bile Acid Synthesis A— Inborn Errors of Cholesterol Synthesis The SmithLemliOpitz syndrome is an autosomal recessive disorder that was described as a clinical entity in 1964 but was only recently fully characterized as a disorder of hepatic cho
Page 173
Figure 4 Origins of major primary and secondary bile acids of humans. Primary bile acids, cholic and chenodeoxycholic, are synthesized de novo from cholesterol. Secondary bile acids, deoxycholic and lithocholic, are produced in the colon by bacterial 7 dehydroxylation.
lesterol synthesis (42). Clinically the disease is characterized by microcephaly, poor growth, dysmorphic facial features, syndactyly, polydactyly, genital and endocrine disorders, and mental retardation (43). After cystic fibrosis, it is the most common autosomal recessive disorder in the United States. Biochemically, the syndrome is characterized by increased tissue concentration of 7dihydrocholesterol due to a defect in 3 hydroxycholesterol 7reductase, the last enzymatic step in cholesterol synthesis (44,45). The deficiency of this enzyme leads to a decrease in serum cholesterol and its replacement with dihydrocholesterol. This combination of biochemical abnormalities leads to the clinical manifestations. A diet high in cholesterol and bile salts can partially correct cholesterol deficiency while suppressing cholesterol synthesis
Page 174
and consequently may improve the biochemical abnormalities associated with the disease (46,47). B— Inborn Errors of Bile Acid Synthesis Several inborn errors of bile acid biosynthesis have been identified. These disorders are very rare and can be divided into a group in which there is a deficiency of enzymes participating in the modification of steroid nucleus and another group in which there is a deficiency in the enzyme participating in the side chain cleavage of cholesterol or bile acid intermediates (Table 1). Clinical disorders have been reported in association with three bile acid biosynthetic enzymes that act on the steroid nucleus: 3 hydroxyC27steroid oxidoreductase (48), 43oxosteroid5 reductase (classic pathway) (49), and oxysterol 7 hydroxylase (alternative pathway) (50). Clinically all three of these defects are associated with cholestasis and tend to progress to cirrhosis and early death in infancy. Cholestatic features associated with these diseases are presumably due to accumulation of hepatotoxic intermediates and metabolites in the liver. Therapy with primary bile acids (chenodeoxycholic or cholic), with the intent of restoring the bile salt pool and suppressing endogenous bile acid synthesis, has been successful in ameliorating the course of disease and leading to histological improvement (51). Deficiency in sterol 27hydroxylase, an enzyme that initiates sidechain oxidation, results in a disease called cerebrotendinous xanthomatosis (CTX). This is an autosomal recessive disorder characterized by xanthomas, progressive neurological dysfunction, premature atherosclerosis, and eventual death (52). These patients have severely reduced bile acid synthesis, a marked increase in biliary secretion of bile alcohols (multiply hydroxylated cholesterol derivatives lacking a carboxylic acid in the side chain), and increased cholesterol synthesis. In patients with CTX, there is also a marked accumulation of cholestanol in the tissues and plasma (52). Recently, the molecular basis of CTX has been established, following cloning and sequencing of sterol 27hydroxylase (33). Patients with CTX have been shown to have a variety of point mutations or deletions of sterol 27hydroxylase, which lead to loss of enzyme activity and are responsible for the observed biochemical abnormalities. CTX can be treated by administration of cholic or chenodeoxycholic acid to repress bile acid synthesis, thereby reducing accumulation of cholesterol and bile alcohols (53). Also, some improvement in the manifestations of disease was reported with the use of lovastatin, an inhibitor of the synthesis of HMGCoA reductase and cholesterol (54). If the disease is recognized and treated early, the accu Table 1 Inborn Errors of Bile Acid Synthesis Disease
Deficient enzyme
3 5C27, hydroxy oxidoreductase deficiency
Same
Cholestatic liver disease
Primary bile acids, liver transplantation
Same
Cholestatic liver disease
Primary bile acids, liver transplantation
Same
Cholestatic liver disease
Liver transplantation
Neurological disorder; premature atherosclerosis
Primary bile acids
43oxosteroid5 reductase Oxysterol 7 hydroxylase
Cerebrotendinous xanthomatosis Sterol 27 (CTX) hydroxylase
Clinical manifestation
Therapy
Page 175
mulation of sterols in the central nervous system as well as neurological damage can be prevented. V— Cholesterol and Bile Salt Homeostasis in Health and Disease Figure 5 summarizes the pathways of cholesterol metabolism in the hepatocyte. Net cholesterol input to the hepatocyte comes from de novo synthesis or uptake of lipoproteins; elimination proceeds via either lipoprotein secretion (VLDL), bile salt synthesis, or biliary secretion of free cholesterol. Within the hepatocyte, cholesterol may be compartmented in various membranes, predominantly the plasma membrane, or may be reversibly stored in lipid droplets in the form of cholesterol esters. Changes in one pathway of cholesterol metabolism frequently lead to compensatory changes in other pathways. Thus, for example, moderate inhibition of cholesterol synthesis with inhibitors of HMGCOA reductase leads to upregulation of this enzyme at the level of gene transcription (55) as well as upregulation of LDL receptors (56). More profound inhibition of cholesterol synthesis in some species may lead to downregulation of cholesterol 7 alpha hydroxylase with suppression of bile acid synthesis (57,58). Another example is the response to feeding of bile acidbinding resins such as cholestyramine. These agents cause increased fecal elimination of bile salts, leading to upregulation of bile acid synthesis. The resultant increase in cholesterol degradation is, in turn, accompanied by upregulation of both HMGCoA reductase and LDL receptor (59). The magnitude of a particular response often varies both between and within species. Genetic variations in regulation of individual pathways of cholesterol homeostasis may explain why some individuals placed on lowcholesterol diets exhibit a decline in circulating LDL
Figure 5 Overview of pathways of hepatic cholesterol metabolism.
Page 176
cholesterol, whereas others show no hypolipidemic response to wide variations in dietary cholesterol (27,60). Genetic variations may also help to explain individual heterogeneity in response to hypolipidemic drugs. Changes in pathways of cholesterol homeostasis that increase the availability of free cholesterol within the hepatocyte frequently lead to increased biliary cholesterol secretion. The mechanisms by which biliary vesicles are assembled and secreted are poorly understood, as are the factors that dictate the absolute rate of biliary cholesterol secretion and the cholesterol: phosphatidylcholine ratio of biliary vesicles. These two parameters are critical in determining the degree to which bile is saturated with cholesterol. It has been proposed that a "metabolically active" pool of free cholesterol in the hepatocyte may be the principal determinant of biliary cholesterol secretion (61,62). Attempts to define this regulatory cholesterol pool by kinetic techniques have yielded inconclusive results. Nevertheless, this hypothesis provides a useful conceptual framework for explaining many of the factors that predispose to cholesterol gallstone disease. A clearer understanding of the regulation of biliary cholesterol in health and disease must await elucidation of the cellular and biochemical mechanisms by which cholesterol is delivered to the canalicular membrane and packaged into vesicles for secretion into bile. VI— Phosphatidylcholine A— Biology of Phosphatidylcholine Phospholipids are the fundamental components of all biological membranes. These compounds are very poorly watersoluble lipids possessing a strongly hydrophilic head group and two strongly hydrophobic hydrocarbon tails. In aqueous systems, phospholipids selfaggregate to form fluid planar bilayers. In these bilayers, the individual phospholipids are oriented perpendicularly with their polar head groups exposed at the bilayer surface and nonpolar tails sequestered (63). Phospholipid bilayers are relatively impermeable to polar molecules, and this property is crucial to their role of demarcating cellular and subcellular compartments with distinct composition and function. Although a wide variety of phospholipids are present in cells, nearly all of the phospholipid in bile is of a single class, phosphatidylcholine (also termed lecithin). The basic structure of phosphatidylcholine (PC) is illustrated in Fig. 6. Glycerol forms a backbone, to which two fatty acids are esterified at adjacent carbons. In general, in mammals the fatty acid on the first carbon (sn1) is saturated, whereas the second (sn2) is unsaturated. The third carbon is esterified to phosphate, which, in turn, is esterified to the quaternary amine choline. Phosphatidylcholine is a zwitterion; at physiological pH, the phosphate group carries a negative charge that balances the positive charge of the choline moiety, and the net charge is thus neutral. B— De Novo Synthesis The great majority of phosphatidylcholine secreted into bile is synthesized de novo in the liver. PC synthesis in hepatocytes begins with the assembly of diacylglycerol. This is followed by addition of choline phosphate, which can occur via two pathways (64), as shown in Fig. 7. In the predominant pathway, choline phosphate is transferred directly to diacylglycerol; in the alternate methylation pathway, phosphatidylethanolamine is synthesized and then converted to PC by addition of 3 N methyl groups. Biosynthesis of diacylglycerol starts with transesterification of an sn1 fatty acid from coenzyme A to glycerol3phosphate. Two distinct glycerol3phosphate acyltransferases are involved, one located in mitochondria and the other in the endoplasmic reticulum. This is followed by addition of a second fatty acid at the sn2 position, catalyzed by a third enzyme to produce phosphatidic acid. The regulation of this process and the mechanisms by which specific fatty acids are selected for incorporation at each step are not well understood. Finally,
Page 177
Figure 6 Structure of phosphatidylcholine. Fatty acyl substituents in this instance include palmitic acid (saturated) in the sn1 position and linoleic acid (unsaturated) in the sn2 position.
phosphatidic acid phosphohydrolase cleaves the sn3 phosphate to release diacylglycerol. Phosphatidic acid phosphohydrolase normally resides in the cytoplasm in its inactive form; in the presence of phosphatidic acid, it translocates to the endoplasmic reticulum and becomes active. Most of the choline employed for phosphatidylcholine biosynthesis derives from dietary sources. Choline is an essential nutrient in the human diet; it is actively taken up from the gut and is transported into the hepatocyte actively by both sodiumdependent and sodiumindependent carriers. In the liver, a fraction of choline is oxidized to betaine and employed as a methyl donor for methionine synthesis; the majority, however, is employed in synthesis of phospholipid. The activation of choline for phospholipid synthesis begins with phosphorylation by choline kinase. This is followed by transesterification with cytidine triphosphate to form CDP choline, catalyzed by the key enzyme CTP: phosphocholine cytidylyltransferase (CCT). In the final step of this pathway, CDPcholine condenses with 1,2 diacylglycerol to yield phosphatidylcholine (64). This reaction is catalyzed by CDPcholine: 1,2diacylglycerol phosphotransferase, a predominantly microsomal enzyme that to date has not been well characterized in mammals. CCT is the major ratelimiting step in PC biosynthesis (65). It is upregulated by CDPcholine (feedforward regulation) and downregulated by PC (feedback inhibition). The major site of regulation is posttranslational. CCT normally resides in the cytoplasm in its inactive form; when phosphorylated, it translocates to the endoplasmic reticulum and becomes active. Phosphorylation is largely mediated via protein kinase C, which, in turn, is activated by diacylglycerol. However, under extreme conditions of phosphatidylcholine depletion, such as partial hepatectomy, upregulation of CCT gene expression has been demonstrated. Recently the existence of two distinct human CCT genes, termed CCT and CCT , has been demonstrated
Page 178
Figure 7 Pathways of synthesis of phosphatidylcholine (PC) in the liver. G3P, glycerol3phosphate; PA, phosphatidic acid; DAG, 1,2diacylglycerol; FA, fatty acyl group; PE, phosphatidylethanolamine; CK, choline kinase; CCT, CTP:phosphocholine cytidyltransferase; CP, CDPcholine:1,2diacylglycerol phosphotransferase; G3PA, glycerol3phosphate acyltransferase; PAPH, phosphatidic acid phosphohydrolase; PEMT, phosphatidylethanolamine methyltransferase.
(66). The latter is present predominantly in placenta and testis; its role, if any, in the liver is not known. The pathway described above exists in virtually all nucleated human cells; the ability to synthesize phosphatidylcholine is critical for cell growth and proliferation. A second pathway to production of phosphatidylcholine is found almost exclusively in the liver. In this alternate pathway, the cell first synthesizes phosphatidylethanolamine, which then is converted to phosphatidylcholine via a series of three methylations. Two phosphatidylethanolamineNmethyltransferases (PEMT) catalyze these steps (67,68). The first, PEMT1, is found on the endoplasmic reticulum; the second, PEMT2, is confined to an ERlike membrane fraction associated with mitochondria. The role and regulation of this alternate pathway are not fully understood. In cell culture, overexpression of PEMT suppresses CCT gene expression (69). PEMT2 overexpression also inhibits cell proliferation by a mechanism that appears to be independent of phosphatidylcholine production (70). However knockout of the PEMT2 gene in mice had no discernible effect on overall phenotype, liver morphology, or plasma or biliary lipid composition (71). C— Uptake, Hydrolysis, and Reacylation Although a large amount of phospholipid is delivered to the liver in lipoproteins, kinetic studies suggest that very little is taken up intact. PC ingested during receptor mediated uptake of LDL and chylomicron remnants is degraded in lysosomes. PC of HDL is hydrolysed by hepatic lipase on sinusoidal endothelial cells, yielding lysolecithin and fatty acid, which are then taken up by the hepatocyte via pathways that are not yet known. Reacylation of lysolecithin in many tissues utilizes a 1acyl glycerophosphocholine:acylCoA acyltransferase that is present in mi
Page 179
crosomes and is specific for polyunsaturated acyl groups (72). Deacylationreacylation permits modification of the fatty acid composition of the PC molecule and may be important in determining the relative proportions of different molecular species of PC within the hepatocyte. Acknowledgments This work was supported in part by grants from the U.S. Department of Veterans Affairs and National Institutes of Health, PO1DK38030. The authors wish to thank Mary Cousins and Cecile Rock for their expert secretarial assistance. References 1. J.M. Dietschy, S.D. Turley, and D.K. Spady. Role of liver in the maintenance of cholesterol and low density lipoprotein homeostasis in different animal species, including humans. J. Lipid Res. 34:1637–1659, 1993. 2. P.L. Yeagle. Cholesterol and the cell membrane. In: P.L. Yeagle, ed. Biology of Cholesterol. Boca Raton, FL: CRC Press, 1988, pp. 121–146. 3. L. Liscum, K.L. Luskey, D.J. Chin, Y.K. Ho, J.L. Goldstein, and M.S. Brown. Regulation of 3hydroxy3methylglutaryl coenzyme a reductase and its mRNA in rat liver as studied with a monoclonal antibody and a cDNA probe. J. Biol. Chem. 258:8450–8455, 1983. 4. M.S. Brown and J.L. Goldstein. Multivalent feedback regulation of HMG CoA reductase, a control mechanism coordinating isoprenoid synthesis and cell growth. J. Lipid Res. 21:505–517, 1980. 5. M.S. Brown, J.L. Goldstein, and J.M. Dietschy. Active and inactive forms of 3hydroxy3methylglutaryl coenzyme A reductase in the liver of the rat: comparison with the rate of cholesterol synthesis in different physiological states. J. Biol. Chem. 254:5144–5149, 1979. 6. M.S. Brown and J.L. Goldstein. The SREBP pathway: regulation of cholesterol metabolism by proteolysis of a membranebound transcription factor. Cell 89:331– 340, 1997. 7. D.M. Small. Lipid classification based on interactions with water. In: D.M. Small, ed. Handbook of Lipid Research: IV. The Physical Chemistry of Lipids. New York: Plenum Press, 1986, pp. 89–96. 8. T.J. Rea, R.B. DeMattos, R. Homan, R.S. Newton, and M.E. Pape. Lack of correlation between ACAT mRNA expression and cholesterol esterification in primary liver cells. Biochim. Biophys. Acta Lipids Lipid Metab. 1299:67–74, 1996. 9. V.L. Meiner, S. Cases, H.M. Myers, E.R. Sande, S. Bellosta, M. Schambelan, R.E. Pitas, J. McGuire, J. Herz, and R.V.J. Farese. Disruption of the acyl CoA:cholesterol acyltransferase gen in mice: evidence suggestion multiple cholesterol esterification enzymes in mammals. Proc. Natl. Acad. Sci. U.S.A. 93:14041– 14046, 1996. 10. P. Oelkers, A. Behari, D. Cromley, J.T. Billheimer, and S.L. Sturley. Characterization of two human genes encoding acyl coenzyme A:Cholesterol acyltransferaserelated enzymes. J. Biol. Chem. 273:26765–26771, 1998. 11. R.A. Anderson, C. Joyce, M. Davis, J.W. Reagan, M. Clark, G.S. Shelness, and L.L. Rudel. Identification of a form of acylCoA:cholesterol acyltransferase specific to liver and intestine in nonhuman primates. J. Biol. Chem. 273:26747–26754, 1998. 12. S. Cases, S. Novak, Y.W. Zheng, H.M. Myers, S.R. Lear, E. Sande, C.B. Welch, A.J. Lusis, T.A. Spencer, B.R. Krause, S.K. Erickson, and R.V.J. Farese. ACAT2, a second mammalian acylCoA:cholesterol acyltransferase: its cloning, expression and characterization. J. Biol. Chem. 273:26755–26764, 1998.
Page 180
13. S. Ghosh, D.H. Mallonee, P.B. Hylemon, and W.M. Grogan. Molecular cloning and expression of rat hepatic neutral cholesteryl ester hydrolase. Biochim. Biophys. Acta 1259:305–312, 1995. 14. S. Ghosh, R. Natarajan, W.M. Pandak, P.B. Hylemon, and W.M. Grogan. Regulation of hepatic neutral cholesteryl ester hydrolase by hormones and changes in cholesterol flux. Am. J. Physiol. Gastrointest. Liver Physiol. 274:G662–G668, 1998. 15. G.R. Thompson, R.P. Naoumova, and G.F. Watts. Role of cholesterol in regulating apolipoprotein B secretion by the liver. J. Lipid Res. 37:439–447, 1996. 16. D.K. Spady. Hepatic clearance of plasma low density lipoproteins. Semin. Liver Dis. 12:373–385, 1992. 17. L. Liscum and N.K. Dahl. Intracellular cholesterol transport. J. Lipid Res. 33:1239–1254, 1992. 18. S. Acton, A. Rigotti, K.T. Landschulz, S. Xu, H.H. Hobbs, and M. Krieger. Identification of scavenger receptor SRB1 as a high density lipoprotein receptor (see comments). Science 271:518–520, 1996. 19. S. Xu, M. Laccotripe, X. Huang, A. Rigotti, V.I. Zannis, and M. Krieger. Apolipoproteins of HDL can directly mediate binding to the scavenger receptor SR B1, and HDL receptor that mediates selective lipid uptake. J. Lipid Res. 38:1289–1298, 1997. 20. K. Fluiter, D.R. van der Westhuijzen, and T.J. van Berkel. In vivo regulation of scavenger receptor B1 and the selective uptake of high density lipoprotein cholesteryl esters in rat liver parenchymal and Kupffer cells. J. Biol. Chem. 273:8434–8438, 1998. 21. I. Bjorkhem and H. Danielsson. 1976. Biosynthesis and metabolism of bile acids in man. Prog. Liver Dis. 5:215–231, 1976. 22. M. Nishimoto, M. Noshiro, and K. Okuda. Structure of the gene encoding human liver cholesterol 7ahydroxylase. Biochim. Biophys. Acta Gene Struct. Express. 1172:147–150, 1993. 23. W.M. Pandak, Y.C. Li, J.Y.L. Chiang, E.J. Studer, E.C. Gurley, D.M. Heuman, Z.R. Vlahcevic, and P.B. Hylemon. Regulation of cholesterol 7ahydroxylase mRNA and transcriptional activity by taurocholate and cholesterol in the chronic biliary diverted rat. J. Biol. Chem. 266:3416–3421, 1991. 24. D.M. Heuman, P.B. Hylemon, and Z.R. Vlahcevic. Regulation of bile acid synthesis: III. Correlation between biliary bile salt hydrophobicity index and activities of enzymes regulating cholesterol and bile acid synthesis in the rat. J. Lipid Res. 30:1161–1171, 1989. 25. R.T. Stravitz, Y.P. Rao, Z.R. Vlahcevic, E.C. Gurley, W.D. Jarvis, and P.B. Hylemon. Hepatocellular protein kinase C activation by bile acids: Implications for regulation of cholesterol 7ahydroxylase. Am. J. Physiol. Gastrointest. Liver Physiol. 271:G293–G303, 1996. 26. M.P. Jones, W.M. Pandak, D.M. Heuman, J.Y.L. Chiang, P.B. Hylemon, and Z.R. Vlahcevic. Cholesterol 7ahydroxylase: evidence for transcriptional regulation by cholesterol or metabolic products of cholesterol in the rat. J. Lipid Res. 34:885–892, 1993. 27. F. Kern Jr. Normal plasma cholesterol in an 88yearold man who eats 25 eggs a day—mechanisms of adaptation. N. Engl. J. Med. 324:896–899, 1991. 28. H.M.G. Princen, P. Meijer, and B. Hofstee. Dexamethasone regulates bile acid synthesis in monolayer cultures of rat hepatocytes by induction of cholesterol 7a hydroxylase. Biochem. J. 262:341–348, 1989. 29. W.M. Pandak, D.M. Heuman, P.B. Hylemon, J.Y.L. Chiang, and Z.R. Vlahcevic. Failure of intravenous infusion of taurocholate to downregulate cholesterol 7a hydroxylase in rats with biliary fistulas. Gastroenterology 108:533–544, 1995. 30. I. Björkhem. Mechanism of degradation of the steroid side chain in the formation of bile acids. J. Lipid Res. 33:455–472, 1992. 31. M. Schwarz, E.G. Lund, R. Lathe, I. Björkhem, and D.W. Russell. Identification and characterization of a mouse oxysterol 7ahydroxylase cDNA. J. Biol. Chem. 272:23995–24001, 1997.
Page 181
32. Z.R. Vlahcevic, S.K. Jairath, D.M. Heuman, R.T. Stravitz, P.B. Hylemon, N.G. Avadhani, and W.M. Pandak. Transcriptional regulation of hepatic sterol 27 hydroxylase by bile acids. Am. J. Physiol. Gastrointest. Liver Physiol. 270:G646–G652, 1996. 33. J.J. Cali, C.L. Hsieh, U. Francke, and D.W. Russell. Mutations in the bile acid biosynthetic enzyme sterol 27hydroxylase underlie cerebrotendinous xanthomatosis. J. Biol. Chem. 266:7779–7783, 1991. 34. S. Ishibashi, M. Schwarz, P.K. Frykman, J. Herz, and D.W. Russell. Disruption of cholesterol 7 hydroxylase gene in mice: 1. Postnatal lethality reversed by bile acid and vitamin supplementation. J. Biol. Chem. 271:18017–18023, 1996. 35. Z.R. Vlahcevic, R.T. Stravitz, D.M. Heuman, P.B. Hylemon, and W.M. Pandak. Quantitative estimations of the contribution of different bile acid pathways to total bile acid synthesis in the rat (see comments). Gastroenterology 113:1949–1957, 1997. 36. U. Andersson, G. Eggertsen, and I. Björkhem. Rabbit liver contains one major sterol 12alphahydroxylase with broad substrate specificity. Biochim. Biophys. Acta 1389:150–154, 1998. 37. Z.R. Vlahcevic, P.B. Hylemon, K. Redford, G. Eggertsen, R.T. Stravitz, D.M. Heuman, and W.M. Pandak. Regulation of sterol 12a hydroxylase (CYP12) and cholic acid synthesis (abstr). Hepatology 28:379A, 1998. 38. A. Fini and A. Roda. Chemical properties of bile acids: IV. Acidity constants of glycineconjugated bile acids. J. Lipid Res. 28:755–759, 1987. 39. D.H. Mallonee, W.B. White, and P.B. Hylemon. Cloning and sequencing of a bile acidinducible operon from Eubacterium sp. strain VP1 12708. J. Bacteriol. 172:7011–7019, 1990. 40. N. Carulli, P. Loria, M. Bertolotti, L.M. Ponz de, D. Menozzi, G. Medici, and I. Piccagili. Effects of acute changes of bile acid pool composition on biliary lipid secretion. J. Clin. Invest. 74:614–624, 1984. 41. F. Berr, G.A. KullakUblick, G. Paumgartner, W. Münzing, and P.B. Hylemon. 7adehydroxylating bacteria enhance deoxycholic acid input and cholesterol saturation of bile in patients with gallstones. Gastroenterology 111:1611–1620, 1996. 42. G.S. Tint, M. Irons, E.R. Elias, A.K. Batta, R. Frieden, T.S. Chen, and G. Salen. Defective cholesterol biosynthesis associated with the SmithLemliOpitz syndrome. N. Engl. J. Med. 330:107–113, 1994. 43. C. Cunniff, L.E. Kratz, A. Moser, M.R. Natowicz, and R.I. Kelley. Clinical and biochemical spectrum of patients with RSH/SmithLemliOpitz syndrome and abnormal cholesterol metabolism. Am. J. Med. Genet. 68:263–269, 1997. 44. G.S. Tint, M. Seller, R. HughesBenzie, A.K. Batta, S. Shefer, D. Genest, M. Irons, E. Elias, and G. Salen. Markedly increased tissue concentrations of 7 dehydrocholesterol combined with low levels of cholesterol are characteristic of the SmithLemliOpitz syndrome. J. Lipid Res. 36:89–95, 1995. 45. G. Xu. Reproducing abnormal cholesterol biosynthesis as seen in the SmithLemliOpitz syndrome by inhibiting the conversion of 7dehydrocholesterol to cholesterol in rats. J. Clin. Invest. 95:76–81, 1995. 46. M. Irons, E.R. Elias, G.S. Tint, G. Salen, R. Frieden, T.M. Buie, and M. Ampola. Abnormal cholesterol metabolism in the SmithLemliOpitz syndrome: report of clinical and biochemical findings in four patients and treatment in one patient. Am. J. Med. Genet. 50:347–352, 1994. 47. N.A. Nwokoro and J.J. Mulvihill. Cholesterol and bile acid replacement therapy in children and adults with SmithLemliOpitz (SLO/RSH) syndrome. Am. J. Med. Genet. 68: 315–321, 1997. 48. E. Jacquemin, K.D.R. Setchell, N.C. O'Connell, A. Estrada, G. Maggiore, J. Schmitz, M. Hadchouel, and O. Bernard. A new cause of progressive intrahepatic cholestasis: 3bhydroxyC27steroid dehydrogenase/isomerase deficiency. J. Pediatr. 125:379–384, 1994. 49. B.L. Shneider, K.D. Setchell, P.F. Whitington, K.A. Neilson, and F.J. Suchy. Delta 43
Page 182
oxosteroid 5 betareductase deficiency causing neonatal liver failure and hemochromatosis. J. Pediatr. 124:234–238, 1994. 50. E.R. Elias and M. Irons. Abnormal cholesterol metabolism in SmithLemliOpitz syndrome. Curr. Opin. Pediatr. 7:710–714, 1995. 51. D.W. Russell and K.D. Setchell. Bile acid biosynthesis. Biochemistry 31:4737–4749, 1992. 52. G. Salen, S. Shefer, F.W. Cheng, B. Dayal, A.K. Batta, and G.S. Tint. Cholic acid biosynthesis: the enzymatic defect in cerebrotendinous xanthomatosis. J. Clin. Invest. 63:38–44, 1979. 53. G. Salen, T.W. Meriwether, and G. Nicolau. Chenodeoxycholic acid inhibits increased cholesterol and cholestanol synthesis in patients with cerrebrotendinous xanthomatosis. Biochem. Med. 14:57–74, 1975. 54. T. Nakamura, Y. Matsuzawa, K. Takemura, M. Kubo, H. Miki, and S. Tarui. Combined treatment with chenodeoxycholic acid and pravastatin improves plasma cholestanol levels associated with marked regression of tendon xanthomas in cerebrotendinous xanthomatosis. Metabolism 40:741–746, 1991. 55. T. Kita, M.S. Brown, and J.L. Goldstein. Feedback regulation of 3hydroxy3methylglutaryl coenzyme A reductase in livers of mice treated with mevinolin, a competitive inhibitor of the reductase. J. Clin. Invest. 66:1094–1100, 1980. 56. P.T. Ma, G. Gil, T.C. Sudhof, D.W. Bilheimer, J.L. Goldstein, and M.S. Brown. Mevinolin, an inhibitor of cholesterol synthesis, induces MRNA for low density lipoprotein receptor in livers of hamsters and rabbits. Proc. Natl. Acad. Sci. U.S.A. 83:8370–8374, 1986. 57. W.M. Pandak, D.M. Heuman, P.B. Hylemon, and Z.R. Vlahcevic. Regulation of bile acid synthesis: IV. Interrelationship between cholesterol and bile acid biosynthesis pathways. J. Lipid Res. 31:79–90, 1990. 58. W.M. Pandak, Z.R. Vlahcevic, J.Y.L. Chiang, D.M. Heuman, and P.B. Hylemon. Bile acid synthesis: VI. Regulation of cholesterol 7 hydroxylase by taurocholate and mevalonate. J. Lipid Res. 33:659–668, 1992. 59. D.K. Spady, E.F. Stange, L.E. Bilhartz, and J.M. Dietschy. Bile acids regulate hepatic low density lipoprotein receptor activity in the hamster by altering cholesterol flux across the liver. Proc. Natl. Acad. Sci. U.S.A. 83:1916–1920, 1986. 60. S.D. Turley, D.K. Spady, and J.M. Dietschy. Identification of a metabolic difference accounting for the hyper and hyporesponder phenotypes of cynomolgus monkey. J. Lipid Res. 38:1598–1611, 1997. 61. S.D. Turley, D.K. Spady, and J.M. Dietschy. Alteration of the degree of biliary cholesterol saturation in the hamster and rat by manipulation of the pools of preformed and newly synthesized cholesterol. Gastroenterology 84:253–264, 1983. 62. B.G. Stone, S.K. Erickson, W.Y. Craig, and A.D. Cooper. Regulation of rat biliary cholesterol secretion by agents that alter intrahepatic cholesterol metabolism. Evidence for a distinct biliary precursor pool. J. Clin. Invest. 76:1773–1781, 1985. 63. D.M. Small. Phospholipids. In: D.M. Small, ed. Handbook of Lipid Research: IV. The Physical Chemistry of Lipids. New York: Plenum Press, 1986, pp. 475– 522. 64. D.E. Vance. Glycerolipid biosynthesis in eukaryotes. In: D.E. Vance and J.E. Vance, eds. Biochemistry of Lipids, Lipoproteins and Membranes. Amsterdam: Elsevier Science, 1986, pp. 153–181. 65. S.L. Pelech, P.H. Pritchard, D.N. Brindley, and D.E. Vance. Fatty acids promote translocation of CTP:phosphocholine cytidylyltransferase to the endoplasmic reticulum and stimulate rat hepatic phosphatidylcholine synthesis. J. Biol. Chem. 258:6782–6788, 1983. 66. A. Lykidis, K.G. Murti, and S. Jackowski. Cloning and characterization of a second human CTP:phosphocholine cytidyltransferase. J. Biol. Chem. 273:14022– 14029, 1998. 67. D.E. Vance, C.J. Walkey, and Z. Cui. Phosphatidylethanolamine Nmethyltransferase from liver. Biochim. Biophys. Acta 1348:142–150, 1997.
Page 183
68. Z. Cui, J.E. Vance, M.H. Chen, D.R. Voelker, and D.E. Vance. Cloning and expression of a novel phosphatidylethanolamine Nmethyltransferase: a specific biochemical and cytological marker for a unique membrane fraction in rat liver. J. Biol. Chem. 268:16655–16663, 1993. 69. Z. Cui, M. Houweling, and D.E. Vance. Expression of phosphatidylethanolamine Nmethyltransferase2 in McArdleRH7777 hepatoma cells inhibits the CDP choline pathway for phosphatidylcholine biosynthesis via decreased gene expression of CTP:phosphocholine cytidylyltransferase. Biochem. J. 312:939–945, 1995. 70. Z. Cui, M. Houweling, and D.E. Vance. Suppression of rat hepatoma cell growth by expression of phosphatidylethanolamine Nmethyltransferase2. J. Biol. Chem. 269:24531–24533, 1994. 71. C.J. Walkey, L.R. Donohue, R. Bronson, L.B. Agellon, and D.E. Vance. Disruption of the murine gene encoding phosphatidylethanolamine Nmethyltransferase. Proc. Natl. Acad. Sci. U.S.A. 94:12880–12885, 1997. 72. P.C. Choy, M. Skrzypczak, D. Lee, and F.T. Jay. AcylGPC and alkenyl/alkylGPC:acylCoA acyltransferases. Biochim. Biophys. Acta 1348:124–133, 1998.
Page 185
9— Cholesterol Crystallization in Bile Fred M. Konikoff Tel Aviv Sourasky Medical Center, Tel Aviv, Israel Joanne M. Donovan Harvard Medical School and Boston VA Medical Center, Boston, Massachusetts I— Introduction In supersaturated bile, cholesterol tends to precipitate and form crystals. A variety of crystal morphologies can be detected by polarizing light microscopy in fresh human gallbladder bile obtained at cholecystectomy. Moreover, typical rhomboid cholesterol monohydrate crystals can form rapidly (within a few days) during ex vivo incubation of lithogenic human bile that has first been cleared from native crystals by filtration or ultracentrifugation (1). This chapter reviews the crystal forms found in cholesterol gallstones and their mechanisms of formation, approaches to assessing crystallization in bile, factors that modulate crystal formation, and, last, touches upon the process of crystal agglomeration that must occur to produce a mature gallstone. In 1674, van Leeuwenhoek, using light microscopy, first observed cholesterol crystals (2). Almost 300 years later, Lyon devised a way of stimulating gallbladder contraction and collecting bile specimens from the duodenum (3). The method was used by Juniper and Burson to demonstrate the significance and usefulness of detecting biliary crystals in the diagnosis of hepatobiliary and pancreatic diseases (4); thus they initiated the modern era of investigating cholesterol crystallization and its relevance for gallstone formation. The central role of cholesterol crystallization in gallstone formation was defined by two sets of observations: first, the ability to define the physical properties of bile that allow crystals to form, and, second, the clinical correlation of cholesterol crystals with gallstone disease. Physicochemical studies revealed that cholesterol supersaturation was essential for biliary cholesterol to nucleate and crystallize (5,6). Despite this basic understanding of the physicochemical forces that govern biliary cholesterol solubilization on one hand and its precipitation on the other hand, it was soon realized that supersaturation is a necessary but not sufficient prerequisite for crystal and stone formation (7). Subsequently, Holzbach and colleagues showed that the propensity of bile to form de novo cholesterol crystals during ex vivo incubation is the most definitive characteristic differentiating the biles of gallstone formers and nonformers (1). In a landmark study, Sedaghat and Grundy showed that cholesterol crystals appeared in gallbladder biles of all patients with cholesterol stones, as opposed to none in the biles of pigment stone patients (8). Cholesterol crystals could also be detected in duodenal bile, although intermittently (9). Thus, gradually accumulating evidence supported the notion that cholesterol crystallization is the key to the cascade leading to gallstone formation.
Page 186
Evidence reviewed elsewhere in this volume describes the panoply of biliary proteins that have been demonstrated to accelerate or inhibit cholesterol crystallization in vitro or in vivo. This chapter focuses on the physicochemical mechanisms by which cholesterol crystallization occurs in bile. II— Crystal Composition of Gallstones Cholesterol gallstones, as the name implies, are composed mainly of cholesterol and can be defined by a weight fraction of cholesterol that is greater than 75 to 80% cholesterol (10), but which, in some cases, approaches 100% (11,12). The composition of gallstones bears archeological witness to the composition of bile during the lengthy period of lithogenesis. In the western world, over threequarters of patients have predominantly cholesterol stones (13,14). Mixed stones, with cholesterol compositions intermediate between those of pigment and cholesterol stones, are thought to occur in biles that have become secondarily infected (15). The remainder of a cholesterol gallstone is a mixture of biliary glycoproteins, calcium salts, and bile pigments (16). Stones with from 25 to 75% cholesterol by weight (16), are likely to derive from secondary bacterial contamination and precipitation of calcium bilirubinate. At the center of most cholesterol gallstones is a nidus of visible pigment (17), which has been determined by infrared spectroscopy to be the calcium salt of the unconjugated bilirubin (UCB) monoanion, Ca(HUCB)2 (18). Small amounts of calcium phosphate and traces of calcium carbonate are also present at the core of most human cholesterol stones (19) and in laminated rings associated with calcium bilirubinate (20). While this central pigment has been hypothesized to be involved in the earliest stages of nucleation, its presence may represent coprecipitation (18) or subsequent diffusion of pigments into existing stones (21). From a materials sciences perspective, the term cholesterol gallstone is, strictly speaking, a misnomer, since these stones are not real ceramic stones but organic concretions (22). As revealed by inspection with the unaided eye and lowpower microscopy, these concretions are agglomerates of a multitude of individual crystals of cholesterol monohydrate, usually oriented with the long axes radiating outward like spokes of a wheel from the center of the stone and held together by an organic matrix of glycoproteins (23). Formation of these concretions requires the presence of a matrix composed of mucin glycoproteins (24,25), and has yet to be duplicated in vitro. Cholesterol crystallization is a pivotal step in cholesterol gallstone formation, irrespective whether the crystallization occurs as a primary event within the bile or secondarily on the surface of a stone. Therefore, the study of cholesterol crystallization has become a central issue in the study of cholesterol gallstone pathogenesis (26,27). III— Crystal Forms of Cholesterol In nature, cholesterol can exist in two different crystalline forms, anhydrous and monohydrate (Fig. 1), both of which are thermodynamically stable under different conditions. In an aqueous environment such as bile at 37°C, the most stable form is cholesterol monohydrate (28). Although cholesterol may also crystallize as esters or other mixed compounds, these are generally not found in bile (29–31). A— Cholesterol Monohydrate Cholesterol monohydrate crystals are triclinic, with eight molecules of cholesterol and eight water molecules in the unit cell, as disclosed by the elegant xray diffraction studies of Craven (32). As shown in Fig. 1, cholesterol molecules are packed such that they form a doublelayer
Page 187
Figure 1 Crystalline structures of cholesterol monohydrate and anhydrous cholesterol. Dark circles represent oxygen atoms from the cholesterol hydroxyl group or from water for the monohydrate form. For cholesterol monohydrate, the eight molecule crystal structure is formed by superimposing the right four molecules (D, E, F, H) over the left four molecules (A, B, C, G). Two eightmember unit cells of lowtemperature anhydrous cholesterol are shown, and are superimposed over each other for the complete crystal structure. For cholesterol monohydrate, note the internal bilayer structure formed by mirror reflections of the steroid nuclei, with the hydrogenbonded oxygen atoms forming an approximate plane. Also, the steroid nuclei are approximately perpendicular to the plane formed by the oxygen atoms. In anhydrous cholesterol, the oxygen atoms are hydrogen bonded, but the planar structure is less distinct. (From Ref. 31.)
Page 188
structure with a thickness of 33.9 Å. The water molecules are located between the double layers, so that the C3 hydroxyl groups of cholesterol and the water molecules produce a hydrogen bonding network between the layers (32). Cholesterol monohydrate crystals have a typical appearance of rhomboid, platelike structures, with characteristic angles (79.2 and 100.8 degrees) (28). These crystals frequently have a tiny corner notch, which has been suggested to derive from a sequence of cholesterol crystallization via a variety of intermediate structures such as arcs, spirals, helices, and tubes (33), as discussed further below. Polarizing light microscopy allows identification of these crystal forms by their characteristic morphology and birefringence properties. Physical identification of cholesterol monohydrate crystals can be made by xray diffraction, calorimetry, or specific gravity measurements. Powder diffraction of cholesterol monohydrate crystals reveals three major reflection peaks at 34.1, 17.6, and 5.9 Å (28). The mass density of cholesterol monohydrate crystals is 1.045 g/dL. With differential scanning calorimetry (DSC), cholesterol monohydrate crystals display two secondary transitions and a primary phase transition (28). During heating from room temperature to 160°C (beyond the melting point of cholesterol), three distinct endotherms, at 86.4, 123.4, and 156.8°C are observed, corresponding to the loss of hydration, transition from crystal to liquid crystal, and melting point, respectively. B— Anhydrous Cholesterol Anhydrous cholesterol also has a triclinic crystal configuration, but with distinct features (34). One striking difference from the monohydrate is that anhydrous cholesterol exists in two different polymorphic forms, as detected by differential scanning calorimetry (35) and xray diffraction (28). At a temperature below 31.6°C, the unit cell consists of eight cholesterol molecules; whereas at higher temperatures (including body temperature), it has 16 molecules. The mass density of anhydrous cholesterol crystals is 1.03 g/dL (34), and the crystal habit is needlelike. The aqueous solubility of anhydrous cholesterol in micellar bile salt solutions exceeds that of cholesterol monohydrate (36) due to the less thermodynamically favorable crystal structure. In the presence of excess water, cholesterol monohydrate is the thermodynamically stable form. Of note, the crystal habit observed visually is not specific for the presence of monohydrate or anhydrous crystals and is determined by additional factors, including hydration and the speed of crystallization. Thus, anhydrous cholesterol crystals may sometimes have a platelike habit, just as the monohydrate may have a needlelike habit (35,37). Unless cholesterol monohydrate crystals are maintained under conditions of high humidity, the water molecules are lost and calorimetry will fail to demonstrate the transition from monohydrate to anhydrous forms at 86°C. Evaporation of the water of hydration may account for some observations of anhydrous crystals in cholesterol gallstones not continuously maintained in an aqueous environment (38,39). C— Additional Crystal Forms in Bile Biliary cholesterol crystallization can yield a variety of crystal forms, or habits, in addition to classic rhomboid plates (33,40,41). These intermediate structures appear transiently between metastable supersaturated systems and thermodynamic equilibrium. Therefore all are thermodynamically unstable with respect to cholesterol monohydrate (i.e., their crystal structures are metastable or less favorable thermodynamically) (42). Figure 2 displays the wide variety of morphologies that can be observed. In a dilute, bile saltrich model bile, the earliest cholesterol crystals are flexible filamentous structures, with a mass density as well as xray diffraction pattern suggestive of initial anhydrous crystallization (33,43). Thin needle structures or arclike crystals form under similar conditions and are be
Page 189
Figure 2 Intermediate crystal structures found in model and native biles. Helical and tubular structures can be seen in a–d, along with a filament in a. Helical markings can be seen on the tubule in c. Panels e and f show fracture of a tubule with subsequent growth of a platelike crystal. (From Ref. 42.)
lieved also to reflect anhydrous crystallization. These structures subsequently undergo a series of morphological transformations while growing in size. Cholesterol can form helical structures of at least two different pitch angles (44). Further crystal growth produces tubule structures, sometimes with characteristic markings attesting to their helical origin. Fracture of tubules can be observed, unfurling a flat crystal with characteristic angles and often notches typical of platelike crystals of cholesterol monohydrate. As discussed below, the composition of bile influences formation of these structures. Although initially observed in a bile saltrich dilute, nonphysiological system (33), these structures have also been observed in concentrated, physiologically relevant model biles as well as native biles. The quantitative contribution of these structures to cholesterol crystallization in human bile in vivo remains unclear. IV— Solubilization of Cholesterol in Bile Bile salts, phosphatidylcholine, and cholesterol aggregate to form different biliary lipid aggregates, depending on their relative concentrations. The amphiphilic nature of these lipids drives formation of these aggregate structures that minimize exposure of hydrophobic portions of the molecules to aqueous solution. Alone, aqueous bile salts form simple micelles, aggregates about 20 to 30 Å in size, above the critical micellar concentration of up to several millimol depending on the bile salt species (45,46). Together with phosphatidylcholine, thermodynamically stable
Page 190
mixed micelles are formed (47). At high relative bile salt contents and total lipid concentrations, mixed micelles are small and globular (40 to 80 Å in diameter) (47); but at low relative bile salt contents and total lipid concentration, rodshaped structures up to several hundred angstroms in length form (48). Cholesterol can be solubilized by simple or mixed micelles, although the capacity of the mixed micelle greatly exceeds that of simple micelles (49). When the micellar solubility of either phosphatidylcholine or cholesterol is exceeded, additional phases can form: a cholesterol crystalline phase and a lamellar phase (50). At equilibrium, the crystal structure is that of cholesterol monohydrate. Bilayer structures containing phosphatidylcholine, cholesterol, and small amounts of bile salts range in size from unilamellar vesicles (250–2000 Å in diameter) composed of a single phosphatidylcholine/cholesterol bilayer to multilamellar structures of a size visible by light microscopy (>2000 Å). A— Equilibrium Phase Diagram Fundamental insights into cholesterol crystallization in bile derive from the equilibriumphase diagram of biliary lipids, which define the conditions under which cholesterol crystal formation can occur in bile (5,51). Because the total lipid concentration of bile varies severalfold, relative rather than absolute concentrations of the three major biliary lipids—bile salts, phosphatidylcholine, and cholesterol—were found to define the limits of cholesterol solubility most precisely (6). As shown in Fig. 3, the equilibriumphase diagram displays relative compositions
Figure 3 Equilibriumphase diagram of model bile at a total lipid concentration of 10 g/dl. See text for details. (From Ref. 52.)
Page 191
of the principal lipids of bile at a physiological concentration, 10 g/dL total lipid concentration. Each triangular apex represents systems made up of a single component: either bile salt, phosphatidylcholine, or cholesterol. Interior points represent biles with differing relative compositions of all three components. Only within the micellar zone at relatively high bile salt concentrations do simple and mixed micelles completely solubilize cholesterol. The cholesterol saturation index is defined as the ratio between the molar percentage of cholesterol of a system and the maximum micellar solubility of cholesterol; it is unity at the micellarphase boundary. Systems with cholesterol contents that exceed the micellarphase boundary (CSI > 1) must contain at least one additional phase at equilibrium, either cholesterol monohydrate crystals or cholesterol/phosphatidylcholine bilayer structures. On the left side of the phase diagram at relatively high bile salt contents, a twophase region contains cholesterolsaturated micelles and cholesterol monohydrate crystals. On the right side of the phase diagram at relatively high phosphatidylcholine contents, another twophase region contains cholesterol/phosphatidylcholine bilayer structures and cholesterolunsaturated micelles. Between these two regions is a threephase region, that contains cholesterol monohydrate crystals, cholesterolsaturated micelles, and cholesterol/phosphatidylcholine vesicles, presumably in a 1:1 ratio (53). Both the micellar phase limit and the boundaries between the multiphase regions depend on total lipid concentration and bile salt hydrophobicity (5,40,54). In dilute biles, the micellar zone is smaller, and the boundaries of the multiphase regions shift to the left. Thus, the twophase zone containing micelles and a lamellar phase is expanded at the expense of the regions containing cholesterol monohydrate crystals at equilibrium. With increasing bile salt hydrophobicity, the micellar zone expands, and the boundaries of the multiphase regions shift to the right. Consequently, the regions containing cholesterol monohydrate crystals at equilibrium are expanded. The relative lipid concentrations, as plotted on the triangular equilibrium phase diagram, have been shown to be important also in determining the crystallization pathways of cholesterol, including the intermediate crystal forms, as is discussed below. B— Metastable Equilibrium Cholesterolsupersaturated biles can exist in metastable states in which cholesterol crystals are not present but will form if sufficient time is allowed for the system to reach equilibrium. Under these conditions, the vesicular cholesterol/phosphatidylcholine ratio exceeds unity, and the coexisting micelles are also cholesterol supersaturated (55–58). The composition of biliary lipid aggregates under these conditions is not fixed and must be determined by an analytical method that does not perturb the composition of the micelles and vesicles (53,59). Biliary mixed micelles and vesicles are in rapid equilibrium with bile salt monomers and simple micelles at a concentration known as the intermixed micellar/intervesicular bile salt concentration (IMC). Utilizing this concentration, biliary vesicles can be isolated by gel filtration chromatography, resulting in their dilution without alterations in their composition (60). As higher concentrations of bile salts more effectively solubilize vesicular phosphatidylcholine than cholesterol (61), higher IMC values are associated with increased vesicular cholesterol/phosphatidylcholine ratios (58). Thus, in supersaturated biles, changes in relative and absolute biliary lipid composition such as increased total lipid concentration and increased bile salt/phosphatidylcholine are associated with higher IMC values, and are consequently associated with increases in vesicular cholesterol/phosphatidylcholine ratio. Cholesterol can equilibrate between lipid bilayers over a period of hours (62), but the presence of bile salts accelerates this process (63). The implication is that in supersaturated biles, both micelles and vesicles are supersaturated, as has been confirmed experimentally (58). Rapid equilibration of cholesterol between noncrystalline phases can therefore allow both micelles and vesicles to serve as sources for cholesterol crystallization.
Page 192
V— Measurement of Cholesterol Crystallization in Bile Measurements of the kinetics of cholesterol crystallization have been an indispensable tool in gallstone research, enabling investigation of factors that promote or retard cholesterol crystallization (1,64,65). In clinical practice, observations of cholesterol crystals in duodenal bile, obtained after gallbladder contraction induced by cholecystokinin, can provide key diagnostic information. The presence of cholesterol crystals as determined by biliary microscopy can diagnose biliary sludge and allow for further therapeutic interventions to prevent complications such as pancreatitis (66,67). Measurement of the kinetics of crystallization in bile obtained from the duodenum has been proposed (68), but the accuracy is limited by the presence of pancreatic phospholipase (69,70), producing free fatty acid in a process known to accelerate nucleation (71). Determination of the nucleation time has been used to predict the likelihood of gallstone dissolution by oral bile salts (72) and recurrence of gallstones after cholecystotomy (73). Despite the limitations of individual assay techniques, these methods provide much of our current understanding of biliary cholesterol crystallization and the factors that modulate crystallization. A— Concept of Critical Nucleus Biliary cholesterol crystallization includes a sequence of distinct steps. Initially, there is rearrangement of cholesterolcontaining lipid aggregates (supersaturated vesicles and micelles) and possible redistribution of cholesterol within and between the aggregates (74). This is believed to result in the formation of a critical nucleus, the earliest form of crystalline cholesterol (75). Detection of this critical nucleus has been so far unsuccessful. The earliest crystals are small and thin, below the resolution limits (about 200 nm) of conventional light microscopy. Further increase in crystal mass and size, which may also involve changes in crystal shape and habit (42), eventually leads to the formation of thermodynamically stable, classic platelike cholesterol monohydrate crystals. While mature birefringent cholesterol crystals are easily recognized by polarizing light microscopy, the precise time point at which cholesterol crystallization occurs has been elusive. Moreover, crystallization may occur in bile from multiple nuclei, as a continuing asynchronous process (75). Hence, attempts to assess biliary cholesterol crystallization quantitatively have intrinsic limitations. Nucleation is classified as homogeneous, which occurs from an isotropic solution, or heterogeneous, which occurs on a separate nidus. Heterogeneous nucleation is believed to predominate in bile (75) and occurs far more readily than homogeneous nucleation. For example, extremely pure filtered water can be supercooled well below the freezing point, but it freezes rapidly once ice crystals or even dust is added. Thus, the presence of microcrystals of cholesterol that are not completely removed can accelerate the observed rate of crystallization (76). B— Direct Light Microscopy Despite methodological limitations, measurement of cholesterol crystallization has become an essential part of gallstone research and the clinical assessment of bile lithogenicity. For measurements of nucleation time, crystals are removed by ultracentrifugation (1) and the isotropic supernatant is incubated at 37°C, while aliquots are removed periodically for examination by polarized light microscopy. Nucleation time is a misnomer and has more recently and more properly been renamed crystal observation time. A variation on this technique is mixing model biles with serial dilutions of native biles, with determination of the maximum dilution affecting crystal observation time (77). The number of crystals provides a semiquantitative measure of the mass of crystalline cholesterol. This procedure is laborious, but can be performed by polarized light microscopy
Page 193
(79). The crystal growth curve provides additional information about crystallization kinetics, but with the disadvantage of not taking into consideration variations in crystal size. Moreover, once the number of crystals becomes large and agglomeration occurs, the technique loses accuracy. Attempts to automate this process with a Coulter counter have not gained wide acceptance because of concerns about its accuracy (80,81). C— TurbidityBased Assays Assays based on the turbidity of suspended cholesterol crystals offer advantages in reproducibility and speed (81). Busch et al. (81) introduced a spectrophotometric method to provide a means of measuring cholesterol crystal growth. A minute amount of seed crystals is added to a model bile to act as a catalyst for crystal growth. At intervals, aliquots of model bile are diluted with excess bile salts to rapidly solubilize bilayer structures, and the turbidity of the resulting solution is measured. Because bile salts dissolve cholesterol crystals relatively slowly (36), the light scattered by the crystals alone can be measured. The development of turbidity with time yields a sigmoidal crystal growth curve that is characterized by an onset time, a rate of increase in turbidity corresponding to crystal growth, and a plateau reflecting maximal crystal concentration at equilibrium. The process of nucleation can thus be characterized by a lag period in which no crystals can be observed, followed by crystal growth. The technique has the theoretical limitation that scattered light is not always directly correlated with the number of crystals due to variability in crystal sizes and shapes. The method is less sensitive than microscopy because several hundreds of crystals are required before detection, but it is less dependent on investigator expertise and easier to perform on a large number of samples. Recently, the assay was modified using microtiter plates and a plate reader, providing a rapid highcapacity method for detecting cholesterol crystal precipitation and growth in model biles (82,83). Unfortunately, biliary pigments limit the use of optical methods for crystallization assays in native biles. However, studies have been extended to develop methods that allow early detection and quantitative measurement of cholesterol monohydrate crystal growth in human native gallbladder bile, based on the combination of spectrophotometry with preparative ultracentrifugation (84) or dilution of the samples (85). D— Assessment of Intermediate Crystal Forms None of the above crystallization assays provide information about the actual mechanism(s) by which cholesterol nucleates and crystals grow, nor do they distinguish between various crystallization pathways (40,86). To overcome this limitation, the crystallization process has been depicted by a semiquantitative graphic method that shows separate crystallization curves for distinct crystal forms (40,86,87). Because of differences in crystal morphology, this format does not necessarily reflect the relative mass of each type of crystal present. A density gradient ultracentrifugation assay has been developed to overcome this limitation (88). E— Measurement of Crystal Mass Crystal mass can be quantified radiochemically using tracer amounts of [3H]cholesterol (80), although this method has not been widely applied or validated. Ultracentrifugation with a density gradient can also be used to separate cholesterol crystals and quantify crystal growth (84). Separation of cholesterol precipitates by sequential ultracentrifugation (88) can provide quantitative as well as qualitative monitoring of the crystallization process. This technique takes advantage of differences in the density (d) of bilayer structures (d < 1.02 g/mL), anhydrous
Page 194
cholesterol (d = 1.03), and cholesterol monohydrate (d = 1.04). Early intermediate forms composed of anhydrous cholesterol can be separated from rhomboidal cholesterol monohydrate plates. However, intermediate forms—such as helices and tubules—approach the density of cholesterol monohydrate and cannot be determined independently. This technique also allows the concomitant monitoring of precipitable cholesterolcontaining multilamellar vesicles during the crystallization process. VI— Mechanisms of Crystallization A— Origins of Biliary Cholesterol Cholesterol is secreted into bile in unilamellar phosphatidylcholine vesicles (56,89,90), as described in detail elsewhere in this volume. Formation of these vesicles depends upon activity of mdr2 (91,92), a canalicular membrane protein of the ATP cassette family that transfers phosphatidylcholine to the exoleaflet of the canaliculus. These nascent vesicles are believed to have a cholesterol/phosphatidylcholine ratio of approximately 0.3, the ratio of these lipids in hepatic bile (Fig. 4). Bilayer structures with this composition are thermodynamically stable and incapable of forming cholesterol crystals (94). Independent active transport of bile salts into the canaliculus by the bile salt export protein (95) mediates transformation of these nascent vesicles. Because the capacity of bile salts to solubilize phosphatidylcholine exceeds their capacity to solubilize cholesterol, mixed micelles are formed, but residual vesicles contain a much higher relative cholesterol concentration (61,89,96). Within the gallbladder, further rearrangement of cholesterolcontaining lipid aggregates occurs before nucleation and crystallization. Due to concentration of bile, micellar cholesterol
Figure 4 Biliary lipid aggregates in bile. Cholesterol is secreted into bile as cholesterol/phosphatidylcholine vesicles with a molar ratio of approximately 0.3. As the bile ducts concentrate bile, bile salts as monomers and micelles selectively solubilize phosphatidylcholine to produce supersaturated cholesterol/phosphatidylcholine vesicles with molar ratios above unity. In the gallbladder, vesicles aggregate and fuse, forming cholesterolrich microdomains that provide a template for formation of crystalline cholesterol. (From Ref. 93.)
Page 195
carrying capacity increases. Concomitantly, vesicles aggregate and fuse, initiating the process of cholesterol nucleation and precipitation in supersaturated bile (97–99). Rhomboidal plates can be seen emerging from aggregated vesicles by phasecontrast microscopy (99). Quantitation of biliary aggregates suggests that the preponderance of precipitated cholesterol derives from vesicles rather than from micelles (100). Further support for this sequence comes from the observation that nucleationpromoting proteins also disrupt vesicles (101) and accelerate vesicle fusion and aggregation (97). On the other hand, apolipoprotein AI, which prolongs crystal observation time (102), prevents fusion of vesicles in model systems (103). The striking similarity in molecular orientation between putative cholesterolrich microdomains in these multilayer structures and the crystal structure of cholesterol monohydrate suggests that multilamellar structures form a template that accelerates cholesterol crystallization (104). Recently, videoenhanced light microscopy has been used in combination with cryogenic transmission electron microscopy (cTEM) to provide direct imaging of microstructural details and evolution during biliary cholesterol nucleation and crystallization (105–108) (Fig. 5). These studies have substantiated the role of micelles and vesicular (uni and multilamellar) aggregates in the nucleation process, and revealed additional early vesicular structures, believed to represent intermediates in a micelle to vesicle transition (105,108). They have, however, failed to support the importance of vesicle fusion in the nucleation process (105,108). Although the critical nucleus has so far evaded even these new powerful observation methods, the growth of early cholesterol crystals by possible feeding from micelles and bilayer structures has been visualized (108). Despite the weight of evidence that vesicle aggregation precedes cholesterol crystal formation, mixed micelles also play a role. First, mixed micelles coexisting with cholesterolrich vesicles are also supersaturated (58). In bile saltcholesterol model systems, cholesterol crystals are formed in the absence of phosphatidylcholine, and cholesterol crystals have been observed to derive from the micellar phase in vivo (109). Because of the difficulty of separating micelles and vesicles and the rapid equilibration of cholesterol between membranes and micelles, the true origin of crystalline cholesterol is ambiguous. B— Pathways of Cholesterol Crystallization Since the initial description of nonplatelike (filamentous, helical, and tubular) cholesterol crystals (33), it has become evident that there are several pathways by which biliary cholesterol can nucleate and grow into ''mature" crystals. As shown in Fig. 6, there appear to be at least five distinct pathways in model bile, as denoted by Wang and Carey from A to E, according to the relative lipid composition as it plots on a triangular phase diagram (40). These pathways differ in terms of cholesterol precipitates involved and their sequence of appearance, as shown schematically in Fig. 7. In pathway A, the first cholesterol precipitates are arclike crystals, followed by helical and tubular crystals. Plates appear only after helices and become predominant only as other crystals disappear. In pathway B, plates are the first precipitates to appear, soon after supersaturation is initiated, followed by transient appearances of arcs, helices, and tubular cholesterol crystals. In pathways C and D, crystal precipitation is slower and is preceded by the appearance of liquid crystals. In both these pathways the first crystal precipitates are platelike. However, in pathway C, the crystallization sequence is similar to that in pathway B (plates arcs helices tubes plates); whereas in pathway D, only plates form. In pathway E, only liquid crystals form, without precipitation of solid cholesterol crystals. Thus all pathways except E lead eventually to the formation of classical platelike cholesterol monohydrate crystals at equilibrium. As shown in Fig. 7, these can be grouped into pathways in which intermediate crystal habits and cholesterol monohydrate crystals appear (A, B, and C), and those in which only cholesterol monohydrate crystals appear. The distinction may be whether the degree of cholesterol supersaturation is sufficient thermodynamically to allow anhydrous cholesterol to precipitate (110).
Page 196
Figure 5 CryoTEM of lipid microstructures appearing in nucleating bile. After dilution totrigger supersaturation, spheroidal micelles are seen immediately along with discoid structures and forming vesicles. Panels B and C show evolution of multilamellar structures with incomplete bilayer patches. (From Ref. 108.)
A major underlying factor that determines the crystallization behavior of biliary cholesterol is the relative phosphatidylcholine concentration of the bile (40). At low phosphatidylcholine molar fractions, cholesterol crystallization occurs in the absence of liquid crystal formation, revealing filamentous and arclike crystals as the earliest crystal forms (pathways A and B). With increasing phosphatidylcholine concentrations, cholesterol crystallization is preceded by liquid crystal (multilamellar vesicle) formation, and platelike crystals precipitate before or even without precipitation of other crystal forms (pathways C, D, and E). Dilution,
Page 197
Figure 6 Ternary phase diagram showing relative concentrations for which various crystallization pathways occur. The shaded areas depict the twophase zones that at equilibrium contain micelles and cholesterol monohydrate crystals, and a lamellar phase and cholesterol monohydrate crystals, respectively. Above the micellar zone is the unshaded threephase zone containing micelles, cholesterol monohydrate crystals, and a lamellar phase at equilibrium. Four distinct sequences of cholesterol crystallization occur, defined by the temporal sequence in which different structures appear. For pathway A, arclike crystals appear, followed by helices, tubules, and plates. For pathway B, plates appear initially, followed by transient appearances of helices, tubules, and plates. In pathway C, formation of microscopically observable vesicles precede the same sequence as in B. In pathway D, liquid crystals precede the development of plate. Pathway E denotes regions of the phase diagram where only liquid crystals are present. Increases in bile salt hydrophobicity or total lipid concentration result in an expansion of all zones to the left. (From Ref. 40.)
cooling, and decreasing bile salt hydrophobicity have an opposite effect to the increasing phosphatidylcholine content (40,41,111). Phospholipid molecular species also influence the crystallization pathways of biliary cholesterol (87,112). The shifts in pathways of crystallization occur in parallel with shifts in the multiphase regions of equilibrium that are observed with changes in total lipid concentration, temperature, and bile salt hydrophobicity (40). Recently, Carey has proposed a tentative molecular explanation for cholesterol crystallization pathways (113). At high phosphatidylcholine molar fractions, an abundance of phosphatidylcholine/cholesterol vesicles aggregate and fuse to form multilamellar vesicles. Within these multilayered structures, cholesterol molecules align themselves in bimolecular layers, resembling the molecular packing in cholesterol monohydrate crystals. When supersaturated,
Page 198
Figure 7 Sequence of structures appearing during various cholesterol crystallization pathways. In pathways A, B, and C, intermediate structures including arcs, helices, and tubules disappear to leave cholesterol monohydrate plates at equilibrium. The relative position of these structures on the horizontal axis depicts the relative time at which they appear during the various pathways. The column on the right displays the final crystal structures. Increases in relative bile salt content, bile salt hydrophobicity, or total lipid concentration result in shifts toward pathway A.
these cholesterolrich layers grow and tend to precipitate as platelike cholesterol monohydrate crystals. At low phosphatidylcholine molar fractions, however, only unilamellar vesicles form. Within these, in the absence of multilayers, cholesterol packs at supersaturation as a condensed core structure, far from water molecules. From these cholesterolrich cores, cholesterol precipitates as anhydrous crystals, with a filamentous or needlelike habit. In bile, anhydrous crystals hydrate slowly (28), while going through a series of morphological transitions to become classic platelike monohydrate crystals. This explanation is, however, at present highly speculative, and needs to be validated by confirmatory experiments. In general, human bile has a composition that falls into the two and threephase regions of the phase diagram, which involve crystallization pathways B, C, or D (40,86). Therefore, in native bile, the earliest cholesterol crystal forms tend to be platelike. However, in most human biles, intermediate crystal forms (arcs, helices, and tubes) should appear during the crystallization process. These predictions have been substantiated by observations in fresh human bile samples as well as in ex vivo incubated biles (41). In interpreting these data, one special point of caution must be remembered. The depiction of cholesterol crystallization pathways is based on information accumulated from model biles and native biles obtained from cholesterol gallstone patients. Both these situations may not adequately represent what happens in vivo, particularly at the beginning of gallstone formation. This, of course, is a general caveat in the study of gallstone formation and must be kept in mind in discussing gallstone pathogenesis. Nevertheless, since similar pathways have also been observed in sequentially studied animal models, these processes likely play a role in cholesterol crystallization in vivo.
Page 199
VII— Factors Modulating Crystallization Both the rate and extent of cholesterol crystal formation can be affected by numerous physiologically relevant factors, including cholesterol content, phosphatidylcholine content and acyl chain composition, bile salt content and hydrophobicity, total lipid concentration, a panoply of biliary proteins, and ionic constituents of bile, as well as in vitro by temperature and ionic strength. The effects of biliary proteins and the local environment in the gallbladder are reviewed in detail elsewhere in this volume. A— Cholesterol Content Initial studies examining crystal observation time in bile focused on cholesterol saturation index as a key parameter controlling crystallization rate (1). Although there was a general trend of decreasing crystal observation time with increasing cholesterol saturation index, a wide variability was observed for any given value. The limitations of cholesterol saturation index as a single parameter were highlighted by observations that cholesterol crystals did not precipitate from highly supersaturated hepatic bile (65). However, at least in concentrated biles, the cholesterol/phosphatidylcholine of the vesicular phase in supersaturated bile was found to correlate inversely with crystal observation time (114,115). Despite the identification of a variety of pro and antinucleating proteins in bile, multivariate analysis has shown that cholesterol saturation was the only variable independently correlated with crystal observation time (116). The equilibriumphase diagram (Fig. 3) shows why cholesterol saturation index is inadequate as a measurement of propensity for cholesterol crystallization. At the micellar phase limit, the cholesterol saturation index is, by definition, unity. However, at low bile salt/phosphatidylcholine ratios, cholesterol crystals cannot exist at cholesterol contents slightly above the micellar phase limit. The concept of cholesterol monomer activity has been proposed to measure the thermodynamic propensity for cholesterol crystallization (109). The concentration of cholesterol monomers in aqueous solution is extremely low and must be below the limit of cholesterol solubility in unsaturated solutions (approximately 3 × 108) (118). By definition thermodynamically, supersaturated solutions that can form cholesterol monohydrate crystals must contain monomeric cholesterol concentrations above this limit. This monomeric cholesterol activity can be measured experimentally by quantitating partitioning of cholesterol into a polymeric phase (110,117,119). Although crystal observation time has been found to correlate inversely with cholesterol monomer activity (117,120), this approach has not been used widely. B— Bile Salt Species Patients with cholesterol gallstones consistently have an increased fraction of the hydrophobic bile salt deoxycholate in their bile acid pool (121–124). Intestinal bacterial transformation of the trihydroxy bile acid cholic acid produces the more hydrophobic dihydroxy bile acid deoxycholic acid. Deoxycholate is a more powerful detergent and can more efficiently extract cholesterol from the canalicular membrane (125,126). When the reservoir function of the gallbladder is impaired in cholesterol gallstone disease (127,128), newly synthesized hepatic bile tends to be secreted into the intestinal lumen via the common bile duct rather than stored in the gallbladder. Since the relative amount of residence time in the intestine increases, a larger fraction is transformed to deoxycholate. In addition to its role in increasing biliary cholesterol content, deoxycholate independently accelerates cholesterol crystal formation in model (54,129) and native (123) biles. This effect appears to be mediated through an increased cholesterol/phosphatidylcholine ratio in vesicles (123).
Page 200
C— Phosphatidylcholine As discussed above, altering the relative phosphatidylcholine/bile salt ratio can affect the pathway of crystallization in model and native biles. Either an increase in the relative phosphatidylcholine content or addition of exogenous phosphatidylcholine prolongs the crystal observation time (129,130). The type of phosphatidylcholine also influences cholesterol crystallization, with more unsaturated acyl chains tending to accelerate nucleation (131,132), especially in the sn2 position (133). In part, this is due to alterations in the size of the micellar zone (134). Individual phosphatidylcholine species are differentially distributed between micelles and vesicles (135), and vesicular lipids are preferentially adsorbed to early cholesterol crystals (87). D— Calcium The precise role of calcium in cholesterol gallstone formation is unclear. Calcium has minimal effect on the equilibrium solubility of cholesterol in bile salt/phosphatidylcholine solutions (40). The presence of calcium at the core of cholesterol gallstones suggests a role of calcium salts in initial stages of gallstone formation, possibly serving as templates for heterogeneous nucleation. Experimental evidence is conflicting: addition of calcium to model biles accelerated cholesterol crystal formation, but only at supraphysiological concentrations (136,137), while calcium chelation did not reduce crystal observation time in native bile (138). On the contrary, calcium appears to accelerate crystal growth disproportionate to any effect on crystal observation time (139,140). Since calcium binds to monomeric bile salts (141), mixed micelles, and vesicles (142), its effect could be mediated through induction of lateralphase separations of membrane lipids, as is known to occur for other phospholipids (143). Calcium accelerates fusion, particularly of negatively charged phospholipid vesicles (143). Calcium interactions with negatively charged bile salts bound to cholesterol/phosphatidylcholine vesicles could have similar effects in bile (142). E— Total Lipid Concentration Both in model and native biliary lipid systems, increases in total lipid concentration accelerate cholesterol crystallization (144,145), despite the enhanced micellar solubility of cholesterol at high total lipid concentrations (5). Increases in total lipid concentration are associated with increased vesicular cholesterol/phosphatidylcholine ratios. F— Biliary Sterols Other Than Cholesterol Cholesterol is the predominant biliary sterol, constituting over 97% of total sterols (146). In gallstones, polar sterols are enriched relative to bile, and have been suggested to be important in biliary cholesterol crystallization (147). However, the similarity of patterns in cholesterol and pigment stones argues against a pathogenic role (148). VIII— Crystal Growth and Stone Formation After initial nucleation and crystal formation, cholesterol crystals continue to grow and aggregate, forming clusters and finally small stones. Despite the importance of these steps in gallstone pathogenesis, surprisingly little has been published in this regard in gallstone research. This may in part be related to the fact that the process of stone formation is a slow and prolonged one, and it has not been successfully reproduced ex vivo. Structural analysis of gallstones obtained at cholecystectomy (and autopsy) have provided indirect data regarding stone formation and growth.
Page 201
Two separate and distinct forms of stone growth appear to occur in humans (12,149). In one form, a single stone grows, often to quite large dimensions. In the second form, many (occasionally thousands) small stones develop simultaneously, without agglomeration into a single stone. Interestingly, after treatment (by lithotripsy, dissolution, or surgery), when the gallbladder is left in place, the initial growth pattern is usually preserved when stones recur. Why these two different growth patterns, presumably representing two distinct disease patterns, occur is at present obscure. Looking at the "anatomy of gallstones," Womack et al. realized over 35 years ago that gallbladder mucin plays a central role in stone formation and growth (17). They noted that even pure cholesterol stones are built within a meshwork of mucous glycoproteins, while the nucleus of the stone also generally contained a focal collection of mucin. Other types of cholesterol stones were suggested to be agglomerates of smaller stones, held together by mucin. Womack et al. suggested that mucin aggregation was the primary event, upon which cholesterol precipitated and grew to form a stone. Although the universality of a mucinbased nidus has been challenged thereafter (18), more recent studies—employing scanning and freezefracture electron microscopy as well as immunostaining—support the general presence of mucin in the matrix of cholesterol gallstones (23,150,151). Which is the primary event—mucin or cholesterol precipitation—cannot be determined by certainty from these studies. In addition to mucin, cholesterol gallstones also contain other matrix substances. Calcium bilirubinate and other calcium salts (e.g., carbonate and palmitate) have been shown to be deposited cyclically during the growth process of cholesterol stones (19,152). Other metal cations, sulfur, and phosphorus are also present (151). An outer shell of pigment and calcium salts may encase some stones (153,154). Several biliary proteins have been shown to be incorporated into gallstones. Using immunostaining and EDAX microanalysis, specific proteins have been localized to different areas within cholesterol gallstones (151). Mucin was identified in a threedimensional network intercalated between deposits of pigment and cholesterol, whereas the biliary calciumbinding protein APF/CBP and the pronucleating protein aminopeptidaseN coated only the calciumrich pigment deposits. No specific topographical localization was found for albumin or IgA. These findings have been interpreted as evidence for an active role for mucin, APF/CBP, and aminopeptidaseN in the formation of cholesterol gallstones. The authors propose that during stone formation, cholesterol crystals and aminopeptidaseN bind directly to mucin, while calcium salts and pigment deposit on APF/GBP bind hydrophobically to mucin (151). The crystalline form of cholesterol within gallstones is generally held to be the monohydrate. However, anhydrous cholesterol crystals have been observed repeatedly in stones (38,39,155,156). The interpretation has generally been technical, in terms of postharvesting dehydration of the stones held or stored in room air. In view of the more recent observations of possible early anhydrous crystallization of cholesterol in bile, these findings have gained renewed interest. It has been implied that anhydrous crystals, which are needlelike and stickier than the monohydrate plates, may precipitate periodically and induce agglomeration of mixtures of cholesterol microliths (113). Gallstones grow quite slowly. It has been estimated by 14Cradiocarbon measurements that the growth rate is about 2.6 mm per year (157). Calculations from a large cohort of cholecystectomies or periodic followup of individual stone growth by ultrasonography have revealed even slower growth rates, between 0.4 and 2 mm per year (158,159). IX— Conclusion Available data regarding how cholesterol crystals are transformed or incorporated into gallstones, as well as how stones grow, are clearly patchy and incomplete. This somewhat neglected field will hopefully be tackled more intensively in the future, applying some of the newer approaches of modern chemical and biocrystallization techniques to gallstone research.
Page 202
Acknowledgments Supported in part by research funding from the Veterans Administration and the Minerva Center for Cholesterol Gallstones and Lipid Metabolism in the Liver. References 1. Holan KR, Holzbach RT, Hermann RE, Cooperman AM, Claffey WJ. Nucleation time: a key factor in the pathogenesis of cholesterol gallstone disease. Gastroenterology 1979; 77:611–617. 2. van Leeuwenhoek A. Letter No. 16, February 11th 1675. In: Geleerden CvN, ed. The Collected Letters of Antoni van Leeuwenhoek. Amsterdam: NV. Swets & Zeitlinger; 1939:265–267. 3. Lyon BBV. Nonsurgical Drainage of the Gall Tract. Philadelphia and New York: Lea & Febiger; 1923. 4. Juniper K Jr, Burson EN Jr. Biliary tract studies: II. The significance of biliary crystals. Gastroenterology. 1957; 32:175–208. 5. Carey MC, Small DM. The physical chemistry of cholesterol solubility in bile: relationship to gallstone formation and dissolution in man. J Clin Invest 1978; 61:998–1026. 6. Admirand WH, Small DM. The physicochemical basis of cholesterol gallstone formation in man. J Clin Invest 1968; 47:1045–1052. 7. Holzbach RT, Marsh M, Olszewski M, Holan K. Cholesterol solubility in bile: evidence that supersaturated bile is frequent in healthy man. J Clin Invest 1973; 52:1467–1479. 8. Sedaghat A, Grundy SM. Cholesterol crystals and the formation of cholesterol gallstones. N Engl J Med 1980; 302:1274–1277. 9. Marks JW, Bonorris G. Intermittency of cholesterol crystals in duodenal bile from gallstone patients. Gastroenterology 1984; 87:622–627. 10. Bills PM, Lewis D. A structural study of gallstones. Gut 1975; 16:630–637. 11. Juvonen T, Kervinen K, Kairaluoma MI, Lajunen LH, Kesaniemi YA. Gallstone cholesterol content is related to apolipoprotein E polymorphism. Gastroenterology 1993; 104:1806–1813. 12. Wolpers C, Hofmann AF. Solitary versus multiple cholesterol gallbladder stones. Mechanisms of formation and growth. Clin Invest 1993; 71:423–434. 13. Sauerbruch T, Stellaard F, Soehendra N, Paumgartner G. Cholesteringehalt von Gallengangssteinen. Dtsch Med Wochenschr 1983; 108:1099–1102. 14. Trotman BW, Ostrow JD, Soloway RD. Pigment vs cholesterol cholelithiasis: comparison of stone and bile composition. Am J Dig Dis 1974; 199:585–590. 15. Wosiewitz U, Schenk J, Sabinski F, Schmack B. Investigations on common bile duct stones. Digestion 1983; 26:43–52. 16. Nakayama F. Quantitative microanalysis of gallstones. J Lab Clin Med 1968; 72:602–611. 17. Womack NA, Zeppa R, Irvin GL III. The anatomy of gallstones. Ann Surg 1963; 157: 670–686. 18. Malet PF, Williamson CE, Trotman BW, Soloway RD. Composition of pigmented centers of cholesterol gallstones. Hepatology 1986; 6:477–481. 19. Been JM, Bills PM, Lewis D. Microstructure of gallstones. Gastroenterology 1979; 76: 548–555. 20. Ruiz de Aguiar A, Medina Nuñez JA, Lopez Domingo MI, Villacorta Patió J, Leguey Jimenez S. Calcium carbonate in cholesterol gallstones. J Hepatol 1988; 6:71–79. 21. Sanabria JR, Gordon ER, Harvey PR, Goresky CA, Strasberg SM. Accumulation of unconjugated bilirubin in cholesterol pellets implanted in swine gallbladders. Gastroenterology 1996; 110:607–613.
Page 203
22. Kumar S, Burns SJ. The crystallization behavior of gallstones grown from cholesterol. J Mater Sci 1993; 4:460–465. 23. Abe A, Tsuchiya Y, Sugiura N, Saisho H, Nishimura K, Takeo K. Ultrastructure of cholesterol gallstones as observed by electron microscopy after freeze fracturing. Tissue Cell 1997; 29:191–7. 24. LaMont JT, Smith BF, Moore JRL. Role of gallbladder mucin in the pathophysiology of gallstones. Hepatology 1984; 4:51S–56S. 25. Sutor DJ, Wooley SE. The organic matrix of gallstones. Gut 1974; 15:487–491. 26. Holzbach RT. Nucleation of cholesterol crystals in native bile. Hepatology 1990:155S–159S. 27. Carey MC, LaMont JT. Cholesterol gallstone formation: 1. Physicalchemistry of bile and biliary lipid secretion. Prog Liver Dis 1992; 10:139–163. 28. Loomis CR, Shipley GG, Small DM. The phase behavior of hydrated cholesterol. J Lipid Res 1979; 20:525–535. 29. Klinkner AM, Waites CR, Kerns WD, Bugelski PJ. Evidence of foam cell and cholesterol crystal formation in macrophages incubated with oxidized LDL by fluorescence and electron microscopy. J Histochem Cytochem 1995; 43:1071–1078. 30. Katz SS, Small DM. Isolation and and partial characterization of the lipid phases of human atherosclerotic plaques. J Biol Chem 1980; 255:9753–9759. 31. Craven BM. Cholesterol crystal structures: adducts and esters. In: Small DM, ed. The Physical Chemistry of Lipids: From Alkanes to Phospholipids. New York: Plenum Press; 1986:149–182. 32. Craven BM. Crystal structure of cholesterol monohydrate. Nature 1976; 260:727–729. 33. Konikoff FM, Chung DS, Donovan JM, Small DM, Carey MC. Filamentous, helical, and tubular microstructures during cholesterol crystallization from bile: evidence that cholesterol does not nucleate classic monohydrate plates. J Clin Invest 1992; 90:1155–1160. 34. Shieh HS, Hoard LG, Nordman CE. Crystal structure of anhydrous cholesterol. Nature 1977; 267:287–289. 35. Hsu LY, Nordman CE. Phase transition and crystal structure of the 37 degrees C form of cholesterol. Science 1983; 220:604–606. 36. Igimi H, Carey MC. Cholesterol gallstone dissolution in bile: dissolution kinetics of crystalling (anhydrate and monohydrate) cholesterol with chenodeoxycholate, ursodeoxycholate, and their glycine and taurine conjugates. J Lipid Res 1981; 22:254–270. 37. Garti N, Karpuj L, Sarig S. Correlation between crystal habit and the composition of solvated and nonsolvated cholesterol crystals. J Lipid Res 1981; 22:785– 791. 38. Sutor DJ, Gaston PJ. Anhydrous cholesterol: a new crystalline form in gallstones. Gut 1972; 13:64–65. 39. Aho AJ, Vilhonen E, Peltola S, Lehtonen A. An xray diffraction study of the crystalline composition of gallstones. Scand J Gastroenterol 1985; 20:901–906. 40. Wang DQH, Carey MC. Complete mapping of crystallization pathways during cholesterol precipitation from model bile: influence of physicalchemical variables of pathophysiologic relevance and identification of a stable liquid crystalline state in cold, dilute and hydrophilic bile saltcontaining systems. J Lipid Res 1996; 37:606–630. 41. Portincasa P, van EK, Jansen A, Renooij W, Gadellaa M, vanBerge HG. Behavior of various cholesterol crystals in bile from patients with gallstones. Hepatology 1996; 23: 738–748. 42. Chung DS, Benedek GB, Konikoff FM, Donovan JM. Elastic free energy of anisotropic helical ribbons as metastable intermediates in the crystallization of cholesterol. Proc Natl Acad Sci USA 1993; 90:11341–11345. 43. Konikoff FM, Carey MC. Cholesterol crystallization from a dilute bile saltrich model bile. J Crystal Growth 1994; 144:79–86.
Page 204
44. Zastavker YV, Asherie N, Lomakin A, et al. Selfassembly of helical ribbons. Proc Natl Acad Sci U S A 1999; 96:7883–7887. 45. Small DM. The physical chemistry of cholanic acids. In: Nair PP, Kritchevsky D, eds. The Bile Acids, Vol. I. New York: Plenum Press; 1971:249–356. 46. Carey MC. Physicalchemical properties of bile acids and their salts. In: Danielsson H, Sjsvall * J, Van Deenen LLM, eds. Sterols and Bile Acids. Amsterdam: Elsevier; 1985: 345–403. 47. Mazer NA, Benedek GB, Carey MC. Quasielastic lightscattering studies of aqueous biliary lipid systems: mixed micelle formation in bile saltlecithin solutions. Biochemistry 1980; 19:601–615. 48. Cohen DE, Chamberlain RA, Thurston GM, Benedek GB, Carey MC. Cylindrical "wormlike" micelles in bile saltlecithin solutions: implications for the earlies events in bile formation. In: Paumgartner G, Stiehl A, Gerok W, eds. Bile Acids as Therapeutic Agents. Lancaster UK: Kluwer Academic; 1991:147–150. 49. Armstrong MJ, Carey MC. Thermodynamic and molecular determinants of sterol solubilities in bile salt micelles. J Lipid Res 1987; 28:1144–1155. 50. Small DM, Bourg SM, Dervichian DG. Ternary and quaternary aqueous systems containing bile salt, lecithin, and cholesterol. Nature 1966; 211:816–818. 51. Cabral DJ, Small DM. Physical chemistry of bile. In: Schultz SG, Forte JG, Rauner BB, eds. Handbook of Physiology—The Gastrointestinal System III, Section 6. Baltimore, MD: American Physiology Society, Waverly Press; 1989:621–662. 52. Donovan JM. Pathogenesis of Gallstones. In: LaRusso NF, ed. Images in Gastroenterology—Gallstones. Philadelphia: Current Medicine; 1996: 8.1–8.16. 53. Donovan JM, Carey MC. Separation and quantitation of cholesterol "carriers" in bile. Hepatology 1990; 12:94S–105S. 54. Donovan JM, Timofeyeva N, Carey MC. Cholesterol monohydrate and liquid crystal formation in model biles: effects of bile salt hydrophobicity and phosphatidylcholine contents, Hepatology 1989; 10:598. 55. Somjen G, Gilat T. Contribution of vesicular and micellar carriers to cholesterol transport in human bile. J Lipid Res 1985; 26:699–704. 56. Ulloa N, Garrido J, Nervi F. Ultracentrifugal isolation of vesicular carriers of biliary cholesterol in native human and rat bile. Hepatology 1987; 7:235–244. 57. Pattinson N, Chapman B. Distribution of biliary cholesterol between mixed micelles and nonmicelles in relation to fasting and feeding in humans. Gastroenterology 1986; 91: 697–702. 58. Donovan JM, Jackson AA. Accurate separation of biliary lipid aggregates requires the correct intermixed micellar/intervesicular bile salt concentration. Hepatology 1998; 27: 641–648. 59. Harvey PRC, Somjen G, Gilat T, Gallinger S, Strasberg SM. Vesicular cholesterol in bile: Relationship to protein concentration and nucleation time. Biochim Biophys Acta 1988; 958:10–18. 60. Donovan JM, Timofeyeva N, Carey MC. Influence of total lipid concentration, bile salt/lecithin ratio and cholesterol content on inter mixed micellar/vesicular (non lecithin associated) bile salt concentrations in model bile. J Lipid Res 1991; 32:1501–1512. 61. Cohen DE, Angelico M, Carey MC. Structural alterations in lecithincholesterol vesicles following interactions with monomeric and micellar bile salts: physical chemical basis for subselection of biliary lecithin species and aggregative states of biliary lipids during bile formation. J Lipid Res 1990; 31:55–70. 62. McLean LR, Phillips MC. Mechanism of cholesterol and phosphatidylcholine exchange or transfer between unilamellar vesicles. Biochemistry 1981; 20:2893– 2900. 63. Vlahcevic ZR, Gurley EC, Heuman DM, Hylemon PB. Bile salts in submicellar concentrations promote bidirectional cholesterol transfer (exchange) as a function of their hydrophobicity. J Lipid Res 1990; 31:1063–1071.
Page 205
64. Holzbach RT, Kibe A, Thiel E, Howell JH, Marsh M, Hermann RE. Biliary proteins: unique inhibitors of cholesterol crystal nucleation in human gallbladder bile. J Clin Invest 1984; 73:35–45. 65. Groen AK, Stout JPJ, Drapers JAG, Hoek FJ, Grijm R, Tytgat GNJ. Cholesterol nucleationinfluencing activity in Ttube bile. Hepatology 1988; 8:347–352. 66. Lee SP, Nicholls JF, Park HZ. Biliary sludge as a cause of acute pancreatitis. N Engl J Med 1992; 326:589–593. 67. Ros E, Navarro S, Bru C, Garciapuges A, Valderrama R. Occult microlithiasis in idiopathic acute pancreatitis—prevention of relapses by cholecystectomy or ursodeoxycholic acid therapy. Gastroenterology 1991; 101:1701–1709. 68. Janowitz P, Swobodnik W, Wechsler JG, Zoller A, Kuhn K, Ditschuneit H. Comparison of gall bladder bile and endoscopically obtained duodenal bile. Gut 1990; 31:1407–1410. 69. Marks JW, Broomfield P, Bonorris GG, Schoenfield LJ. Factors affecting the measurement of cholesterol nucleation in human gallbladder and duodenal bile. Gastroenterology 1991; 101:214–219. 70. Petroni ML, Jazrawi RP, Ahmed HA, Finch PJ, Dormandy J, Northfield TC. Cholesterol nucleation time measurement in nasobiliary or nasoduodenal bile— comparison with surgical bile. Scand J Gastroenterol 1993; 28:803–808. 71. Luk AS, Kaler EW, Lee SP. PhospholipaseCinduced aggregation and fusion of cholesterollecithin small unilamellar vesicles. Biochemistry 1993; 32:6965– 6973. 72. Janowitz P, Wechsler JG, Janowitz A, Kuhn K, Swobodnik W, Ditschuneit H. Nucleation time, cholesterol saturation index, and biliary bile acid pattern: a comparison in responders and nonresponders to systemic litholysis with bile acids. Scand J Gastroenterol 1991; 26:367–373. 73. Jungst D, del PR, Dolu MH, Schneeweiss SG, Frimberger E. Rapid formation of cholesterol crystals in gallbladder bile is associated with stone recurrence after laparoscopic cholecystotomy. Hepatology 1997; 25:509–513. 74. FudimLevin E, Bor A, Kaplun A, Talmon Y, Lichtenberg D. Cholesterol precipitation from cholesterolsupersaturated bile models. Biochim Biophys Acta 1995; 1259: 23–28. 75. Small DM. Cholesterol nucleation and growth in gallstone formation. N Engl J Med 1980; 302:1305–1307. 76. Harvey PR, Upadhya GA, Strasberg SM. Cholesterol microcrystals associated with concanavalin Abinding glycoproteins contribute artifactually to nucleating activity assays. J Lipid Res 1995; 36:2661–2669. 77. Drapers JAG, Groen AK, Stout JPJ, et al. Quantification of cholesterol nucleation promoting activity in human gallbladder bile. Clin Chim Acta 1987; 165:295– 302. 78. Lee TJ, Smith BF. Bovine gallbladder mucin promotes cholesterol crystal nucleation from cholesteroltransporting vesicles in supersaturated model bile. J Lipid Res 1989; 30:491–498. 79. Levy PF, Smith BF, LaMont JT. Human gallbladder mucin accelerates nucleation of cholesterol in artificial bile. Gastroenterology 1984; 87:270–275. 80. Whiting M J, Watts JM. Cholesterol crystal formation and growth in model bile solutions. J Lipid Res 1983; 24:861–868. 81. Busch N, Tokumo H, Holzbach RT. A sensitive method for determination of cholesterol growth using model solutions of supersaturated bile. J Lipid Res 1990; 31:1903–1909. 82. Harvey PR, Upadhya GA. A rapid, simple high capacity cholesterol crystal growth assay. J Lipid Res 1995; 36:2054–2058. 83. Somjen GJ, Ringel Y, Konikoff FM, Rosenberg R, Gilat T. A new method for the rapid measurement of cholesterol crystallization in model biles using a spectrophotometric microplate reader. J Lipid Res 1997; 38:1048–1052. 84. Corradini SG, Cantafora A, Capocaccia L, Dellaguardia P, Giacomelli L, Angelico M.
Page 206
Development and validation of a quantitative assay for cholesterol crystal growth in human gallbladder bile. BbaLipid Lipid Metab 1994; 1214:63–72. 85. Ohya T, Tazuma S, Hatsushika S, et al. An estimation of human bile metastability: clinical application of a sensitive cholesterol crystal growth assay. J Gastroenterol Hepatol 1994; 9:223–227. 86. Wang DQ, Carey MC. Characterization of crystallization pathways during cholesterol precipitation from human gallbladder biles: identical pathways to corresponding model biles with three predominating sequences. J Lipid Res 1996; 37:2539–2549. 87. Konikoff FM, Cohen DE, Carey MC. Phospholipid molecular species influence crystal habits and transition sequences of metastable intermediates during cholesterol crystallization from bile SaltRich model bile. J Lipid Res 1994; 35:60–70. 88. Konikoff FM, Laufer H, Messer G, Gilat T. Monitoring cholesterol crystallization from lithogenic model bile by timelapse density gradient ultracentrifugation. J Hepatol 1997; 26:703–710. 89. Cohen DE, Carey MC. Physical chemistry of biliary lipids during bile formation. Hepatology 1990:143S–147S. 90. Cohen DE, Angelico M, Carey MC. Quasielastic light scattering evidence for vesicular secretion of biliary lipids. Am J Physiol 1989; 257:G1–G8. 91. Elferink RPJO, Ottenhoff R, Vanwijland M, Smit JJM, Schinkel AH, Groen AK. Regulation of biliary lipid secretion by mdr2 pglycoprotein in the mouse. J Clin Invest 1995; 95:31–38. 92. Crawford JM, Mockel GM, Crawford AR, et al. Imaging biliary lipid secretion in the rat: ultrastructural evidence for vesiculation of the hepatocyte canalicular membrane. J Lipid Res 1995; 36:2147–2163. 93. Donovan JM. Physical and metabolic factors in gallstone pathogenesis. Gastroenterol Clin North Am 1999; 28:75–98. 94. Collins JJ, Phillips MC. The stability and structure of cholesterolrich codispersions of cholesterol and phosphatidylcholine. J Lipid Res 1982; 23:291–298. 95. Gerloff T, Steiger B, Hagenbuch B, et al. The sister of Pglycoprotein represents the canalicular bile salt export pump of mammalian liver. J Biol Chem 1998; 173:10046–10050. 96. Carey MC, Cohen DE. Update on physical state of bile. Ital J Gastroenterol 1995; 27:92–100. 97. Afdhal NH, Niu N, Nunes DP, et al. Mucinvesicle interactions in model bile: evidence for vesicle aggregation and fusion before cholesterol crystal formation. Hepatology 1995; 22:856–865. 98. Halpern Z, Dudley MA, Lynn MP, Nader JM, Breuer AC, Holzbach RT. Vesicle aggregation in model systems of supersaturated bile: relation to crystal nucleation and lipid composition of the vesicular phase. J Lipid Res 1986; 27:295–306. 99. Halpern Z, Dudley MA, Kibe A, Lynn MP, Breuer AC, Holzbach RT. Rapid vesicle formation, and aggregation in abnormal human biles: a timelapse video enhanced contrast microscopy study. Gastroenterol 1986; 90:875–885. 100. Peled Y, Halpern Z, Eitan B, Goldman G, Konikoff F, Gilat T. Biliary micellar cholesterol nucleates via the vesicular pathway. Biochim Biophys Acta 1989; 1989:246–249. 101. de Bruijn MAC, Goldhoorn BG, Zijlstra AIM, Tytgat GNJ, Groen AK. Interaction of cholesterolcrystallizationpromoting proteins with vesicles. Biochem J 1995; 305:93–96. 102. Kibe A, Holzbach RT. Inhibition of cholesterol crystal formation by apolipoproteins in supersaturated model bile. Science 1984; 225:514–516. 103. Tao S, Tazuma S, Kajiyama G. Apolipoprotein AI stabilizes phospholipid lamellae and thus prolongs nucleation time in model bile systems: an ultrastructural study. Biochim Biophys Acta 1993; 1166:25–30. 104. Phillips MC. Cholesterol packing, crystallization and exchange properties in phosphatidylcholine vesicle systems. Hepatology 1990; 12:75S–80S; discussion 80S–82S.
Page 207
105. Gantz DL, Wang DQ, Carey MC, Small DM. Cryoelectron microscopy of a nucleating model bile in vitreous ice: formation of primordial vesicles. Biophys J 1999; 76:1436–1451. 106. Kaplun A, Talmon Y, Konikoff FM, et al. Direct visualization of lipid aggregates in native human bile by light and cryotransmission electron microscopy. FEBS Lett 1994; 340:78–82. 107. Kaplun A, Konikoff FM, Eitan A, et al. Imaging supramolecular aggregates in bile models and human bile. Microsc Res Tech 1997; 39:85–96. 108. Konikoff FM, Danino D, Weihs D, Rubin M, Talmon Y. Microstructural evolution of lipid aggregates in nucleating model and human biles visualized by cryo transmission electron microscopy. Hepatology 2000; 31:261–268. 109. Ahrendt SA, Foxtalbot K, Kaufman HS, Lillemoe KD, Pitt HA. Cholesterol nucleates rapidly from mixed micelles in the prairie dog. BbaLipid Lipid Metab 1994; 1211:7–13. 110. Donovan JM, Jackson AA, Yuet PK, Blankschtein D. Cholesterol thermodynamic activity determines cholesterol crystal habit in model bile. Gastroenterology 1996; 110:A454. 111. Juste C, Catala I, Henry R, et al. Influence of bile salt molecular species on cholesterol crystallization from supersaturated model biles. BbaLipid Lipid Metab 1995; 1254:89–97. 112. Konikoff FM, Cohen DE, Carey MC. Filamentous crystallization of cholesterol and its dependence on lecithin species in bile. Mol Cryst Liq Cryst 1994; 248:291–296. 113. Carey MC. Formation and growth of cholesterol gallstones: the new synthesis. In: Fromm H, Leuschner U, eds. Bile AcidsCholestasisGallstones. Dordrecht: Kluwer Academic; 1996:147–175. 114. Harvey PRC, Rupar CA, Gallinger S, Petrunka CN, Strasberg SM. Quantitative and qualitative comparison of gallbladder mucous glycoprotein from patients with and without gallstones. Gut 1986; 27:374–381. 115. Somjen GJ, Rosenberg R, Gilat T. Gel filtration and quasielastic light scattering studies of human bile. Hepatology 1990:123S–128S. 116. Miquel JF, Nunez L, Amigo L, et al. Cholesterol saturation, not proteins or cholecystitis, is critical for crystal formation in human gallbladder bile. Gastroenterology 1998; 114:1016–1023. 117. Chijiiwa K, Kiyosawa R, Nakayama F. Cholesterol monomer activity correlates with nucleation time in model bile. Clin Chim Acta 1988; 178:181–192. 118. Renshaw PE, Janoff AS, Miller KW. On the nature of dilute aqueous cholesterol suspensions. J Lipid Res 1983; 24:47–51. 119. Jain UK, Higuchi WI, Liu CL, Lee PH, Mazer NA. Cholesterol (thermodynamic) activity determinations in bile saltlecithincholesterol systems and cholesterol rich liquid crystalline mesophase formation. Pharm Res 1992; 9:792–799. 120. Noshiro H, Chijiiwa K, Hirota I. Activity of cholesterol in human gallbladder bile in relation to nucleation of cholesterol monohydrate crystals. Clin Chim Acta 1992; 205:167–179. 121. Marcus SN, Heaton KW. Intestinal transit, deoxycholic acid and the cholesterol saturation of bile—three interrelated factors. Gut 1986; 27:550–558. 122. Marks JW, Bonorris GG, Schoenfield LJ. Roles of deoxycholate and arachidonate in pathogenesis of cholesterol gallstones in obese patients during rapid loss of weight. Digest Dis Sci 1991; 36:957–960. 123. Hussaini SH, Pereira SP, Murphy GM, Dowling RH. Deoxycholic acid influences cholesterol solubilization and microcrystal nucleation time in gallbladder bile. Hepatology 1995; 22:1735–1744. 124. VanBerge Henegouwen GP, van der Werf SDJ, Ruben AT. Fatty acid composition of phospholipids in bile in man: Promoting effect of deoxycholate on arachidonate. Clin Chim Acta 1987; 165:27–37.
Page 208
125. Carulli N, Loria P, Bertolotti M, et al. Effects of acute changes of bile acid pool composition on biliary lipid secretion. J Clin Invest 1984; 74:614–624. 126. Gurantz D, Hofmann AF. Influence of bile acid structure on bile flow and biliary lipid secretion in the hamster. Am J Physiol 1984; 247:G736–G748. 127. Pomeranz IS, Shaffer EA. Abnormal gallbladder emptying in a subgroup of patients with gallstones. Gastroenterology 1985; 88:787–791. 128. van der Werf SDJ, van Berge Henegouwen GP, Palsma DMH, Ruben AT. Motor function of the gallbladder and cholesterol saturation of duodenal bile. Neth J Med 1987; 30:160–171. 129. Jungst D, Lang T, Huber P, Lange V, Paumgartner G. Effect of phospholipids and bile acids on cholesterol nucleation time and vesicular/micellar cholesterol in gallbladder bile of patients with cholesterol stones. J Lipid Res 1993; 34:1457–1464. 130. Holzbach RT, Marsh M. Transient liquid crystals in human bile analogues. Mol Cryst Liq Cryst 1974; 28:217. 131. Tao S, Tazuma S, Kajiyama G. Fatty acid composition of lecithin is a key factor in bile metastability in supersaturated model bile systems. Biochim Biophys Acta 1993; 1167:142–146. 132. Tazuma S, Ochi H, Teramen K, et al. Degree of fatty acyl chain unsaturation in biliary lecithin dictates cholesterol nucleation and crystal growth. BbaLipid Lipid Metab 1994; 1215:74–78. 133. Ringel Y, Somjen GJ, Konikoff FM, Rosenberg R, Gilat T. Increased saturation of the fatty acids in the sn2 position of phospholipids reduces cholesterol crystallization in model biles. Biochim Biophys Acta 1998; 1390:293–300. 134. Halpern Z, Moshkowitz M, Laufer H, Peled Y, Gilat T. Effect of phospholipids and their molecular species on cholesterol solubility and nucleation in human and model biles. Gut 1993; 34:110–115. 135. Cohen DE, Carey MC. Acyl chain unsaturation modulates distribution of lecithin molecular species between mixed micelles and vesicles in model bile— implications for particle structure and metastable cholesterol solubilities. J Lipid Res 1991; 32:1291–1302. 136. Kibe A, Dudley MA, Halpern Z, Lynn MP, Breuer AC, Holzbach RT. Factors affecting cholesterol monohydrate crystal nucleation time in model systems of supersaturated bile. J Lipid Res 1985; 26:1102–1111. 137. Toor EW, Evans DF, Cussler EL. Cholesterol monohydrate growth in model bile solutions. Proc Natl Acad Sci USA 1978; 75:6230–6234. 138. Gallinger S, Harvey PRC, Petrunka CN, Strasberg SM. Effect of binding of ionised calcium on the in vitro nucleation of cholesterol and calcium bilirubinate in human gall bladder bile. Gut 1986; 27:1382–1386. 139. Berenson MM, Cardinal JR. Calcium accelerates cholesterol phase transitions in analog bile. Experientia 1985; 41:1328–1330. 140. Konikoff FM, Lechene de la Porte P, Laufer H, Domingo N, Lafont H, Gilat T. Calcium and the anionic polypeptide fraction (APF) have opposing effects on cholesterol crystallization in model bile. J Hepatol 1997; 27:707–715. 141. Moore EW. The role of calcium in the pathogenesis of gallstones: Ca++ electrode studies of model bile salt solutions and other biologic systems: with a hypothesis on structural requirements for Ca++ binding to proteins and bile acids. Hepatology 1984; 4:228S–243S. 142. Donovan JM, Leonard MR, Batta AK, Carey MC. Calcium affinity for biliary lipid aggregates in model biles: complementary importance of bile salts and lecithin. Gastroenterology 1994; 107:831–846. 143. Leckband DE, Helm CA, Israelachvili J. Role of calcium in the adhesion and fusion of bilayers. Biochemistry 1993; 32:1127–1140. 144. van Erpecum KJ, vanBerge Henegouwen GP, Stoelwinder B, Schmidt YMG, Willekens FLH. Bile concentration is a key factor for nucleation of cholesterol crystals and cho
Page 209
lesterol saturation index in gallbladder bile of gallstone patients. Hepatology 1990; 11:1–6. 145. van Erpecum KJ, Stolk MFJ, van den Broek AMWC, Renooij W, van de Heijning BJM, van Berge Henegouwen GP. Bile concentration promotes nucleation of cholesterol monohydrate crystals by increasing the cholesterol concentration in the vesicles. Eur J Clin Invest 1993; 23:283–288. 146. Miettinen TA, Kesaniemi YA, Jarvinen H, Hastbacka J. Cholesterol precursor sterols, plant sterols, and cholestanol in human bile and gallstones. Gastroenterology 1986; 90:858–864. 147. Boldrini P. CC2W is the physical cause of cholesterol gallstones. Physiol Chem Phys 1979; 11:303–316. 148. Miettinen TE, Kesaniemi YA, Gylling H, Jarvinen H, Silvennoinen E, Miettinen TA. Noncholesterol sterols in bile and stones of patients with cholesterol and pigment stones. Hepatology 1996; 23:274–280. 149. Jungst D, Lang T, Vonritter C, Pratschke E, Paumgartner G. Cholesterol nucleation time in gallbladder bile of patients with solitary or multiple cholesterol gallstones. Hepatology 1992; 15:804–808. 150. Bogren HG, Mutvei H, Renberg G. Scanning electron microscope studies of human gallstones after plasma etching. Ultrastruct Pathol 1995; 19:447–453. 151. de la Porte PL, Domingo N, Vanwijland M, Groen AK, Ostrow JD, Lafont H. Distinct immunolocalization of mucin and other biliary proteins in human cholesterol gallstones. J Hepatol 1996; 25:339–348. 152. Malet PF, Weston NE, Trotman BW, Soloway RD. Cyclic deposition of calcium salts during growth of cholesterol gallstones. Scan Electron Microsc 1985; 11 (Pt 2):775–779. 153. Kaufman HS, Magnuson TH, Pitt HA, Frasca P, Lillemoe KD. The distribution of calcium salt precipitates in the core, periphery and shell of cholesterol, black pigment and brown pigment gallstones. Hepatology 1994; 19(5):1124–1132. 154. Ostrow JD. Unconjugated bilirubin and cholesterol gallstone formation. Hepatology 1990; 12:219S–224S. 155. Sutor DJ, Wooley SE. Xray diffraction studies of the composition of gallstones from English and Australian patients. Gut 1969; 10:681–683. 156. Bogren H, Larsson K. An xray diffraction study of crystalline cholesterol in some pathological deposits in man. Biochim Biophys Acta 1963; 75:65–69. 157. Mok HYI, Druffel ERM, Rampone WM. Chronology of cholelithiasis: dating gallstones from atmospheric radiocarbon produced by nuclear bomb explosions. N Engl J Med 1986; 314:1075–1077. 158. Lowenfels AB, Walker AM, Althaus DP, Townsend G, Domello L. Gallstone growth, size, and risk of gallbladder cancer: an interracial study. Int J Epidemiol 1989; 18:50–54. 159. Nudelman I. On the growth rate of gallstones in the human gallbladder. J Crystal Growth 1993; 130:1–5.
Page 211
10— Gallbladder Mucin Gwynneth D. Offner Boston University Medical Center, Boston, Massachusetts I— Introduction Epithelial surfaces lining the gastrointestinal, respiratory, and genitourinary tracts are covered with a layer of mucus comprised of water, electrolytes, proteins, and DNA. This layer forms a viscous barrier that protects the underlying epithelium from desiccation, mechanical injury, and chemical and microbial assault while allowing active absorption and secretion by mucosal cells. The principal protein components of the mucous layer are called mucins. These proteins are made up of approximately 15 to 20% protein and 80% carbohydrate, present largely in the form of Olinked glycans (1–4). They are well suited for their protective functions in that mucin monomers can form multimers, with apparent molecular weights greater than 20 to 40 million Da. The hydrophilic mucin multimers expand rapidly in the presence of water molecules shortly after secretion to form the cytoprotective gel layer. Because of the unique physiological environment within the gallbladder, mucins secreted by this epithelium have been the subject of much active research over the past three decades. Gallbladder mucins must protect the gallbladder epithelium from a variety of potentially harmful and cytotoxic substances present in bile, such as bile acids, lysophospholipids, and oxygen metabolites. Perhaps more importantly, however, ample evidence has accumulated that, in addition to their protective functions, gallbladder mucins also have a major role in the pathogenesis of cholesterol gallstone disease. This chapter reviews the structural features of gallbladder mucins, the nature of their interactions with biliary lipids, and factors that regulate synthesis and secretion. This information is correlated with the multiple functions of mucins in the healthy and pathological gallbladder. II— General Properties of Mucins The very properties that enable mucins to function effectively as barrier molecules have made study of these proteins by classic biochemical techniques a formidable challenge. Their large size and extensive glycosylation have made it nearly impossible to obtain primary amino acid sequence information. In addition, mucins display considerable heterogeneity in both the protein and carbohydrate moieties (1–3). Early studies that focused on the overall structural and rheological properties of mucins demonstrated that these macromolecules were made up of monomeric subunits that polymerized into rodlike structures, presumably by endto end linkage (5,6). The ability to polymerize is crucial for the viscoelastic properties of mucins and the
Page 212
formation of mucin gels. That disulfide bonding between mucin monomers could account for their ability to polymerize was shown by treatment with reducing agents, which led to depolymerization (7,8). A— Mucin Glycans While the mucin polypeptide backbone has been difficult to study, characterization of mucin glycans by classic techniques has revealed that most are Olinked, although several early reports have suggested the presence of Nlinked glycans as well (1). Molecular cloning studies have now shown that virtually all mucins contain the consensus Nglycosylation sites AsnXSer and AsnXThr. In general, mucin Oglycans range in length from 8 to 20 sugar residues and contain Nacetyl galactosamine (GalNAc), Nacetyl glucosamine (GlcNAc), galactose (Gal), fucose (Fuc), and sialic acid (NeuNAc) (9). The glycan structure can be divided into a core region containing GalNAc attached to serine or threonine residues in the mucin polypeptide, a backbone region, and a peripheral region. Six types of simple core structures have been identified that can either be directly terminated with sialic acid residues or can serve as the attachment sites for the backbone sugar residues. Two common types of backbone structures have been identified: type 1, Gal (1–3)GlcNAc, and type 2, Gal (1–4)GlcNAc. The backbone structures can be branched and sometimes sulfated or terminated with peripheral sugars Fuc, GalNAc, Gal, and NeuNAc. The peripheral regions of many mucin glycans are rich in sialic acid and sulfate esters, which confer an overall negative charge to the sugar moieties and to the molecule as a whole. The complexity of these glycan structures is amplified by the fact that many different side chains can be found on a single mucin molecule. Furthermore, side chains can be terminated prematurely during synthesis, owing in part to the availability of specific glycosyltransferases or substrate sugars in different Golgi compartments. B— Mucin Primary Structure It was only after molecular cloning techniques were applied to the study of human mucins that the first information about the primary structure of the polypeptide backbone became available. These studies are discussed in greater detail in Sec. IV.A below, but they showed that, in general, all human mucins share several common structural features. The mucin protein backbone can be divided into two distinct regions. The first—enriched with respect to serine, threonine, and sometimes proline—contains tandemly repeated sequences ranging from 8 to 169 amino acids in length. In some cases, these sequences may be repeated hundreds of times with little variation. In many mucins, this domain occupies the central region of the molecule and serves as the attachment site for Olinked glycans. The sequences flanking the central tandem repeat domain are frequently enriched with respect to cysteine and are only sparsely glycosylated with both O and Nlinked sugars. The polymerization of mucin monomers into multimers occurs through intermolecular disulfide linkages between these cysteinerich domains. III— Gallbladder Mucin and the Pathogenesis of Gallstone Disease It is now well recognized that the development of gallstones is multifactorial (10,11), involving three primary defects: (a) hepatic secretion of cholesterol supersaturated bile; (b) accelerated nucleation of cholesterol monohydrate crystals within the gallbladder; and (c) impaired gallbladder motility with stasis, allowing growth and agglomeration of cholesterol crystals into gallstones. Studies performed in our laboratory and elsewhere have shown that gallbladder mucin is intimately involved in the latter two abnormalities; it accelerates the nucleation of
Page 213
cholesterol crystals and, as a consequence of hypersecretion, forms a mucin gel layer that provides an ideal environment for the growth of cholesterol crystals into mature stones. A— Gallbladder Mucin Is Found in Cholesterol Gallstones Some of the earliest evidence for the involvement of gallbladder mucin in gallstone disease came from the observation that a glycosylated substance could be detected in the core of cholesterol gallstones as well as in the stone matrix (12,13). This led to the speculation that this glycoprotein might serve as a scaffold upon which cholesterol, calcium, and other organic molecules could be deposited. Subsequent studies identified this glycoprotein as mucin by demonstrating that the glycoprotein isolated from gallstones had a composition similar to that of the highmolecularweight mucin isolated from gallbladder bile (14,15). More recent work using specific antibodies has localized mucin to the core of cholesterol gallstones as well as to a threedimensional fibrillar network intercalated with cholesterol crystals (16). B— Mucin Hypersecretion Precedes the Formation of Stones Numerous studies have now shown that mucin hypersecretion occurs prior to the formation of cholesterol crystals and gallstones in a variety of animal models. One of the most widely used models is the prairie dog, which, in response to cholesterol feeding, rapidly develops cholesterolsaturated bile and stones. In a seminal study, Lee and colleagues examined the rate of mucin secretion in explants from gallbladders removed 3 to 14 days after initiating the cholesterolenriched diet (17). Increased mucin secretion was first noted on day 3, although no crystals could be detected until day 5, at which time mucin synthesis reached a maximum. As discussed below, the cholesterol crystals were embedded in the thick layer of mucin covering the gallbladder epithelium. Hypersecretion of mucin before the appearance of cholesterol crystals and stones has also been observed in mice (18,19), hamsters (20), and ground squirrels (21,22) fed a lithogenic diet. Numerous studies in humans have also shown that mucin levels are increased in the gallbladders of patients with cholesterol gallstone disease (14,23–26). While no temporal relationship between mucin secretion and stone formation could be established by these observations, a series of recent studies on morbidly obese subjects has provided a unique opportunity to investigate changes in the bile composition of patients who developed gallstones during rapid weight reduction. Patients in these studies had a sample of gallbladder bile removed at the time of gastric bypass surgery and again at the time of cholecystectomy for symptomatic gallstones. Mucin levels increased 18 to 50fold over the levels found prior to stone formation (27,28). C— Mucin Forms a Gel Layer in Which Cholesterol Crystals Nucleate While concentrations of mucin in the bulk phase of bile range from 1 to 4 mg/mL, hypersecretion can result in concentrations as high as 10 to 20 mg/mL in the gel layer adherent to the gallbladder epithelium. Examination of gallbladders removed at cholecystectomy from patients with cholesterol stones revealed that crystals were found predominantly within the mucous gel layer, indicating that high local concentrations of gallbladder mucin provided an ideal environment for stone formation (29,30). These findings have been extended by the demonstration of cholesterol crystals in biliary sludge—a complex mixture of calcium bilirubinate, mucin, and other proteins—which is recognized as a precursor to the development of gallstones in highrisk individuals such as patients on longterm parenteral nutrition or those on rapidweightreducing diets (31,32). As discussed above, Lee and colleagues demonstrated that in prairie dogs fed a highcholesterol diet, hypersecretion of gallbladder mucin preceded the appearance of cholesterol
Page 214
crystals (17). It was also noted that in this animal model, crystals were first found within the mucous gel layer of the gallbladder. Similar findings were obtained in mice (18,19), Syrian hamsters (20), and ground squirrels (21,22) fed a lithogenic diet. In a more recent study, Syrian hamsters treated with estradiol and medroxyprogesterone showed marked hypersecretion of mucin, which was associated with crystallike deposits viewed by transmission and scanning electron microscopy (33). Taken together, these findings in both humans and animal models have shown that interactions between mucin and biliary lipids are promoted in the thick mucin gel lining the gallbladder, much in the same way that gels have been shown to promote crystallization and crystal growth in industrial applications (34). D— Role of Mucin in Nucleation and Growth of Cholesterol Crystals In order to dissect the mechanisms by which gallbladder mucin promotes gallstone formation, in vitro models were developed to examine different stages in the pathway of cholesterol crystallization. In early work, purified human gallbladder mucin was added to model bile containing lithogenic concentrations of bile salts (usually sodium taurocholate), phospholipid (eggyolk phosphatidylcholine) and cholesterol with cholesterol saturation index (CSI) 1.3–1.4. In a typical ''nucleation" assay, aliquots of the solution were examined daily by polarizing light microscopy for the appearance of cholesterol crystals. These studies showed that mucin at concentrations of 1 to 4 mg/mL were capable of accelerating the appearance of crystals in a time and dosedependent manner (35,36). Similar results were obtained when purified mucin was added back to bile from cholesterol gallstone patients from which the mucin had been removed by ultracentrifugation and ultrafiltration (37). These early studies led to several important observations. First, they showed that mucin purified from control gallbladders nucleated as effectively as mucin purified from the gallbladders of patients with gallstone disease (35). This suggested that there are no qualitative differences in the gallbladder mucins isolated from these two groups of individuals and that such differences could not be used to explain the propensity of some individuals, families, and ethnic groups to develop gallstone disease. It was concluded that in cholesterol gallstone patients, the increased quantity of gallbladder mucin but not the properties of mucin itself was important for accelerating cholesterol crystallization both in vivo and in vitro. However, in view of what is now known about the complex expression pattern of mucin genes in the gallbladder, it is tempting to speculate that alterations in the levels of one or more of these genes could contribute to the development of gallstone disease (See Sec. VI). The second key observation provided by these studies was that gallbladder mucin contained two distinct domains, one heavily glycosylated and the other minimally or nonglycosylated. Both bovine and human gallbladder mucin were shown to bind a variety of hydrophobic ligands including bilirubin (38), hydrophobic dyes (36,39), cholesterol (36), and phospholipids (36). Binding of these ligands could be abolished by prior treatment of the mucin with pronase, which could remove only the nonglycosylated portions of the molecule, leaving the heavily glycosylated portion intact (36,39). In addition, Smith showed that pronasetreated mucin lost its ability to promote cholesterol crystallization in vitro (36). These experiments demonstrated that the proteasesensitive, nonglycosylated domains of mucin were critical for its interactions with biliary lipids. In addition to its role in promoting cholesterol crystal nucleation, gallbladder mucin has also been shown to promote crystal growth. Bovine gallbladder mucin was added to supersaturated model bile preseeded with cholesterol crystals of a defined size. Daily analysis of the incubation mixture by polarizing light microscopy showed a time and concentrationdependent increase in crystal area beginning at day 3. This increase in crystal mass was accompanied by a concomitant decrease in the CSI of the model bile with no detectable increase in the number of crystals present (40). Since few crystal aggregates were detected, the results of this study suggest that mucin promotes the growth of crystals by accelerating the transfer of cholesterol from either smaller microcrystals or from cholesterolrich vesicles.
Page 215
IV— Identification of Gallbladder Mucins It is now clear that, in humans, a number of different mucin genes are expressed in the gallbladder. Each of these genes is expressed in other tissues in the gastrointestinal and respiratory tracts and, as yet, a unique gallbladder mucin gene has not been identified. The identification of the major gallbladder mucins has come only recently following a decade of research in which molecular cloning techniques were applied to the study of human and other mammalian mucin genes. A— Human Mucin Genes To date, the partial or complete sequences of nine human mucins have been determined (2,3,41,42). Mucin genes have been given the name MUC and have been numbered MUC1 to MUC4, MUC5AC, MUC5B, and MUC6 to MUC8, in the order of their discovery. Each of these genes is expressed in a unique but overlapping tissuespecific pattern. Human mucins can be subdivided into membraneassociated and secreted forms and into high and lowmolecularweight forms. In addition, mucins have also been classified as either epitheliumassociated or endotheliumleukocyte associated glycoproteins. The latter group includes adhesion molecules, some of which function as ligands for selectins and integrins (42). Because these molecules lack many of the characteristic structural features of epithelial mucins, they are not discussed further here. Many of the mucins described below were identified using a common technique. Mucin was isolated from the appropriate tissue and purified using a combination of sizeexclusion chromatography and cesium chloride densitygradient ultracentrifugation. The purified protein was deglycosylated using hydrogen fluoride and antibodies were raised against the deglycosylated protein. These antibodies were used to screen a cDNA expression library prepared with mRNA isolated from the corresponding tissue. In most cases, this method resulted in the identification of cDNA clones encoding mucin tandem repeats, since the deglycosylated repeat sequences are highly immunogenic. Clones containing the sequences flanking the tandem repeats were more difficult to obtain; frequently, genomic cloning or 5' and 3' rapid amplification of cDNA ends (RACE) were used to obtain complete mucin sequences. The properties of each of the human mucins identified to date are listed in Table 1 and are discussed briefly below. 1— MUC1 MUC1 is expressed almost ubiquitously in normal epithelia, including the gallbladder, and in neoplastic tissues (43,44). It was first isolated nearly simultaneously from mammary gland and pancreas (45–47) and was the first mucin identified that contains a membranespanning domain, through which it is anchored to the cell surface. This mucin, also referred to as episialin, contains wellconserved 20amino acid tandem repeats and encodes a 300kDa mucin gene product. MUC1 contains a 69 amino acid cytoplasmic domain that is thought to interact with intracellular actin, a transmembrane domain, and an extracellular domain (43,48). MUC1 is not a gelforming mucin, but it has been shown to exist in both soluble and membraneassociated forms. These forms can result from alternative splicing of MUC1 mRNAs (49,50), or from a novel recycling pathway in which the membraneassociated form can be internalized and reprocessed in the Golgi apparatus with further glycosylation and removal of the cytoplasmic tail (51). The recycled protein can then be secreted from the cell as a different species (52). The function of MUC1 in normal epithelia is not well established, but it has recently been shown to have an important role in the enhancement of tumor progression and metastasis. Increased levels of MUC1 expression in carcinoma cells inhibit cellcell adhesion and interactions with cytotoxic T lymphocytes (53,54) as well as reducing integrin mediated contacts with the extracellular matrix (55).
Page 216 Table 1 Human Mucin Genes Gene
Source of cDNA
Expression in GB ++
Tandem repeat
MUC1
Pancreatic/Mammary
MUC2
Intestine
20 aa GSTAPPAHGVTSAPDTRPAP
MUC3
Intestine
+++
17 aa HSTPSFTSSITTTETTS
MUC4
Trachea
16 aa TSSASTGHATPLPVTD
MUC5AC
Trachea, stomach
+
MUC5B
Trachea, gallbladder
MUC6
23 aa PTTTPITTTTTVTPTPTPTGTQT
8 aa TSTTSAP
+++
29 aa SSTPGTAHTLTVLTTTATTPTATGSTATP
Stomach
+
169 aa SPFSSTGPMTATSFQTTTTYPTPSHPQTT LPTHVPPFSTSLVTPSTGTVITPTHAQMA TSASIHSTPTGTIPPPTTLKATGSTHTAPP MTPTTSGTSQAHSSFSTAKTSTSLHSHTS STHHPEVTPTSTTTITPNPTSTGTSTPVAH TTSATSSRLPTPFTTHSPPTGS
MUC7
Salivary Gland
MUC8
Trachea
a
Degenerate tandem repeats.
23 aa TTAAPPTPSATTPAPPSSSAPPE 13/41 aa TSCPRPLQEGTRVa
Page 217
2— MUC2, MUC5AC, MUC5B, and MUC6 MUC2, MUC5AC, MUC5B, and MUC6 are all secreted highmolecularweight mucins. The genes for these mucins are clustered at 11p15 (56,57) and all have been shown to have a similar structural organization. However, each of these mucins displays a unique pattern of tissuespecific expression. MUC2 was first cloned from a human small intestinal cDNA library screened with antibodies to deglycosylated colonic mucin (58). It is expressed primarily in small intestine, colon, and respiratory epithelium (44,59,60) and was the first highmolecularweight mucin to be sequenced completely (61,62). This mucin is >5100 amino acids in length and contains a central domain made up of 23 amino acid repeats that is flanked by cysteinerich regions at the Nand Ctermini (61,62). The amino terminal domain contains D1, D2, and D3 domains analogous to those found in human prepro von Willebrand factor (63,64), while the carboxylterminal domain contains a D4 domain followed by a C1 domain and an extreme Cterminal cystine knot motif. The cystine knot is a higherordered structure which is found in transforming growth factor beta (TBF ) and related growth factors (65). This structure contains three pairs of disulfidebonded cysteine residues with an internal free cysteine which has been implicated in the formation of homo and heterodimers (66). This domain is necessary for the dimerization and efficient secretion and packaging of von Willebrand factor monomers (63,67) and well as porcine submaxillary mucin monomers (68). In von Willebrand factor, the aminoterminal D domains are involved in the higherordered polymerization of protein dimers in a process thought to be selfcatalyzed. MUC5AC (originally called MUC5) was first isolated from a tracheal cDNA library screened with antibodies to deglycosylated tracheobronchial mucin (69). MUC5AC is expressed primarily in the bronchial mucosa (59) as well as the stomach (70) and conjuctiva (71). The overall structure of MUC5AC is similar to that described for MUC2 except that its central tandem repeat region is interrupted by domains containing cytsteinerich sequences (72,73). The amino and carboxyl terminal regions of MUC5AC contain cysteinerich sequences similar to those in MUC2 and von Willebrand factor (72,74–77). MUC5B was also first isolated from a tracheobronchial cDNA library (78) and is expressed primarily in the respiratory epithelium (59,79), gallbladder (80), female reproductive tract (81,82), colon (78,83), and salivary glands (84–86). The central region of this mucin contains domains of 29amino acid tandem repeats, which, like those of MUC5AC, are interrupted by short cysteinerich sequences (87) and it, like MUC2 and MUC5AC, has aminoand carboxylterminal cysteinerich von Willebrand factorlike sequences (80,88–90). MUC5B is one of the major mucin gene products expressed in the gallbladder (80); more detail about its structural properties is given in Sec. IV.C. MUC6 was first identified in a gastric cDNA library, and this mucin is expressed primarily in this tissue with lesser amounts in the gallbladder, colon, and terminal ileum (91). MUC6 contains the longest tandem repeats described in any mucin, each containing 169 amino acids. MUC6 and MUC5AC expression have been localized to the mucous neck cells and the surface mucous cells of the stomach, respectively, suggesting that these mucins have distinct functional roles in the stomach (70). Interestingly, the carboxylterminal sequence of MUC6 lacks a von Willebrand factorlike D4 domain but contains a cystine knot motif at the extreme Cterminal end (92). The aminoterminal region of MUC6 has not been described. 3— MUC3 MUC3 was first cloned from a human small intestinal cDNA library using antibodies against deglycosylated small intestinal mucin (93). The gene for MUC3 is located at 7q22 (94), and this mucin is expressed primarily in the small intestine (93) as well as in the gallbladder (44). MUC3 contains 17amino acid tandem repeats, which are flanked by a nonrepetitive region and a cysteinerich sequence at the Cterminal end (95). Unlike the cysteinerich D domain like sequences, the cysteinerich sequence in MUC3 resembles an epidermal growth factor
Page 218
(EGF) motif, similar to those found in a number of proteins with ligandbinding properties. Whether this region in MUC3 has a ligandbinding function and the nature of the potential ligands remains unclear. While there are as yet no sequence data for the aminoterminal region of MUC3, this mucin appears to have one of the most complicated structural organizations of any known mucin. Restriction fragment analysis indicates that there are at least two regions in the molecule containing the 17 amino acid tandem repeats (95) and recent work from another laboratory has identified yet another tandem repeat region containing 59amino acid repeats (96). There has been speculation that MUC3 may be another membraneassociated mucin like MUC1. While no transmembrane domain has as yet been identified, two animal homologues, mouse and rat MUC3, both contain putative membranespanning regions (97,98). 4— MUC4 MUC4 was first isolated from a tracheobronchial cDNA library (99). It is expressed primarily in respiratory epithelium (99) and female reproductive tract (81,82), with lesser expression in the colon (99) and salivary glands (100). The gene for MUC4 is located at 3q29 (99,101), and this mucin appears to have yet another unique structural organization. It contains a central tandem repeat region comprised of 16amino acid repeats (99) flanked by an Nterminal region that is nearly devoid of cysteine (102). The Cterminal region is cysteinerich and contains two EGF repeats similar to the one in MUC3 (103). MUC4 displays a high degree of homology to the heterodimeric rat sialomucin complex, which consists of a membraneassociated mucin subunit (ASGP1) and a transmembrane subunit (ASGP2) (104,105). It has been suggested that MUC4 may bind the receptor tyrosine kinase ErbB2, thereby functioning as both an epithelial mucin and a cellsignaling molecule. 5— MUC7 MUC7 is the only known lowmolecularweight secreted mucin and is expressed primarily in the submandibular and sublingual glands, with lesser expression in the trachea (79,106,107). The gene for MUC7 is located at 4q1321 and encodes a protein containing five (or six) 23amino acid tandem repeats flanked by a N terminal region containing only two cysteines and a Cterminal region devoid of cysteine (108). MUC7 appears to be a small secretory protein that does not form intermolecular disulfide linkages and probably exists as monomers in salivary secretions. 6— MUC8 MUC8 was identified in a tracheal cDNA library screened with an antibody to deglycosylated tracheobronchial mucin. The sequences of several clones revealed that this mucin contained a tandem repeat domain comprising 41 base repeats. The fact that this number is not divisible by three, coupled with several nucleotide deletions, produces alternating tandem repeats with either 41 or 13 amino acids. MUC8 has been localized to 12q24 and its expression has only been detected in the respiratory epithelium and stomach (109,110). B— Identification of Gallbladder Mucin Genes Until the complexity of the mucin gene family was discovered by molecular cloning studies, it was widely thought that gallbladder mucin was a single biochemical entity, which, like other mucins isolated from gastrointestinal tissues, displayed heterogeneity primarily in its carbohydrate moieties. This was supported by histochemical studies using classic techniques that could distinguish between neutral mucins and both sulfated and sialic acidcontaining acid mucins. Epithelial cells in the body of the gallbladder were found to contain primarily sulfomucins, with staining observed on both the luminal surface and within cytoplasmic secretory
Page 219
granules. Mucous glands in the neck of the gallbladder stained for both sulfo and sialomucins (111–114). Similar heterogeneous staining was observed when gallbladder tissue sections were probed with a panel of lectins (111). In addition, Groen and colleagues have shown that mucins isolated from gallbladder bile vary widely in their reactivity toward both lectins and carbohydratereactive antimucin antibodies (115). Taken together, these studies suggested either that there was great diversity in the carbohydrate side chains of a single gallbladder mucin polypeptide or that distinct mucin polypeptides with different glycan structures were expressed in the gallbladder. As cDNA probes for the human mucin genes became available, it was clear that the latter possibility was true. A complex and sometimes contradictory expression pattern of gallbladder mucin genes emerged, owing in part to the fact that different studies employed different techniques and used normal and diseased gallbladder tissue interchangeably. Some of the first studies used Northern blot analysis to demonstrate that MUC2 (116), MUC3 (44), and MUC6 (91) mRNAs were expressed in the gallbladder. Subsequently, the expression of MUC1, MUC2, MUC5AC, and MUC5B apomucins were examined in gallbladder epithelium using a panel of specific monoclonal and polyclonal antibodies (117). All antisera reacted to sections of gallbladder epithelium, with the strongest reactivity seen with MUC1 and MUC5B. Subsequently, Campion and colleagues examined RNA isolated from freshly isolated gallbladder epithelial cells by Northern analysis and found that the cells expressed high levels of MUC3, MUC5AC, and MUC5B mRNAs with lower levels of MUC1 and MUC4 mRNA expression and barely detectable levels of MUC2 mRNA (118). Several years later, members of the same group used in situ hybridization and Northern analysis to demonstrate high levels of MUC5B, MUC3, and MUC6 mRNA and weaker expression of MUC1, MUC5AC, and MUC2 mRNA in the gallbladder (119). The discrepancies in these findings led us to try to identify the major human gallbladder mucins by expression cloning. A human gallbladder cDNA expression library was prepared and screened with an antibody directed against deglycosylated human gallbladder mucin (80). Analysis of positive clones showed that greater than 95% encoded MUC5B tandem repeats. Since the antibody used was raised against all mucins present in gallbladder mucosal scrapings, these results suggested that MUC5B was the major mucin polypeptide expressed in the gallbladder. This was somewhat surprising in view of the fact that when Northern blots of human gallbladder mRNA were hybridized with a series of mucinspecific probes, high levels of both MUC5B and MUC3 transcripts were observed with lower levels of MUC5AC and MUC6 transcripts and almost undetectable levels of MUC2 and MUC4 (Fig. 1). The failure to detect MUC3 clones in the library suggests either that MUC3 mRNA is not translated into protein at high levels in the gallbladder or, more likely, that the MUC3 polypeptide was not present in the highmolecularweight mucin fraction used to prepare the antiserum. Taken together, these studies show that at least four mucin genes are expressed in the gallbladder and that human gallbladder mucin is likely to contain a mixture of the corresponding mucin gene products. C— Structural Organization of the Major Gallbladder Mucin, MUC5B At the time MUC5B was identified as a major gallbladder mucin, nothing was known about the primary structure of this mucin except for the sequence of the tandem repeats (78). It was therefore of interest to determine the sequences of the amino and carboxylterminal regions flanking the tandem repeat domain, since these were likely to contain the proteasesensitive functional domains of human gallbladder mucin identified earlier. The complete sequence of the carboxylterminal region of MUC5B was deduced from the sequences of cDNA and genomic clones and was found to contain approximately 800 amino acids (80). This sequence was enriched with respect to cysteine residues, nearly all of which occurred in identical positions in the carboxyl terminal sequences of MUC2 (61), MUC5AC (72,76), and the D4, C1, and extreme Cterminal domain of human von Willebrand factor (120,121). Remarkably, the positions of exon/intron boundaries in the carboxylterminal
Page 220
Figure 1 Northern blot analysis of human gallbladder mRNA hybridized with a panel of mucin cDNA probes (A) or with a series of control cDNA probes (B). All of the mucin probes were specific for tandem repeat sequences and all blots were hybridized and washed under identical conditions and exposed to Xray film for 18 h. The positions of the 28S and 18S ribosomal RNAs are indicated. These data show that MUC5B and MUC3 transcripts are abundant in gallbladder RNA, with significantly lower levels of MUC5AC and MUC6 RNA and almost undetectable levels of MUC2 and MUC4 RNA. The polydisperse signals observed on these Northern blots are typical of the hybridization patterns usually seen with mucin mRNAs. The reason for this pattern is incompletely understood, but could result from rapid turnover of mucin mRNAs, partial degradation of large mRNAs, alternative splicing or heterogeneity of mRNA species synthesized. (B). Stripping and rehybridization of blots with control probes for 28 S ribosomal RNA (28S), actin or glyceraldehyde 3phosphate dehydrogenase (GAPDH) showed a discrete signal in each case, demonstrating the integrity of the RNA preparations used in this study.
regions of MUC5B were nearly identical to those in von Willebrand factor, as were the type of splice junctions (types 0, 1, 2), suggesting that these proteins were derived from a common ancestral gene. It is widely held that introns were inserted into existing genes late in evolution and that the presence of introns has played a major role in the evolution of mammalian protein families
Page 221
by promoting exon shuffling between genes (122). The shuffling of exons encoding different protein modules has been especially important in the evolution of cell surface and ligandbinding proteins (123). Exons encoding these shuffled modules are commonly bordered by type 1 introns at their 5 and 3 ends (124). As in other mucin genes, the tandem repeat domain of MUC5B is encoded in a single exon, which is separated from the first exon encoding the carboxylterminal domain with a type 1 intron (80,87). This suggests that MUC5B may have been assembled by shuffling of the tandemrepeat exon into a von Willebrand factorlike precursor molecule. In a broader sense, the mechanism for evolution of mucin genes with distinct tandemrepeat arrays may have involved the insertion and subsequent duplication of exons encoding tandem repeats into the primordial gene for a mucinlike cellsurface protein. The MUC5B tandemrepeat domain contains 19 separate subdomains of three distinct types (Fig. 2). Subdomains containing the MUC5B 29 amino acid repeats (RI RV) and other conserved subdomains (R01,R02) are interrupted by cysteinerich subdomains containing approximately 100 amino acids (Cys1 to Cys7). The organization of these sequences results in a pattern that has been referred to as a superrepeat (87) in that the subdomains are organized in manner in which they too are repeated four times. The aminoterminal region of MUC5B contains 1321 amino acids, and like the carboxylterminal region is enriched with respect to cysteine (88). This cysteinerich sequence is organized into D1, D2, and D3 domains, similar to those in von Willebrand factor. Interestingly, the degree of sequence identity between the amino terminal regions of MUC5B, MUC5AC, and MUC2 is much higher than that between the corresponding carboxylterminal regions, suggesting that structural features required for mucin assembly or function have been conserved. Each of the D domains contains the consensus sequence CGLCG, which resembles the sequence at the active site of protein disulfide isomerase (125) and is thought to be involved in the selfcatalyzed polymerization of von Willebrand factor monomers (126). In addition, analysis of the D domains showed the presence of a number of structural motifs found on extracellular and ligandbinding proteins and not previously identified in mammalian mucins. For example, the D3 domain contains consensus motifs for Ctype lectin domains, which have been shown to bind calcium, protein ligands, and carbohydrates (127,128) as well as selectin complementbinding repeats, which mediate the interactions between Pselectin and leukocytes (129). In addition, sequences similar to lowdensitylipoprotein (LDL)receptor class A repeats were identified. These repeats are present on the LDL receptor family proteins megalin and the 2 macroglobulin and have been shown to bind a variety of ligands including apolipoprotein J, lipoprotein lipase, lactoferrin, calcium, and polycationic drugs (161).
Figure 2 Schematic of the structural organization of MUC5B. MUC5B is organized into an Nterminal, a tandem repeat (TR) and a Cterminal region. The Nterminal region contains von Willebrand factorlike D1, D2 and D3 domains (gray boxes). The TR domain contains alternating cysteinerich subdomains (Cys1Cys7; white boxes) and TR domains (RIRV; black boxes). Other semiunique sequences present in the TR domain are not shown here. The Cterminal region contains a von Willebrand factorlike D4 domain (gray box) and a cystine knot at the extreme Cterminal end (hatched box).
Page 222
V— Relationship between Gallbladder Mucin Structure and Function As discussed above, early studies performed with bovine and human gallbladder mucins clearly showed that these molecules are comprised of two distinct domains, a heavily glycosylated, proteaseinsensitive domain and a poorly glycosylated proteasesensitive domain, the latter necessary for the binding of biliary lipids and for promoting cholesterol crystallization. Recent studies on the cloning of both bovine (130) and human gallbladder mucin (80) have identified the structural basis for these findings. As noted above, both of these mucins contains cysteinerich sequences that bear some resemblance to the ligandbinding domains on a variety of extracellular proteins. We have attempted to further define the sequences of the lipidbinding domains on gallbladder mucin with the goal of developing therapeutic modalities that could prevent the interactions between gallbladder mucin and biliary lipids. In these studies, bovine gallbladder mucin was used as a model system since, despite the heterogeneity observed in human gallbladder mucin, only one mucin gene product has been identified in the bovine gallbladder. This mucin has an overall structural organization similar to that of the major human gallbladder mucin, MUC5B, in that it contains alternating glycosylated tandem repeat and cysteinerich sequences (130). These cysteinerich sequences display little similarity to the cysteinerich sequences in MUC5B, but bear a striking resemblance to the scavenger receptor cysteinerich (SRCR) domains found on a number of extracellular ligand binding proteins, including the macrophage scavenger receptor (131), CD5 (132), and CD6 (133). Constructs containing cysteinerich repeats were expressed in Escherichia coli and the recombinant bovine gallbladder mucin (rGBM) purified. rGBM was shown to bind cholesterol and hydrophobic dyes in a manner similar to that of native mucin (134). It also accelerated the appearance of cholesterol crystals in a "nucleation" time assay. These studies show directly that the nonglycosylated regions of gallbladder mucin are responsible for its interactions with biliary lipids. Furthermore, they show that the formation of a gel is not required for the ability of mucin to bind cholesterol or promote cholesterol crystallization. These studies also demonstrate the feasibility of using recombinant proteins to investigate the function of individual mucin domains. As noted above, it has been suggested that unlike the secreted, gelforming mucins MUC5B, MUC5AC, and MUC2, MUC3 may be anchored to the cell membrane like MUC1 and MUC4 (95). This observation is supported by the identification of immunoreactive MUC3 in intrahepatic bile ducts (119) and suggests that in the gallbladder, MUC5B and MUC3 in intrahepatic bile ducts (119) and suggests that in the gallbladder, MUC5B and MUC3 may have distinct functions. It is not as yet possible to separate individual highmolecularweight mucins from one another due to the overall similarity in their physical properties. Therefore, in order to determine whether MUC5B, MUC3, and other mucins have unique functions in the gallbladder, recombinant polypeptides containing cysteinerich domains from these mucins are currently being examined in lipidbinding and nucleation experiments such as those described above. These studies may show that different mucin gene products perform different functions in the gallbladder and may provide important insights into the role of these proteins in the pathogenesis of gallstone disease. VI— Expression of Mucin Genes in Gallstone Disease Histological and histochemical examination of gallbladder tissue from patients with cholesterol gallstones consistently shows changes in both the morphological appearance of the gallbladder epithelium and in the staining pattern of gallbladder mucins (111,112). Intestinaltype metaplasia with increased numbers of goblet cells has been correlated with increased staining for nonsulfated acid mucin and neutral mucin. Gastric metaplasia has also been noted in some specimens with the appearance of antraltype glands and gastrictype epithelium and alterations
Page 223
in the mucin staining pattern. In animal models of gallstone disease, similar changes in the histochemical properties of hypersecreted mucin have been identified. Gallbladders removed from cholesterolfed ground squirrels demonstrated a reduction in the percentage of sialic acidcontaining mucins when compared with control gallbladders (22). While it is not possible to correlate the histochemical staining pattern with the presence of a particular mucin gene product, studies in both humans and animals suggest that alterations in mucin gene expression, perhaps associated with inflammatory or metaplastic changes, occur in cholesterol gallstone disease. We have recently attempted to investigate this question by comparing the pattern of mucin gene expression in control gallbladders and in gallbladder specimens obtained at cholecystectomy from patients with cholesterol gallstone disease. RNA transcripts for MUC2, MUC3, MUC4, MUC5AC, MUC5B, and MUC6 were measured in slotblot experiments and by reverse transcriplase polymerase chain reaction (RTPCR). When normalized to actin mRNA expression, levels of MUC3 and MUC5B were significantly increased in gallstone patients, whereas levels of the other mucin mRNAs were unchanged (135). Similar increases in the level of MUC5B and MUC3 transcripts in gallstone patients were also noted by Kano and collaborators who showed in addition, that MUC2 and MUC6 mRNA levels were also increased (136). These studies suggest that changes in the relative expression of mucin genes does occur in patients with cholesterol gallstones, although it is not possible to identify any one gene product as the "lithogenic" mucin. VII— Regulation of Mucin Secretion As noted above, mucin hypersecretion precedes gallstone formation in a number of animal models and increased mucin levels have been found in gallbladder tissue and bile from patients with cholesterol gallstone disease. The mediators and intracellular signaling pathways leading to increased mucin secretion remain unclear, but several potential mechanisms are discussed below. In the prairie dog, the profound increase in mucin secretion resulting from cholesterol feeding can be prevented by ligation of the cystic duct (17), suggesting that the stimulus for mucin hypersecretion is carried to the gallbladder in hepatic bile. Whether cholesterol supersaturation of bile is the stimulus or whether other substances are released into the bile by the liver in response to cholesterol feeding is not known, but similar studies in the ground squirrel suggest that this response is rapid and occurs within hours of cholesterol feeding (21,22). A— Prostaglandins It is possible that cholesterolsupersaturated bile induces a local inflammatory response in the gallbladder epithelium. Proinflammatory substances such as prostaglandins have been shown to mediate mucin secretion in both the gallbladder and other mucinsecreting tissues such as the stomach and trachea (137,138). Administration of aspirin to cholesterolfed prairie dogs (139) and ground squirrels (22) prevented mucin hypersecretion and stone formation, suggesting that increased prostaglandin synthesis by the gallbladder might mediate mucin secretion. In the prairie dog, increases in gallbladder prostaglandin levels in response to cholesterol feeding were found to precede the hypersecretion of gallbladder mucin (140), and exogenous arachidonic acid has been shown to increase mucin secretion in prairie dog gallbladder explants (141). In another study, intraperitoneal treatment of guinea pigs with prostaglandin E2 (PGE2) also led to a significant increase in mucin secretion despite the fact that these animals were fed a control diet and did not develop cholesterolsupersaturated bile (142). Finally, using cultured dog gallbladder epithelial cells, Kuver and colleagues demonstrated that prostaglandins and other substances that increased the level of intracellular cAMP resulted in increased secretion of mucin (143). Despite this considerable body of evidence, the effect of prostaglandins
Page 224
on mucin secretion remains controversial, since additional studies have shown that prostaglandins had no effect on mucin secretion in both prairie dogs and hamsters (144,145). Less is known about the effect of prostaglandins on human gallbladder mucin secretion. We have demonstrated increased levels of mucin secretion in a human gallbladder epithelial cell line treated with PGE2 (146). In addition, treatment of human gallbladder explants with aspirin was shown to result in an inhibition of mucin synthesis (147). The latter result led to a series of clinical studies in which nonsteroidal antiinflammatory drugs (NSAIDs) were administered to patients prior to cholecystectomy (148), to highrisk patients on rapidweightreducing diets (149), and to patients after gallstone dissolution therapy (150). Again, the results of these studies have not established a conclusive link between NSAID use and inhibition of mucin secretion or gallstone formation (151,152), although there is some evidence for a reduction in gallbladder mucin secretion in patients with a history of chronic NSAID use (25). B— Other Mediators Other biliary constituents that have been suggested to contribute to the mucin secretory response are lysophosphatidylcholine (LPC), the proinflammatory cytokine tumor necrosis factoralpha (TNF ), and increased concentrations of free bile acids. In the cholesterolfed prairie dog, LPC levels in the gallbladder increased prior to the hypersecretion of mucin and stone formation and LPC was shown to stimulate mucin release in guinea pig gallbladder explants (140). We and others have shown that treatment of gallbladder epithelial cells with TNF leads to an increase in mucin secretion (153,154). This cytokine has been identified in both gallbladder and hepatic bile (155) although its origin and mechanism of secretion remain unknown. Similar effects of TNF on mucin secretion have been observed in tracheal epithelial cells (156,157). A series of studies performed with cultured dog gallbladder epithelial cells has shown that bile salts stimulate mucin secretion (158,159) in a manner independent of the detergent effects of the bile salts on the cell membrane. A more recent study with primary cultures of human gallbladder epithelial cells has also shown a bilesalt stimulated increase in mucin secretion, which appears to act by increasing the level of intracellular calcium through activation of calcium/calmodulin kinase II (160). The latter study also demonstrated the involvement of the protein kinase C (PKC) pathway in mediating mucin secretion in response to substances such as phorbol esters. Interestingly, these authors found no significant stimulation in mucin secretion in response to activation of protein kinase A (PKA) by increased intracellular levels of cAMP which would be generated by mediators such as prostaglandins, vasoactive intestinal peptide (VIP), and secretin. It has also been suggested that increased concentrations of cholesterol itself may alter the fluidity of gallbladder epithelial cell membranes leading to a secretory response, perhaps through the generation of NO, a known mucin secretogogue in other tissues (157,162). This effect would be analogous to the generation of NO in vascular endothelial cells in patients with hypercholesterolemia. It is of interest in this regard that iNOS has recently been identified in human gallbladder epithelial cells (163). VIII— Regulation of Mucin Gene Expression in the Gallbladder There are several levels at which mediators such as those described above could lead to increased secretion of gallbladder mucin. At one level, these mediators could act as a stimulus for the release of stored mucin secretory granules. They could also initiate a signaltransduction cascade leading to increased transcription of mucin genes or could act at the posttranscriptional level by influencing factors such as mRNA stability. At present, nothing is known about the
Page 225
effects of mucin secretagogues on rates of transcription and steadystate levels of individual gallbladder mucin mRNAs. The availability of the MUC5B gene promoter (88) will allow us to begin to elucidate the pathways by which different mediators activate specific transcription factors and to identify the sequence elements to which these factors bind. It is clear that a detailed understanding of how each of the gallbladder mucin genes is regulated is critical to an understanding of the role of these important proteins in the healthy and diseased gallbladder. In future, this information could lead to the development of pharmaceuticals that could selectively modulate transcription of mucin genes and prevent the formation of cholesterol gallstones in susceptible individuals. Acknowledgment Supported in part by grant DK44619 from the National Institutes of Health. References 1. Strous G, Dekker J. Mucintype glycoproteins. Crit Rev Biochem Mol Biol 1992; 27:57–92. 2. Gum JR. Mucin genes and the proteins they encode. Am J Respir Cell Mol Biol 1992; 7:557–564. 3. Gendler SJ, Spicer AP. Epithelial mucin genes. Annu Rev Physiol 1995; 57:607–634. 4. Amerongen AN, Bolscher JGM, Bloemena E, Veerman ECI. Sulfomucins in the human body. Biol Chem 1998; 379:1–18. 5. Sheehan JKK, Oates K, Carlstedt I. Electron microscopy of cervical, gastric and bronchial mucus glycoproteins. Biochem J 1986; 239:147–153. 6. Allen A. Mucus glycoproteins of the normal gastrointestinal tract. Eur J Gastroenterol Hepatol 1993; 5:193–199. 7. Snary D, Allen A, Pain RH. Structural studies on gastric mucus glycoproteins; lowering of molecular weights after eduction with 2mercaptoethanol. Biochem Biophys Res Comm 1970; 40:844–850. 8. Sellers LA, Allen A, Morris ER, RossMurphy SB. Mucus glycoprotein gels: role of glycoprotein polymeric structure and carbohydrate side chains in gel formation. Carbo Res 1988; 178:93–110. 9. Devine PL, McKenzie IFC. Mucins: Structure, function, and associations with malignancy. Bioessays 1992; 14:619–625. 10. Carey MC. Pathogenesis of gallstones. Am J Surg 1993; 165:410–419. 11. Carey MC, Lamont JT. Cholesterol gallstone formation: 1. Physical chemistry of bile and biliary lipid secretion. Prog Liver Dis 1992; 10:139–163. 12. Womack NA, Zeppa R, Irvin GL. The anatomy of gallstones. Ann Surg 1963; 157:670–686. 13. Maki T, Matsushiro T, Suzuki N, Nakamura N. Role of sulfated glycoprotein in gallstone formation. Surg Gynecol Obstet 1971; 132:846–854. 14. Lee SP, Lim TH, Scott AJ. Carbohydrate moities of glycoproteins in human ehpatic and gallbladder bile, gallbladder mucosa and gallstones. Clin Sci 1979; 56:533–538. 15. Smith BF, LaMont JT. Identification of gallbladder mucinbilirubin complex in human cholesterol gallstone matrix: effects of reducing agents on in vitro dissolution of matrix and intact gallstones. J Clin Invest 1985; 76:439–445. 16. Lechene de la Porte P, Domingo N, vanWijland M, Groen AK, Ostrow JD, Lafont H. Distinct immunolocalization of mucin and other biliary proteins in human cholesterol gallstones. J Hepatol 1996; 25:339–348.
Page 226
17. Lee SP, LaMont JT, Carey MC. Role of gallbladder mucus hypersecretion in the evolution of cholesterol gallstones. J Clin Invest 1981; 67:1712–1723. 18. Lee SP. Hypersecretion of mucus glycoprotein by the gallbladder epithelium in experimental cholelithiasis. J Pathol 1981; 134:199–207. 19. Lee SP, Scott AJ. The evolution of morphologic changes in the gallbladder before stone formation in mice fed a cholesterolcholic acid diet. Am J Pathol 1982; 108:1–8. 20. Malet PF, Deng SQ, Soloway RD. Gallbladder mucin and cholesterol and pigment gallstone formation in hamsters. Scand J Gastroenterol 1989; 24:1055–1060. 21. Pemsingh RS, MacPherson BR, Scott GW. Mucus hypersecretion in the gallbladder epithelium of ground squirrels fed a lithogenic diet. Hepatology 1987; 7:1267–1271. 22. MacPherson BR, Pemsingh RS. Ground squirrel model for cholelithiasis: role of epithelial glycoproteins. Microsc Res Tech 1997; 39:39–55. 23. Bouchier IAD, Cooperband SR, Kodsi BM. Mucous substances and viscosity of normal and pathological human bile. Gastroenterology 1965; 49:343–353. 24. Mingrone G, Greco AV, Finotti E, Passi S. Free fatty acids: a stimulus for mucin hypersecretion in cholesterol gallstone biles. Biochim Biophys Acta 1988; 958:52–59. 25. Sterling RK, Shiffman ML, Sugerman HJ, Moore EW. Effect of NSAIDs on gallbladder bile composition. Dig Dis Sci 1995; 40:2220–2226. 26. Swobodnik W, Wenk H, Janowitz P, Hagert N, Kratzer W, Berghold J, Zhang Y, Bittner R, Schusdziarra V, Ott R, Kuhn K, Classen M. Total biliary protein, mucus glycoproteins, cyclic AMP, and apolipoproteins in the gallbladder bile of patients with cholesterol stones and stonefree controls. Scand J Gastroenterol 1991; 26:771–778. 27. Shiffman ML, Sugerman HJ, Kellum JM, Moore EW. Changes in gallbladder bile composition following gallstone formation and weight reduction. Gastroenterology 1992; 103:214–221. 28. Shiffman ML, Shamburek RD, Schwartz CC, Sugerman HJ, Kellum JM, Moore EW. Gallbladder mucin, arachidonic acid, and bile lipids in patients who develop gallstones during weight reduction. Gastroenterology 1993; 105:1200–1208. 29. Hulten O. Formation of gallstones. II. Acta Chir Scand 1968; 134:557–561. 30. Hulten O. Formation of gallstones I. Acta Chir Scand 1968; 134:125–130. 31. Lee SP. Pathogenesis of biliary sludge. Hepatology 1990; 12:200S–203S; discussion, 203S–205S. 32. Carey MC, Cahalane MJ. Whither biliary sludge? Gastroenterology 1988; 95:508–523. 33. Gilloteaux J, Karkare S, Don AQ, Sexton RC. Cholelithiasis induced in the Syrian hamster: evidence for an intramucinous nucleating process and down regulation of cholesterol 7 alphahydroxylase (CYP7) gene by medroxyprogesterone. Microsc Res Tech 1997; 39:56–70. 34. Henisch HK. Crystal Growth in Gels. University Park, PA: Pennsylvania State University, 1970. 35. Levy PF, Smith BF, LaMont JT. Human gallbladder mucin accelerates nucleation of cholesterol in artificial bile. Gastroenterology 1984; 87:270–275. 36. Smith BF. Human gallbladder mucin binds biliary lipids and promotes cholesterol crystal nucleation in model bile. J Lipid Res 1987; 28:1088–1097. 37. Gallinger SA, Taylor RD, Harvey PR, Petrunka CN, Strasberg SM. Effect of mucous glycoprotein on nucleation time of human bile. Gastroenterology 1985; 89:648–658. 38. Smith BF, LaMont JT. Bovine gallbladder mucin binds bilirubin in vitro. Gastroenterology 1983; 85:707–712. 39. Smith BF, LaMont JT. Hydrophobic binding properties of bovine gallbladder mucin. J Biol Chem 1984; 259:12170–12177. 40. Afdhal NH, Niu N, Gantz D, Small DM, Smith BF. Bovine gallbladder mucin accelerates cholesterol monohydrate crystal growth in model bile. Gastroenterology 1993; 104:1515–1523. 41. Seregni E, Botti C, Massaron S, Lombardo C, Capobianco A, Bogni A, Bombardieri E. Structure, function and gene expression of epithelial mucins. Tumori 1997; 83:625–632.
Page 227
42. Van Klinken BJW, Dekker J, Buller HA, Einerhand AWC. Mucin genestructure and expression: protection: protection vs. adhesion. Am J Physiol 1995; 269:G613–G627. 43. Gendler SJ, Spicer AP, Lalani EN, Duhig T, Peat N, Burchell J, Pemberton L, Boshell M, TaylorPapadimitriou J. Structure and biology of a carcinoma associated mucin, MUC1. Am Rev Respir Dis 1991; 144:S42–S47. 44. Ho SB, Niehans GA, Lyftogt C, Yan PS, Cherwitz DL, Gum ET, Dahiya R, Kim YS. Heterogeneity of mucin gene expression in normal and neoplastic tissues. Cancer Res 1993; 53:641–651. 45. Gendler SJ, Burchell JM, Duhig T, Lamport D, White R, Parker M, TaylorPapadimitriou J. Cloning of partial cDNA encoding differentiation and tumor associated mucin glycoproteins expressed by human mammary epithelium. Proc Natl Acad Sci USA 1987; 84:6060–6064. 46. Lan MS, Bhatra SK, Qi W, Metzgar RS, Hollingsworth MA. Cloning and sequencing of a human pancreatic tumor mucin cDNA. J Biol Chem 1990; 265:15294–15299. 47. Gendler SJ, Lancaster CA, TaylorPapadimitriou J, Duhig T, Peat N, Burchell J, Pemberton L, Lalani E, Wilson D. Molecular cloning and expression of the human tumorassociated polymorphic epithelial mucin, PEM. J Biol Chem 1990; 265:15286–15293. 48. Pemberton L, TaylorPapadimitriou J, Gendler SJ. Antibodies to the cytoplasmic domain of the MUC1 mucin show conservation throughout mammals. Biochem Biophys Res Commun 1992; 185:167–175. 49. Ligtenberg MJL, Vos HL, Gennissen AMC, Hilkens J. Episialin, a carcinomaassociated mucin, is generated by a polymorphic gene encoding splice variants with alternative aminotermini. J Biol Chem 1990; 5573–5578. 50. Ligtenberg MJ, Gennissen AM, Vos HL, Hilkens J. A single nucleotide polymorphism in an exon dictates allele dependent differential splicing of episialin mRNA. Nucleic Acids Res 1991; 19:297–301. 51. Boshell M, Lalani EN, Pemberton L, Burchell J, Gendler S, TaylorPapadimitriou J. The product of the human MUC1 gene when secreted by mouse cells transfected with the fulllength cDNA lacks the cytoplasmic tail. Biochem Biophys Res Commun 1992; 185:1–8. 52. Litvinov SV, Hilkens J. The epithelial sialomucin, episialin, is sialylated during recycling. J Biol Chem 1993; 268:21364–21371. 53. Ligtenberg MJL, Buijs F, Vos HL, Hilkens J. Suppression of cellular aggregation by high levels of episialin. Cancer Res 1992; 52:2318–2324. 54. van de Wielvan Kemenade E, Ligtenberg MJ, de Boer AJ, Buijs F, Vos HL, Melief CJ, Hilkens J, Figdor CG. Episialin (MUC1) inhibits cytotoxic lymphocyte target cell interaction. J Immunol 1993; 151:767–776. 55. Wesseling J, Van der Valk SW, Vos HL, Sonnenberg A, Hilkens J. Episialin (MUC1) overexpression inhibits integrinmediated cell adhesion to extracellular matrix components. J Cell Biol 1995; 129:255–265. 56. Pigny P, GuyonnetDuperat V, Hill AS, Pratt WS, GalliegueZoutitina S, h'Hooge MC, Laine A, VanSeuningen I, Degand P, Gum JR, Kim YS, Swallow DM, Aubert JP, Porchet N. Human mucin genes assigned to 11p15.5: identification and organization of a cluster of genes. Genomics 1996; 38:340–352. 57. Desseyn JL, Buisine MP, Porchet N, Aubert JP, Degand P, Laine A. Evolutionary history of the 11p15 human mucin gene family. J Mol Evol 1998; 46:102–106. 58. Gum JR, Byrd JC, Hicks JW, Toribara NW, Lamport DTA, Kim YS. Molecular cloning of human intestinal mucin cDNAs. J Biol Chem 1989; 264:6480–6487. 59. Audie JP, Janin A, Porchet N, Copin C, Gosselin B, Aubert JP. Expression of human mucin genes in the respiratory, digestive and reproductive tracts ascertained by in situ hybridization. J Histochem Cytochem 1993; 41:1479–1485. 60. Chang SK, Dohrman AF, Basbaum CB, et al. Localization of mucin (MUC2 and MUC3) messenger RNA and peptide expression in human normal intestine and colon cancer. Gastroenterology 1994; 107:28–36.
Page 228
61. Gum JR Jr, Hicks JW, Toribara NW, Rothe EM, Lagace RE, Kim YS. The human MUC2 intestinal mucin has cysteinerich subdomains located both upstream and downstream of its central repetitive region. J Biol Chem 1992; 267:21375–21383. 62. Gum JR Jr, Hicks JW, Toribara NW, Siddiki B, Kim YS. Molecular cloning of human intestinal mucin (MUC2) cDNA: identification of the amino terminus and overall sequence similarity to preprovon Willebrand factor. J Biol Chem 1994; 269:2440–2446. 63. Sadler JE. Biochemistry and genetics of von Willebrand factor. Annu Rev Biochem 1998; 67:395–424. 64. Sadler JE, SheltonInloes BB, Sorace JM, Titani K. Cloning of cDNA and genomic DNA for human von Willebrand factor. Cold Spring Harb Symp Quant Biol 1986; 51:515–523. 65. Sun PD, Davis DR. The cystineknot growth factor superfamily. Annu Rev Biophys Biomol Struct 1995; 24:269–291. 66. Meitinger T, Meindl A, Bork P, Rost B, Sander C, Haasemann M, Murken J. Molecular modeling of the Norrie disease protein predicts a cystine knot growth factor tertiary structure. Nature Genet 1993; 5:376–380. 67. Voorberg J, Fontijn R, Calafat J, Janssen H, van Mourik JA, Panekoek H. Assembly and routing of von Willebrand factor variants: the requirements for disulfide linked dimerization reside within the carboxyterminal 151 amino acids. J Cell Biol 1991; 113:195–205. 68. PerezVilar J, Hill RL. The carboxylterminal 90 residues of porcine submaxillary mucin are sufficient for forming disulfidebonded dimers. J Biol Chem 1998; 273:6982–6988. 69. Aubert JP, Porchet N, Crepin M, DuterqueCoquilland M, Vergnes G, Mazzuca M, Debuire B, Petiprez D, Degand D. Evidence for different human tracheobronchial mucin peptides deduced from nucleotide cDNA sequences. Am J Respir Cell Mol Biol 1991; 5:178–185. 70. Ho SB, Roberton AM, Shekels LL, Lyftogt CT, Niehans GA, Toribara NW. Expression cloning of gastric mucin complementary DNA and localization of mucin gene expression. Gastroenterology 1995; 109:735–747. 71. Inatomi T, SpurrMichaud S, Tisdale AS, Zhan Q, Feldman ST, Gipson IK. Expression of secretory mucin genes by human conjunctival epithelia. Invest Ophthalmol Vis Sci 1996; 37:1684–1692. 72. Meerzaman D, Charles P, Daskal E, Polymeropoulos MH, Martin BM, Rose MC. Cloning and analysis of cDNA encoding a major airway glycoprotein, human tracheobronchial mucin (MUC5). J Biol Chem 1994; 269:12932–12939. 73. Guyonnet Duperat V, Audie JP, Debailleul V, Laine A, Buisine MP, GaliegueZouitina S, Pigny P, Degand P, Aubert JP, Porchet N. Characterization of the human mucin gene MUC5AC: a consensus cysteinerich domain for 11p15 mucin genes? Biochem J 1995; 305:211–219. 74. van de Bovenkamp JH, Hau CM, Strous GJ, Buller HA, Dekker J, Einerhand AW. Molecular cloning of human gastric mucin MUC5AC reveals conserved cysteinerich Ddomains and a putative leucine zipper motif. Biochem Biophys Res Commun 1998; 245:853–859. 75. Li D, Gallup M, Fan N, Szymkowski DE, Basbaum CB. Cloning of the aminoterminal and 5 flanking region of the human MUC5AC mucin gene and transcriptional upregulation by bacterial exoproducts. J Biol Chem 1998; 273:6812–6820. 76. Lesuffleur T, Roche F, Hill AS, Lacasa M, Fox M, Swallow DM, Zweibaum A, Real FX. Characterization of a mucin cDNA clone isolated from HT29 mucus secreting cells: the 3 end of MUC5AC? J Biol Chem 1995; 270:13665–33673. 77. Buisine MP, Desseyn JL, Porchet N, Degand P, Laine A, Aubert JP. Genomic organization of the 3 region of the human MUC5AC mucin gene: additional evidence for a common ancestral gene for the 11p15.5 mucin gene family. Biochem J 1998; 332:729–738.
Page 229
78. Dufosse J, Porchet N, Audie JP, GuyonnetDuperat V, Laine A, Van Seuningen I, Marrakchi S, Degand P, Aubert JP. Degenetate 87 base pair tandem repeats create hydrophilic/hydrophobic alternating domains in human mucin peptides mapped to 11p15. Biochem J 1993; 293:329–337. 79. Sharma P, Dudus L, Nielsen PA, Clausen H, Yankasas JR, Hollingsworth MA, Englehardt JF. MUC5B and MUC7 are differentially expressed in mucous and serous cells of submucosal glands in human bronchial airways. Am J Respir Cell Mol Biol 1998; 19: 30–37. 80. Keates AC, Nunes DP, Afdhal NH, Troxler RF, Offner GD. Molecular cloning of a major human gall bladder mucin: complete Cterminal sequence and genomic organization of MUC5B. Biochem J 1997; 324:295–303. 81. Gipson IK, SpurrMichaud S, Moccia R, Zhan Q, Toribara N, Ho SB, Gargiulo AR, Hill JA. MUC4 and MUC5B transcripts are the prevalent mucin messenger ribonucleic acids of the human endocervix. Biol Reprod 1999; 60:58–64. 82. Gipson IK, Ho SB, SpurrMichaud SJ, Tisdale AS, Zhan Q, Torlakovic E, Pudney J, Anderson DJ, Toribara NW, Hill JA. Mucin genes expressed by human female reproductive tract epithelia. Biol Reprod 1997; 56:999–1011. 83. van Klinken BJ, Dekker J, van Gool SA, van Marle J, Buller HA, Einerhand AW. MUC5B is the prominent mucin in human gallbladder and is also expressed in a subset of colonic goblet cells. Am J Physiol 1998; 274:G871–G878. 84. Troxler RF, Iontcheva I, Oppenheim FG, Nunes DP, Offner GD. Molecular characterization of a major high molecular weight mucin from human sublingual gland. Glycobiology 1997; 7:965–973. 85. Troxler RF, Offner GD, Zhang F, Iontcheva I, Oppenheim FG. Molecular cloning of a novel high molecular weight mucin (MG1) from human sublingual gland. Biochem Biophys Res Commun 1995; 217:1112–1119. 86. Nielsen PA, Bennett EP, Wandall HH, Therkildsen MH, Hannibal J, Clausen H. Identification of a major human high molecular weight salivary mucin (MG1) as tracheobronchial mucin MUC5B. Glycobiology 1997; 7:413–419. 87. Desseyn JL, GuyonnetDuperat V, Porchet N, Aubert JP, Laine A. Human mucin gene MUC5B, the 10.7kb large central exon encodes various alternate subdomains resulting in a superrepeat: structural evidence for a 11p15.5 gene family. J Biol Chem 1997; 272:3168–3178. 88. Offner GD, Nunes DP, Keates AC, Afdhal NH, Troxler RF. The aminoterminal sequence of MUC5B contains conserved multifunctional D domains: implications for tissuespecific mucin functions Biochem Biophys Res Commun 1998; 251:350–355. 89. Desseyn JL, Aubert JP, Van Seuningen I, Porchet N, Laine A. Genomic organization of the 3 region of the human mucin gene MUC5B. J Biol Chem 1997; 272:16873–16883. 90. Desseyn JL, Buisine MP, Porchet N, Aubert JP, Laine A. Genomic organization of the human mucin gene MUC5B. cDNA and genomic sequences upstream of the large central exon. J Biol Chem 1998; 273:30157–30164. 91. Toribara NW, Roberton AM, Ho SB, Kuo WL, Gum E, Hicks JW, Gum JR, Byrd JC, Siddiki B, Kim YS. Human gastric mucin: identification of a unique species by expression cloning. J Biol Chem 1993; 268:5879–5885. 92. Toribara NW, Ho SB, Gum E, Gum JR Jr, Lau P, Kim YS. The carboxylterminal sequence of the human secretory mucin, MUC6: analysis of the primary amino acid sequence. J Biol Chem 1997; 272:16398–16403. 93. Gum JR, Hicks JW, Swallow DM, Lagace RL, Byrd JC, Lamport DTA, Siddiki B, Kim YS. Molecular cloning of cDNAs derived from a novel human intestinal mucin gene. Biochem Biophys Res Commun 1990; 171:407–415. 94. Fox MF, Lahbib F, Pratt W, Attwood J, Gum J, Kim Y, Swallow DM. Regional localization of the intestinal mucin gene MUC3 to chromosome 7q22. Ann Hum Genet 1992; 56:281–287.
Page 230
95. Gum JR Jr, Ho JJL, Pratt WS, Hicks JW, Hill AS, Vinall LE, Roberton AM, Swallow DM, Kim YS. MUC3 human intestinal mucin: analysis of gene structure, the carboxyl terminus, and a novel upstream repetitive region. J Biol Chem 1997; 272:26678–26686. 96. Van Klinken BJ, Van Dijken TC, Oussoren E, Buller HA, Dekker J, Einerhand AW. Molecular cloning of human MUC3 cDNA reveals a novel 59 amino acid tandem repeat region. Biochem Biophys Res Commun 1997; 238:143–148. 97. Shekels LL, Hunninghake DA, Tisdale AS, Gipson IK, Kieliszewski M, Kozak CA, Ho SB. Cloning and characterization of mouse intestinal MUC3 mucin: 3 sequence contains epidermalgrowthfactorlike domains. Biochem J 1998; 330:1301–1308. 98. Khatri IA, Forstner GG, Forstner JF. The carboxylterminal sequence of rat intestinal mucin RMuc3 contains a putative transmembrane region and two EGFlike motifs. Biochim Biophys Acta 1997; 1326:7–11. 99. Porchet N, Nguyen VC, Dufosse J, Audie JP, GuyonnetDuperat V, Gross MS, Denis C, Degand P, Bernheim A, Aubert JP. Molecular cloning and chromosomal localization of a novel human tracheobronchial mucin cDNA containing tandemly repeated sequences of 48 base pairs. Biochem Biophys Res Commun 1991; 175:414–422. 100. Liu B, Offner GD, Nunes DP, Oppenheim FG, Troxler RF. MUC4 is a major component of salivary mucin MG1 secreted by the human submandibular gland. Biochem Biophys Res Commun 1998; 250:757–761. 101. Gross MS, GuyonnetDuperat V, Porchet N, Bernheim A, Aubert JP, Nguyen VC. Mucin 4 (MUC4) gene: regional assignment (3q29) and RFLP analysis. Ann Genet 1992; 35:21–26. 102. Nollet S, Moniaux N, Maury J, Petiprez D, Degand P, Laine A, Porchet N, Aubert JP. Human mucin gene MUC4: organization of its 5'region and polymorphism of its central tandem repeat array. Biochem J 1998; 322:739–748. 103. Moniaux N, Nollet S, Porchet N, Degand P, Laine A, Aubert JP. Complete sequence of the human mucin MUC4: a putative cell membraneassociated mucin. Biochem J 1999; 338:325–333. 104. Wu K, Fregien N, Carraway KL. Molecular cloning and sequencing of the mucin subunit of a heterodimeric, bifunctional cell surface glycoprotein complex of ascites rat mammary adenocarcinoma cells. J Biol Chem 1994; 269:11950–11955. 105. Sherblom AP, Carraway KL. A complex of two cell surface glycoproteins from ascites mammary adenocarcinoma cells. J Biol Chem 1980; 255:12051–12059. 106. Bobek LA, Tsai H, Biesbrock AR, Levine MJ. Molecular cloning, sequence, and specificity of expression of the gene encoding the low molecular weight human salivary mucin (MUC7). J Biol Chem 1993; 268:20563–20569. 107. Bobek LA, Liu J, Sait SN, Shows TB, Bobek YA, Levine MJ. Structure and chromosomal localization of the human salivary mucin gene, MUC7. Genomics 1996; 31:277–282. 108. Biesbrock AR, Bobek LA, Levine MJ. MUC7 gene expression and genetic polymorphism. Glycoconj J 1997; 14:415–422. 109. Shankar V, Pichan P, Eddy RL Jr, Tonk V, Nowak N, Sait SN, Shows TB, Schultz RE, Gotway G, Elkins RC, Gilmore MS, Sachdev GP. Chromosomal localizatioin of a human mucin gene (MUC8) and cloning of the cDNA corresponding to the carboxy terminus. Am J Respir Cell Mol Biol 1997; 16:232–241. 110. Shankar V, Gilmore MS, Elkins RC, Sachdev GP. A novel human airway mucin cDNA encodes a protein with unique tandem repeat organization. Biochem J 1994; 300:295–298. 111. Madrid JF, Hernandez F, Ballesta J. Characterization of glycoproteins in the epithelial cells of human and other mammalian gallbladder: a review. Microsc Res Tech 1997; 38: 616–630. 112. Laitio M. Morphology and histochemistry of nontumorous gallbladder epithelium. Pathol Res Pract 1980; 167:335–345.
Page 231
113. Frierson HF. The gross anatomy and histology of the gallbladder, extrahepatic bile ducts, valerian system and minor papilla. Am J Surg Pathol 1989; 13:146– 162. 114. Esterly JR, Spicer SS. Mucin histochemistry of human gallbladder: changes in adenocarcinoma, cystic fibrosis and cholecystitis. J Natl Cancer Inst 1968; 40:1– 11. 115. Klinkspoor JH, van Wijland MJ, Koeleman CA, van Dijk W, Tytgat GN, Groen AK. Heterogeneity of human biliary mucin: functional implications. Clin Sci 1994; 86:75–82. 116. Jany BH, Gallup MW, Yan PS, Gum JR, Kim YS, Basbaum CR. Human bronchus and intestine express the same mucin gene. J Clin Invest 1991; 87:77–82. 117. Carrato C, Balague C, de Bolos C, Gonzalez E, Gambus G, Planas J, Perini JM, Andreu D, Real FX. Differential apomucin expression in normal and neoplastic human gastrointestinal tissues. Gastroenterology 1994; 107:160–172. 118. Campion JP, Porchet N, Aubert JP, L'Helgoualc'h A, Clement B. UWpreservation of cultured human gallbladder epithelial cells: phenotypic alterations and differential mucin gene expression in the presence of bile. Hepatology 1995; 21:223–231. 119. Vandenhaute B, Buisine MP, Debailleul V, Clement B, Moniaux N, Dieu MC, Degand P, Porchet N, Aubert JP. Mucin gene expression in biliary epithelial cells. J Hepatol 1997; 27:1057–1066. 120. SheltonInloes BB, Titani K, Sadler JE. cDNA sequences for human von Willebrand factor reveal five types of repeated domains and five possible protein sequence polymorphisms. Biochemistry 1986; 25:3164–3171. 121. Titani K, Kumar S, Takio K, Ericsson LH, Wade RD, Ashida K, Walsh KA, Chopek MW, Sadler JE, Fukikawa K. Amino acid sequence of human von Willebrand factor. Biochemistry 1986; 25:3171–3184. 122. Rogers JH. The role of introns in evolution. FEBS Lett 1990; 268:339–343. 123. Baron M, Norman DG, Campbell ID. Protein modules. Trends Biochem Sci 1991; 16:13–17. 124. Patthy L. Introndependent evolution: preferred types of exons and introns. FEBS Lett 1987; 214:1–7. 125. Mayadas TN, Wagner DD. Vicinal cysteines in the prosequence play a role in von Willebrand factor multimer assembly. Proc Natl Acad Sci USA 1992; 89:3531–3535. 126. Dong Z, Thoma RS, Crimmins DL, McCourt DW, Tuley EA, Sadler JE. Disulfide bonds required to assemble functional von Willebrand factor multimers. J Biol Chem 1994; 269:6753–6758. 127. Aspberg A, Miura R, Bourdoulous S, Shimonaka M, Heinegard D, Schachner M, Ruoslahti E, Yamaguchi Y. The Ctype lectin domains of lecticans, a family of aggregating chondroitin sulfate proteoglycans, bind tenascinR by proteinprotein interactions independent of carbohydrate moiety. Proc Natl Acad Sci USA 1997; 94:10116–10121. 128. Hosoi T, Imai Y, Irimura T. Coordinated binding of sugar, calcium, and antibody to macrophage Ctype lectin. Glycobiology 1998; 8:791–798. 129. RuchaudSparagano MH, Malaud E, Gayet O, Chignier E, Buckland R, McGregor JL. Mapping the epitope of a functional Pselectin monoclonal anibody (LYP20) to a short complementlike repeat (SCR 4) domain: use of humanmouse chimaera and homologuereplacement mutagenesis. Biochem J 1998; 332:309– 314. 130. Nunes DP, Keates AC, Afdhal NH, Offner GD. Bovine gallbladder mucin contains two distinct tandem repeating sequences: evidence for scavenger receptor cysteinerich repeats. Biochem J 1995; 310:41–48. 131. Kodama T, Freeman M, Rohrer L, Zabrecky J, Matsudaira P, Krieger M. Type I macrophage scavenger receptor contains helical and collagenlike coiled coils. Nature 1990; 343:531–535. 132. Jones NH, Clabby ML, Dialynas DP, Huang HS, Herzenberg LA, Strominger JL. Isolation of complementary DNA clones encoding the human lymphocyte glycoprotein T1/Leu1. Nature 1986; 323:346–349.
Page 232
133. Aruffo A, Melnick MB, Linsley PS, Seed B. The lymphocyte glycoprotein CD6 contains a repeated domain structure characteristic of a new family of cell surface and secreted proteins. J Exp Med 1991; 174:949–952. 134. Nunes DP, Afdhal NH, Offner GD. A recombinant bovine gallbladder mucin polypeptide binds biliary lipids and accelerates cholesterol crystal appearance time. Gastroenterology 1999; 116:936–942. 135. Ofner GD, Nunes DP, Zhang F, McAneny DB, Afdhal NH. Alterations in gallbladder mucin gene expression in patients with cholesterol gallstones. Gastroenterology 1996; 110:A1282. 136. Kano M, Shoda J, Irimura T, Ueda T, Iwasaki R, Urasaki T, Kawauchi Y, Asano T, Matsuzaki Y, Tanaka N. Effects of longterm ursodeoxycholate administration on expression levels of secretory lowmolecularweight phospholipases A2 and mucin genes in gallbladders and biliary composition in patients with multiple cholesterol stones. Hepatology 1998; 28:302–313. 137. McCool DJ, Marcon MA, Forstner JF, Forsmer GG. The T84 human colonic adenocarcinoma cell line produces mucin in culture and releases it in response to various secretagogues. Biochem J 1990; 267:491–500. 138. Forstner G, Zhang Y, McCool D, Forstner J. Mucin secretion by T84 cells: stimulation by PKC, Ca2+, and a protein kinase activated by Ca2+ ionophore. Am J Physiol 1993, 264:G1096–G1102. 139. Lee SP, Carey MC, LaMont JT. Aspirin prevention of cholesterol gallstone formation in prairie dogs. Science 1981; 211:1429–1431. 140. LaMorte WW, LaMont JT, Hale W, Booker ML, Scott TE, Turner B. Gallbladder prostaglandins and lysophospholipids as mediators of mucin secretion during cholelithiasis. Am J Physiol 1986; 251:G701–G709. 141. LaMont JT, Turner BS, DiBenedetto D, Handin R, Schafer AI. Arachidonic acid stimulates mucin secretion in prairie dog gallbladder. Am J Physiol 1983; 245:G92–G98. 142. Sasaki H, Tazuma S, Kajiyama G. Effects of 16,16dimethyl prostaglandin E2 on biliary mucous glycoprotein and gallstone formation in guinea pigs. Scand J Gastroenterol 1993; 28:495–499. 143. Kuver R, Savard C, Oda D, Lee SP. PGE generates intracellular cAMP and accelerates mucin secretion by cultured dog gallbladder epithelial cells. Am J Physiol 1994; 267:G998–G1003. 144. Cohen BI, Mosbach EH, Ayyad N, Yoshi M, McSherry CK. Aspirin does not prevent cholesterol cholelithiasis in two established animal models. Gastroenterology 1991; 101:1106–1109. 145. O'Leary DP, LaMorte WW, Scott TE, Booker ML, Stevenson J. Inhibition of prostaglandin synthesis fails to prevent gallbladder mucin hypersecretion in the cholesterolfed prairie dog. Gastroenterology 1991; 101:812–820. 146. Offner GD, Nunes DP, Moore EW, Afdhal NH. Prostaglandin E1 stimulates mucin secretion and increases MUC3 mRNA levels in cultured human gallbladder epithelial cells. Hepatology 1995; 22:110A. 147. Rhodes M, Allen A, Lennard TWJ. Mucus glycoprotein biosynthesis in the human gallbladder: inhibition by aspirin. Gut 1992; 33. 148. Rhodes M, Allen A, Dowling RH, Murphy G, Lennard TW. Inhibition of human gallbladder mucus synthesis in patients undergoing cholecystectomy. Gut 1992; 33:1113–1117. 149. Broomfield PH, Chopra R, Sheinbaum RC, Bonorris GG, Silverman A, Schoenfield LJ, Marks JW. Effects of ursodeoxycholic acid and aspirin on the formation of lithogenic bile and gallstones during loss of weight. N Engl J Med 1988; 319:1567–1572. 150. Hood K, Gleeson D, Ruppin DC, Dowling RH. Prevention of gallstone recurrence by nonsteroidal antiinflammatory drugs. Lancet 1988; 2:1223–1225. 151. Pazzi P, Scagliarini R, Sighinolfi D, Govoni M, La Corte R, Gullini S. Nonsteroidal
Page 233
antiinflammatory drug use and gallstone disease prevalence: a casecontrol study (see comments). Am J Gastroenterol 1998; 93:1420–1424. 152. Kurata JH, Marks J, Abbey D. One gram of aspirin per day does not reduce risk of hospitalization for gallstone disease. Dig Dis Sci 1991; 36:1110–1115. 153. Eder MI, Nunes DP, Afdhal NH, Offner GD, TNF stimulates mucin secretion, but not mucin synthesis, in human gallbladder epithelial cells. Gastroenterology 1998; 114:A1238. 154. Cheung CH, Moore EW, Prystowsky JB, Rege RV. TNF stimulates mucus secretion in gallbladder epithelial cells. Gastroenterology 1998; 114:A1224. 155. Jackson GDF, Dai Y, Fung C, Hansen PGC. Tumour necrosis factor alpha—A constitutive protein of bile. Adv Exp Med Biol 1995; 371B:1083–1086. 156. Levine SJ, Larivee P, Logun C, Angus CW, Ognibene FP, Shelhamer JH. Tumor necrosis factoralpha induces mucin hypersecretion and MUC2 gene expression by human airway epithelial cells. Am J Respir Cell Mol Biol 1995; 12:196–204. 157. Fischer BM, Rochelle LG, Voynow JA, Akley NJ, Adler KB. Tumor necrosis factoralpha stimulates mucin secretion and cyclic GMP production by guinea pig tracheal epithelial cells in vitro. Am J Respir Cell Mol Biol 1999; 20:413–422. 158. Klinkspoor JH, Kuver R, Savard CE, Oda D, Azzouz H, Tytgat GN, Groen AK, Lee SP. Model bile and bile salts accelerate mucin secretion by cultured dog gallbladder epithelial cells. Gastroenterology 1995; 109:264–274. 159. Klinkspoor JH, Yoshida T, Lee SP. Bile salts stimulate mucin secretion by cultured dog gallbladder epithelial cells independent of their detergent effect. Biochem J 1998; 332:257–262. 160. DrayCharier N, Paul A, Combettes L, Bouin M, Mergey M, Malladur P, Capeau J, Housset C. Regulation of mucin secretion in human gallbladder epithelial cells: predominant role of calcium and protein kinase C. Gastroenterology 1997; 112:978–990. 161. Orlando RA, Exner M, Czekay RP, Yamazaki H, Saito A, Ullrich R, Kerjaschki D, Farquhar MG. Identification of the second cluster of ligandbinding repeats in megalin as a site for receptorligand interactions. Proc Natl Acad Sci USA 1997; 94:2368–2373. 162. Adler KB, Fischer BM, Li H, Choe NH, Wright DT. Hypersecretion of mucin in response to inflammatory mediators by guinea pig tracheal epithelial cells in vitro is blocked by inhibition of nitric oxide synthase. Am J Respir Cell Mol Biol 1995; 13:526–530. 163. Keaveny AP, Offner GD, Afdhal NH. Inducible nitric oxide synthase (iNOS) is the principal isoform expressed in human gallbladder. Gastroenterology 1998; 114:A526.
Page 235
11— Role of Proteins in Cholesterol Crystallization in Bile A. Andre van den Berg and Albert K. Groen Academic Medical Center, Amsterdam, The Netherlands I— Introduction Until the midseventies, cholesterol supersaturation of bile was considered to be the primary and perhaps only factor responsible for cholesterol gallstone formation. In the early eighties, several reports appeared (1–4) that demonstrated cholesterol supersaturation in bile from persons without cholesterol stones and sometimes undersaturated bile in patients with stones (4), with much overlap. Rather, those with gallstones were distinguished by the much more rapid crystallization of cholesterol when the filtered bile was studied in vitro. These phenomena were considered to be incompatible with an exclusive role of cholesterol supersaturation, and proteins were considered to be responsible for the anomalous behavior. In 1984, a landmark study of Holzbach et al. (5) appeared, for the first time showing inhibition of cholesterol crystallization by a fraction of bile that contained primarily proteins. Somewhat later, Strasberg's group suggested that bile also contained proteins promoting cholesterol crystallization (6). Since these pioneering studies, a multitude of biliary proteins have been isolated and claimed to be important factors in the pathogenesis of cholesterol gallstones. In this chapter we review the characteristics, mechanism, and significance of the different proteins. II— Origin of Protein To a great extent, the protein composition of the bile reflects that of serum (7). It has therefore been assumed that most biliary protein originates from the serum compartment and passes into bile via the tight junctions between both the hepatocytes and the biliary epithelial cells. Exceptions to the rule suggest, however, that many proteins synthesized in the liver are transported directly into bile either via selective active transport mechanisms or in a selective mode through cotransport. A case in point is albumin; Saucan and Palade (8) demonstrated that, at least in rats, this most abundant biliary protein is secreted in bipolar fashion both at the sinusoidal and canalicular side of the membrane. Mucin is the second most abundant biliary protein, followed by IgA and APF/CBP (see Chap. 10) (9) (Table 1). Apart from mucin, no important differences have been observed so far between the relative composition of proteins in gallbladder bile and hepatic bile. Interestingly, in control patients (without gallstones), no difference has been observed in the concentrations of protein between gallbladder bile and hepatic bile despite the concentrative function
Page 236 Table 1 Important Proteins in Gallbladder Bile
Concentration (mg/mL)
Reference
Albumin
0.4–2.5
17,74
Mucin
0.2–10
17,18,32,75,76
APF/CBP
0.6–1
77,78
Immunoglobulin A
0.13
17,18,27
Immunoglobulin G
0.05–0.62
17,18,27
Immunoglobulin M
0.05–0.14
17,18,27
0.05–0.07
17,18,48
0.04–0.05
17,18,47
1acid glycoprotein Haptoglobin Aminopeptidase N
0.13
17
of the gallbladder, which should have increased the protein content to a similar extent as the biliary lipids—i.e., about fivefold (10). Since protease activity in gallbladder bile is minor, one has to conclude that there is substantial uptake of protein by the gallbladder epithelium (11). In patients with gallstones, the situation is different. An increased protein content compared to controls, which has been reported in most but not all studies (12–16), would suggest a defect in the absorption of protein by the gallbladder. Recent studies have shed more light on this issue. Miquel et al. confirmed the increased protein content of gallbladder bile from cholesterol stone patients compared with samples from controls (17). Interestingly, this increase was due to only a few proteins: albumin, mucin, and IgG. In line with these observations, Keulemans et al., studying cholesterol gallstone patients, found that only albumin, mucin and IgG were relatively enriched in gallbladder bile as compared with hepatic bile (18). These observations suggest that, under normal conditions, uptake of all biliary proteins takes place in the gallbladder. This process is largely intact in gallstone patients, but there appears to be selective impairment of absorption and/or secretion of certain proteins, perhaps due to local inflammatory processes in the gallbladder mucosa. III— Definition of Nucleation Many studies use the term nucleation time to describe the first measurable appearance of crystals in human or model bile during incubation in vitro. The unit cell of a cholesterol crystal comprises eight cholesterol molecules (9). The size of such a crystal nucleus is a few angstroms—too tiny to be visualized by the methodology available today. Thus, it is crystal detection time (CDT) and crystal growth that is actually measured, so that it is inappropriate to use the terms nucleation time or nucleation factors. We will therefore employ a more general terminology and describe the effects exerted by proteins as crystallizationpromoting or inhibiting. In almost all studies described in this chapter, model biles have been used to characterize the influence of proteins. These model biles are composed of physiological total concentrations of bile salts, phospholipid, and cholesterol. In general, taurocholate is employed as the bile salt and eggyolk lecithin as the phospholipid. These differ from the bile salts and phospholipids in human bile, which not only influences crystallization behavior but also may affect the way proteins influence the system. To circumvent these problems, the group of Strasberg (19) has used heatinactivated bile derived from gallstone patients. The advantage of this system is that the lipid and ion content of such bile fully reflects the in situ situation. The disadvantages are the lack of fixed concentrations of nonprotein components and the presence of heatdenatured proteins.
Page 237
IV— Inhibitors of Crystallization Table 2 shows a list of biliary proteins that have been demonstrated to exert crystallizationinhibiting activity. Since the bile of patients without gallstones was frequently found to be supersaturated with cholesterol, the activity of this class of proteins was expected to be present in such control biles. In 1984, Holzbach et al. (5) fractionated bile by gel permeation chromatography and found indeed a fraction that inhibited cholesterol crystallization and contained a multitude of proteins. Subsequently apolipoproteins AI and AII from this fraction were shown to be good candidates to account for the inhibiting activity (20). Busch et al. (21) confirmed that Apo AI both increases the time of onset of visible cholesterol crystallization and also inhibits crystal growth. The concentration in gallbladder bile, however, does not differ between patients with or without gallstones (22). To our knowledge, the protein has not been isolated from bile, rendering it uncertain whether biliary Apo AI is intact and exerts activity or might differ qualitatively in bile from patients with gallstones versus those without. Subsequently Holzbach's group has isolated a potent inhibiting fraction by its affinity for Helix pomatia lectin (23). The main protein in this fraction was a 128kDa protein composed of 58 and 63kDa subunits, each of which yielded an identical polypeptide of 35 kDa after deglycosylation (23). A preliminary report of partial amino acid sequence showed homology to a cytokeratin, but this work has not yet appeared in definitive form, nor has the concentration of the protein been compared between stone and nonstone patients. More recently the same group reported the presence of yet another crystallization inhibitor in human gallbladder bile. This 15kDa protein, which forms dimers and tetramers, was isolated by affinity chromatography using bound antibodies against Apo AI. The protein was also present in serum and appeared to be unique on N terminal amino acid sequencing. When this protein was added to model biles to a final concentration of 10 µg/mL (24), it only mildly affected crystal detection time and the crystal mass formed. Busch et al. (25) isolated another inhibitory protein fraction that coprecipitated with the cholesterol crystals formed upon incubation in supersaturated model biles of the protein fraction, which bound to the H. pomatia lectin. The four coprecipitated proteins, purified via SDSpolyacrylamide gel electrophoresis (SDSPAGE), each inhibited crystallization of cholesterol from model bile systems. Aminoterminal sequencing revealed the 74, 63, and 28kDa proteins to be parts of IgA (26). The identity of the 16kDa protein was not established, but the molecular weight suggested this protein to be the J chain of IgA (26). The 74, 63, and 28kDa subunits each increased onset time and decreased the final extent of crystallization; the 28kDa light chain exhibited the highest inhibitory activity. Since the subunits probably denatured during isolation from SDSPAGE gels, it is not clear whether the activity observed has relevance for the in vivo situation. No reconstitution experiments were performed comparing the activity of the combined subunits to the intact (native) IgA fraction. Busch et al. did test the activity of native (polymeric) IgA, isolated by immunoaffinity purification from an H. pomatiabound fraction. A complex pattern of inhibition was observed. At very low concentrations (1 and 10 µg/mL), the protein did have an inhibiting effect, but this was attenuated Table 2 List of Crystallization Inhibitors Inhibitor
Reference
Apolipoprotein AI
20
Apolipoprotein AII
20
128kDa Helix pomatia binding protein IgA 15kDa Protein
23 25,26 24
Page 238
as concentrations of IgA were increased; at 300 µg/mL, no effect was observed (26). By contrast, Upadhya et al. (27) found a modest dosedependent increase of crystallization when 62.5 to 625 µg/mL of purified biliary IgA was added to heat inactivated bile samples from gallstone patients; very low concentrations were not tested, however. Ahmed et al. (28), on addition of IgA derived from human colostrum, reported a dose dependent increase in crystallization of cholesterol from model bile. In their hands, 5 µg/mL decreased crystal detection time by 50%. What can be the explanation for these varying results? First, the three groups used different detection systems; particularly the system of heated abnormal bile used by Upadhya et al. (27) could behave differently, since this system already contains endogenous IgA. Second, different fractions of IgA were used in the three studies; of note, the immunopurified protein obtained by Busch et al. (26) from the H. pomatiabinding fraction contains only a minor subfraction of biliary IgA. It is not yet clear whether the activity of IgA varies with subclass. If all components of the protein indeed are inhibitory, probably the (variable) antigen binding site is not involved. Clearly more work is required to clarify these issues. V— CrystallizationPromoting Proteins Table 3 lists the proteins that have been reported to promote cholesterol crystallization. The first evidence for the presence of promoters was reported by Burnstein et al. (6), who showed that the addition of small portions of gallbladder bile from patients with cholesterol gallstones dramatically shortened the crystal detection time of bile from patients without stones. Although the presence of microcrystals in the pathological biles could not be excluded, passage of the activity through a filter with a 300kDa cutoff was highly suggestive for a role of proteins smaller than mucins. We later demonstrated potent crystallizationpromoting activity of a concanavalin A bound biliary glycoprotein fraction that was devoid of mucins (29). Levy et al. (30) did report promoting activity for mucin at physiological concentrations. By contrast, Gallinger et al. (19) demonstrated that removal of mucin from native bile by filtration and ultracentrifugation did not change crystal detection time, suggesting a minor role for mucin. In this study, however, potential effects on crystal growth were not determined, nor could it be excluded that microcrystals were still present in the ultracentrifuged samples. The investigators did confirm the procrystallizing activity of mucin; when added to heatinactivated bile, the protein did accelerate crystallization. Gallinger et al. also found no difference in activity between mucin from patients with stones compared to that from controls (19). This result was confirmed by Klinkspoor et al. (31). Harvey et al. found a twofold higher mucin content in Table 3 Pronase Resistancy of Crystallization Promoters Crystallization promoter
Pronaseresistant
Reference
Mucin
No
79
Aminopeptidase N
No
37,38
Immunoglobulin G
No
27,43
Immunoglobulin M
No
27,43
No
72
No
47
IAcid glycoprotein Haptoglobin 1Antichymotrypsin
38
Yes
80
Fibronectin
No
41
Lowdensity pronucleator
Yes
52
Yes
Phospholipase C
Page 239
cholesterol stone patients compared to controls and pigment stone patients, but the difference was not significant (32). Nonetheless, as during the isolation of the mucins, their quaternary structures are distorted; the equality of the activities might be imposed by the isolation procedure. Differences in the qualities of the hydrophobic domains of native mucin between gallstone patients and controls might still exist and influence the crystal detection time. A simple and reliable isolation procedure has recently been described by Miquel et al. (33) and applied to measure the mucin content of gallbladder bile in a large group of Chilean patients. Qualitatively, the results compared well with those of Harvey et al. (32), but the twofold difference in mucin content between cholesterol stone patients and controls was highly significant. The values in the Chilean patients were threefold higher, which might reflect technology but also ethnic differences (33). Similar results were found by other groups, indicating that the lack of significance in the study of Harvey et al. was probably due to a type 2 error. Using lectin affinity chromatography with concanavalin A (Con A) sepharose, a nonmucin biliary glycoprotein fraction was isolated that contained potent crystallizationpromoting activity (29). Later, Harvey et al. (34) demonstrated that lentil lectin and wheat germ agglutinin likewise could bind glycoproteins with crystallizationpromoting activity. Like Con A, lentil lectin is selective for glycoproteins containing saccharide side chains with mannose or glucose, whereas wheat germ aggulutinin is selective for Nacetylglucosamine. Since the Con Abinding fraction (CABF) seemed to contain the most potent activity in most studies, this fraction was used for further purification. In 1988, we reported that the activity in CABF was resistant to prolonged treatment with pronase, a potent protease (29). This was important, because Pattinson and Willis demonstrated no effect of prolonged pronase treatment on the crystal detection time (CDT) of gallbladder bile from patients with cholesterol gallstones (35); we have confirmed this result (unpublished data). After pronase treatment, a 130kDa protein was isolated from CABF by native polyacrylamide gel electrophoresis (36); this protein was identified later as aminopeptidaseN (37). Offner et al. (37) could not confirm the resistance of this protein to pronase, and subsequent studies by us confirmed the pronase sensitivity of isolated aminopeptidaseN (38). Probably, in native bile, the protein is partly protected because it incorporates into phospholipid/cholesterol vesicles (39,40). A number of other proteins in CABF were reported to be crystallization promoters. Chijiiwa et al. (41) suggested a role for fibronectin. However, their results could not be confirmed (42). The group of Strasberg suggested immunoglobulin G and M to be the most active candidates in the Con Abinding fraction (27,43). When isolated from bile, these proteins indeed shortened CDT in heatinactivated bile, and others have confirmed these results in model biles (28,44). IgG concentrations were found to be increased in gallbladder bile of cholesterol stone patients (27,43) and up to fivefold in (45) in bile derived from Chilean patients. It remains unclear, however, whether the immunoglobulins indeed account for the rapid crystallization in gallstone patients. Extraction of immunoglobulins from bile did not influence crystallization activity (45), and the immunoglobulins are not resistant to pronase even under the conditions prevailing in the Con Abinding fraction. The group of Holzbach reported crystallizationpromoting activity by other proteins in the Con Abinding fraction—the acutephase proteins 1acid glycoprotein and haptoglobin (46,47). The concentration of 1acid glycoprotein was reported to be increased somewhat in the bile of cholesterol stone patients (48). Extraction of 1acid glycoprotein by immunoaffinity chromatography reduced the crystallizationpromoting activity of CABF by about 30% (44). We have not been able to confirm this result. Pattinson and Willis found phospholipase Clike activity in bile, which was resistant to pronase treatment, and the enzyme is a potent crystallization promoter when added to model bile (40–51). Abei et al. (44), however, were unable to immunoprecipitate the phospholipase Clike activity by immunoaffinity chromatography and suggested that the activity was caused by something else. It could well be, however, that the phospholipase C isolated by Pattinson and Willis (49) is an as yet uncharacterized isoform that does not crossreact with available antibodies. To identify the proteaseresistant crystallizationpromoting proteins, Zijlstra et al. carried out a prolonged and repeated incubation
Page 240
with pronase and subsequently fractionated the digested CABF on a superose gelpermeation column. Two peaks with activity emerged; one in the void volume the other at about 60kDa (38). Pronase treatment did not alter the elution profile of the activity, indicating that promoters remained intact and were truly protease insensitive. The void volume peak contained a biliary lipoprotein consisting of small amounts of free fatty acid, cholesterol, phospholipid, and an 85kDa protein (52). The 60kDa peak contained a number of proteins, as determined by Nterminal amino acid sequencing, which revealed the more prominent components to be derived from the protease inhibitors 1antitrypsin and 1antichymotrypsin (28). Immunoextraction of these proteins from bile showed that 1antichymotrypsin accounted for most of the activity; 1antitrypsin was not active. De Bruijn (53) attempted to quantify the contribution of the biliary lipoprotein particle, which contains the 85kDa protein, to the 1antichymotrypsincontaining fraction. She separated the two activities by densitygradient ultracentrifugation, which generates a baseline separation. Unfortunately, it proved to be impossible to produce a reliable quantification, since the isolation procedure altered the activity of the promoters, producing a recovery of higher than 100%. Harvey et al. (54) suggested that CABF contained cholesterol microcrystals that account for part of the activity and in principle could explain the excess of activity found by De Bruijn (53). Two observations argue against the importance of contaminating cholesterol crystals. First, after biotinylation of the proteins in CABF, almost all activity bound to agarose bead coated with streptavidin. Second, filtration of mixtures of CABF with model bile over a 0.2µm filter did not remove activity, which excludes a role for the type of crystal reported by Harvey et al. (54). Patients carrying the E4 isoform of apolipoprotein E (apo E) have been implicated to be at risk for gallstone disease. A higher cholesterol content of their gallstones has been reported in Finnish patients (55) and an increased cholesterol saturation index (CSI) in Spanish patients (56). In Chileans the association was, however, absent (57), and also in Dutch patients no association between apo E4 subtype and CSI or CDT was found (58). Whether the isolated apo E4 shows crystallization promoting activity has not been reported. VI— Relevance of Proteins to Crystallization in Native Bile Differences in CDTs between biles from patients with versus those without gallstones are quite dramatic. Therefore, for a given bile component, clearcut qualitative or quantitative differences can be expected if such a factor truly contributes significantly to the more rapid crystal formation in bile of gallstone patients. However, the fact that isolated biliary factors often display significant effects on the crystallization of cholesterol when applied as a single agents in model biles and show significant changes in concentration during formation of gallstones in laboratory animals provides no proof of their relevance in complete human biles. The role of an individual protein in native human bile is best assessed by investigating the effect of specific extraction or addition on the crystallization of cholesterol. Gallinger et al. (19) were the first to perform such a study. In their hands, extraction of mucin did not affect crystallization. In principle, this is convincing evidence for a lack of importance of mucins in the bile samples tested. The effect of addition of mucins isolated from the same biles was not tested. In our lab the effect of adding isolated CABF to native bile was tested. In 5 of the 7 samples, CABF increased crystallization in native bile (29). Keulemans et al. (59) determined the effect of direct extraction of CABF from native bile sample. To this purpose, native bile was incubated with Con Asepharose beads, and bile incubated with sepharose beads served as control. After 2 h of incubation, the beads were centrifuged and crystallization was determined in the supernatant. Surprisingly, extraction of CABP, inhibited crystallization only in fastcrystallizing biles (<4 days), and the effect was more pronounced on crystal growth than on crystal detection time. Extraction never induced increased crystallization, indicating that inhibitors that bind to ConA do not play a role in the regulation of crystallization in bile from
Page 241
patients with gallstones. Interestingly, the content of proposed promoters or inhibitors ( 1acidglycoprotein, IgA, IgM, IgG, haptoglobin) was not different between slow and fastcrystallizing samples. In addition, removal of all these factors from slowcrystallizing samples did not influence crystallization, indicating that these factors, at least in combination, do not control crystallization. Van Erpecum et al. investigated whether the content of known crystallization promoters could account for the differences in CDT between patients with single stones and those with multiple stones. No difference in the content of 1acidglycoprotein, IgA, IgM, IgG, haptoglobin, and mucin was observed. Since biles from singlestone patients in general belong to the slowcrystallizing group of gallstone patients with CDT >4 days, this observation is in line with the data of Keulemans et al. (59), who also did not find a difference in the content of the above mentioned promoters between slow and fastcrystallizing biles. The question remains which factor then accounts for the activity in the fast crystallizing samples. A good candidate is the LDL particle, but no direct quantifying method for this factor is yet available. In a subsequent study, Keulemans et al. (56) investigated whether the CABF played a role in the rapid crystallization of cholesterol from gallbladder biles from patients with Crohn's disease. Indeed, extraction of CABF shortened CDT in all but one of the crystallizing samples. In contrast, in biles from patients with ulcerative colitis, extraction of CABF increased crystallization in 7 of the 9 crystallizing samples, indicating activity of crystallization inhibitors. Subjecting supersaturated samples from patients with and without stones to the same procedure will have to reveal differences. As yet, no firm evidence has been provided that any individual biliary protein contributes significantly to the difference between the fast crystallization of supersaturated biles from patients with cholesterol gallstones and the slower crystallization of similarly supersaturated biles from patients without cholesterol gallstones. VII— Possible Mechanism by Which Proteins Affect Crystallization The molecular mechanisms involved in the crystallization of cholesterol are discussed in greater detail in Chap. 5 of this book. Here a limited interpretation is given to provide a basis for understanding how biliary proteins can influence the formation of cholesterol crystals. Supersaturated biles carry small amounts of monomeric cholesterol in the aqueous phase but primarily in mixed micelles and vesicles. Theoretically, these three separate but interequilibrating forms might serve as precursors for the formation of cholesterol crystals. In bile supersaturated with cholesterol, the bulk of excess cholesterol is probably carried in vesicles, and most studies indicate an important role for the vesicles in the onset of crystallization (60–63). A clear negative correlation between the time of detectable onset of crystallization (recorded crystal detection time) and the presence of large multilamellar vesicles has been suggested by several studies. Studies on pure vesicle suspensions demonstrated that vesicles with high cholesteroltophospholipid ratios participate more actively in cholesterol crystallization (see Chap. 5). Some studies haved clearly demonstrated a relationship between the CDT and the vesicular fraction of bile (64,65). This has been taken to indicate that crystal nuclei that form in model and native biles probably originate from cholesterol in vesicles. Aggregation and fusion of small unilamellar vesicles into large multilamellar vesicles seems to enhance formation of the crystal nucleus as well as its subsequent growth into larger crystals that can be detected by light microscopy. Nevertheless, it has not been excluded that the nuclei originate from supersaturated micelles or from monomeric free cholesterol in the aqueous phase. The distribution of cholesterol between the different phases is determined by the bile salts and phospholipids. In native bile, an additional phenomenon plays a role— e.g., direct nucleation of cholesterol on small foreign bodies or precipitates of calcium salts, a process called epitaxy. Epitaxis for cholesterol in bile has been demonstrated to occur (9).
Page 242
Pronucleating agents in bile enhance the crystallization of cholesterol, either by stimulating the formation of crystal nuclei and/or the subsequent growth of these nuclei to the size of visible crystals. The former can be accomplished either by increasing the cholesterol to the phospholipid ratio of vesicles and/or by promoting the aggregation and/or fusion of vesicles into larger multilamellar vesicles. Because the basal unit of the cholesterol crystal is an octameric flat box with two layers of four molecules in square orientation, one could envision that the probability of layering of cholesterol in such an orientation increases when cholesterolrich domains are present in several stacked membranes. This probably explains why aggregation and fusion of vesicles into multilamellar vesicles promotes cholesterol crystallization. The vesicles could be the target of both promoters and inhibitors of crystallization—the former by destabilizing vesicles or promoting fusion and the latter by stabilization or prevention of fusion of vesicles. Ahmed et al. (28) suggested the effect of proteins to be aspecific and just a function of their hydrophobicity. In their hands, hydrophilic proteins were promoters and hydrophobic proteins inhibitors. Although this is an attractive concept, data from many others do not support this contention. For instance, Ahmed et al. (28) found that a strong promoting effect was exerted by albumin in bile, which has not been seen in other studies. In addition, albumin is pronasesensitive, and this concept would not explain the resistance of the CDT to pronase. VIII— Crystallization Inhibitors A— APo A1 and A2 Yamashita and coworkers (66) studied the crystallizationinhibiting activity of Apo A1. When commercially available Apo A1 was eluted from a sepharose CL6B column, the retention time was altered if it was first incubated in model bile, suggesting that an interaction with biliary lipids occurred. After addition to model bile to a concentration of 30 g/mL, Apo A1 (within the physiological range of human gallbladder biles), Yamashita et al. observed only a slight inhibition of crystal formation in a photometric crystal growth assay. More direct evidence for a stabilizing role of apolipoproteins toward bile has come from a sequential transmission electron microscopy study by Tao et al. (67). In this study, lipid particles in model bile displayed much slower fusion of phospholipid vesicles into multilamellar structures and subsequent nucleation of cholesterol crystals in the presence of Apo A1. After elution from a gelpermeation column, the Apo A1 was consistently recovered from the fractions that contained small phospholipid vesicles, again supporting the hypothesis that apolipoproteins bind to small vesicles and inhibit their fusion. Another study from Japan likewise reported that both Apo A1 and Apo A2 retarded the development of maximal turbidity of supersaturated model biles. Since the maximal turbidity is likely related to multilamellar vesicles and coincides with the onset of crystallization, these results also suggest that apolipoproteins inhibit crystallization by slowing the fusion of unilamellar vesicles (68). In summary, the data favor a mechanism in which apolipoproteins bind to or are incorporated into the smaller vesicular structures of bile, preventing their aggregation and/or fusion into the larger multilamellar vesicles from which crystals nucleate. The recently described nonperturbing techniques of cryotransmission electron microscopy or videoenhanced light microscopy should now be applied to directly estimate the effects of various biliary apolipoproreins on the average size of vesicles in bile and the time of onset of crystal formation (69). B— LectinBinding Inhibitory Glycoproteins The supposed mechanism whereby the bile glycoproteins inhibit crystallization and bind to H. pomatia lectin is completely different from that of the apolipoproteins. The former are hypothesized to function like the glycoproteins in the serum of polar fish, preventing formation of ice in the blood (23,25). These glycoproteins inhibit crystal growth by binding to and
Page 243
poisoning the crystal surface—i.e., they are antitaxis factors. The proteins apparently bind preferentially to the sites where crystal growth is fastest, very effectively arresting growth. The study by Busch and colleagues (26) is strong support for a similar mechanism involved in the inhibition of cholesterol crystal growth by the four secretory IgA glycoproteins. These IgA subunits were shown to bind to cholesterol crystals, change the morphology of the crystals, and retard the growth rate, resulting in a decreased crystal mass. This mechanism has also been proposed for the inhibition of crystal formation by the 120kDa glycoprotein and its subunits (23). This seems less likely, as these glycoproteins did not change growth kinetics. IX— Crystallization Promoters A— Mucin Biliary mucins (described in detail in Chap. 6) are among the most potent in vitro pronucleating agents. Afdhal and coworkers (70) showed that mucins from various sources all accelerate both vesicle aggregation and vesicle fusion and stimulate the formation of cholesterol crystals in model biles. Using fluorescently labeled bovine gallbladder mucins in model biles with fluorescently labeled vesicles, they demonstrated further that mucin has a biphasic effect. The glycan domains reversibly promote vesicle aggregation. The hydrophobic stretches of the mucin polypeptide backbone facilitated an irreversible fusion of vesicles. These authors hypothesized further that a large multilamellar vesicular structure, with mucin bound to it, is the true crystal precursor in systems with mucin present, but no evidence has been provided that this complex generates crystals more readily than normal multilamellar vesicles in the absence of mucin. B— Concanavalin ABinding Fraction CABF has multiple effects on crystallization. It strongly decreases crystal appearance time, increases the rate of crystal growth, and increases the magnitude of the final plateau of crystallization. In true equilibrium, the quantity of crystals at the plateau is determined exclusively by the degree of supersaturation of the system. Since it is difficult to imagine how relatively small amounts of proteins could control the thermodynamics of cholesterol solubilization, it is likely that only metastable intermediary plateaus of crystallization are influenced by CABF. De Bruijn et al. loaded small vesicles with high (selfquenching) concentrations of the fluorescent dye carboxyfluorescein (71). Addition of CABF lead to immediate fluorescence, indicating release of the carboxyfluorescein in the surrounding medium. The CABF thus disrupted the vesicles directly without their prior aggregation and fusion into large multilamellar vesicles. This mechanism has also been shown to be part of the pronucleating activity of mucins. Which of the individual proteins in CABF is responsible for these phenomena is not yet clear, but De Bruijn (53) showed differential effects of the low and highdensity fractions in the crystal growth assay. The lowdensityparticle fraction mainly shortened CDT and had very little effect on crystal growth, whereas the mixture of proteins in the highdensity fraction mainly increased crystal growth. Data from the group of Holzbach (72) suggest that the glycan moieties of the proteins determine activity, since only 1acid glycoprotein, with a high Con Abinding affinity, significantly promoted activity. X— Role of Proteins in Gallstone Growth So far we have limited our discussion to the effect of biliary proteins on cholesterol crystallization on a time scale of days, but growth of a gallstone appears to occur over many years. The role of pro and antinucleating proteins in stone growth has been little studied, since it has always been assumed that factors promoting the crystallization of cholesterol likewise
Page 244
Figure 1 Growth of small human cholesterol gallstones, 15 to 25 mg each, in 2 mL of model biles supersaturated with cholesterol. The model biles were composed of tauro and deoxytaurocholic acids, eggyolk lecithin, and cholesterol (molar ratios 8:2:1) with total lipids 12 g/dL, CSI 1.6. A. The increase in mass was set to 100% in each experiment. B to D. Relative mass increases in model biles where crystallization was stimulated with 330 g/mL CABF added (B); 660 g/mL CABF added (C); 1 mg/mL of human hepatic mucin and calcium phosphate precipitates added (D). E. Mass increases in model biles where crystallization of cholesterol was completed before gallstones were added.
enhance the formation and growth of cholesterol gallstones. This contention has never been proven; no one has yet been able to grow cholesterol gallstones de novo in vitro. We have approached this problem by studying the growth of human small gallstones in model systems and native bile. A summary of our recent data is given in Fig. 1. Small cholesterol gallstones incubated in 2 mL of fresh model biles supersaturated with cholesterol increased their mass from 3 to 18% within 2 weeks. This growth rate was positively correlated with the saturation index but was negatively correlated with the rate of crystallization. Thus, when the formation of cholesterol crystals in model biles was increased either by the addition of hepatic human mucins, calcium salt precipitates or the addition of Con Abinding glycoprotein fractions of biles from gallstone patients, the growth of the stones was very much reduced. Stones incubated in model biles containing massive amounts of cholesterol crystals but with far lower excess cholesterol in the fluid phase never increased more than 2% in mass, suggesting that the stones did not grow by aggregation of performed crystals. It thus seems that cholesterol carried in excess in bile can either accrete to the surface of a stone or selfaggregate and nucleate as a crystal, and these processes appeared to be competitive in vitro. Whether such cholesterol that accretes to stone surface is of monomeric, micellar, or vesicular origin requires further investigation. We hypothesize that soluble protein factors that induce crystallization of cholesterol reduce stone growth in vivo as well. One could reason that during inflammation of the gallbladder, when contraction is reduced and bile becomes more supersaturated, a mechanism is activated to prevent stone formation. The physicological function of these pronucleators, many of which are acutephase proteins and immunoglobulins, would then be to limit growth of cholesterol gallstones through rapid growth of cholesterol crystals. It suggests a rationale for why the human body would produce pro and antinucleating proteins. XI— Concluding Remarks The major conclusion from this chapter is that biliary proteins do play a role in cholesterol crystallization and particularly account for the rapid crystal formation often seen in bile of
Page 245
patients with gallstones. Which specific factors are responsible for this phenomenon still remains elusive. None of all the putative promoters that have been isolated as yet fulfill the criteria formulated by Harvey and Strasberg (73): (a) the substance should be present in fastcrystallizing bile with either a quantitative or qualitative difference from normal bile; (b) upon extraction of the substance from fastcrystallizing bile, the crystallization should be delayed; and (c) when the substance is added to slowly crystallizing bile, the crystallization should be accelerated. We add a fourth criterion: such a protein should be resistant to protease treatment. Of the promoters listed in Table 3, only three candidates fulfill the latter criterion; phospholipase C, 1antichymotrypsin, and the lowdensity particle from CABF. Regarding the role of inhibitors, the situation is even more uncertain. An inhibiting influence of proteins has not yet been demonstrated except in bile of patients with ulcerative colitis. It has been postulated by many authors that an imbalance between procrystallizing and anticrystallizing factors is decisive whether or not stones are formed. Our recent data on the effect of proteins on gallstone growth makes one wonder whether this hypothesis is correct. The true relevance of pro and antinucleating protein factors to the overall cholelithiasis in native bile, especially to the growth of gallstones, remains to be explored. References 1. Holzbach RT, Marsh M, Oleszewski M, Holan K. Cholesterol solubility in bile: evidence that supersaturated bile is frequent in healthy man. J Clin Invest 1973; 53:1467–1479. 2. Holan KR, Holzbach RT, Hermann RE, Cooperman AM, Claffey WJ. Nucleation time: a key factor in the pathogenesis of cholesterol gallstone disease (abstr). Gastroenterology 1979; 77:611–617. 3. Sedaghat A, Grundy SM. Cholesterol crystals and the formation of cholesterol gallstones. N Engl J Med 1980; 302:1274–1277. 4. Gollish SH, Burnstein MD; Ilson RG, Petrunka CN, Strasberg SM. Nucleation of cholesterol monohydrate crystals from hepatic and gallbladder bile of patients with cholesterol gallstones. Gut 1983; 24:836–844. 5. Holzbach RT, Kibe A, Thiel E, Howell JH, Marsh M, Hermann RE. Biliary proteins: unique inhibitors of cholesterol crystal nucleation in human gallbladder bile. J Clin Invest 1984; 73:35–45. 6. Burnstein MJ, Ilson RG, Petrunka CN, Taylor RD, Strasberg SM. Evidence for a potent nucleating factor in the gallbladder bile of patients with cholesterol gallstones. Gastroenterology 1983; 85:801–807. 7. He C, Fischer S, Meyer G, Muller I, Jungst D. Twodimensional electrophoretic analysis of vesicular and micellar proteins of gallbladder bile. J Chromatogr 1997; 776:109–115. 8. Saucan L, Palade GE. Differential colchicine effects on the transport of membrane and secretory proteins in rat hepatocytes in vivo—bipolar secretion of albumin. Hepatology 1992; 15:714–721. 9. Craven BM. Crystal structure of cholesterol monohydrate. Nature 1976; 260:727–729. 10. Gallinger S, Harvey PRC, Petrunka CN, Ilson RG, Strasberg SM. Biliary proteins and the nucleation defect in cholesterol cholelithiasis. Gastroenterology 1987; 92:867–875. 11. Harvey PRC, Aravinda Upadhya G, Toth JL, Strasberg SM. Fluorometric assay of protein in native human bile. Clin Chim Acta 1989; 183:147–154. 12. Sahlin S. Total protein content of human gallbladder bile: relation to cholesterol gallstone disease and effects of treatment with bile acids and aspirin. Eur J Surg 1996; 162:463–469. 13. Jungst D, Lang T, von Ritter C, Paumgartner G. Role of high total protein in gallbladderbile in the formation of cholesterol gallstones. Gastroenterology 1991; 100:1724–1729.
Page 246
14. Chijiiwa K, Hirota I, Noshiro H. High vesicular cholesterol and protein in bile are associated with formation of cholesterol but not pigment gallstones. Dig Dis Sci 1993; 38:161–166. 15. Strasberg SM, Toth JL, Gallinger S, Harvey PRC. High protein and total lipid concentration are associated with reduced metastability of bile in an early stage of cholesterol gallstone formation. Gastroenterology 1990; 98:739–746. 16. Yamazaki K, Powers SP, LaRusso NF. Biliary proteins: assessment of quantitative techniques and comparison in gallstone and nongallstone subjects. J Lipid Res 1988; 29:1055–1063. 17. Miquel JF, Nunez L, Amigo L, Gonzalez S, Raddatz A, Rigotti A, Nervi F. Cholesterol saturation, not proteins or cholecystitis, is critical for crystal formation in human gallbladder bile. Gastroenterology 1998; 114:1016–1023. 18. Keulemans YCA, Mok KS, Dewit LT, Gouma DJ, Groen AK. Hepatic bile versus gallbladder bile—a comparison of protein and lipid concentration and composition in cholesterol gallstone patients. Hepatology 1998; 28:11–16. 19. Gallinger S, Taylor RD, Harvey PRC, Petrunka CN, Strasberg SM. Effect of mucous glycoprotein on the nucleation of time of human bile: Gastroenterology 1985; 89:648–658. 20. Kibe A, Holzbach RT, LaRusso NF, Mao ST. Inhibition of cholesterol crystal formation by apolipoproteins AI and AII in model systems of supersaturated bile: implications for gallstone pathogenesis in man. Science 1984; 225:514–516. 21. Busch N, Matiuck N. Sahlin S, Pillay SP, Holzbach RT. Inhibition and promotion of cholesterol crystallization by protein fractions from normal human gallbladder bile. J Lipid Res 1991; 32:695–702. 22. Sewell RB, Mao SJT, Kawamoto T, LaRusso NF. Apolipoproteins of high, low, and very low density lipoproteins in human bile. J Lipid Res 1983: 24:391–401. 23. Ohya T, Schwarzendrube J, Busch N, Gresky S, Chandler K, Takabayashi A, Igimi H, Egami K, Holzbach RT. Isolation of a human biliary glycoprotein inhibitor of cholesterol crystallization. Gastroenterology 1993; 104:527–538. 24. Secknus R, Yamashita G, Corradini SG, Chernosky A, Williams C, Hays L, Secknus MA, Holzbach RT. Purification and characterization of a novel human 15 kd cholesterol crystallization inhibitor protein in bile. J Lab Clin Med 1996; 127:169–178. 25. Busch N, Lammert F, Marschall HU, Matern S. A new subgroup of lectinbound biliary proteins binds to cholesterol crystals, modifies crystal morphology, and inhibits cholesterol crystallization. J Clin Invest 1995; 96:3009–3015. 26. Busch N, Lammert F, Matern S. Biliary secretory immunoglobulin a is a major constituent of the new group of cholesterol crystalbinding proteins. Gastroenterology 1998; 115:129–138. 27. Upadhya CA, Harvey PRC, Strasberg SM. Effect of human biliary immunoglobulins in the nucleation of cholesterol. J Biol Chem 1993; 268:5193–5200. 28. Ahmed HA, Petroni ML, AbuHamdiyyah M, Jazrawi RP, Northfield TC. Hydrophobic/hydrophilic balance of proteins: a major determinant of cholesterol crystal formation in model bile. J Lipid Res 1994; 35:211–219. 29. Groen AK, Stout JPJ, Drapers JAG, Hoek FJ, Grijm R, Tytgat GNJ. Cholesterol nucleationinfluencing activity in Ttube bile. Hepatology 1988; 8:347–352. 30. Levy PF, Smith BF, Lamont JT. Human gallbladder mucin accelerates nucleation of cholesterol in artificial bile. Gastroenterology 1984; 87:270–275. 31. Kinkspoor JH, Van Wijland MJA, Koeleman CAM, Van Dijk W, Tytgat GNJ, Groen AK. Heterogeneity of human biliary mucin: functional implications. Clin Sci 1994; 86:75–82. 32. Harvey PRC, Rupar CA, Gallinger S, Petrunka CN, Strasberg SM. Quantitative and qualitative comparison of gallbladder mucus glycoprotein from patients with and without gallstones. Gut 1986; 27:374–381.
Page 247
33. Miquel JF, Groen AK, Vanwijland MJA, Delpozo R, Eder MI, Vonritter C. Quantification of mucin in human gallbladder bile: a fast, specific, and reproducible method. J Lipid Res 1995; 36:2450–2458. 34. Harvey PRC, Toth JL Sr. Lectin binding characteristics of a cholesterol nucleation promoting protein. Clin Chim Acta 1989; 185:185–189. 35. Pattinson NR, Willis KE. Nucleation of cholesterol crystals from native bile and the effect of protein hydrolysis. J Lipid Res 1991; 32:215–221. 36. Groen AK, Noordam C, Drapers JA, Egbers P, Jansen PLM, Tytgat GN. Isolation of a potent cholesterol nucleationpromoting activity from human gallbladder bile: role in the pathogenesis of gallstone disease. Hepatology 1990; 11:525–533. 37. Offner GD, Gong DH, Afdhal NH. Identification of a 130kilodalton human biliary concanavalin A binding protein as aminopeptidase N. Gastroenterology 1994; 106:755–762. 38. Zijlstra AIM, Offner GD, Afdhal NH, Vanoverveld M, Tytgat GNJ, Groen AK. The pronase resistance of cholesterolnucleating glycoproteins in human bile. Gastroenterology 1996; 110:1926–1935. 39. Rigotti A, Nunez L, Amigo L, Puglielli L, Garrido J, Santos M, Gonzalez S, Mingrone G, Greco A, Nervi F. Biliary lipid secretion: immunolocalization and identification of a protein associated with lamellar cholesterol carriers in supersaturated rat and human bile. J Lipid Res 1993; 34:1883–1894. 40. Nunez L, Amigo L, Mingrone G, Rigotti A, Puglielli L, Raddatz A, Pimentel F, Greco AV, Gonzalez S, Garrido J, Miquel JF, Nervi F. Biliary aminopeptidaseN and the cholesterol crystallization defect in cholelithiasis. Gut 1995; 37:422–426. 41. Chijiiwa K, Koga A, Yamasaki T, Shimada K, Noshiro H, Nakayama F. Fibronectin: a possible factor promoting cholesterol monohydrate crystallization in bile. Biochim Biophys Acta 1991; 1086:44–48. 42. Miquel JF, Vonritter C, Delpozo R, Lange V, Jungst D, Paumgartner G. Fibronectin in human gallbladder bile: cholesterol pronucleating and/or mucin ''link" protein? Am J Physiol 1994; 267:G393–G400. 43. Harvey PRC, Upadhya GA, Strasberg SM. Immunoglobulins as nucleating proteins in the gallbladder bile of patients with cholesterol gallstones. J Biol Chem 1991; 266:13996–14003. 44. Abei M, Schwarzendrube J, Nuutinen H, Broughan TA, Kawczak P, Williams C, Holzbach RT. Cholesterol crystallizationpromoters in human bile: comparative protencies of immunoglobulins, alpha 1acid glycoprotein, phospholipase C, and aminopeptidase N1. J Lipid Res 1993; 34:1141–1148. 45. Hajri T, Elliottbryant R, Sipe JD, Liang JS, Hayes KC, Cathcart ES. The acute phase response in apolipoprotein a1 knockout mice—apolipoprotein serum amyloid a and lipid distribution in plasma high density lipoproteins. Biochim Biophys Acta Lipids Lipid Metab 1998; 1394:209–218. 46. Thompson RP, Hofmann AF. Separation of bilirubin and its conjugates by thin layer chromatography. Clin Chim Acta 1971; 35:517–519. 47. Abei M, Nuutinen H, Kawczak P, Schwarzendrube J, Pillay SP, Holzbach RT. Identification of human biliary alpha(1)acid glycoprotein as a cholesterol crystallization promoter. Gastroenterology 1994; 106:231–238. 48. Nuutinen H, Abei M, Schwarzendrube J, Corradini SG, Walsh M, Kawczak P, Holzbach RT. Biliary alpha(1)acid glycoprotein concentrations in gallstonefree controls and in patients with multiple or solitary cholesterol gallstones. Dig Dis Sci 1995; 40:1786–1791. 49. Pattinson NR, Willis KE. Effect of phospholipase C on cholesterol solubilization in model bile: a concanavalin Abinding nucleationpromoting factor from human gallbladder bile. Gastroenterology 1991; 101:1339–1344. 50. Pattinson NR, Willis KE. Phospholipase C and diacylglycerol lipase in human gallbladder and hepatic bile. Gastroenterology 1990; 99:1798–1806. 51. Pattinson NR. Identification of a phosphatidylcholine active phospholipase C in human
Page 248
gallbladder bile [published erratum appears in Biochem Biophys Res Commun 1988 Mar 15;151(2):955]. Biochem Biophys Res Commun 1988; 150:890–896. 52. De Bruijn MAC, Mok KS, Nibbering CP, Out T, Vanmarle J, Stellaard F, Tytgat GNJ, Groen AK. Characterization of the cholesterol crystallizationpromoting lowdensity particle isolated from human bile. Gastroenterology 1996; 110:1936–1944. 53. De Bruijn MAC. Crystallization Promoting Factors in Cholesterol Gallstone Formation. Utrecht, Holland: Elinkwijk, 1994. 54. Harvey PRC, Upadhya GA, Strasberg SM. Cholesterol microcrystals associated with concanavalin abinding glycoproteins contribute artifactually to nucleating activity assays. J Lipid Res 1995; 36:2661–2669. 55. Juvonen T, Kervinen K, Kairaluoma MI, Lajunen LH, Kesaniemi YA. Gallstone cholesterol content is related to apolipoprotein e polymorphism. Gastroenterology 1993; 104:1806–1813. 56. Keulemans YCA, Mok KS, Slors JFM, Gouma DJ, Tytgat GNJ, Groen AK. The biliary concanavalin Abinding fraction explains increased cholesterol crystallization in bile from patients with Crohn's disease compared to patients with ulcerative colitis (abstr). Gastroenterology 1995; 110:A461. 57. Mella JG, Miquel JF, Rollan A, Covarrubias C, Nervi F. ApolipoproteinE polymorphism in patients with cholesterol gallstones in a high risk population (abstr). Gastroenterology 1996; 112:A1332. 58. Vanerpecum KJ, VanBergeHenegouwen GP, Eckhardt ERM, Portincasa P, Vandeihjning BJM, Dallingathie GM, Groen AK, Cholesterol crystallization in human gallbladder bile—relation to gallstone number, bile composition, and apolipoprotein e4 isoform. Hepatology 1998; 27:1508–1516. 59. Keulemans YCA, Mok KS, Gouma DJ, Groen AK. The role of the concanavalin Abinding fraction in cholesterol crystallization in native human bile. J Hepatol 1997; 27:1041–1050. 60. Halpern Z, Dudley, MA, Lynn MP, Nader JM, Breuer AC, Holzbach RT. Vesicle aggregation in model systems of supersaturated bile: relation to crystal nucleation and lipid composition of the vesicular phase. J Lipid Res 1986: 27:295–306. 61. Lee SP, Park HZ, Madani H, Kaler EW. Partial characterization of a nonmicellar system of cholesterol solubilization in bile. Am J Physiol 1987; 252:G374– G384. 62. Harvey PRC, Somjen G, Lichtenberg MS, Petrunka C, Gilat T, Strasberg SM. Nucleation of cholesterol from vesicles isolated from bile of patients with and without cholesterol gallstones. Biochim Biophys Acta 1987; 921:198–204. 63. Peled Y, Halpern Z, Baruch R, Goldman G, Gilat T. Cholesterol nucleation from its carriers in human bile. Hepatology 1988; 8:914–918. 64. Ahrendt SA, FoxTalbot MK, Kaufman HS, Lillemoe KD, Lipsett PA, Pitt HA. Characterization of a small vesicular cholesterol carrier in human gallbladder bile. Ann Surg 1994; 220:635–643. 65. Harvey PRC, Somjen G, Gilat T, Gallinger S, Strasberg SM. Vesicular cholesterol in bile: relationship to protein concentration and nucleation time. Biochim Biophys Acta 1988; 958:10–18. 66. Yamashita Y, Tazuma S, Kajiyama G. Method for quantitative assessment of transformation of nonmicellar cholesterol carriers in model bile systems. J Gastroenterol Hepatol 1996; 11:864–869. 67. Tao S, Tazuma S, Kajiyama G. Fatty acid composition of lecithin is a key factor in bile metastability in supersaturated model bile systems. Biochim Biophys Acta 1993; 1167:142–146. 68. Yamashita G, Secknus R, Chernosky A, Krivacic KA, Holzbach RT. Comparison of haptoglobin and apolipoprotein AI on biliary lipid particles involved in cholesterol crystallization. J Gastroenterol Hepatol 1996; 11:738–745. 69. Kaplun A, Talmon Y, Konikoff RM, Rubin M, Eitan A, Tadmor M, Lichtenberg D. Direct
Page 249
visualization of lipid aggregates in native human bile by light and cryotransmission electronmicroscopy. FEBS Lett 1994; 340:78–82. 70. Afdhal NH, Niu N, Nunes DP, Bansil R, Cao XX, Gantz D, Small DM, Offner GD. Mucinvesicle interactions in model bile: evidence for vesicle aggregation and fusion before cholesterol crystal formation. Hepatology 1995; 22:856–865. 71. De Bruijn MAC, Goldhoorn BG, Zijlstra AIM, Tytgat GNJ, Groen AK. Interaction of cholesterolcrystallizationpromoting proteins with vesicles. Biochem J 1995; 305:93–96. 72. Yamashita G, Corradini SG, Secknus R, Takabayashi A, Williams C, Hays L, Chernosky AL, Holzbach RT. Biliary haptoglobin, a potent promoter of cholesterol crystallization at physiological concentrations. J Lipid Res 1995; 36:1325–1333. 73. Harvey PR, Strasberg SM. Will the real cholesterolnucleating and antinucleating proteins please stand up? (editorial; comment). Gastroenterology 1993; 104:646–650. 74. Cohen S, Kapplan M, Gottlieb L, Patterson J. Liver disease and gallstones in regional enteritis. Gastroenterology 1971; 60:237–245. 75. Lee SP, Nicholls JF. Nature and composition of biliary sludge. Gastroenterology 1986; 90:677–686. 76. Broomfield PH, Chopra R, Sheinbaum RV, Bonorris GG, Silverman A, Schoenfield LJ, Marks JW. Effects of ursodeoxycholic acid and aspirin on the formation of lithogenic bile and gallstones during loss of weight. N Engl J Med 1988; 319:1567–1572. 77. Ostrow JD. APF/CBP, an anionic polypeptide in bile and gallstones that may regulate calcium salt and cholesterol precipitation from bile (editorial). Hepatology 1992; 16:1493–1496. 78. Domingo N, Lafont H, Halpern Z, Pele Y, Grosclaude J, Gilat T. Anionic polypeptide fraction in bile of patients with and without gallstones. Hepatology 1993; 17:778–780. 79. Afdhal NH, Niu N, Gantz D, Small DM, Smith BE Bovine gallbladder mucin accelerates cholesterol monohydrate crystal growth in model bile. Gastroenterology 1993; 104:1516–1523. 80. Pattinson NR. The possible role of phospholipase in gallstone pathogenesis; reply. Gastroenterology 1991; 101:593.
Page 251
12— Normal Gallbladder Motor Functions R.P. Jazrawi St. George's Hospital Medical School, London, England I— Introduction Gallbladder (GB) motor functions play a vital role in the maintenance of an intact enterohepatic circulation in health, and it is evident that abnormalities of GB motor functions have serious clinical implications. Despite the significant improvement in the techniques used for measuring GB motor functions, the old concept that the GB empties gradually after meals and fills between meals is still held by many. The goals of this chapter are to give a brief overview of the physiology of GB motor functions, to describe the techniques used in measurement and discuss the information provided by these techniques, to highlight controversies in the available data, and to present a new concept of GB motor functions in health and its role in disease. II— The Role of the Gallbladder in Health: A Historical Review The representative eighteenthcentury view of GB function may be summarized as follows: "it receives the bile when the stomach, being empty, has no call for it, that afterwards it may be able to return it in greater plenty, when we principally want it for the digestion of aliments now flowing in great quantity into the duodenum" (1). This view, which is strikingly similar to our current understanding of GB function, was further elaborated in the nineteenth century by physiologists like Foster, who said that "The act of secretion of bile from the liver must not be confounded with the discharge of bile from the bile duct into the duodenum. When the acid contents of the stomach are poured over the orifice of the biliary duct, a gush of bile takes place. . . . The discharge is due to a contraction of the muscular walls of the GB and the ducts, accompanied by a relaxation of the sphincter of the orifice; both acts are probably of a reflex nature . . ."(2). Boyden, in a series of studies on animals and humans (3–8), established that GB emptying has both hormonal [cholecystokinin(CCK)mediated] and neural (vagus mediated) components and that during GB emptying, the GB contracts and the sphincter of Oddi relaxes. He also established sex and age differences in the emptying of the human GB. III— Control of Gallbladder Motility GB motility parameters are controlled by both neural and endocrine mechanisms. A list of the hormones and neurotransmitters that affect GB motility is presented in Table 1. As the evolution
Page 252 Table 1 Hormones and Neurotransmitters That Affect Gallbladder Motility Compound
GB motility
Hormones Cholecystokinin
Up
Motilin
Up
Gastrin
Up Up
Cerulin Secretin
Glucagon
Down
Somatostatin
Down
a
Neurotransmitters
Vasoactive intestinal peptide Down Pancreatic polypeptide
Down
Peptide YY
Down
Neuropeptide Y
Up
Substance P
Up
Gastrinreleasing peptide
Up
Calcitonin
Down
Neurotensin
Down
Histamine
Up (H1), down (H2)b
Opiates
Down
a
Potentiates effect of CCK but has no effect by itself on GB motility. b H1 and H2 are histamine receptors 1 and 2.
of our understanding of these control mechanisms progresses, the distinction between them becomes less defined, as it is now established that several peptides function as hormones or neurotransmitters under different circumstances. Even CCK, the main hormone promoting GB contraction, has been identified in nerve fibers within the GB wall and has been proposed as a parasympathetic neurotransmitter. A— Neural Control The GB is innervated extrinsically by parasympathetic and sympathetic nerves and intrinsically by an intramural plexus involving different cholinergic, catecholinergic, serotonergic, and peptinergic neurones. The central nervous system may have a modulating effect on the enteric neural circuits that influence the digestive state. Thus GB motility in humans is under the influence of cephalic regulation, as demonstrated by GB contraction in response to sham feeding (9,10) and by the fact that this emptying is not accompanied by a concomitant increase in circulating cholecystokinin (9). It has been suggested that direct vagal stimulation is involved in the GB response to sham feeding. There is also evidence that vagal activity contributes to the normal GB tone. The physiological significance of sympathetic innervation of the GB remains unclear, although in balance the effect of sympathetic stimulation is to inhibit GB motility (11). There is evidence that adrenergic agents produce GB relaxation (12) via a beta adrenoreceptormediated effect (13).
Page 253
B— Hormonal Control The endocrine factors include both intrinsic and extrinsic hormones and/or peptides that act as hormones (neurotransmitters or neuromodulators). A summary of the effects of these compounds on GB motility is shown in Table 1. The intrinsic neurotransmitters include acetylcholine, vasoactive intestinal peptide, nitric oxide, gastrin releasing peptide, and substance P. These are present in the autonomic plexuses within the GB wall. Of the extrinsic hormones, the most important is CCK; however, others like somatostatin, motilin, and pancreatic polypeptide also play an important role in regulating GB motility. CCK is a peptide hormone (33 amino acids), that was first extracted from the small intestinal mucosa and given its name by Ivy and Oldberg (14). Its Cterminal sequence of eight amino acids (CCK octapeptide) was later identified as the active segment (15). Its release from the intestinal mucosa is stimulated by luminal H+, fat, and amino acids (16) and inhibited by pancreatic secretion (especially trypsin) and, in a negativefeedback fashion, by bile salts (17). CCK produces a dose dependent GB contraction in vitro (18,19) and in vivo (20). Its action is mediated by binding to a specific receptor on the smooth muscle cells of the GB (21). Blockade of this receptor by a specific CCK antagonist loxiglumide completely abolishes CCKinduced GB contraction (22). CCK acts directly on the GB smooth muscle and its effect is not significantly altered by cholinergic or adrenergic blockade (13,23). It has also been identified within vagal neurons in the GB intramural plexus and may act as a parasympathetic neurotransmitter (12). Gastrin has some structural similarity to CCK (24) and some cholecystokinetic activity, but at pharmacological rather than physiological doses (19,25). Gastrin releasing peptide immunoreactive nerve fibers and functional receptors have also been demonstrated in the GB wall of the pig, and their stimulation has been shown to cause GB contraction. However, its role in the regulation of GB motility remains unclear. Motilin is a polypeptide produced in the upper gastrointestinal mucosa; it has been shown to cause GB contraction both in vivo (26) and in vitro on isolated smooth muscle cells from the GB (27). Spontaneous fasting GB contractions have been shown to be associated with increased endogenous serum motilin levels (28,29). Secretion is also produced in the upper gastrointestinal mucosa in response to luminal acidic contents. On its own, it has no effect on the GB; but it can protentiate the effects of CCK (30–34). Somatostatin, a 28amino acid peptide released by the intestine in response to a fatty meal, is a potent inhibitor of GB contraction, whether mealstimulated, vagally induced, or CCKinduced (35). Female sex hormones (estrogen and progesterone receptors) have been identified GB (36), but no direct effect of these compounds on the GB has been demonstrated. During pregnancy, increased resting GB volume (37,38) as well as impaired GB emptying in response to food (38,39) have been demonstrated. C— Other Peptides and Neurotransmitters With regard to intrinsic neurotransmitters, acetylcholine plays an important role in controlling interdigestive GB motility (40,41), specifically via muscarinic receptors (42). Vasoactive intestinal peptide (VIP)immunoreactive nerve fibers have been demonstrated in the GB wall of different species (43), and its stimulation causes GB relaxation (44–46). However in none of these studies have the plasma levels of VIP been measured to distinguish between the physiological and pharmacological actions of this peptide. The GB is supplied by three types of nerve fibers: cholinergic, CCKergic and VIPergic. The vagal regulation of GB tone and contraction is, therefore, the net result of the interplay between the stimulatory actions of acetylcholine and CCK and the inhibitory actions of VIP. Nitric oxide acts as a neurotransmitter in nonadrenergic, noncholinergic pathways of the gastrointestinal tract including the GB (47). This compound has been found to inhibit GB tone and contraction in both animal and human studies (48–51). Pancreatic polypeptide, peptide
Page 254
YY, and neuropeptide Y are all 36amino acid peptides with considerable sequence homology. Pancreatic polypeptide and peptide YY both cause relaxation of the GB and may be important in facilitating postprandial Gb refilling (52,53). Neuropeptide Y, unlike the other two, has been shown to promote GB contraction (54). A number of other peptides have known effects on GB motility, but their physiological significance is unclear. Substance P causes GB contraction (55). Calcitonin generelated peptide, pancreastatin, and neurotensin all inhibit CCKstimulated GB contraction or cause GB relaxation in vivo (56–58). Histamine can cause both GB contraction by binding to H1 receptors and GB relaxation mediated by H2 receptors (59). Prostaglandins have also been shown to have potent effects on enhancing GB motility in animals and humans (60–62). These effects are inhibited by opiates and tetradotoxin but not by hexamethonium or atropine (61), suggesting that prostaglandins act on intrinsic noncholinergic, nonadrenergic nerves. Opiates are produced endogenously by neuroendocrine cells of the gut mucosa and myenteric plexus. They have been found to inhibit CCKinduced GB emptying and to increase fasting GB volume (63). Control of GB motility is therefore a complex process involving both the enteric and autonomic nervous systems as well as an interplay between several hormones. The specific role of each system is difficult to investigate because of mutual regulatory mechanisms involving different nervous circuits, neurotransmitters, and hormones. IV— Role of the Gallbladder in the Enterohepatic Circulation of Bile Acids The integrity of the enterohepatic circulation depends on the intact function of two physical pumps (GB and intestinal motility) and two chemical pumps (active bile acid uptake by the terminal ileum and by the liver). During fasting, a large proportion of the bile acid pool collects in the GB and is concentrated there by removal of water and electrolytes; consequently the organic constituents become concentrated by a factor of 5 to 10 (64). The proportion of hepatic bile handled in the GB prior to delivery into the duodenum has been estimated to be 35 to 50% in the baboon (65) and about 75 to 100% in humans (20,66). Besides receiving, storing, and concentrating bile, the function of the GB is to react to the stimulus of eating by contracting and discharging its content into the duodenum. In contracting, the GB acts as as motor driving the enterohepatic circulation (67). When the GB is inert, as is the case in patients with celiac disease, the enterohepatic circulation is said to be sluggish. The distribution of the bile acid pool changes during the day. This phenomenon is illustrated by the diurnal pattern in bile acid concentration in blood and in bile. Increased postprandial bile acid concentration, by comparison with the fasting state, has been reported in humans in portal (68–69) and peripheral blood (68, 70–73). These observations imply a role for the mechanical pumps (GB and intestinal motility) in the diurnal variation of the bile acid pool distribution. Indeed the role of GB emptying in regulating the size of the bile acid pool in humans was elegantly demonstrated by Duane and Hanson (74), who found a significant inverse correlation between GB emptying and the pool sizes of both cholic and chenodeoxycholic acids. They attributed that to variations in fractional turnover rates but not synthesis rates for the two primary bile acids, as only the former but not the latter correlated with GB emptying. Prolonged increase in GB emptying induced by CCK octapeptide injections has also been shown to cause a reduction in the size of the bile acid pool and an increase in fractional turnover rate but no change in the synthesis rate of the two primary bile acids (75). Everson et al. (76) demonstrated that more complete GB emptying and faster intestinal transit increased hepatic secretion and enterohepatic cycling of all biliary lipids and reduced the molar percent of cholesterol in bile. The above studies have provided the rationale that GB motor functions are important for maintaining the integrity of the enterohepatic circulation and that abnormal
Page 255
ities of GB motor functions are instrumental in the pathogenesis of gallstone disease. This is best illustrated by the fact that 50% of patients with acromegaly who are on octreotide therapy develop gallstones (77). V— Measurement of Gallbladder Motor Functions Four techniques have been used for measuring GB motor functions. These are oral cholecystography, ultrasonography, cholescintigraphy, and duodenal perfusion techniques. The advantages and disadvantages of each of these techniques are discussed separately. A— Oral Cholecystography This technique was first applied by Boyden (4,5) to the systematic study of GB motor function; the method he devised for the measurement of GB volume (5) was later improved by DePaula and Silva (78). It was still the principal technique used for measuring GB volume and emptying in the 1960s (79–81). GB volume is usually estimated by direct measurement from oral cholecystogram images. It is no longer used as a research method due to the radiation hazards and pitfalls in the method itself (the volume measurements being based on planimetric geometry and on several theoretical assumptions). The distance of the subject and the xray film from the xray tube can lead to large magnification errors and thus variable errors in volume estimation. This can probably account for the many conflicting results from early studies on GB emptying using oral cholecystography (82). Keeping the distance between xray film and xray tube constant although still produces a constant magnification error, but it can get rid of some of the variability in repeated volume measurements (83). B— Ultrasonography In 1978, the use of ultrasonography in measuring GB emptying was first reported by Ornstein et al. (84). It quickly became the method of choice for measuring GB volume because, unlike cholecystography, it involves no radiation hazards and can therefore be used to carry out repeated measurements (85,86). Conventionally, by using an ultrasound probe situated over the abdomen, the GB is visualized on a screen. A caliper is used to determine the largest longitudinal and transverse diameters. The probe is then positioned at a right angle to its first position and a third diameter is calculated. The three diameters are used to calculate GB volume. GB images can be either filmed using an attached camera or stored on a linked computer for later analysis. Quantitatively, the information provided by both techniques above is similar, as they both measure fluctuations in GB volume—as shown by Everson et al. (87)—but measurements by ultrasonography are more accurate than by cholecystography due to lack of magnification errors. The specific disadvantge of ultrasonography, however, is its dependence on personal operator skills, resulting in high intra and interobserver error (87). Disadvantages inherent to both techniques are as follows: 1. GB volume calculations are geometrydependent, requiring assumptions to be made regarding GB shape. Thus standardizing the position of the subject with regard to the xray tube during cholecystography, as well as the angle of the probe and the timing of the measurement with regard to inspiration or expiration during ultraso nography, are important in minimizing errors due varying GB geometry. 2. Changes in volume without any change in bile flow in and out of the GB may occur because of net secretion or absorption of fluid across the GB wall. 3. If GB emptying and refilling both occur between one observation period and the next, cholescintigraphy and ultrasonography can detect only the net result of both
Page 256
events. For example, if an emptying of 10 mL and refilling of 4 mL both occurred between two observations, cholescintigraphy and ultrasonography will detect an emptying (reduction in GB volume) of 6 mL. 4. The information obtained may differ according to the length of the observation period and to the frequency of observations. For example, measurements made at 20 or 30min intervals may provide a different value for maximum GB emptying from those carried out at 5 to 10min intervals. Similarly, studies lasting 30 min may provide a different picture of GB emptying from those lasting 120 min or longer. 5. Obesity, intraabdominal gas, blocked cystic duct, and thickened GB wall are among many factors that can cause poor GB visualization by either or both techniques and hence may introduce errors in volume calculations. Furthermore, cholecystography is indirect, requiring hepatic metabolism and excretion of contrast material and concentration by the GB. It is necessary, therefore, for one to be prudent in interpreting or comparing results of studies using these techniques. C— Cholescintigraphy In 1966, Englert and Chiu (88) were the first to report that the use of a gammalabeled radioisotope (113mIiopanoic acid), secreted specifically into bile, in combination with external scanning using a scintillation counter, could overcome many of the problems inherent in other methods. In 1975, Harvey et al. (89) described the use of 99mTcHIDA, one in a family of improved hepatobiliary agents, which—due to their high specificity for hepatic uptake and excretion, short half life, and low radiation exposure—rapidly became the most suitable substances for hepatobiliary scintigraphy. The transport of 99mTcHIDA across the hepatocyte follows a carriermediated organic anion pathway, similar to the hepatic handling of bilirubin (90). Furthermore, the use of a gamma camera linked to a computer system has enabled studies to be recorded on tape or disk and then played back to delineate precise regions of interest and exclude adjacent structures (91–93). More recently, a synthetic gammalabeled bile acid analogue, 75SeHCAT, has become available for hepatobiliary scintigraphy. This substance behaves like a natural bile acid with regard to its overall enterohepatic circulation, ileal absorption, and hepatic uptake (94–97). With regard to measuring GB motor functions, the main difference between 99mTcHIDA and 75SeHCAT is that the former is not reabsorbed in the intestine whereas the latter is reabsorbed in the terminal ileum and undergoes enterohepatic recycling (98). Thus, following a bolus injection of both isotopes, the postprandial GB activity measurement of 99mTcHIDA provides an index of GB emptying only, whereas that of 75ScHCAT provides an index of GB emptying plus refilling. The conventional procedure for 99mTcHIDA GB scintigraphy is to inject 99mTcHIDA as an intravenous bolus, then allowing sufficient time for it to be excreted by the liver and for a proportion to get partitioned into the GB. 99mTcHIDA activity over a period of time is then monitored over the GB region of interest, following administration of a stimulus to contract the GB. Immediately prior to GB contraction, the proportion of 99mTcHIDA in the GB out of the total abdominal activity is used to determine fasting GB filling. Following GB contraction, the percentage reduction in fasting GB activity is used to determine absolute GB emptying. It is called absolute because no 99mTcHIDA activity is present in the hepatic bile entering the GB postprandially. 99mTcHIDA can also be administered as a continuous infusion at a constant rate (99). Using this procedure, one can measure a postprandial GB change in activity, which is a net result between that leaving and that entering the GB. This in a way is similar to the information obtained with ultrasonography. The main advantages of cholescintigraphy is that the technique is noninvasive, quantitative, and reproducible and has a low interobserver error (100). Furthermore, 99mTcHIDA is a
Page 257
highenergy, shorthalflife compound, allowing a small dose with negligible radiation hazard to enable accurate observations to be carried out at a high sampling frequency. The main disadvantages of cholescintigraphy are as follows: 1. When it is carried out in the conventional way, the fasting GB volume is not known; thus all measurements are a fractional percentage of an unknown starting volume and are usually expressed as a percent of the starting 99mTcHIDA GB activity. 2. The main assumption in this technique is that when 99mTcHIDA enters the GB with hepatic bile, it mixes homogeneously and immediately with GB contents. Although there is some evidence suggesting no stratification of GB contents in health (101), the same cannot be said in gallstone disease; there is no information on other disease categories. 3. The concentration of 99mTcHIDA activity differs between hepatic bile and GB bile even when a constant hepatic infusion of 99mTcHIDA is used. Indeed, hepatic bile should theoretically contain very little or no 99mTcHIDA activity if a bolus injection is used and sufficient time is allowed for it to clear the liver. Thus, in the context of expulsion of GB contents, a reduction in GB 99mTcHIDA activity of, for example, "20%" at the beginning of the emptying phase (a large amount of emptying accompanied by a small amount of filling), has a different meaning from a "20%" reduction at the end of the emptying phase (a small amount of emptying accompanied by a large amount of filling). 4. Varying the distance between the region of interest and the gamma camera crystal, whether situated anteriorly or posteriorly, can introduce errors in radioactivity counting. Thus, in a large GB, if an anterior gamma camera crystal were used, the same amount of radioactivity would give a higher count if it were situated at the topmost part of the GB than if it were placed at the bottom of the GB (101,102). Ideally, a combination of anterior and posterior recording of activity and calculation of the geometric mean should be carried out (101,103). For continuous imaging, this would require a double gamma camera, which is not always available. D— Duodenal PerfusionTechniques These techniques provide an alternative method of measuring GB motor functions; they are not influenced by GB absorption or secretion and therefore specifically indicate GB storage or emptying. Whether it provides net or absolute of GB storage of emptying depends on the experimental conditions and particularly whether a hepatic bile marker, a GB bile marker, or both are used in addition to a duodenal recovery marker. Conventionally, this technique enables recovery of an endogenous (bilirubin) or exogenous (indocyanine green) hepatic bile marker to be measured by comparison with nonabsorbable recovery markers infused intraduodenally (104,105). Since these hepatic markers are excreted by the liver at a known and constant rate, net GB emptying is measured when the duodenal recovery of the hepatic marker exceeds its hepatic secretion rate; net GB storage is being measured when the duodenal recovery is lower than the hepatic secretion rate (66). The main disadvantages of these techniques are as follows: 1. They are quite invasive, requiring continuous intravenous infusions over long periods of time (12 to 24 h), intraabdominal catheters using xray screening situated at specific parts of the stomach and duodenum and checked radiologically to ensure that they have not changed position. 2. Sampling frequency is low because a representative sample must be collected to ensure that correction for incomplete recovery does not introduce large errors. Errors can also occur due to incomplete aspiration or inadequate mixing of GB contents with gastric and duodenal secretions.
Page 258
3. Aspiration techniques assume a steady state and are unable to differentiate in the duodenal aspirate between duodenal and GB bile unless a dualmarker technique is employed (66). 4. The hepatic marker indocyanine green can itself alter GB motor functions by its choleretic effect (106). Similarly, depletion of the duodenal bile acids during aspiration and reinfusion of part of the aspirate as boluses may affect GB motility through its marked effect on intestinal motility (107,108). VI— Parameters of Gallbladder Motor Functions A— Gallbladder Volume Measurement of GB volume forms the basis for the assessment of GB motor functions by cholecystography and ultrasonography. B— Gallbladder Filling and Storage GB filling is determined by the rate of bile secretion from the liver and the resistance to flow through the lower end of the bile duct produced by the sphincter of Oddi. This latter factor is the main regulator of bile flow into the GB or duodenum, although there is some evidence that the cystic duct displays sphincterlike properties (18) and responds to pharmacological and hormonal stimuli (109). Bile that enter the GB is concentrated by transport of solute and water across the GB mucosa. The amount (or mass) of compound stored in the GB is the product of the volume of bile within the GB and the concentration of the compound. Thus, GB storage of a compound is influenced by the GB motor functions that regulate GB volume and its concentration capacity. In this chapter, the term GB filling refers to the shortterm partitioning of bile or its constituents in into the GB, whereas GB storage refers to the longterm (overnight) storage capacity of a compound that has been affected by the concentration process in the GB. In vivo, GB filling is measured by selecting a test compound and determining the amount of the compound that partitions into the GB. Filling can be quantitatively expressed as percent filling or filling rate. These have been conventionally measured by shortterm (60 to 90min) cholescintigraphic techniques. Using these techniques, filling rate (i.e., the rate of 99mTcHIDA accumulation in the GB expressed as a percent of the maximum activity of the isotope in the GB on completion of the filling phase) has been reported to average 2 to 2.5%/min (20). Percent filling (usually taken as the GB activity expressed as a percent of the total abdominal activity of 99mTcHIDA) has been reported to average 40 to 70% (20,101,110). The variations can in part be accounted for by methodological differences. Shaffer et al. (20) and van der Werf et al. (110) used one (anterior) gamma camera scanning head only for assessing 99mTcHIDA activity, whereas Jazrawi et al. (101) used two scanning heads (anterior and posterior) and found, with this latter technique, that the error in radioactivity counting due to variable distance of the GB from the scanning head is about 8%, compared with an error of up to 300% when one scanning head is used. GB refilling following administration of a solid meal has been reported by Lawson et al. (111) to start at about 250 min after the meal and to be 70% complete at about 340 min when ultrasonography is used to assess changes in GB volume. The longterm storage function of the GB has been measured by using duodenal perfusion techniques (104,105) and reported to be 50 to 58% of the bile acid pool. Another technique to assess longterm GB storage is to measure the mass of bile acids within the GB and express that as a proportion of the total size of the bile acid pool (102,112). Such studies have yielded similar values to those obtained by perfusion studies. It therefore appears that shortterm GB filling studies, although hardly representative of GB storage function during a physiological period of fasting, agree with longterm studies in indicating that
Page 259
the GB on average stores about 50% of hepatic bile during the interdigestive period. Animal studies carried out in animals (65,113) also agree with this quantitative estimate of GB storage function. C— Gallbladder Emptying All the different techniques described above have been used to assess GB emptying. With cholescintigraphy and ultrasonography, GB volumes are determined in the fasting state and at different intervals following stimulation of GB contraction. The reduction in poststimulus GB volumes, by comparison with fasting GB volume, has been used to describe different GB emptying parameters as follows: 1. Lag time, which is the time between application of stimuli and the start of GB emptying. 2. Maximum emptying, which is the difference between the smallest poststimulus and fasting GB volumes. Results have been expressed in terms of milliliters or in percentage terms, whereby fasting GB volume is considered as 100%. 3. Ejection fraction, which is calculated at different time periods from the fractional reduction in fasting GB volume at the time of observation. It is expressed either in percentage terms considering fasting GB volume as 100%, or in milliliters (ejection volume). This calculation has been the most popular, because several measurements can be made at different observation times and plotted against time to demonstrate an emptying pattern. 4. Emptying rate, which is calculated by dividing the ejection fractions or volumes by the time period between start of stimulus and observation. Results can again be expressed in terms of percent per unit time or milliliters per unit time. Using cholescintigraphy, the GB volume cannot be measured. However, all the above kinetic parameters can be calculated exactly as described above, but using GB counts (of the radioisotope) in the fasting and poststimulus states instead of the GB volumes and expressing the results in percentage terms, considering the fasting GB count to be 100%. Continuous scintigraphic imaging has the advantage over ultrasonography that observations can be made at very frequent time intervals (<1 min) and that radioisotope counting by a gamma camera is more sensitive than volume calculation on ultrasound. Duodenal perfusion techniques, when used in the conventional way, can measure emptying rates but not emptying volumes or ejection fractions. With the use of continuous gamma camera imaging, there is now a consensus that the pattern of GB emptying approximates a double exponential function: a rapid fast exponential where about 50% emptying occurs in around 30 min (114,115) followed by a slower exponential. A similar biphasic pattern for GB emptying has been observed using ultrasonography (111). D— Mixing Function of the Gallbladder Mixing of freshly secreted hepatic bile with GB content is supported by the observation that both lipidsoluble contrast media and watersoluble cholescintigraphic agents are evenly distributed within the GB. Although it is conceivable that the turbulence caused by GB emptying may favor mixing of GB contents postprandially, little is known of the mechanism involved in this effect in the fasting state. Shaffer et al. (1978), cited by Howard (116), studied the mechanism of GB uptake of intravenously injected contrast media radiologically. They reported early collection of the contrast media in the infundibulum 15 min after its intravenous injection, followed by its spreading within the GB from the wall toward the fundus, with complete mixing after 2 h. Van der Linden and Kempi (117) studied the kinetics of the mixing process using
Page 260
computerassisted cholescintigraphy. They reported a pattern characterized first by the appearance of activity along the long axis of the GB, with subsequent lateral spreading. They constructed timeactivity curves for 99mTcHIDA activity in the ''proximal" and "distal" parts of the GB. Using this technique, they almost always observed simultaneous increase of activity in both distal and proximal. Jazrawi et al. (101) measured the ratio of 99mTcHIDA activity to bile acid concentration in successive GB bilerich duodenal samples aspirated following GB stimulation. The 99mTcHIDA had been injected as an intravenous bolus 2 h earlier. They found a constant ratio despite large differences in the bile acid concentrations in the successive samples. They argued that the successive samples represented bile from different areas in the GB, and that the constant ratio suggested homogenous mixing of 99mTcHIDA or by implication hepatic bile with the GB contents in healthy subjects. The driving forces involved in the exchange mechanism between different parts of the GB are unknown, but one possibility is that the turbulence created by rapid alternations in filling and emptying in the interdigestive and postprandial. period may play a role (66). Impairment in the filling and emptying functions of the GB could therefore lead to stratification of GB contents, which might contribute to the pathogenesis of gallstone disease. VII— Variability of Gallbladder Motor Functions in Health and Disease In health, there is no established physiological range for any of the kinetic parameters of GB motor functions described above. Furthermore, studies on gallbladder motor function in patients with gallstones have yielded conflicting results. Increased (118–120), normal (121), and impaired gallbladder emptying have all been described (122–129). Several factors are implicated in the wide variability in results obtained in the different studies. These factors fall into three categories: (a) subjectrelated differences, (b) methodological differences, and (c) conceptual differences. Each of these categories is discussed separately. A— SubjectRelated Differences 1— Intraindividual Variation In cholescintigraphic studies in particular, intraindividual variability makes it difficult to discriminate between normal and abnormal GB emptying. In order to overcome this problem, Mackie et al. (130) attempted to, construct a database for GB emptying in asymptomatic subjects to be used as a reference in clinical cholescintigraphy. They. found percent 99mTcHIDA remaining in the GB, 50 min after administration of 300 mL milk had a value of 21 ± 58% (mean ± 2 SD), and the authors stated that the range of responses was too broad for normal value purposes. Similar wide variations in fasting and residual GB volumes and in GB contraction have been observed using ultrasonography in healthy subjects (131). In an attempt to explain the variability between individual subjects, Baxter et al. (114) suggested the possibility of subpopulations of healthy subjects for GB emptying. These authors, by measuring simultaneously gastric emptying and GB emptying using two different gammalabeled isotopes, were able to divide their healthy controls into two groups: a group with GB emptying in relation to eating and a second group with GB emptying unrelated to eating. The former group was also separated into two subgroups with a high and low efficiency in GB emptying in terms of both t1/2 and ejection fraction, respectively. The differences between the groups could not be accounted for by age, sex, or body weight. Similar observations have been reported in gallstone subjects with radiologically functioning GBs (124,132,133). In these latter studies two populations of gallstone patients have been reported: "contractors" and "noncontractors." Although the proposal of a nonnormal distribution of GB emptying in the general population may be attractive, it must be born in mind that results in the studies reported above
Page 261
have been based upon arbitrary separation of the subgroups and a small number of subjects. A true bimodal distribution of GB emptying parameters will need epidemiological studies for confirmation. 2— Effect of Age, Sex, and Obesity Results on comparisons between young and old subjects are conflicting, with reduced, similar, and increased GB motor functions reported (28,91,130,134–136). Similarly, with regard to the effect of sex, some authors have reported reduced GB emptying in males as compared with females, whereas others have found no difference (91,111,130). In a large population studied by ultrasonography (136), the authors found that males had larger GB volumes than females at all ages, but those authors did not compare GB emptying in the two sexes. In females, the ovulatory cycle is thought to influence GB motor functions to only a minor degree (38,39,137). Progesterone, however, can cause impairment in GB motor functions, as has been shown from both in vivo and in vitro studies (137,138). There is some evidence that obesity may influence GB motor functions (139–141) and that obese subjects have larger GB volumes than nonobese subjects (136). The balance of evidence, however, suggests that obesity per se has no effect on GB motor functions (142,143). The increase in gallstone prevalence in obese populations (144–146) is thought to be due to increased hepatic cholesterol synthesis and secretion rather than impaired GB motor functions. B— Methodological Differences 1— Type of Stimulus A wide variety of stimuli have been used in different studies on GB motor functions. These have included hormones (CCK, CCKoctapeptide, motilin, cerulin); prokinetic agents (cisapride, erythromycin, nitric oxide synthase inhibitors); relaxing agents (octreotide, loxiglumide, nitric oxide synthase stimulators) or meals of different caloric values; constituency (solid, liquid, or mixed) or composition (amino acid or fat content). This has to be taken into consideration when comparisons between different studies are made, as it has been demonstrated, for example, that meal composition can affect GB emptying (76,147–149). Liquids empty quickly from the stomach and stimulate immediate CCK release from the upper intestine to induce rapid GB contraction, whereas solids empty more slowly, maintain prolonged CCK release, and induce tonic GB contraction. Gastric emptying, which influences GB contraction, is itself dependent on the physical state of the meal (liquid or solid) as well as on its fat content (148). 2— Dose and Route of Administration A doseresponse effect has been reported for cholecystokinin (91,92) and also for synthetic cholecystokinin analogues (150). It is therefore important to take into consideration whether a physiological or pharmacological dose of a stimulus or whether a low or highcaloricvalue meal is administered. A different pattern of GB emptying has been reported for intravenous infusion by comparison with either intravenous or intramuscular bolus injection of hormones (150). Because meals are given orally, they may cause GB contraction via a cephalic phase (114). 3— Techniques Used for Assessing Gallbladder Motor Functions Difficulties may arise from the differences in methods used for the assessment of GB emptying and for expression of results. Thus time lag measured by continuous 1 minframe cholescintigraphy (20,114) is not comparable with that measured by duodenal recovery of bile marker (74) because of the delay in the recovery of duodenal contents and the longer time intervals between consecutive samples in using the latter method. Results of emptying rates as measured
Page 262
by cholescintigraphy (114,123) are usually expressed in terms of t 1/2 (time for the gammalabeled GB bile marker to half its activity over the GB region of interest) or as time for maximum emptying. These results are not comparable to those obtained by ultrasonography or by cholecystography (5,76). The main reason is that cholescintigraphic techniques are not affected by GB filling and can only detect "absolute" GB emptying, whereas both ultrasonographic and radiological techniques measure changes in GB volume and thus are affected by GB filling; therefore they measure "net" GB emptying (66). The duration of procedures can vary widely, especially with ultrasound studies. Some studies have lasted as little as 30 min, whereas others run to 3 to 4 h. Similarly, the observation frequency has varied between 1/min and 1 to 2/h. 4— Expression of Results Results form previous studies especially on GB emptying have been expressed in a number of ways. Terms such as ejection fraction, ejection rate, ejection volume, % emptying, rate of emptying, emptying volume, time to maximum emptying, time to 50% emptying, slope of emptying, area under the curve for emptying, and so on are among ones used in both ultrasound
Figure 1 The old concept of the behavior of the GB in response to a stimulus, which is in the form continuous emptying over a period of time followed by continuous filling. The new concept of rapidly alternating short periods of emptying and refilling occurring during the whole postprandial period.
Page 263
or cholescintigraphybased studies. This wide variation in the expression of results adds to the difficulties encountered in trying to compare previous studies. C— Conceptual Problems Qualitatively, the response of the GB to food or hormones has been conventionally described as a continuous smooth emptying curve (Fig. 1A). This pattern has been reported by all the different techniques, including cholecystography (151,152), ultrasonography (87,133, 134,153,154), and cholescintigraphy (10,35,93,114,130) as well as intestinal perfusion techniques (74,104,105). All the above techniques had two unifying methodological aspects: First, for the cholecystographic and ultrasonographic studies, time intervals between observations were relatively long (15 min or more), thus they were unable to detect what happens during the 15min intervals. Second, for the cholescintigraphic and perfusion studies, only one marker of GB or hepatic bile was used. Thus only one pattern of GB motor function (emptying or filling) could be observed. In a few studies, when certain modifications to conventional methodology were applied (mainly more frequent measurements), there were clues to a different pattern of postprandial GB response: that of a rapid alternation between emptying and filling (5,155,156). Further evidence of this new pattern of GB motor function has been provided by several animal studies (26,157,158). VIII— Gallbladder Motor Functions: Concepts and Methods The assumption that in humans the GB empties progressively after meals and refills progressively between meals was originally challenged by studies using dual isotope scintigraphy (98), which demonstrated that both emptying and refilling occurred simultaneously during the postprandial period (Fig. 2). This observation was later confirmed by measuring simultaneous postprandial emptying and refilling using a combination of duodenal perfusion and scintigraphy (66). The observation of intermittent postprandial emptying and refilling has also been reported in humans using minutebyminute ultrasonography (120). On the basis of these observations, a new concept of GB response to a stimulus was proposed (Fig. 1B), in which the continuous emptying and refilling phases shown in Fig. 1A are composed of several small, rapidly alternating phases of emptying and refilling. None of the previous studies of gallbladder motor function in health and gallstone disease, however, have been designed to enable assessment of both emptying and refilling, since neither ultrasonography nor scintigraphy used alone is capable of assessing both events simultaneously. In fact, ultrasonography measures changes in gallbladder volume, which are dependent on refilling, whereas scintigraphy measures the reduction in total GB isotope counts, which is independent of refilling (Fig. 3). Jazrawi et al. in 1995 (115) employed both ultrasonography and scintigraphy simultaneously to measure GB emptying in healthy subjects and gallstone patients. This enabled them to quantify for the first time postprandial GB refilling, and to calculate turnover of bile within the GB (being the sum of postprandial GB emptying and refilling). They observed that GB refilling started immediately after a meal, increased slowly during the first 40 min of the postprandial period and faster afterwards. This pattern is not surprising as both emptying and refilling takes place in a rapid alternating fashion through a single channel (cystic duct). It is therefore logical to assume that there is a limit to the overall traffic of bile (input plus output; see Fig. 3) through this channel. Hence, a large output (emptying) during the fast emptying phase would lead to a small input (refilling) and vice versa. The finding in the healthy subjects of GB refilling of 41 mL at 90 min (115) confirms the observation (66) that the majority of hepatic bile enters the GB before reaching the duodenum even in the postprandial period. The large discrepancy between cumulative refilling and residual GB volume suggests that the he
Page 264
Figure 2 Timeactivity curves generated over the GB area for 99mTcHIDAe and 75 SeHCAT using simultaneous dualisotope recording. Following an initial rapid emptying phase, where the activities of both isotopes decrease in parallel, the activity of the absorbable isotope 75SeHCAT starts to increase, whereas that of the nonabsorbable 99mTcHIDA continues to decrease. This suggests that at the latter part of the emptying phase, both emptying and filling of the GB were occurring in the same time.
patic bile that enters the gallbladder in the postprandial period leaves quickly, thus confirming the "washout effect" (120) postulated in a previous study. Jazrawi et al. (115) also reported that the cumulative turnover of bile in the GB (the volume of bile handled by the GB in the postprandial period) was more than five times the fasting GB volume in health, suggesting that the GB is a dynamic organ With complex motor functions and not a mere storage sac for bile that empties postprandially. Lanzini et al. (66), have demonstrated that rapid alternations in GB filling and emptying occur not only postprandially but also during fasting. They suggested that these rapid alternations contribute to mixing of GB contents. The results of Jazrawi et al. (115) confirmed that these rapid alternations are indeed associated with high turnover of bile within the GB (i.e.; a "washout effect") postprandially. They are therefore likely to have a similar effect in the fasting state. The same authors also demonstrated that in gallstone patients both GB refilling and turnover were markedly impaired in gallstone patients (115) and that they are useful measures to distinguish between those with and those without gallstone recurrence (159). IX— Prospective Studies A— Techniques Although there has been a vast improvement in the techniques for measuring GB motor functions in the past decades, there is still scope for further improvement. Three dimensional (3D) ultrasonography is not influenced by differences in GB position due to the respiratory cycle,
Page 265
Figure 3 This diagram postulates a bidirectional traffic of bile in the cystic duct during fasting (large filling, small emptying) and the postprandial period (initial large emptying accompanied by small filling, followed by reduced emptying and increased filling). It also shows the different pattern of results obtained by ultrasound and scintigraphy, suggesting that these techniques do not measure the same parameters of GB motor functions.
thus it has the advantage over conventional ultrasonography of more accurately measuring GB volume. Furthermore, it should, in theory, allow more frequent measurements to be carried out because it is not restricted by the respiratory cycle. If the 3D probe can be secured to the abdominal wall, 24h monitoring of fluctuations in GB volume should become possible. With regard to scintigraphy, longhalflife gammalabeled isotopes should enable the retention of GB contents to be measured. In the context of gallstone formation, it is of less importance to know how much the GB empties than how long what remains in the GB stays there. B— Methods It is now recognized that postprandial GB filling and turnover play an important role in the pathogenesis of primary and recurrent gallstones. Future studies should enable the assessment of these parameters in highrisk patients (Table 2) like the obese; those on weightreducing diets, parenteral nutrition, or female sex hormones; diabetics; and patients with Crohn's disease. It should be possible to resolve some of the controversies in the results obtained with regard to the effect of certain prokinetic agents like motilin agonists (160,161), antikinetic agents like nitric oxide (47–49,51), or octreotide (162,163). Ursodeoxycholic acid (UDCA) has long been established in the nonsurgical treatment of gallstones, but there is still a lot of controversy regarding its effect on GB emptying (164–167). Indeed, most studies have shown that UDCA impairs emptying (166,167). Most of these studies are based on ultrasound, which measures net differences in gallbladder volume and thus an increase in refilling, which is expected with a choleretic agent like UDCA, might be detected as reduced emptying by ultrasound.
Page 266 Table 2 Conditions Associated with Abnormalities in Gallbladder Motor Functions Condition
GB motility
Gallstone disease
Down
Gastrointestinal diseases Crohn's diseasea
Down
b
IBS
Down
Celiac disease
Down
Idiopathic slow transit constipationa
Down
Surgical procedures Biliopancreatic bypass
a
Down
Gastrectomya
Down
Vagotomya
Down
Sphincterotomy
Up
a
Colectomy
Down
Proctocolectomy plus ileal pouch
Up
Other conditions a
Down
Diabetes mellitus a
Down
Cirrhosis
Enteral nutrition
Up a
Parenteral nutrition
Down
Sickle hemoglobinopathy
Down
Pregnancy
Down
a
Down
Obesity
a
Rapid weight loss a
Down
Associated with increased risk of gallstones. Irritable bowel syndrome.
b
X— Summary The GB plays an integral part in the enterohepatic circulation of bile acids, and normal GB motor functions are of vital importance to health. Control of GB motor functions involves a constant interplay between a large number of stimulatory and inhibitory hormone and neurotransmitters. The concept of GB emptying during meals and refilling between meals is not correct. GB response to a stimulus is more complex, involving rapid alternation of emptying and refilling during the postprandial period. Present methodology (ultrasonography or scintigraphy alone) is not capable of evaluating both emptying and refilling in a quantitative manner, and previous studies employing these techniques have yielded large variations in results in healthy persons and conflicting results in gallstone patients. Hence there is a need for improved methodology. Postprandial refilling and turnover of bile are important parameters that need to be assessed in addressing gallbladder motor function and its role in the pathogenesis of cholesterol gallstone disease. References 1. Anatomy and Physiology Lectures, 2nd ed. Edinburgh, Scotland: University of Edinburgh, 1787, p 404 (quoted by Boyden, 1926). 2. Textbook of Physiology, 6th ed. Part II. Book II. 1895 p 495 (quoted by Boyden, 1926).
Page 267
3. Boyden EA. The effect of natural foods on the distention of the gallbladder with a note of the change in pattern of the mucosa as it passes from distension to collapse. Anat Rec 1925; 30:333–363. 4. Boyden EA. A study of the behaviour of the human gallbladder in response to the ingestion of food, together with some observations on the mechanism of expulsion of bile in experimental animals. Anat Rec 1926; 40:201–256. 5. Boyden EA. An analysis of the reaction of the human gallbladder to food. Anat Rec 1928; 40:147–189. 6. Boyden EA, Birch CL. Reaction of gall bladder to stimulation of gastrointestinal tract: 1. Response to substances injected into the duodenum. Am J Physiol 1930; 92:287–300. 7. Boyden EA, Fuller AH. Anatomy and physiology of the gall bladder in children. Am J Dis Child 1934; 48:565–576. 8. Boyden EA, Grantham SA Jr. Evacuation of Gall Bladder in Old Age. Surg Gynecol Obstet 1936; 62:34–42. 9. Hopman WPM, Jansen JBMJ, Rosenbusch G, Lamers CBHW. Cephalic stimulation of gallbladder contraction in humans: role of cholecystokinin and the cholinergic system. Digestion 1987; 38:197–203. 10. Fisher RS, Rock E, Malmud LS. Gallbladder emptying response to sham feeding in humans. Gastroenterology 1986; 90:1854–1857. 11. Tierney S, Pitt HA, Lillemoe KD. Physiology and pathophysiology of gallbladder motility. Surg Clin North Am 1993; 73:1267–1290. 12. Strah KM, Melendez RL, Pappas TN, et al. Interactions of vasoactive intestinal polypeptide and cholecystokinin octapeptide on the control of gallbladder contraction. Surgery 1986; 99:469–473. 13. Amer MS. Studies with cholecystokinin in vitro: III. Mechanism of the effect on the isolated rabbit gallbladder strips. J Pharmacol Exp Ther 1972; 183:527–534. 14. Ivy AC, and Oldberg E. A hormone mechanism for gallbladder contraction and evacuation. Am J Physiol 1928; 86:599–613. 15. Jorpes JE, Mutt V, Toczko K. Further purification of cholecystokinin and pancreozymin. Acta Chem Scand 1964; 18:2408–2410. 16. Thompson JC, Fender HR, Ramus NI, et al. Cholecystokinin metabolism in man and dogs. Ann Surg 1975; 182:496–504. 17. Gomez G, Liuis F, Guo YS, et al. Bile inhibits release of cholecystokinin and neurotensin. Surgery 1986; 100:363–368. 18. Doyle JS, Farrar JT. A sphincteric mechanism in the cystic duct of dogs. Irish J Med Sci 1969; 2:109–113. 19. Yau WM, Maklouf GM, Edward LE, et al. Mode of cholecystokinin and related peptides on gallbladder muscle. Gastroenterology 1973; 65:451–456. 20. Shaffer EA, McOrmond P, Duggan H. Quantitative cholescintigraphy: assessment of gallbladder filling and emptying and duodenogastric reflux. Gastroenterology 1980; 79:899–906. 21. Schjoldager B, Molero X, Miller LJ. Functional and biochemical characterization of the human gallbladder muscularis cholecystokinin receptor. Gastroenterology 1987; 96:1119–1125. 22. Schmidt WE, Creutzfeldt W, Schleser A, et al. Role of CCK in regulation of pancreaticobiliary functions and GI motility in humans: effects of loxiglumide. Am J Physiol 1991; 260:G197–G206. 23. Hedner P. Effect of the Cterminal octapeptide of cholecystokinin on guinea pig ileum and gallbladder in vitro. Acta Physiol Scand 1970; 78:232–235. 24. Dockray GJ. Molecular evolution of gut hormones: application of comparative studies on the regulation of digestion. Gastroenterology 1978; 72:344–358. 25. Nakano M, McCloy RE, Gin AC, et al. Effect of prostaglandins E1, E2, F2 alpha, and pentagastrin on the gallbladder pressure in dogs. Eur J Pharmacol 1975; 30:107–112.
Page 268
26. Takahashi I, Suzuki T, Aizawa I, et al. Comparison of gallbladder contractions induced by motilin and cholecystokinin in dogs. Gastroenterology 1982; 82:419– 424. 27. Yamasaki T, Chijiiwa K, Chijiiwa Y. Direct contractile effect of motilin on isolated smooth muscle cells human gallbladder. J Surg Res 1994; 56:89–93. 28. Peeters TL, Vantrappen G, Janssens J. Bile acid output and the interdigestive migrating complex in normals and in cholecystectomized patients. Gastroenterology 1980; 79:678–681. 29. Vantrappen G, Janssens J, Hellemans J, et al. Motilin and the interdigestive migrating motor complex in man. Dig Dis Sci 1979; 24:497–500. 30. Cameron AJ, Phillips SF, Summerskill WH. Effect of cholecystokinin, gastrin secretin, and glucagon on human gallbladder in vitro. Proc Soc Exp Biol Med 1969; 131:149–153. 31. Jansson R, Svanvik J. Effects of intravenous secretin and cholecystokinin on net water absorption and motility in the cat. Gastroenterology 1977; 72:639–643. 32. Grace PA, Poston GJ, Williamson RCN. Biliary motility. Gut 1990; 31:571–582. 33. Ryan J, Cohen S. Interaction of gastrin I, cholecystokinin on gallbladder smooth muscle. Am J Physiol 1976; 230:553–556. 34. Ryan J, Cohen S. Pressurevolume response to gastrointestinal hormones. Am J Physiol 1976; 230:1461–1465. 35. Fisher RS, Rock E, Levin G, et al. Effects of somatostatin on gallbladder emptying. Gastroenterology 1987; 92:885–890. 36. Messa C, Maselli MA, Cavallini A, Caruso ML, Pezzolla F, Di Leo A. Sex steroid hormone receptors and human gallbladder motility in vitro. Digestion 1990; 46:214–219. 37. Redberg G, Asztely M, Cantor P, Rehfield JR, JarnfeldtSamsioe, Svanvik J. Gastric and gallbladder emptying in relation to the secretion of cholecystokinin after a meal in late pregnancy. Digestion 1989; 42:174–180. 38. Braverman DZ, Johnson ML, Kern F. Effect of pregnancy and contraceptive steroids on gallbladder function. N Engl J Med 1980; 302:362–364. 39. Everson GE, McKinley C, Lawson M, Johnson M, Kern F. Gallbladder function in the human female: effect of the ovulatory cycle, pregnancy, and contraceptive steroids. Gastroenterology 1982; 82:711–719. 40. Ura K, Sarna SK, Condon RE. Antral control of gallbladder cyclic motor activity in the fasting state. Gastroenterology 1992; 102:295–302. 41. Magee DF, Naruse S, Pap A. Vagal control of gallbladder contraction. J Physiol 1984; 355:65–70. 42. Yegen B, Biren T, Onat F, Tankurt E, Gurmen N, Oktay S, Chey WY, Ulusoy NB. Modulation of gallbladder contraction by pirenzepine in humans. Am J Gastroenterol 1995; 90:1489–1494. 43. Sundler F, Alumets J, Hakanson R, Ingemansson S, Fahrenkrug J, Schaffalitzky de Muckadell O. VIP innervation of the gallbladder. Gastroenterology 1977; 72:1375–1377. 44. Jansson R, Steen G, Svanvik J. Effects of intravenous vasoactive intestinal peptide (VIP) on gallbladder function in the cat. Gastroenterology 1978; 75:47–50. 45. Vagne M, Troitskoja V. Effects of secretin, glucagon, and VIP on gallbladder contraction. Digestion 1976; 14:62–67. 46. Ryan JP, Cohen S. Effect of vasoactive intestinal polypeptide on basal and cholecystokinin induced gallbladder pressure. Gastroenterology 1977; 73:870–872. 47. Sanger P, Schneider H, Hanisch E. Nonadrenergic noncholinergic regulation of gallstone containing and gallstone free human gallbladders. Zentralbl Chir 1997; 122:418–424. 48. Mourelle M, Guarner F, Molero X, Moncada S, Malagelada JR. Regulation of gallbladder motility by the argininenitric oxide pathway in guinea pigs. Gut 1993; 34:911–915.
Page 269
49. Salomons H, Keaveny AP, Henihan R, et al. Nitric oxide and gallbladder motility in prairie dogs. Am J Physiol 1997; 272:G770–G778. 50. Pauletzki JG, Xu QW, Shaffer EA. Inhibition of gallbladder emptying decreases Cholesterol saturation in bile in the Richardson ground squirrel. Hepatology 1995; 22:325–331. 51. Konturek JW, Konturek SJ, Pawlik T, Domschke W. Physiological role of nitric oxide in gallbladder emptying in men. Digestion 1997; 58:373–378. 52. Conter R, Roslyn JJ, Muller EL, et al. Effect of pancreatic polypeptide on gallbladder filling. J Surg Res 1985; 38:461–467. 53. Conter RL, Roslyn JJ, Taylor IL. Effect of peptide YY on gallbladder motility. Am J Physiol 1987; 252:736–741. 54. Lillemoe KD, Webb TH, Pitt HA. Neuropeptide Y: a candidate neurotransmitter for biliary motility. J Surg Res 1988; 45:254. 55. Lonovics J, Varro V, Thompson JC. The effect of cholecystokinin and substance P antagonist on cholecystokininsubstance Pstimulated gallbladder contraction (abstr). Gastroenterology 1985; 88:1480. 56. Hashimoto T, Poston GJ, Gomez G, et al. The effect of pancreastatin on cholecystokinin stimulated gallbladder contraction in vivo and in vitro (abstr). Gastroenterology 1988; 94:A176. 57. Hashimoto T, Poston GJ, Greeley GH Jr, et al. CGRP inhibits gallbladder contractility. Surgery 1988; 104:419–423. 58. Walker JP, Khalil T, Wiener J, et al. The role of neurotensin in human gallbladder motility. Ann Surg 1985; 201:678–683. 59. LaMorte WW, Shoetz DJ, Birkett DH, Williams LF. The role of the gallbladder in the pathogenesis of cholesterol gallstones. Gastroenterology 1979; 77:580– 592. 60. Thornell E, Svanvik J, Wood JR. Effects of intraarterial PGE2 on gallbladder fluid transport, motility and hepatic bile flow in the cat. Scand J Gastroenterol 1981; 16:1083–1088. 61. Jivegard L, Svanvik J. The influence of morphine, loperamide and naloxone on gallbladder response to protaglandin E2 in anaesthetized cat. Acta Physiol Scand 1986; 127:275–279. 62. Kotwall CA, Clanachan AS, Baer HP. Effect of prostaglandins on motility of gallbladder removed from patients with gallstones. Arch Surg 1984; 119:709–712. 63. Worobetz LJ, Baker RJ, McCallum JA, et al. The effect of naloxone, morphine, and an enkephalin analogue on cholecystokinin, octapeptidestimulated gallbladder emptying. Am J Gastroenterol 1982; 77:7:506–511. 64. Davenport HW. Physiology of the Digestive Tract, 2nd ed. Chicago: Year Book, 1966. 65. O'Brien JJ, Shaffer EA, Williams LF, Small DM, Lynn J, Wittenberg J. A physiological model to study gallbladder function in primates. Gastroenterology 1974; 67:119–125. 66. Lanzini A, Jazrawi RP, Northfield TC. Simultaneous, quantitative measurements of absolute gallbladder storage and emptying during fasting and eating in humans. Gastroenterology 1987; 92:852–861. 67. LowBeer TS, Heaton KW, Heaton ST, Read AE. Gallbladder inertia and sluggish enterohepatic circulation of bile salts in coeliac disease. Lancet 1971; 1:991– 994. 68. Lindblad L, Lundholm K, Schersten T. Bile acid concentration in systemic and portal serum in presumably normal man and in cholestatic and cirrhotic conditions. Scand J Gastroenterol 1977; 12:395–400. 69. Angelin B, Bjorkhem I, Einarsson K, Ewerth S. Hepatic uptake of the bile acids in man: fasting and postprandial concentrations of individual bile acids in portal venous and systemic blood serum. Clin Invest 1982; 70:724–731. 70. Hepner GW, Deemers LM. Dynamics of the enterohepatic circulation of the glycine conjugates of cholic, chenodeoxycholic, deoxycholic and sulfolithocholic acid in man. Gastroenterology 1977; 72:499–501.
Page 270
71. Roda E, Aldini A, Mazzella G, Roda A, Sama C, Festi D, Barbara L. Enterohepatic circulation of bile acids after cholecystectomy. Gut 1978; 19:640–649. 72. Pennington CR, Ross PE, Bouchier IAD. Influence of the gallbladder on serum bile acids. J Clin Pathol 1982; 35:754–756. 73. Setchell KDR, Lawson AM, Blackstock RJ, Murphy GM. Diurnal changes in serum unconjugated bile acids in normal man. Gut 1982; 23:637–642. 74. Duane WC, Hanson KC. Role of gallbladder emptying and small bowel transit in regulation of bile acid pool size in man. J Lab Clin Med 1978; 92:858–872. 75. Jazrawi RP, Northfield TC. Effects of a pharmacological dose of cholecystokinin on bile acid kinetics and biliary cholesterol saturation in man. Gut 1986; 27:355– 362. 76. Everson TG, Lawson MJ, McKinley C, Showalter R, Kern F Jr. Gallbladder and small intestinal regulation of biliary lipid secretion during intraduodenal infusion of standard stimuli. J Clin Invest 1983; 71:596–603. 77. Hussaini SH, Pereira SP, Veysey MJ, et al. Roles of gallbladder emptying and intestinal transit in the pathogenesis of octreotide induced gallbladder stones. Gut 1996; 38:775–783. 78. DePaula E, Silva GS. A simple method for computing the volume of human gallbladder. Radiology 1949; 52:94–102. 79. Glanville JN, Duthie HL. Contraction of the gallbladder before and after total abdominal vagotomy. Clin Radiol 1964; 15:350–354. 80. Rudrick J, Hutchinson JSF. Effects of vagal nerve section on the biliary system. Lancet 1964; 1:579–581. 81. Ingberg MV, Vuorio M. Human gallbladder function after selective gastric and total abdominal vagotomy. Acta Chir Scand 1969; 135:625–633. 82. Baxter JN. Gallbladder emptying. Gastroenterol Hepatol 1989; 4:353–372. 83. Jazrawi RP, Pigozzi G, Lanzini A, Galatola G, Kupfer RM, Northfield TC. Optimum bile acid therapy for rapid gallstone dissolution. Gut 1992; 33:381–386. 84. Ornstein MH, Palframan A, Meire H. Real time ultrasound—a new method for investigating gallbladder dynamics (abstr). Gut 1978; 19:971. 85. Lanzini A, Northfield TC. Gallbladder motor function in man. In: Northfield T, Jazrawi R, ZentlerMunro P, eds. Bile Acids in Health and Disease. Dordrecht: Kluwer, 1988, pp 83–86. 86. Dodds W, Groh W, Darweesh R, Lawson T, Kishk S, Kern M. Sonographic measurement of gallbladder volume. Am J Roentgenol 1985; 145:1009–1011. 87. Everson GT, Braverman DZ, Johnson L, Kern F Jr. A critical evaluation of realtime ultrasonography for the study of gallbladder volume and contraction. Gastroenterology 1980; 79:40–46. 88. Englert E, Chiu VSW. Quantitative analysis of human biliary evacuation with a radioisotope technique. Gastroenterology 1966; 50:506–518. 89. Harvey E, Loberg M, Cooper M. Tc99mHIDA: A new radiopharmaceutical for hepatobiliary imaging. J Nucl Med 1975; 16:533–538. 90. Harvey E, Loberg M, Ryan J, Sikorski S, Faith W, Cooper M. Hepatic clearance mechanism of Tc99mHIDA and its effect on quantitation of hepatobiliary function. J Nucl Med 1979; 20:310–313. 91. Spellman SJ, Shaffer EA, Rosenthal L. Gallbladder emptying in response to cholecystokinin: a cholescintigraphic study. Gastroenterology 1979; 77:115–120. 92. Krishnamurthy GT, Bobba VR, Kingston E, Turner F. Measurement of gallbladder emptying sequentially using a single dose of 99mTc labelled hepatobiliary agent. Gastroenterology 1982; 83:773–776. 93. Fisher RS, Rock E, Malmud LS. Cholinergic effects of gallbladder emptying in humans. Gastroenterology 1985; 98:716–722. 94. Jazrawi RP, Ferraris R, Bridges C, Northfield TC. Kinetics of the synthetic bile acid 75SeHCAT in man: Comparison with 14C taurocholate. Gastroenterology 1988; 95:164–169.
Page 271 75
95. Jazrawi RP, Ferraris R, Northfield TC. SeHCAT: an overview of its physiological properties and clinical value. In: Northfield TC, Jazrawi RP, ZentlerMunro PL, eds. Bile Acids in Health and Disease. Dordrecht: Kluwer Academic Publishers, 1988, pp 277–289. 96. Galatola G, Jazrawi RP, Bridges C, Joseph AEA, Northfield TC. Direct measurement of firstpass ileal clearance of a bile acid in humans. Gastroenterology 1991: 100:1100–1105. 97. Galatola G, Jazrawi RP, Bridges C, Joseph AEA, Northfield TC. Hepatic handling of the synthetic glabeled bile acid (75SeHCAT). Gastroenterology 1988; 94:771–778. 98. Jazrawi RP, Lanzini A, Britten A, Meller ST, Northfield TC. Dynamic of gallbladder function and of the enterohepatic circulation of bile acids studied by a gamma labelled bile acid. Clin Sci 1984; 66:10P. 99. Qvist N. Motor activity of the gallbladder and gastrointestinal tract as determinants of the enterohepatic circulation: A scintigraphic and manometric study. Danish Med Bull 1995; 42:426–440. 100. Krishnamurthy GT, Bobba VR, Kingston E. Radionuclide ejection fraction: a technique for quantitative analysis of motor function of the human gallbladder. Gastroenterology 1981; 80:482–490. 101. Jazrawi RP, Kupfer RM, Bridges C, Northfield TC. Isotopic assessment of gallbladder storage functions in man. Clin Sci 1983; 65:185–191. 102. Jazrawi RP, Bridges C, Joseph AEA, et al. Effects of artificial depletion of the bile acid pool in man. Gut 1986; 27:771–777. 103. OsterJorgensen E, Gerner T, Pedersen SA. The determination of gastric emptying rate. Eur J Surg 1991; 564 (suppl):32–43. 104. Mok HYI, Von Bergman K, Grundy SM. Kinetics of the enterohepatic circulation during fasting: biliary lipid secretion and gallbladder storage. Gastroenterology 1980; 78:1023–1033. 105. Van Berge Henegouwen G, Hofmann AF. Nocturnal gallbladder storage and emptying in gallstone patients and healthy controls. Gastroenterology 1978; 75:879–885. 106. Loeb PM, Berk RN, LoboFrenkel A, Barnhart JL. The biliary and urinary excretion and the choleretic effect of ioglycamide in dogs. Invest Radiol 1976; 11:449–458. 107. Penagini R, Misiewicz JJ, Frost PG. Effect of jejunal infusion of bile acids on small bowel transit and fasting jejunal motility in man. Gut 1988; 29:789–794. 108. Nilsson BI, Svenberg T, Tollstrom T, Hellstrom PM, Samuelson K, Schnell PO. Relationship between interdigestiVe gallbladder emptying, plasma motilin and migrating motor complex in man. Acta Physiol Scand 1990; 139:55–61. 109. Doty JE, Pitt HA, PorterFink V, et al. The effect of intravenous fat and total parenteral nutrition on biliary physiology. J Parenteral Enteral Nutr 1984; 8:263. 110. Van der Weft SDJ, van Berge Henegouwen GP, Palsma DMH, Ruben AT. Motor function of the gallbladder and cholesterol saturation of duodenal bile. Neth J Med 1987; 30:160–171. 111. Lawson M, Everson GT, Klingensmith W, Kern F. Coordination of gastric and gallbladder emptying after ingestion of a regular meal. Gastroenterology 1983; 85:866–870. 112. Jazrawi RP, Galatola G, Lanzini A, Northfield TC. Biliary lipid mass in the gallbladder in health and in cholesterol gallstone disease. Eur J Gastroenterol Hepatol 1993; 5:373–381. 113. Pellegrini CA, Ryan T, Broderick W, Way LW. Gallbladder filling and emptying during cholesterol gallstone formation in the prairie dog: a cholescintigraphic study. Gastroenterology 1986; 90:143–149. 114. Baxter JN, Grime JS, Critchley M, Shields R. Relationship between gastric emptying of solids and gallbladder emptying in normal subjects. Gut 1985; 26:342– 351. 115. Jazrawi RP, Pazzi P, Petroni ML, Prandini N, Paul C, Adam JA, Gullini S, Northfield TC. Postprandial gallbladder motor function: refilling and turnover of bile in health and cholestasis. Gastroenterology 1995; 109:582–591.
Page 272
116. Howard PJ. Motor aspects of the enterohepatic circulation and the role of biliary tract and intestinal motor activity in the enterohepatic circulation of bile acids. M.D. thesis, University of London, 1988. 117. Van der Linden W, Kempi W. Filling of the gallbladder as studied by computer assisted Tc99m HIDA scintigraphy: concise communication. J Nucl Med 1984; 25:292–298. 118. Maudgal DP, Kupfer RM, ZentlerMunro PL, Northfield TC. Postprandial gallbladder emptying in patients with gallstones. BMJ 1980; 280:141–143. 119. Pauletzki J, Cicala M, Holl J, Sauerbruch T, Schafmayer A, Paumgartner G. Correlation between gallbladder fasting volume and postprandial emptying in patients with gallstones and healthy controls. Gut 1993; 34:1443–1447. 120. Howard PJ, Murphy GM, Dowling RH. Gallbladder emptying patterns in response to a normal meal in healthy subjects and patients with gallstones: ultrasound study. Gut 1991; 32:1406–1411. 121. Van Erpecum KJ, Van Berge Henegouwen GP, Stolk MFJ, Hopman WPM, Jansen JBMJ, Lamers CBHW. Effect of ursodeoxycholic acid on gallbladder contraction and cholecystokinin releases in gallstone patients and normal subjects. Gastroenterology 1990; 3:836–842. 122. Krishnamurthy GT, Bobba VR, McConnell D, Mesgarzadeh M, Kingston E. Quantitative biliary dynamics: introduction of a new noninvasive scintigraphic technique. J Nucl Med 1983; 24:217–223. 123. Forgacs IC, Maisey MN, Murphy GM, Dowling RH. Influence of gallstones and ursodeoxycholic acid therapy on gallbladder emptying. Gastroenterology 1984; 37:299–307. 124. Pomeranz IS, Shaffer EA. Abnormal gallbladder emptying in a subgroup of patients with gallstones. Gastroenterology 1985; 88:787–791. 125. Sylvestrowics TA, Shaffer EA. Gallbladder function during gallstone dissolution: effect of bile acid therapy in patients with gallstones. Gastroenterology 1988; 95:740–748. 126. Masclee AAM, Jansen JBMJ, Driessen WMM, Geuskens LM, Lamers CBHW. Plasma cholecystokinin and gallbladder responses to intraduodenal fat in gallstone patients. Dig Dis Sci 1989; 34:353–359. 127. Festi D, Frabboni R, Bazzoli F, et al. Gallbladder motility in cholesterol gallstone disease: effect of ursodeoxycholic acid administration and gallstone dissolution. Gastroenterology 1990; 99:1779–1785. 128. Spengler U, Sackmann M, Sauerbruch T, Holl J, Paumgartner G. Gallbladder motility before and after extracorporeal shockwave lithotripsy. Gastroenterology 1989; 96:860–863. 129. Kishk SMA, Darweesh RMA, Dodds WJ, Lawson TL, Stewart ET, Kern MK, et al. Sonographic evaluation of resting gallbladder volume and postprandial emptying in patients with gallstones. Am J Roentgenol 1987; 148:875–880. 130. Mackie CR, Baxter JN, Grime JS, Hulks G, Cushieri A. Gallbladder emptying in normal subjects a data base for clinical cholescintigraphy. Gut 1987; 28:137– 141. 131. Donald JJ, Fache JS, Buckley AR, Burhenne HJ. Gallbladder contractility: variation in normal subjects 1991; 157:753–756. 132. Thompson JC, Gried GM, Ogden WD, Fagan CJ, Inoue K, Wiener I, Watson LC. Correlation between release of cholecystokinin and contraction of the gallbladder in patients with gallstones. Ann Surg 1982; 195:670–675. 133. Upp JR, Nealon WH, Singh P, Fagan AS, Greely GH, Thompson JC. Correlation of cholecystokinin receptors with gallbladder contractility in patients with gallstones. Ann Surg 1987; 205:641–648. 134. Khalil T, Walker JP, Weiner I, Fagan CJ, Townsend CM, Greeley GH, Thompson JC. Effect of ageing on gallbladder contraction and release of cholecystokinin33 in humans. Surgery 1985; 98:423–429. 135. Portis SA, King JC. The gastrointestinal of the gallbladder tract in the aged. JAMA 1952; 148:73–79.
Page 273
136. Palasciano G, Serio G, Portincassa P, et al. Gallbladder volume in adults, and relationship to age, sex, body mass index, and gallstones: a sonographic population study. Am J Gastroenterol 1992; 87:493–497. 137. Shaffer EA, Taylor PJ, Logan K, Gadomski S, Corenblum B. The effect of progestin on gallbladder function in young women. Am J Obstet Gynecol 1984; 148:504–507. 138. Keane P, Colwell D, Baer HP, et al. Effects of age, gender and female sex hormones upon contractility of the human gallbladder in vitro. Surg Gynecol 1986; 163:555–560. 139. Marzio L, Capone F, Neri M, Mezzetti M, De Angelis C, Cuccurullo F. Gallbladder kinetics in obese patients: effect of regular meal and lowcalorie meal. Dig Dis Sci 1988; 33:4–9. 140. Bonfissuto G, Soresi M, Amato S, et al. Ultrasonographic assessment of gallbladder motility in obese subjects. Recent Prog Med 1996; 87:338–341. 141. Gourtsoyiannis NC, Damilakis JE, Charoulakis NZ, et al. Relationship of gallbladder contour, fasting volume and emptying to body size indices in normal subjects and patients with gallstones. Digestion 1995; 56:395–399. 142. Vezina WC, Paradis RL, Grace DM, et al. Increased volume and decreased emptying of the gallbladder in large (morbidly obese, tall muscular normal) people. Gastroenterology 1990; 98:1000–1007. 143. Stone BG, Gaveler JS, Belle SH, et al. Impairment of gallbladder emptying in diabetes mellitus. Gastroenterology 1988; 95:170–176. 144. Barbara L, Sama C, Leabate AMM, et al. A population study on the prevalence of gallstone disease: the Sirmione study. Hepatology 1987; 7:913–917. 145. Bennion LJ, Grundy SM. Risk factors for the development of cholelithiasis in man. N Engl J Med 1978; 299:1161–1167, 1221–1227. 146. Kern F Jr, Everson GT, DeMark B, et al. Biliary lipids, bile acids, and gallbladder function in the human female. J Clin Invest 1981; 68:1229–1242. 147. Go VLW, Hofmann AF, Summerskill WHJ. Pancreozymin bioassay in man based on pancreatic enzyme scretion: potency of specific amino acids and other digestive products. J Clin Invest 1970; 49:1558–1564. 148. Fisher RS, Rock E, Malmud LS. Effects of meal composition on gallbladder and gastric emptying in man. Dig Dis Sci 1987; 32:1337–1344. 149. Fried M, Jansen JB, Harpole T, et al. Pancreatobiliary response to an intragastric amino acid meal: comparison to albumin, dextrose, a maximal cholecystokinin stimulus. Gastroenterology 1989; 97:1544–1549. 150. Sarva RP, Shreiner DP, VanThiel D, Vingvorapant N. Gallbladder function: methods of measuring filling and emptying. J Nucl Med 1985; 26:140–144. 151. Sacchetti G, Mandelli V, Roncoroni R, Motanari C. Influence of age and sex on gallbladder emptying induced by a fatty meal in normal subjects. Am J Roentgenol Radium Ther Nucl Med 1973; 119:40–45. 152. Morewood DJW, Whitehouse GH. Ceruletide cholecystography doseresponse and gallbladder function. Br J Radiol 1984; 57:439. 153. Hansen WE, Maurer H, Vollmar J, Brauning C. Guar gum and bile: effects on postprandial gallbladder contraction and on serum bile acids in man. Hepatogastroenterol 1983; 30:131–133. 154. Hopman WPM, Rosenbusch G, de Long AJL, Lamers CBHW. Gallbladder contraction: effects of fatty meals and cholecystokinin. Radiology 1985; 157:37– 39. 155. Boyden EA, Bergh GS, Layne JA. An analysis of the reaction of the human gallbladder and sphincter of Oddi to magnesium sulphate. Surgery 1943;723–733. 156. Torsoli A, Ramorino ML, Colagrande C, Demaio G. Experiments with cholecystokinin. Acta Radiol 1960; 55:193–205. 157. Traynor OJ, Dozois RR, DiMagno EP. Canine interdigestive and postprandial gallbladder motility and emptying. Am J Physiol 1984; 246:G426–G431. 158. Takahashi I, Kern MK, Dodds WJ, Hogan WJ, Sarna SK, Soergel KH, Itoh Z. Con
Page 274
traction pattern of opossum gallbladder during fasting and after feeding. Am J Physiol 1986; 250:G227–G235. 159. Pazzi P, Petroni ML, Prandini N, Adam JA, Gullini S, Northfield TC, Jazrawi RP. Postprandial refilling and turnover: Specific gallbladder motor function defects in patients with gallstone recurrence. Eur J Gastroenterol Hepatol, 1999. In press. 160. Pilot MA. Macrolides in roles beyond antibiotic therapy. Br J Surg 1994; 81:1423–1429. 161. Wehrmann R, Pfeltzer C, Caspary WF. Effect of erythromycin on human biliary motility. Aliment Pharmacol Ther 1996; 10:421–426. 162. Redfern JS, Fortuner WJ II. Octreotideassociated biliary tract dysfunction and gallstone formation: pathophysiology and management. Am J Gastroenterol 1995; 90:1042–1052. 163. Stolk MF, van Erpecum KJ, Koppeschaar HP, et al. Effect of octreotide on fasting gallbladder emptying, antroduodenal motility, and motilin release in acromegaly. Gut 1995; 36:755–760. 164. Portincassa P, DiCiaula A, Palmieri V, et al. Tauroursodeoxycholic acid, ursodeoxycholic acid and gallbladder motility in gallstone patients and healthy subjects. Ital J Gastroenterol 1996; 28:111–113. 165. Van de Heijning BJM, van de Meeberg PC, Portincassa P, et al. Effects of ursodeoxycholic acid therapy on in vitro gallbladder contractility in patients with cholesterol gallstones. Dig Dis Sci 1999; 44:190–196. 166. Sailer C, Pauletzki J, Klueppelberg UG, et al. Acute effect of ursodeoxycholic acid on gallbladder volume in healthy subjects. Scand J Gastroenterol 1996; 31:512–516. 167. Xu QW, Freedman SM, Shaffer EA. Inhibitory effect of bile salts on gallbladder smooth muscle contractility in the guinea pig in vitro. Gastroenterology 1997; 112:1699–1706.
Page 275
13— Gallbladder Motility and Gallstones Ralph R.S.H. Greaves St. Bartholomew's and the Royal London School of Medicine and Dentistry, London, England Luke J.D. O'Donnell Mayo General Hospital, Castlebar, Ireland I— Physiology of Gallbladder Motility A— Gallbladder Function The function of the gallbladder is to deliver concentrated bile to the duodenum after a meal. This requires a complex coordination of the motor, secretory, and absorptive functions of the gallbladder and gastrointestinal tract, which is orchestrated by a variety of neural, endocrine, paracrine, and autocrine factors. This section describes the physiology of gallbladder motility, with special reference to the human gallbladder. B— Embryology and Structure The gallbladder develops as an evagination of the duodenal mucosa (1–3) and shares similarities in structure and innervation with the small intestine. Gallbladder smooth muscle consists of a interwoven arrangement of longitudinal, transverse, and circular muscle layers (4,5). C— Nerve Innervation—Extrinsic The gallbladder is extrinsically innervated by both parasympathetic and sympathetic pathways. Parasympathetic fibers reach the gallbladder through the hepatic branch of the left vagus nerve (6) and terminate in the ganglia of the lamina propria and the smooth muscle (7). Sympathetic fibers originate from the seventh to tenth thoracic segments, reaching the gallbladder via the celiac ganglia (8) and the right phrenic nerve (2). These fibers innervate smooth muscle and interconnect with nonadrenergic nerve cells. D— Nerve Innervation—Intrinsic Nerve ganglia are present in all three main layers in the gallbladder, comprising the subserosal, the myenteric, and mucosal plexuses (9,10). The network of these intramural plexuses receives input from the parasympathetic and sympathetic nervous systems. Several neurotransmitter groups have been identified within the gallbladder wall, including cholinergic (7), adrenergic (7,8), peptidergic (11), serotonergic (12), enkephalinergic (13) and nitrergic neurotransmitters
Page 276
(14,15). Peptides identified in the plexuses include cholecystokinin (16), vasoactive intenstinal polypeptide (17), and neuropeptide Y (18). Although pharmacological studies demonstrate that these neurotransmitters may affect gallbladder function, their physiological role remains for the most part unclear. E— Normal Pattern of Gallbladder Motility 1— Fasting Gallbladder Motility In the fasting state, the human gallbladder is not at rest. Small contractions and relaxations occur constantly, emptying up to 20% of its contents every hour. Isotope emptying studies suggest frequent alterations in absolute gallbladder storage and emptying, analogous to the action of a bellows (19). 2— Postprandial Gallbladder Motility Under physiological conditions, food is the main stimulus to gallbladder emptying. However, it is clear that gallbladder emptying is more complex than a simple mass expulsion after a luminal trigger (20–22). Approximately 15% of gallbladder emptying occurs before the onset of gastric emptying, and refilling occurs after the expulsion of 85% of the gastric contents (21,23). Recent simultaneous scintigraphic and ultrasonographic studies on healthy subjects show that postprandial gallbladder emptying is a dynamic response, combining simultaneous emptying and refilling. Gallbladder refilling begins immediately postprandially, then accelerates after 40 min (24). In addition, there are superimposed small alternating contractions and relaxations of the gallbladder, likened to bellows movements, seen during fasting (19,24). After a liquid meal containing fat, minimum gallbladder volume is reached in 30 to 45 min, after which refilling starts (20). After a solid meal, it may take up to 4 h for the gallbladder to reach its minimum volume (25). The time taken for the gallbladder to refill after a meal depends on the composition of the meal. Following a liquid meal, the gallbladder may start to refill within 30 min. After a solid meal, however, refilling may not occur for several hours, and only after most of the meal has been emptied from the stomach (21). Two patterns of gallbladder refilling have been described: (1) a steady rate and (2) filling in a stepwise manner (24). F— Regulation of Fasting Gallbladder Motility Gallbladder motility in the fasting state depends on a functioning cholinergic pathway and the hormone cholecystokinin (CCK). Cholinergic blockade with an infusion of atropine distends the fasting gallbladder, implying that cholinergic mechanisms have a role in maintaining fasting tone (26,27). Similarly, CCK antagonists increase fasting gallbladder volume, implying a ''tickover" tonic CCK effect on the resting gallbladder (28–30). The regular, small, bellowslike emptying and refilling movements in the fasting state are related to phase II of the duodenal migrating motor complex (31–34). Retrograde neural tracer studies in the Australian brushtailed possum demonstrate long projections between the duodenum and gallbladder, and it seems likely that these are responsible for the coordination of duodenal and gallbladder motility (35). During fasting, the gut peptide motilin is likely to be responsible for the coordination of gallbladder emptying and the migrating motor complex in the dog (36–38), although in humans this appears not to be the case (39). Motilin has no direct effect on in vitro human gallbladder muscle strips (40). G— Neural Regulation of Postprandial Gallbladder Motility Postprandial gallbladder emptying may be divided into cephalic, gastric, and intestinal phases. Sham feeding may produce emptying of up to 65% of gallbladder contents (41,42). This
Page 277
cephalic phase is predominantly a muscarinic M1mediated response, as it is blocked by pirenzipine, an M1receptor antagonist (43). The gastric phase is dependent on antral distention, triggering a cholinergic vagovagal pylorocholecystic reflex (44). Meals high in polyunsaturated fat are the most powerful stimuli to emptying, although meals containing protein and carbohydrate also stimulate some emptying (8,45,46). The cephalic and gastric phases are predominantly cholinergicmediated and independent of a rise in CCK (47). Cholinergic blockade with atropine reduces the rate of gallbladder emptying after a liquid or solid meal, supporting a cholinergic role in gallbladder emptying (42). In contrast, the intestinal phase of gallbladder emptying is largely CCKdependent (43). The physiological role of the adrenergic nerve system in human gallbladder motility is unclear. Beta agonists relax rabbit and human gallbladder muscle strips (48,49). Splanchnic nerve stimulation or noradrenaline infusion produces gallbladder relaxation in the dog and cat (49,50,52); however, other studies have failed to confirm these findings (53,54). There are no published data on the effect of the adrenergic system in the control of human gallbladder motility in vivo. H— Endocrine Regulation of Postprandial Gallbladder Motility The main hormonal regulator of gallbladder contraction is CCK. CCK is a peptide hormone, synthesized and stored in the duodenal I cells as a large preprohormone, CCK58 (55). It is cleaved to CCK8, CCK33, and an intermediate form (56). The terminal eight amino acid residues, containing a sulfated tyrosyl residue at position 7, are mainly responsible for its biological activity (57). CCK is released into the circulation upon ingestion of a meal. Unsaturated fats, proteins, and amino acids are the most potent nutrients producing CCK release (56,58–60). Specific luminal peptides have a modulating role in controlling the release of CCK. Monitor peptide, a 61amino acid protein homologous to the pancreatic trypsin inhibitor family of proteins (61), is produced by the pancreas and released into the intestinal lumen, where it stimulates CCK release (62). This acts in conjunction with a trypsinsensitive CCKreleasting peptide that is intestinal in origin (63,64). In addition to stimulatory factors, there is also evidence for luminal CCKinhibitory factors in the gut such as trypsin, chymotrypsin, elastase and bile salts (65–70). The receptors for CCK in peripheral tissues can be classified into two subtypes CCKA and CCKB, the latter being identical to the gastrin receptor (71,72). The human gallbladder expresses only CCKA receptors (73,74). The receptor status is unrelated to age, gender, body weight, or pathological state of the gallbladder (73,75). CCK can act directly on gallbladder CCK receptors to cause a dosedependent contraction (76). The mechanism of action of CCK at the cellular level has been extensively studied in the isolated pancreatic acinar preparation (77–79) and more recently the isolated gallbladder smooth muscle cell (80,81). CCKreceptors belong to the G proteincoupled family of receptors and act by generation of intracellular messengers, primarily calcium and diacylglycerol (DAG) (82). Production of these messengers is increased by activation of a phosphoinositidespecific phospholipase C enzyme that cleaves phosphotidylinositol 4,5 biphosphate (PIP2) to produce inositol 1,4,5triphosphate (IP2)and DAG IP2 acts to release calcium from intracellular stores and promote calcium influx, thus increasing intracellutar calcium. Calcium and DAG act by regulating a number of kinases and phosphatases. There are two major concentrationdependent intracellular pathways. CCK activates the protein kinase C pathway at low concentrations, whereas it activates the calmodulin pathway at high concentrations, which, in turn, inhibits the activation of protein kinase C (80). Infusion of CCK into the systemic circulation produces dosedependent gallbladder contraction in humans (83,84). Systemic circulating concentrations of CCK are highly correlated with the rate of human gallbladder emptying (56,85).
Page 278
Specific CCKA receptor antagonists have been shown to inhibit CCK and mealstimulated gallbladder contraction, suggesting that CCK is the major mediator of postprandial gallbladder emptying (28–30,86,87). CCK and vagal cholinergic neurotransmission are intimately related. In animals, CCK acts via cholinergicmediated intrinsic neurons, and it is likely that this is the predominant physiological pathway (16,88–92). In humans, cholinergic blockade with atropine reduces gallbladder emptying in response to exogenous CCK (93– 95). Similarly, vagotomy reduces the release of CCK in response to intraduodenal fat (96). I— Autocrine and Paracrine Factors in the Gallbladder As well as the classic endocrine and neurally mediated pathways of gallbladder motility regulation, there is increasing evidence that several bioactive agents, produced and acting locally, also have dramatic effects on motility. These extracellular signals can be classified as paracrine or autocrine and are described in the following sections. The term paracrine was proposed by Feyter to describe a network of cells distributed throughout the gut and other organs close to nerve endings and blood vessels (97). He postulated that these cells were "peripheral endocrine glands" and speculated that, in addition to their distant endocrine actions, these cells might also have local "paracrine" effects in cell regulation. Thus, in paracrine signaling, the target cell is close to the signaling cell, and release of the mediator produces a local response. Many paracrine systems have been described to date—for instance, the local effect of prostaglandins on the uterus at parturition (98), the action of nitric oxide in a number of physiological systems and pathophysiological states (99), the role of endothelins on the peripheral vasculature (100), and the release of vasoactive intestinal peptide by the feline gallbladder in response to vagal stimulation (101). In 1980 Sporn and Todaro added the concept of autocrine secretion to explain the endogenous production of autostimulatory growth factors by transformed cells. Although it was initially postulated that autocrine action was limited to malignant cells, it became clear that autocrine action is central to normal cell biology (102). In autocrine signaling, therefore, cells respond to substances that they themselves release. The autocrine/paracrine systems described here in relation to the regulation of gallbladder motility, and the pathogenesis of gallstones are the prostanoids, nitric oxide, vasoactive intestinal peptide/pituitary adenylate cyclaseactivating polypeptide (VIP/PACAP), and endothelin. 1— Prostaglandins Prostaglandins are potent biological mediators found in several tissues of many species, with diverse physiological and pathophysiological roles. All major prostaglandin subgroups are synthesized in the gallbladders of guineapig (103–105), prairie dog (105–110), and rabbit (111–113). Gallbladder muscle strips from various animal species contract in response to exogenous prostaglandins in vitro. With the guinea pig gallbladder, the rank order of potency is U46619 > PGE2 > PGF2;a (104,114,115). The cat gallbladder will contract upon addition of PGE1, PGE2, and PGF2a (114). The isolated dog gallbladder will contract on the addition of PGD2, PGE1, PGE2, and PGF2a (116). Several in vivo models demonstrate gallbladder contraction with the addition of exogenous prostaglandins. Intraarterial PGE2, produces contraction of the feline gallbladder (114,117). Intravenous administration of PGE2 and PGF2a produces contraction of the canine gallbladder (118). Inhibition of PG synthesis with nonsteroidal antiinflammatory drugs (NSAIDs) also affects in vivo animal gallbladder motility. Administration of indomethacin to guinea a pigs inhibits gallbladder hypostasis during a highcholesterol lithogenic diet (119). PGE2, PGF2 , and U46619 are potent contractile agents in normal human gallbladder muscle strips, with a rank order of potency of PGE2 > U46619 > PGF2a (120).
Page 279
The synthetic prostaglandin E1 derivative misoprostol has been studied in healthy subjects, in whom it had no effect on fasting gallbladder volume or mealinduced gallbladder emptying (121,122). Both aspirin and indomethacin—potent inhibitors of prostaglandin synthesis—have been investigated in normal subjects (122–124). In these studies, aspirin (124) and indomethacin (122,123) had no significant effect on basal gallbladder volume or gallbladder emptying. Prostaglandins are produced in all species of mammalian gallbladders studied thus far. Exogenous application of prostaglandins can affect gallbladder smooth muscle motility. However, the exact physiological role of prostaglandins in normal gallbladders remains undefined. 2— Nitric Oxide NOScontaining neurons have been demonstrated in both the gallbladder (14,15,125), and sphincter of Oddi (125,126) of several animals. NOproducing neurons have been characterized in the human gallbladder (14,15), both in the fibromuscular layer and in the mucosal layer (127). There is colocalization with the neurotransmitters VIP and PACAP, among others (127). Inhibition of NOS in in vitro gallbladder strips from guinea pigs shows a significant enhancement of the contractile response to CCK (128). In addition, inhibition of NOS in guineapig gallbladder in vivo increases basal gallbladder pressure, indicating that there is a continuous tickover production of NO in the gallbladder which contributes to the maintenance of gallbladder volume in the fasting state (128). Pharmacological blockade of NOS in the cat in vivo has no effect on gallbladder motility (125). In healthy humans, glyceryl trinitrate, acting as an NO donor, produces gallbladder dilatation in the fasting state and reduces postprandial gallbladder emptying, suggesting that nitric oxide mechanisms may be operative in the human gallbladder in vivo (129). Inhibition of NOS in animal sphinctor of Oddi both in vitro and in vivo increases sphincter tone, implying a role for NO in regulating sphincter function (125,126,130,131). The effects of NO on human sphincter of Oddi have not been reported. 3— Vasoactive Intestinal Peptide VIP belongs to a family of structurally related peptides that includes secretin and glucagon (132). VIP is localized to the gallbladder of several species, including humans where it is often colocalized with NOproducing neurons (15,127,133,134). VIP is also highly represented in the sphincter of Oddi of several species (134– 136). Evidence for a paracrine neurotransmitter role for VIP in the gallbladder comes from studies demonstrating that the cat gallbladder in vivo releases VIP at a basal rate, and the release is increased in response to activation of noncholinergic fibers in the vagus nerve (101). VIP produces doserelated decreases in resting tension, spontaneous activity, and contractile response to CCK both with isolated guinea pig gallbladder and gallbladder strips (137–139). VIP intravenous infusion in the guineapig, opossum, cat, and dog in vivo reduces basal tone and CCKstimulated gallbladder contraction (17,135,140–142). The effects of VIP on the human gallbladder both in vitro and in vivo have not been documented. 4— Pituitary Adenylate Cyclase Activating Polypeptide PACAP was isolated first from ovine hypothalamus (143). The primary structure indicates that PACAP is a member of the VIP/glucagon/secretion family of peptides, most homologous to VIP. There are two molecular forms, PACAP27 and PACAP38 (144). The Nterminal 28 residues of PACAP show a 68% homology with VIP (144). Recent immunohistochemical work has demonstrated the existence of PACAPcontaining neurons in the fibromuscular and mucosal layers of the human gallbladder, with colocalization with VIP and NOS (127). PACAP27 and PACAP38 have no effect on the resting tension and CCKstimulated contraction of canine gallbladder muscle strips (142). The effect of PACAP on guinea pig gallbladder smooth muscle strips has not been described.
Page 280
PACAP27 and PACAP38 produce transient and tonic gallbladder contractions in the dog in vivo, both in the digestive and interdigestive states, approximately one tenth as potent as contractions induced by CCK on a molar basis (142). There are no data on the effect of PACAP on human gallbladder either in vitro or in vivo. The effects of VIP and PACAP on the gallbladder are likely to be closely related. Both would appear to be synthesized locally, with paracrine and neuroendocrine effects on gallbladder motility. 5— Endothelins Endothelins are potent vasoconstrictor peptides produced in a variety of tissues, where they act at modulators of vasomotor tone, cell proliferation, and hormone production (100,145). There are no recorded data on the release of endothelins from animal gallbladder; however, human gallbladder epithelial cells in primary culture secrete endothelins, and highperformance liquid chromatography of cultured supernatant shows a single peak of endothelin1 (ET1). Synthesis in cultured human gallbladder epithelial cells is stimulated by physiological concentrations of CCK, and this has an inhibitory effect on gallbladder secretion by an autocrine/paracrine mechanism (146,147). ET1 and 2 can be detected in the normal human gallbladder by radioimmunoassay, and ET2 is likely to be the physiologically significant endothelin isopeptide expressed (148). Both ETA and ETB receptors have been characterized in the guinea pig gallbladder by pharmacological techniques, although additional receptors not conforming to the ETAETB classification may be present (149,150). ETA receptors appear to predominate in the human gallbladder (148). Receptor status in the human gallbladder has not been systematically described. ET1, ET2, and ET3 all produce dosedependent contraction in guinea pig gallbladder muscle strips (149,151). The effect of endothelins on human gallbladder motility has not been described. II— Impaired Gallbladder Motility and Gallstones A— Introduction The most common disorder affecting the gallbladder is gallstone formation. Gallstones occur in about onethird of postmenopausal women (152). Although most are asymptomatic, the natural history suggests that approximately onehalf of subjects with asymptomatic gallstones will eventually require a cholecystectomy (153). Consequently, cholecystectomy is the most common elective surgical procedure in the western world. The three major determinants of human cholesterol gallstone formation are cholesterol supersaturation of bile (154), impaired gallbladder emptying (155), and pro aggregatory nucleating factors in the bile (156). Although Meckel von Hemsbach in 1856 suggested that impaired gallbladder emptying was the primary factor (157), much of the work this century has concentrated on bile composition. However, it has only been possible to accurately measure gallbladder motility in humans since the advent of ultrasonographic screening and isotope scintigraphy in the late 1970s and the 1980s. In animal models of gallstone pathogenesis, a reduction in gallbladder emptying precedes gallstone formation, implying that this is a primary pathogenic factor (158– 161). Moreover, enhanced periodic emptying of the gallbladder by administration of a duodenal lavage with a proteinlipid mixture or exogenous CCK prevents gallstone formation (162,163). Animal in vitro work has shown a reduction in maximal muscle strip contraction in response to spasmogens of greater than 50%, compared to controls, in strips obtained from gallbladders containing gallstones (161,164). The specific defect in muscle contraction is elusive, as membrane excitation and excitationcontraction coupling are not impaired and total contractile protein is unchanged (165). However, some workers have failed to demonstrate the decrease in muscle contraction in this model (166).
Page 281
B— Human Studies of Gallbladder Dysmotility and Gallstone Pathogenesis The majority of studies using reliable methodologies demonstrate that at least a subset of patients with gallstones have impaired gallbladder motility. Impaired gallbladder emptying after a fatty meal has been demonstrated in patients with gallstones by both isotope and ultrasound scanning techniques (20,24,167–172). Gallbladder emptying is also reduced after intravenous CCK in gallstone subjects compared to controls (20,167,173). In studies of large numbers of gallstone patients, contraction is impaired in approximately half of the subjects (20,167,170,174). In patients with gallstones who have undergone successful extracorporeal lithotripsy, the motility defect persists (173), and reduced ejection fraction predicts which subjects will develop recurrent gallstones (155). All these studies indicate that impaired gallbladder emptying is an important factor in the development of gallstones. C— Miscellaneous Conditions Associated with Impaired Gallbladder Motility and Gallstone Formation In a number of patient groups with either temporary or permanent impairment of gallbladder motility, there is an increased predisposition to stone formation, which also indicates that the motility defect plays an important role in the pathogenesis of gallstones. Patients who have had major abdominal surgery with associated fasting for more than 48 h have an increased incidence of gallstones (175). This is probably as a result of bile stasis and sludge formation that accompanies pre and postoperative fasting (176,177). Patients receiving total parenteral nutrition do not empty their gallbladders regularly (25) and have a high incidence of both biliary sludge and gallstones (178,179). The regular administration of parenteral CCK in this setting reduces the incidence of biliary sludge and may be a useful prophylactic therapy (180). Women who have had several pregnancies have an increased prevalence of gallstones (181,182). In the last trimester of pregnancy, as the serum progesterone rises, the fasting gallbladder volume is increased and emptying is impaired (183,184). Although bile becomes more supersaturated with cholesterol in the last trimester (185), gallbladder stasis is also likely to be an important factor in the development of pregnancyassociated gallstones. Patients treated with somatostatin (186,187) or octreotide, a somatostatin analogue (188), have impaired gallbladder emptying and have an increased prevalence of gallstones. This is due to the somatostatinlike action on the gallbladder, which produces dilatation and inhibition of CCKinduced emptying (186,187). Patients with spinal cord injuries and after truncal vagotomy, both conditions causing impaired gallbladder emptying, also have an increased prevalence of gallstones (6,189,190,191). D— The Role of CCK in Gallbladder Dysmotility Associated with Gallstones The exact role of CCK in the gallbladder dysmotility associated with gallstones has not been determined. Subjects with gallstones have increased levels of CCK in the duodenal mucosa compared to healthy controls (192); however, studies of the release of CCK into the plasma in subjects with gallstones and healthy controls have produced conflicting results. Studies demonstrate a similar (172,193), reduced (96), and increased release (194) of CCK in subjects with gallstones compared to healthy controls. Human gallbladder muscle strips show a reduction in isometric tension in response to CCK in cholesterol gallstonecontaining gallbladder compared to pigment stone containing gallbladder (195). This finding has been replicated using isolated smooth muscle cells as well as muscle strips (81).
Page 282
The human gallbladder in vivo shows a variable response to exogenous CCK. Most studies demonstrate that exogenously administered CCK produces less contraction in gallstonecontaining gallbladders than in normal gallbladders (167,173), although a single study demonstrated an equivalent response (168). Whether these differing responses are due to an alteration in CCK receptor status in subjects with gallstones is unclear. Although early studies suggested a reduction in CCK receptors (75,196), these studies were uncontrolled. More recent work with appropriate controls has shown that gallstonecontaining human gallbladder is able to bind circulating CCK with equal affinity to gallstonefree gallbladder. Moreover, the size of the CCK binding subunit is similar between the two groups, implying normal function of the CCK receptors (73). In conclusion, the relationship between CCK release and gallbladder smooth muscle response in the dysmotility found in subjects with gallstones is complex and incompletely understood. Nevertheless, the balance of evidence indicates that there is likely to be an endorgan reduction in response to circulating CCK. E— Prostaglandins and Animal Models of Gallstone Production A highcholesterol diet is lithogenic in several animal models. This diet stimulates the production of PGE2, PGF2a, PGI2, and thromboxane A2 in the prairie dog (105– 108,197), and PGE2 and PGI2 in rabbit gallbladders (111,113). This is unlike the response in the guinea pig gallbladder, where enhanced PGI2 production alone is seen (105). In the prairie dog fed on a highcholesterol lithogenic diet, a reduction in gallbladder emptying correlates with the increased synthesis of PGE2 and PGF2a in the gallbladder (106). In this model, there was no increase in PGI2 synthesis. Administration of salicylate in the same model inhibits gallbladder stasis and the formation of gallstones (198). However, others have found that although aspirin in the cholesterolfed prairie dog inhibits endogenous microsomal prostaglandin synthesis, there is no subsequent reduction in gallstone formation (107). This, however, was a 2week study, which may have been too brief to identify gallstone formation. In addition, the same group showed that in the prairie dog, aspirin has no effect on the reduction in CCKstimulated muscle strip contractility observed with the cholesterol diet (110). However, aspirin inhibited PGE2 synthesis solely, with no inhibition of PGF2a, PGI2, and TXA2 synthesis; this is therefore at variance with the rest of the published work in this field. It is likely that local prostaglandin production is stimulated by cholesterol supersaturation in bile. Cholesterol is absorbed into the gallbladder mucosa in both the guinea pig and human gallbladder (199–203). Recent work has demonstrated that the degree of absorption is proportional to biliary cholesterol saturation (204). F— Prostaglandins and Gallstone Pathogenesis in Humans Human gallbladder in vitro produces increased levels of PGE2 and PGI2, which correlate with the degree of histological inflammation (205–207). PGE2, PGF2a, PGI2, and U46619 (a stable analogue of thromboxaneA2) produce contractions in human gallbladder obtained from patients with gallstones (115). Kotwall, again using gallstonecontaining gallbladders, showed a dosedependent contraction to PGD2, PGE1, PGE2, and PGF2a. The spontaneous rhythmic contractions were inhibited by indomethacin, suggesting a continuous release of endogenous prostaglandin (208). In an epidemiological study, NSAID usage appeared to confer some protection against the development of gallstones, but the protective mechanism is poorly understood (209). Both aspirin and indomethacin have been investigated in subjects with gallstones (123,124). In patients with gallstones, administration of aspirin (124) improved the percentage of postprandial gallbladder emptying from 62 to 74%. Similarly, patients who had received successful extracorporeal lithotripsy for gallstone disease also showed improved postprandial gallbladder emptying after administration of indomethacin (123). This differential effect of NSAIDs on the
Page 283
postprandial emptying of normal and diseased gallbladders is intriguing and may reflect a localised role for PGI2, since PGI2, relaxes only gallstonecontaining gallbladders (120). Since gallbladders containing cholesterolsupersaturated bile produce increased PGI2 (206,207), this PGI2induced relaxation may be a determinant of the impaired gallbladder motility of gallstone disease. Whether cholesterol absorption in the human gallbladder has an effect on local prostaglandin production has not been conclusively demonstrated. However obese patients on a weightreducing diet produce increased amounts of biliary PGE2 as biliary cholesterol saturation increases (210), and there is an association between biliary cholesterol saturation and biliary PGE2 production in postmenopausal women (211). G— Other Autocrine/Paracrine Factors and Gallstone Pathogenesis NO is a potent relaxant of nonvascular smooth muscle. Electrical activation of NANC neurons produces relaxation of diseased human gallbladder in vitro that is mediated by NO (212). Since glyceryl trinitrate—acting as an NO donor—has a potent inhibitory effect on human gallbladder motility in vivo (129), it is possible that local alterations in NO production will modify gallbladder motility and gallstone pathogenesis. Similarly, VIP, PACAP, and endothelins each have potent effects on gallbladder motility and may play a role in gallstone pathogenesis. These possibilities, however, have not been investigated to date. H— Molecular Mechanisms Underlying Gallbladder Dysmotility and Cholesterol Gallstone Pathogenesis Isolated muscle cells from gallbladders with cholesterol gallstones contract less well to CCK, acetylcholine, and potassium chloride than muscle cells from gallbladders containing pigment gallstones, which were used as controls (81), confirming previous work in the human (195) and prairie dog gallbladder (161). This suggests that the contractility defect in human gallbladder muscle contraction is in part intrinsic to the muscle cell, as confounding factors such as mucosal edema or fibrosis have been excluded. Impaired muscle contraction cannot be fully explained by damage to CCK or acetylcholine receptors, because the impaired contraction is observed not only in response to CCK and acetylcholine but also in response to potassium chloride, which contracts cells by receptorindependent mechanisms. When cholesterol gallstone muscle cells are permeabilized with the detergent saponin and the second messenger substrate inositol triphosphate is added, the difference in contractility is eliminated (81). This indicates that the postmembrane pathways and the actinmyosin complex are functionally intact, confirming earlier work on the cholesterolfed prairie dog model of cholesterol gallstone (165). Further work has shown a reduced production of the second messengers inositol triphosphate and diacylglycerol (213). In this study, the difference in contraction in the cholesterol gallstone group and bile pigment stone group normalized with the addition of G protein activators. These results indicate that CCK cannot fully activate the intracellular pathways in smooth muscle derived from gallstonecontaining gallbladders; however, the pathways can be fully activated by circumventing membrane receptors with either second messengers or Gprotein activators. The defect may be located in the receptor itself or in the interaction between receptors and G proteins. Behar and colleagues speculate that cholesterol supersaturation alters membrane fluidity and "stiffens" the cell membrane, thus impairing the interaction between receptors and G proteins and reducing contractility (213). Behar and colleagues have also recently demonstrated that isolated gallbladder muscle cells from both prairie dogs and patients with cholesterol gallstones have an increased membrane cholesterol/phospholipid ratio and decreased membrane fluidity, resulting in impaired muscle contractility (214,215).
Page 284
Decreased contractility of the smooth muscle cell due to an abnormal membraneCCK receptor relationship, as demonstrated by Behar and colleagues using isolated gallbladder cells, is clearly important. However, other nonreceptor factors may also be important in this gallbladder dysmotility, as cholesterolrich gallbladders have impaired contraction to potassium chloride, which is receptorindependent. Altered local production of autocrine/paracrine factors in the gallbladder wall—such as prostaglandins, NO, and VIP—due to cholesterol infiltration is highly likely and also might alter the contractility of the gallbladder in response to CCK and other stimulants. I— Gallstone Prevention by Improving Gallbladder Motility The prevention of gallstone formation in animals (163) and sludge formation in humans (180) by promoting gallbladder emptying raises the possibility that oral prokinetic agents may prevent stone formation in humans. Several pharmacological agents modulate gallbladder motility. Indomethacin enhances gallbladder emptying in patients after extracorporeal lithotripsy (123). Similarly, aspirin has a prokinetic effect on gallbladder emptying, which is restricted to patients with gallstones (124). Prevention of gallstone formation by NSAIDs (209) may therefore be the result of their prokinetic effect. A number of other agents have a prokinetic effect on the gallbladder, including erythromycin (216), cholestyramine (217), and intravenous amino acids (59,218). The effect of cisapride is controversial, with early studies demonstrating a prokinetic effect (219,220) and subsequent studies failing to show this (221,222). The sole agent that has been demonstrated to prevent gallbladder sludge formation in humans is daily administration of intravenous CCK in patients receiving total parenteral nutrition (180). Several of the above agents may well find a role in preventing stone formation in highrisk groups such as dieting obese subjects, patients on longterm parenteral nutrition, and those who have had their gallstones removed by nonsurgical techniques (bile acid dissolution, extracorporeal lithotripsy); however, the evidence is unavailable at present. III— Summary and Conclusion Impaired gallbladder motility is an important factor in gallstone formation. The cause of the impaired motility is not fully defined, but cholesterol infiltration of the gallbladder smooth muscle layer with consequent alterations in prostaglandin production and action, as well as cholesterol stiffening of the smooth muscle membrane, may be important. Improving motility in animals prevents gallstones, and in humans prevents the formation of gallbladder sludge and may prevent gallstones. Gallbladder prokinetic agents such as NSAIDs and erythromycin may find a future role in the prevention of gallstone formation in highrisk subjects. References 1. Moore KL. The Developing Human: Clinically Oriented Embryology. Philadelphia: Saunders, 1973. 2. Williams PL, WendellSmith CP, Treadgold S. Basic Human Embryology, 2nd ed. Philadelphia: Lippincott, 1969. 3. Cai W, Gu J, Huang W. Peptide immunoreactive nerves and cells of the guinea pig gallbladder and biliary pathways. Gut 1983; 24:1186–1193. 4. McPherson BR, Pemsingh RS, Scott GW. Experimental cholelithiasis in the ground squirrel. Lab Invest 1987; 56:138–144.
Page 285
5. Wegstapel H, ChessWilliams R, Stephenson TJ, Majeed AW, Bird NC, Johnson AG. Is there a dominant axis of contraction in the human gallbladder? Gut 1992; 33:S58. 6. Clave RA, Gaspar MR. Incidence of gallbladder disease after vagotomy. Am J Surg 1969; 118:169–176. 7. Kyosola K, Pentilla O. Adrenergic innervation of the human gallbladder. Histochemistry 1977; 54:209–218. 8. Ryan JP. Motility of the gallbladder and biliary tree. In: Johnson L, ed. Physiology of the Gastrointestinal Tract. New York: Raven Press, 1981, pp 473–476. 9. Burnett W, Gairns FW, Bacsich P. Some observations on the innervation of the extrahepatic biliary system in man. Ann Surg 1964; 159:8–26. 10. Sutherland SD. The neurons of the gallbladder and gut. J Anat 1967; 101:701. 11. Bauer AJ, Hanani M, Muir TC, Szurszewski JH. Intracellular recordings from the gallbladder ganglia of opossums. Am J Physiol 1991; 260:G299. 12. Mawe GM. The role of cholecystokinin in ganglionic transmission in the guinea pig gallbladder. J Physiol 1991; 439:89–102. 13. Polak JM, Bloom SR, Sullivan SN, Facer P, Pearse AGE. Enkephalinlike immunoreactivity in the human gastrointestinal tract. Lancet 1977; 1:972–974. 14. De Giorgio R, Parodi JE, Brecha NC, Brunicardi FC, Becker JM, Go VLS. Nitric oxide producing neurons in the monkey and human digestive system. J Comp Neurol 1994; 342:619–627. 15. Talmage EK, Mawe GM. NADPHdiaphorase and VIP are colocalized in neurons of gallbladder ganglia. J Auton Nerv Syst 1993; 43:83–89. 16. Strah KM, Pappas TN, Melendez RL, Debas HT. Contrasting cholinergic dependence of pancreatic and gallbladder responses to cholecystokinin. Am J Physiol 1986; 250: G665–G669. 17. Strah KM, Melendez RL, Pappas TN, Debes HT. Interactions of vasoactive intestinal polypeptide and cholecystokinin octapeptide on the control of gallbladder contraction. Surgery 1986; 99:469–473. 18. Allen M, Gu J, Adrian TE, Bloom SR. Neuropeptide Y in the guinea pig biliary tract. Experientia 1984; 40:765. 19. Lanzini A, Jazrawi P, Northfield TC. Simultaneous quantitative measurements of absolute gallbladder storage and emptying during fasting and eating in humans. Gastroenterology 1987; 92:852–861. 20. Forgacs IC, Maisy MN, Murphy GM, Dowling RH. Influence of gallstones and ursodeoxycholic acid therapy on gallbladder emptying. Gastroenterology 1984; 87:299–307. 21. Lawson M, Everson GT, Klingensmith W, Kern F. Coordination of gastric and gallbladder emptying after ingestion of a regular meal. Gastroenterology 1983; 85:866–870. 22. Howard PJ, Murphy GM, Dowling RH. Gallbladder emptying patterns in response to a normal meal in healthy subjects and patients with gallstones. Gut 1991; 32:1409–1411. 23. Baxter JN, Grime JS, Critchley M, Shields R. Relationship between gastric emptying of solids and gallbladder emptying in normal subjects. Gut 1985; 26:342– 351. 24. Jazrawi RP, Pazzi P, Petroni ML, Prandini N, Paul C, Adam JA, Gullini S, Northfield TC. Postprandial gallbladder motor function: refilling and turnover of bile in health and in cholelithiasis. Gastroenterology 1995; 109:582–591. 25. Cano C, Cicero F, Ranieri F, Martin J, Di Contanzo J. Ultrasonographic study of gallbladder motility during parenteral nutrition. Gastroenterology 1986; 91: 313– 317. 26. Marzio L, Capone F, Neri M, Di Felice F, Celiberti V, Mezzetti AGD, Cuccurullo F. Effect of cholinergic agonists and antagonists on gallbladder volume in fasting man. Eur J Clin Pharmacol 1987; 33:151–153. 27. Svenberg T, Christofides ND, Fitzpatrick ML, AreolaOrtiz F, Boom SR, Welbourn RB. Interdigestive biliary output in man: relationship to fluctuations in plasma motilin and effect of atropine. Gut 1982; 23:1024–1028. 28. Niederau C, Heintges T, Rovati L, Strohmeyer G. Effects of loxiglumide on gallbladder emptying in healthy volunteers. Gastroenterology 1989; 97:1331–1336.
Page 286
29. Corazziari E, Ricci R, Biliotti D, Bontempo I, De Medici A, Pallota N, Torsoli A. Oral administration of loxiglumide inhibits post prandial gallbladder contraction without affecting gastric emptying. Dig Dis Sci 1990; 35:50–54. 30. Meyer BM, Werth BA, Beglinger C, Hildebrand P, Jansen JB, Zach D, Rovati LC, Stalder GA. Role of cholecystokinin in regulation of gastrointestinal motor functions. Lancet 1989; 2:12–15. 31. Tooli J, Bushell M, Stephenson G, Dent J, Wycherley A, Iannos J. Gallbladder emptying in man related to fasting duodenal migrating motor contractions. Aust NZ J Surg 1986; 56:147–151. 32. Kraglund K, Hjermind J, Jensen FT, StodkildeJorgensen H, OsterJorgensen E, Pedersen SA. Gallbladder emptying and gastrointestinal cyclic activity in humans. Scand J Gastroenterol 1984; 19:990–994. 33. Quist N, OsterJorljensen E, Rasmussen L, Kraglund K, Pedersen SA. The relationship between gallbladder mechanics and the migrating motor complex in fasting healthy subjects. Scand J Gastroenterol 1988; 23:562–566. 34. Marzio L, Neri M, Capone F, Di Felice F, De Angelis C, Mezzetti ACF. Gallbladder contraction and its relationship to interdigestive duodenal motor activity in normal human subjects. Dig Dis Sci 1988; 33:540–544. 35. Furness JB, Kuramoto H, Baker RA, Toouli J. Direct neural projections from the duodenum to the gallbladder and sphincter of Oddi. Gastroenterology 1990; 98:A379. 36. Itoh Z, Aizawa I, Takeuchi S, Couch EF. Proceedings of the Fifth International Symposium on Gastrointestinal Motility. Herentals, Belgium: Typoff, 1975, pp 48– 55. 37. Wingate DL, Ruppin H, Thompson HH. 13Norleucine motilin versus pentagastrin: contrasting and competitive effects on gastrointestinal myoelectrical activity in the conscious dog. Acta Hepatogastroenterol 1975; 22:409–410. 38. Takahasi I, Suzuki T, Aizawa I, Itoh Z. Comparison of gallbladder contractions induced by motilin and choalecystokinin in dogs. Gastroenterology 1982; 82:419– 424. 39. Svenberg T, Nilsson I, Samuelson K, Welbourn RD. Studies on the causal relationship between gallbladder emptying and motilin release in man. Acta Chir Scand 1984; 520(suppl):59–61. 40. Pomeranz IS, Davison JS, Shaffer EA. In vitro effects of pancreatic polypeptide and motilin on contractility of human gallbladder. Dig Dis Sci 1983; 28:539–544. 41. Ellenbogen S, Grime JS, Mackie CR, Calam J, Springer C, Jenkins SA. Mechanisms responsible for the cephalic phase of gallbladder emptying. Gut 1986; 27:A621. 42. Fisher RS, Rock E, Malmud LS. Gallbladder emptying response to sham feeding in humans. Gastroenterology 1986; 90:1854–1857. 43. Tankurt E, Yegen BC, Biren T, Gurmen N, Ulosoy NB. Influence of pirenzipine on gallbladder contraction in man induced by sham feeding or an intraduodenal meal. Digestion 1992; 51:103–109. 44. Debas HT, Yagamashi T. Evidence for a pylorocholecystic reflex for gallbladder contraction. Ann Surg 1979; 190:170–176. 45. Everson GT, Lawson MJ, McKinley C, Showalter R, Kern F. Gallbladder and small intestine regulation of biliary lipid secretion during intraduodenal infusion of standard stimuli. J Clin Invest 1983; 71:596–603. 46. Fried M, Jansen JB, Harpole T, Taylor IL, Lamers CB, Reedy T, Elsahoff J, Meyer JH. Pancreatobiliary response to an intragastric amino acid meal; comparison to albumin, dextrose and a maximal cholecystokinin stimulus. Gastroenterology 1989; 97:1544–1549. 47. Ellenbogen S, Jenkins SA, Grime JS, Critchley M, Mackie CR, Baxter JN. Preduodenal mechanisms in initiating gallbladder emptying in man. Br J Surg 1988; 75:940–945. 48. Amer MS. Studies with cholecystokinin in vitro: mechanism of the effect on the isolated rabbit gallbladder strips. J Pharmacol Exp Ther 1972; 183:527–534. 49. Persson CGA. Adrenoceptors in the gallbladder. Acta Pharmacol Toxicol 1972; 31:177–185.
Page 287
50. Bainbridge FA, Dale HH. The contractile mechanism of the gallbladder and its extrinsic nervous control. J Physiol Lond 1905; 33:138–155. 51. Persson CGA. Dual effects on the sphincter of Oddi and gallbladder induced by stimulation of the right great splanchnic nerve. Acta Physiol Scand 1973; 87:334– 343. 52. Bjork S, Jansson R, Svanvik J. Adrenergic influence on concentrating function in the feline gallbladder. Gut 1982; 23:1019–1023. 53. Winkelstein A, Achsner PW. The pressure factors in the biliaryduct system of the dog. Am J Med Sci 1924; 168:812. 54. Shoetz DJ, Birkett DH, Williams LF. Gallbladder motor function in the intact primate: autonomic pharmacology. J Surg Res 1978; 24:514. 55. Eysselein VE, Eberlein GA, Schaeffer M, Grandt D, Goebell H, Niedal W, Rosenquist GL, Meyer HE, Reeve JR. Characterisation of the major form of cholecystokinin in human intestine. Am J Physiol 1990: 258:G253–G260. 56. Liddle RA, Goldfine ID, Rosen MS, Taplitz RA, Williams JA. Cholecystokinin bioactivity in human plasma: molecular forms, responses to feeding and relationship to gallbladder contraction. J Clin Invest 1985; 75:1144–1152. 57. Turkelson CM, Solomon TE. Molecular forms of cholecystokinin in rat intestine. Am J Physiol 1990; 259:G364–G371. 58. Beardshall K, Morarji Y, Bloom SR, Frost G, Domin J, Calam J. Saturation of fat and cholecystokinin release: implications for possible pancreatic carcinogenesis. Lancet 1989; 2:1008–1010. 59. Nealon WH, Upp JR, Alexander RW, Gomez G, Townsend CM, Thompson JC. Intravenous amino acids stimulate human gallbladder emptying and hormone release. Am J Physiol 1990; 259:G173–G178. 60. Hopman WPM, DeJong AJL, Rosenbusch G, Jansen JBMJ, Lamers CBHW. Elemental diet stimulated gallbladder contraction and secretion of cholecystokinin and pancreatic polypeptide in man. Dig Dis Sci 1987; 32:45–49. 61. Lin YZ, Isaac DD, Tam JP. Synthesis and properties of cholecystokininreleasing peptide (monitor peptide), a 61residue trypsin inhibitor. Int J Peptide Protein Res 1990; 36:433–439. 62. Iwai K, Fukuoka T, Fushiki T, Kodaira T, Ikei N. Elevation of plasma cholecystokinin concentration after intestinal administration of a pancreatic enzyme secretionstimulating peptide purified from rat bilepancreatic juice: analysis with Nterminal region specific radioimmunoassay. Biochem Biophys Res Commun 1986; 136:701–706. 63. Lu L, Louie D, Owyang C. A cholecystokinin releasing peptide mediates feedback regulation of pancreatic secretion. Am J Physiol 1989; 256:G430–G435. 64. Miyasaka K, Guan R, Liddle RA, Green GM. Feedback regulation by trypsin: evidence for intraluminal cholecystokininreleasing peptide. Am J Physiol 1989; 257:G175–G181. 65. Liddle RA, Goldfine ID, Williams JA. Bioassay of plasma cholecystokinin in rats: effects of food, trypsin inhibitor and alcohol. Gastroenterology 1984; 87:542– 549. 66. Louie DS, May P, Miller P, Owyang C. Cholecystokinin mediates feedback regulation of pancreatic enzyme secretion in rats. Am J Physiol 1986; 250:G252– G259. 67. Owyang CD, Louie DS, Tatum D. Feedback regulation of pancreatic enzyme secretionsuppression of cholecystokinin release by trypsin. J Clin Invest 1986; 77:2042–2047. 68. Folsch URP, Cantor HM, Wilms A, Schafmayer A, Becker HD, Creutzfeldt W. Role of cholecystokinin in the negative feedback control of pancreatic enzyme secretion in conscious rats. Gastroenterology 1987; 92:449–458. 69. Liener IE, Goodale RL, Deshmukh A, Satterberg TL, Ward G, DiPietro CM, Bankey PE, Borner JW. Effect of a trypsin inhibitor from soybeans on the secretory activity of the human pancreas. Gastroenterology 1988; 94:419–427. 70. Gomez G, Lluis F, Guo YS, Greelay GH, Townsend CM, Thompson JC. Bile inhibits release cholecystokinin and neurotensin. Surgery 1986; 100:363–368.
Page 288
71. Muller LJ. Heterogeneity of CCK receptors: classification and characterization. In: Adler G, ed. CCK Antagonists in Gastroenterology. Berlin: SpringerVerlag, 1991, pp 27–34. 72. Wank SA, Harkins R, Jensen RT, Shapira H, DeWerth A, Slattery T. Purification, molecular cloning, and functional expression of the cholecystokinin receptor from rat pancreas. Proc Natl Acad Sci USA 1992; 89:3125–3129. 73. Schjoldager B, Molero X, Miller LJ. Functional and biochemical characterization of the human gallbladder muscularis cholecystokinin receptor. Gastroenterology 1989; 96: 1119–1125. 74. Tang C, Biemond I, Lamers CBHW. Cholecystokinin receptors in human pancreas and gallbladder muscle: a comparative study. Gastroenterology 1996; 111:1621–1626. 75. Upp JP, Nealon WH, Singh P, Fagan C J, Jonas AS, Greeley GH, Thompson JL. Correlation of cholecystokinin receptors with gallbladder contractility in patients with gallstones. Ann Surg 1987; 205:641–647. 76. Yau WM, Makhlouf GM, Edwards LE, Farrar JT. Mode of action of cholecystokinin and related peptides on gallbladder muscle. Gastroenterology 1973; 65:451–456. 77. Peikin SRA, Rottman S, Batzri S, Gardner JD. Kinetics of amylase release by dispersed acini prepared from guinea pig pancreas. Am J Physiol 1978; 235:E743– E749. 78. Schultz GS, Sarras MP, Gunther GR, Hull BE, Alicea HA, Gorelick FS, Jamieson JD. Guinea pig pancreatic acini prepared with purified collagenase. Exp Cell Res 1980; 130:49–62. 79. Williams JA, Korc M, Dormer RL. Action of secretagogues on a new preparation of functionally intact, isolated pancreatic acini. Am J Physiol 1978; 235:E517– E524. 80. Yu P, De Petris G, Biancani P, Amaral J, Behar J. Cholecystokinincoupled intracellular signalling in human gallbladder muscle. Gastroenterology 1994; 106:763– 770. 81. Behar J, Rhim BY, Thompson W, Biancani P. Inositol trisphosphate restores impaired human gallbladder motility associated with cholesterol stones. Gastroenterology 1993; 104:563–568. 82. Williams JA, Yule DI. Stimulumsecretion coupling in pancreatic acinar cells. In: The Exocrine Pancreas: Biology, Pathobiology and Disease. 2nd ed. New York: Raven Press 1993. 83. Spellman SJ, Shaffer EA, Rosenthall L. Gallbladder emptying in response to cholecystokinin: a cholescintographic study. Gastroenterology 1979; 77:115–120. 84. Hopman WP, Kerstens PJ, Jansen JB, Rosenbusch G, Lamers CB. Effect of graded physiologic doses of cholecystokinin on gallbladder contraction measured by ultrasonography: determination of threshold, doseresponse relationships and comparison with intraduodenal bilirubin output. Gastroenterology 1985; 89:1242–1247. 85. Wiener I, Inoue K, Fagan CJ, Lilja P, Watson LC, Thompson JC. Release of cholecystokinin in man: correlation of blood levels with gallbladder contraction. Ann Surg 1981; 194:321–327. 86. Liddle RA, Gertz BJ, Kanayama S, Beccaria L, Coker LD, Turnbull TA, Morita ET. Effects of a novel cholecystokinin receptor antagonist, MK329, on gallbladder contraction and gastric emptying in humans: implications for the physiology of CCK. J Clin Invest 1989; 84:1220–1225. 87. Malesci A, Defazio C, Festorazzi S, Bonato C, Valenti A, Tacconi M, Rovati L, Setnikar I. Effect of loxiglumide on gallbladder contractile response to caerulein and food in humans. Gastroenterology 1990; 98:1307–1310. 88. Behar J, Biancini P. Pharmacology of the biliary tract. In: Scultz SG, Wood JD, Rauner BB, eds. Handbook of Physiology: Section 6. The Gastrointestinal System. New York: Oxford University Press, 1989, pp 1103–1130. 89. Hanyu N, Dodds WJ, Layman RD, Hogan WJ, Chey WY, Takahashi I. Mechanism of cholecystokinininduced contraction of the opossum gallbladder. Gastroenterology 1990; 98:1299–1306. 90. Brotschi EA, Pattavino J, Williams LF. Intrinsic nerves affect gallbladder contraction in the guinea pig. Gastroenterology 1990; 99:826–830.
Page 289
91. Pozo MJ, Salido MD, Madrid JA, Salido GM. In vitro effect of pirenzipine on motility of canine gallbladder. J Pharm Pharmacol 1990; 42:89–93. 92. Takahashi T, May D, Owyang C. Cholinergic dependence of gallbladder response to cholecystokinin in the guinea pig in vivo. Am J Physiol 1991; 261:G565– G569. 93. Gullo L, Bolondi L, Scarpignato C, Priori P, Casnova P, Labo G. Effect of somatostatin and thyrotropinreleasing hormone on cholecystokinininduced gallbladder emptying. Dig Dis Sci 1986; 31:1345–1350. 94. Fisher RS, Rock E, Malmud LS. Cholinergic effects on gallbladder emptying in humans. Gastroenterology 1985; 89:716–722. 95. Hopman WP, Jansen JB, Rosenbusch G, Lamers CB. Role of Cholecystokinin and the cholinergic system in intestinal stimulation of gallbladder contraction in man. Hepatology 1990; 11:261–265. 96. Masclee AA, Jansen JMBJ, Driessen WM, Geuskens LM, Lamers CBHW. Effect of truncal vagotomy on cholecystokinin release, gallbladder contraction, and gallbladder sensitivity to cholecystokinin in humans. Gastroenterology 1990; 98:1338–1344. 97. Feyter F. Uber die These von den peripheren endokrinen Druesen. Wien Z Inn Med 1946; 27:9–38. 98. Olofsson J, Leung CK. Auto/paracrine role of prostaglandins in corpus luteum function. Mol Cell Endocrinol 1994; 100:87–91. 99. Moncada S, Palmer RMJ, Higgs EA. Nitric oxide: physiology, pathophysiology and pharmacology. Pharmacol Rev 1991; 43:109–142. 100. Yanagisawa M, Kurihara H, Kimura S, Tomobe Y, Kobayashi M, Masaki Y. A novel potent vasoconstrictor peptide produced by vascular endothelial cells. Nature 1988; 32:411–415. 101. Bjork S, Fahrenkrug J, Jivegard L, Svanvik J. Release of immunoreactive vasoactive intestinal peptide from the gallbladder in response to vagal stimulation. Acta Physiol Scand 1986; 128:639–642. 102. Sporn MB, Todaro GJ. Autocrine secretion and malignant transformation of cells. N Eng J Med 1980; 303:878–880. 103. Booker ML, LaMorte WW. Prostaglandin release from in vitro guineapig gallbladder. Prostaglandins 1983; 25:143–153. 104. Nakata K, Osumi Y, Fujiwara M. Prostaglandins and the contractility of the guinea pig biliary system. Pharmacology 1981; 22:24–30. 105. LaMorte WW, Booker ML, Scott TE, Williams LF. Increases in gallbladder prostaglandin synthesis before the formation of cholesterol gallstones. Surgery 1985; 98:445–451. 106. Chapman WC, Peterkin GA, LaMorte WW, Williams LF. Alterations in biliary motility correlate with increased gallbladder prostaglandin synthesis in early cholelithiasis in prairie dog. Dig Dis Sci 1989; 34:1420–1424. 107. Myers SI, Li YF, Weisbrodt NW, Russell DH, Moody F. Endogenous gallbladder prostaglandin biosynthesis is not related to gallstone formation in the prairie dog fed a highcholesterol diet. Surg Forum 1989; 40:157–158. 108. LaMorte WW, LaMont JT, Hale W, Booker ML, Scott TE, Turner B. Gallbladder prostaglandins and lysophospholipids as mediators of mucin secretion during cholelithiasis. Am J Physiol 1986; 251:G701–G709. 109. LaMont JT, Turner BS, DiBenedetto D, Handin R, Schafer AI. Arachidonic acid stimulates mucin secretion in prairie dog gallbladder. Am J Physiol 1983; 245:G92–G98. 110. Li YF, Russell DH, Myers SI, Weisbrodt NW, Moody FG. Gallbladder contractility in aspirin and cholesterolfed prairie dogs. Gastroenterology 1994; 106:1662–1667. 111. Myers S, Bartula L, KalleyTaylor B. The role of prostaglandin I2 and biliary lipids during evolving cholecystitis in the rabbit. Gastroenterology 1993; 104:248– 255. 112. Myers SI, HaleyRussell D, Bartula L Nabzdyk. Longterm cholesterol feeding stimulates increased bile phospholipids and gallbladder prostanoid synthesis. Surgery 1991; 109:656–662.
Page 290
113. England DW, Jaffe BM, Webb H, Hoover E. The prostaglandin response to cholesterol feeding by the rabbit gallbladder in vitro. Surgery 1988; 103:69–73. 114. Andersson KE, Andersson R, Hedner P, Persson CGA. Parallelism between mechanical and metabolic responses to cholecystokinin and prostaglandin E2 in extrahepatic biliary tract. Acta Physiol Scand 1973; 89:571–579. 115. Wood JR, Saverymuttu SH, Ashbrooke AB, Stamford IF. Effects of various prostanoids on gallbladder muscle. Adv Prostaglandin Thromboxane Leuko Res 1980; 8:1569–1571. 116. Mroczka J, Baer HP, Scott GW. Effects of prostaglandins on isolated dog gallbladder and cystic duct. In: Wienbeck M. ed. Motility of the Digestive Tract. New York: Raven Press, 1982, pp 421–426. 117. Svanvik J, Thornell E, Wood JR. Effects of prostaglandin E2 on gallbladder function in the cat. J Physiol 1980; 305:104P. 118. Nakano J, McCloy RE, Gin AC, Nakano SK. Effect of prostaglandins E1, E2 and F2a and pentagastrin on the gallbladder pressure in dogs. Eur J Pharm 1975; 30:107–112. 119. Brotschi EA, LaMorte WW, Williams LF. Effect of dietary cholesterol and indomethacin on cholelithiasis and gallbladder motility in guinea pig. Dig Dis Sci 1984; 29:1050–1056. 120. Greaves RRSH, O'Donnell LJD, Farthing MJG. Differential effect of prostacyclin on motility of normal and diseased human gallbladder in vitro. Gastroenterology 1996; 110: A456. 121. Jonderko G, Jonderko K, Konca A, Polko M. Effect of synthetic prostaglandin E1 derivative, misoprostol, on fasted gallbladder volume and on mealinduced gallbladder emptying in man. Am J Gastroenterol 1989; 84:1375–1377. 122. Murray FE, Stinchcombe SJ, Hawkey CJ. Effect of indomethacin and misoprostol on fasted gallbladder volume and mealinduced gallbladder contractility. Dig Dis Sci 1992; 37:128–131. 123. O'Donnell LJD, Wilson P, Guest P, Catnach SM, McLean A, Wickham JEA, Fairclough PD. Indomethacin and postprandial gallbladder emptying. Lancet 1992; 339:269–271. 124. Das A, Baijal SS, Sarawat VA. Effect of aspirin on gallbladder motility in patients with gallstone disease. Dig Dis Sci 1995; 40:1782–1785. 125. Thune A, Delbros DS, Nisson B, Friman S, Svanvik J. Role of nitric oxide in motility and secretion of the feline hepatobiliary tract. Scand J Gastroenterol 1995; 30:715–720. 126. Kaufmann HS, Shermak MA, May CA, Pitt HA, Lillemoe JD. Nitric oxide inhibits resting sphincter of Oddi activity. Am J Surg 1993; 165:74–80. 127. Uemura S, Pompolo S, Furness JB, Hardy KJ. Nitric oxide synthase in neurons of the human gallbladder and its colocalization with neuropeptides. J Gastroenterol Hepatol 1997; 12:257–265. 128. Mourelle M, Guarner F, Molero X, Moncada S, Malagelada JR. Regulation of gall bladder motility by the argininenitric oxide pathway in guinea pigs. Gut 1993; 34:911–915. 129. Greaves RRSH, Miller J, O'Donnell LJD, McLean A, Farthing MJG. Effect of the nitric oxide donor, glyceryl trinitrate, on human gallbladder motility. Gut 1998; 42:410–413. 130. Mourelle M, Guarner F, Moncada S, Malagelada JR. The arginine/nitric oxide pathway modulates sphincter of Oddi motor activity in guinea pigs and rabbits. Gastroenterology 1993; 105:1299–1305. 131. Pauletzki JG, Sharkey KA, Davison JS, Bomzon A, Shaffer EA. Involvement of Largininenitric oxide pathways in neural relaxation of the sphincter of Oddi. Eur J Pharmacol 1993; 232:263–270. 132. Bondanszky M, Klausner YS, Said S. Biological activities of synthetic peptides corresponding to fragments of and to the entire sequence of the vasoactive intestinal peptide. Proc Natl Acad Sci USA 1973; 70:382–384. 133. Sundler F, Alumets J, Hakanson R, Ingemanson S, Fahrekrug J, Shaffalitzky de Muckadell O. VIP innervation of the gallbladder. Gastroenterology 1977; 72:1375–1377.
Page 291
134. Sand J, Tainio H, Nordback I. Neuropeptides in pig sphincter of Oddi, bile duct, gallbladder, and duodenum. Dig Dis Sci 1993; 38:694–700. 135. Dahlstrand C, Edin R, Dahlstrom A, Ahlman H. Adrenergic and VIPergic relaxatory mechanisms of the feline extrahepatic biliary tree. J Auton Nerv Syst 1989; 26:97–106. 136. Alumets J, Fahrekrug J, Hakanson R, Shaffalitzky de Muckadell O, Sundler F, Uddman R. A rich VIP nerve supply is characteristic of sphincters. Nature 1979; 280:155–156. 137. Ryan JP, Ryave S. Effect of vasoactive intestinal peptide on gallbladder smooth muscle in vitro. Am J Physiol 1978; 234:E44–E46. 138. Wood JR, Brennan LJ, Hormbrey JM. Comparisons of the effects of VIP, secretin, GIP and glucagon on gallbladder function. Regul Pept 1982; 3:87. 139. Piper P, Said S, Vane JR. Effects on smooth muscle preparations of unidentified vasoactive peptides from muscle and lung. Nature 1970; 225:1144–1146. 140. Vagne M, Troitskaja V. Effect of secretin, glucagon and VIP on gallbladder contraction. Digestion 1976; 14:62–67. 141. Ryan JP, Cohen S. Effect of vasoactive intestinal peptide on basal and cholecystokinininduced gallbladder pressure. Gastroenterology 1977; 77:870–872. 142. Mizumoto A, Fujimura M, Ohtawa M, Ueki S, Hayashi N, Itoh Z, Fujino M, Arimura A. Pituitary adenylate cyclase activating polypeptide stimulates gallbladder motility in conscious dogs. Regul Pep 1992; 42:39–50. 143. Miyata A, Arimura A, Dahl RR, Minamino N, Uehara A, Jiang L, Culler M, Coy DH. Isolation of a novel 38residue hypothalamic polypeptide which stimulates adenylate cyclase in pituitary cells. Biochem Biophys Res Commun 1989; 164:567–574. 144. Miyata A, Jiang L, Dahl RR, Kitada C, Kubo K, Fujino M, Minamino N, Arimura A. Isolation of a neuropeptide corresponding to the Nterminal 27 residues of the pituitary adenylate cyclase activating polypeptide with 38 residues. Biochem Biophys Res Commun 1990; 170:643–648. 145. Hickey KA, Rubayani GM, Paul RJ, Highsmith RF. Characterization of a coronary vasoconstrictor produced by cultured endothelial cells. Am J Physiol 1985; 248:C550–C556. 146. Housset C, Carayon A, Housset B, Legendre C, Hannoun L, Poupon R. Endothelin1 secretion by human gallbladder epithelial cells in primary culture. Lab Invest 1993; 69:750–755. 147. Fouassier L, Chinet T, Robert B, Carayon A, Balladur P, Mergey M, Paul A, Poupon R, Capeau J, Barbu V, Housset C. Endothelin1 is synthesised and inhibits cyclic adenosine monophosphatedependent anion secretion by an autocrine/paracrine mechanism in gallbladder epithelial cells. J Clin Invest 1998; 101:2881– 2888. 148. McCartney SA, Greaves RRSH, Warner TD, Domizio P, Farthing MJG. Endothelin content but not expression is reduced in inflamed gallbladders. Gastroenterology 1997; 112:A517. 149. Battistini B, O'Donnel JD, Warner TD, Fournier A, Farthing MJG, Vane JR. Characterization of endothelin (ET) receptors in the isolated gallbladder of the guinea pig: evidence for an additional ET receptor subtype. Br J Pharmacol 1994; 112:1244–1250. 150. Cardozo AM, D'OrleansJuste P, Yano M, Frank PA, Rae GR. Influence of endothelin A and endothelin B receptor antagonists on endothelininduced contractions of the isolated guinea pig gallbladder. Regul Pept 1997; 69:15–23. 151. Moummi C, Gullikson GW, Gaginella TS. Effect of endothelin1 on guinea pig gallbladder smooth muscle in vitro. J Pharmacol Exp Ther 1992; 260:549–553. 152. Heaton KW, Braddon FEM, Mountford RA, Hughes AO, Emmett PM. Symptomatic and silent gallstones in the community. Gut 1991; 32:316–320. 153. Attili AF, De Santis A, Capri R, Repice AM, Maselli S. The natural history of gallstones: the GREPCO experience: the GREPCO Group. Hepatology 1995; 21:655–660. 154. Admirand WH, Small DM. The physicochemical basis of cholesterol gallstone formation in man. J Clin Invest 1968; 47:1043–1052.
Page 292
155. Pauletzki J, Althaus R, Holl J, Sackmann M, Paumgartner G: Gallbladder emptying and gallstone formation: a prospective study on gallstone recurrence. Gastroenterology 1996; 111:765–771. 156. Holan KR, Holzbach RT, Hermann RE, Cooperman AM, Claffey WJ. Nucleation time: a key factor in the pathogenesis of cholesterol gallstone disease. Gastroenterology 1979; 77:611–617. 157. Von Hemsbach JH. Mikrogeologie—Ueber die Concemente in Theirischen Organismus: Nach des Vertassers Tode Herausgegeben und Bevortet von Dr. Theodor Billroth. Berlin: Reimer, 1856, pp 1–275. 158. Doty JE, Pitt HA, Kuchenbecker SL, DenBesten L. Impaired gallbladder emptying before gallstone formation in the prairie dog. Gastroenterology 1983; 85:168–174. 159. Pellegrini CA, Ryan T, Broderick W, Way L. Gallbladder filling and emptying during cholesterol gallstone formation in the prairie dog: a cholescintigraphic study. Gastroenterology 1986; 90:143–149. 160. Meyer PD, DenBesten L, Gurll NJ. Effects of cholesterol gallstone induction on gallbladder function and bile salt pool size in the prairie dog model: Surgery 1978; 83:599–604. 161. Fridhandler TM, Davison JS, Shaffer EA. Defective gallbladder contractility in the ground squirrel and prairie dog during the early stages of cholesterol gallstone formation. Gastroenterology 1983; 85:168–174. 162. Roslyn JJ, DenBesten L, Thompson JE. The effect of periodic gallbladder emptying on gallbladder function and cholesterol gallstone formation. Surg Forum 1979; 30:403–406. 163. Roslyn JJ, DenBesten L, Pitt HA, Kuckenbecker S, Polarek JW. Effects of cholecystokinin on gallbladder stasis and cholesterol gallstone formation. Surg Res 1981; 30:200–204. 164. Li YF, Moody FG, Weisbrodt NW, Zalewsky CA, Coelho JC, Senninger N. Decrease of contractility of prairie dog gallbladder muscle strips following cholesterol feeding. Surg Forum 1984; 35:224–226. 165. Li YF, Weisbrodt NE, Moody FG, Coelho JC, Gouma DJ. Calciuminduced contraction and contractile protein content of gallbladder smooth muscle after highcholesterol feeding of prairie dogs. Gastroenterology 1987; 92:746–750. 166. Ono K, Nakayoshi A, Suzuki K, Scott GW, Clanachan AS. Gallbladder contractility and gallstone formation in the Richardson ground squirrel. Gastroenterol Jpn 1990; 25:95–103. 167. Pomeranz IS, Shaffer EA. Abnormal gallbladder emptying in a subgroup of patients with gallstones. Gastroenterology 1985; 88:787–791. 168. Fisher RS, Stelzer F, Rock E, Malmud LS. Abnormal gallbladder emptying in patients with gallstones. Dig Dis Sci 1982; 27:1019–1024. 169. Shaffer EA, McOrmond P, Duggan H. Quantitative cholescintigraphy: assessment of gallbladder filling and emptying and duodenogastric reflux. Gastroenterology 1980; 79:899–906. 170. Portincasa P, Di Ciaula A, Baldassarre G, Palmieri V, Gentile A, Cimmino A, Palasciano G. Gallbladder motor function in gallstone patients: sonographic and in vitro studies on the role of gallstones, smooth muscle function and gallbladder wall inflammation. J Hepatol 1994: 21:430–440. 171. Stolk MFJ, Van Erpecum KJ, Renooij W, Portincasa P, Van de Heijning BJM, VanBergeHenegouwen GE Gallbladder emptying in vivo, bile composition, and nucleation of cholesterol crystals in patients with cholesterol gallstones. Gastroenterology 1995; 108:1882–1888. 172. Van Erpecum KJ, van Berge Henegouwen GP, Stolk MFJ, Hopman WPM, Jansen JBMJ, Lamers C. Fasting gallbladder volume, postprandial emptying and cholescystokinin release in gallstone patients and normal subjects. J Hepatol 1992; 14:194–202.
Page 293
173. Spengler U, Sackmann M, Sauerbruch T, Holl J, Paumgartner G. Gallbladder motility before and after extracorporeal shockwave lithotripsy. Gastroenterology 1989; 96:860–863. 174. Festi D, Frabboni R, Bazzoli F, Sangermano A, Ronchi M, Rossi L, Parini P, Orsini M, Primerano AMM, Mazzella G, Aldini R, Roda E. Gallbladder motility in cholesterol gallstone disease. Gastroenterology 1990; 99:1779–1785. 175. Little JM, Avramovic J. Gallstone formation after major abdominal surgery. Lancet 1991; 337:1135–1137. 176. Lee SP, Maher K, Nicholas JF, Origin and fate of biliary sludge. Gastroenterology 1988; 94:170–176. 177. Bolondi L, Gaiani S, Testa S, Labo G. Gallbladder sludge formation during prolonged fasting after gastrointestinal surgery. Gut 1985; 26:734–738. 178. Roslyn JJ, Pitt HA, Mann LL, Ament ME, Den Besten L. Gallbladder disease in patients on longterm parenteral nutrition. Gastroenterology 1983; 84:148–154. 179. Messing B, Bories C, Kuntslinger F, Bernier JJ. Does total parenteral nutrition induce gallbladder sludge formation and lithiasis? Gastroenterology 1983; 84:1012–1019. 180. Sitzmann JV, Pitt HA, Steinborn PA, Pasha ZR, Sanders RC. Cholecystokinin prevents parenteral nutrition induced biliary sludge in humans. Surg Gynecol Obstet 1990; 170:25–31. 181. Scragg RKR, McMichael AJ, Seamark RF. Oral contraceptives, pregnancy, and endogenous oestrogen in gallstone disease—a casecontrol study. BMJ 1984; 288:1795–1799. 182. Barbara L, Sama C, Labate AMM. A population study on the prevalence of gallstone disease: the Sirmione study. Hepatology 1987; 7:913–917. 183. Everson GT, McKinley C, Lawson M, Johnson M, Kern F. Gallbladder function in the human female: effect of the ovulatory cycle, pregnancy and contraceptive steroids. Gastroenterology 1982; 82:711–719. 184. Braverman DZ, Johnson ML, Kern F. Effects of pregnancy and contraceptive steroids on gallbladder function. N Engl J Med 1980; 302:362–364. 185. Kern F, Everson GT, DeMark B, McKinley C, Showalter R, Erfling W, Braverman DZ, SzczepanikVan Leeuwen P, Klein PD. Biliary lipids, bile acids and gallbladder function in the human female: effects of the ovulatory cycle. J Clin Invest 1981; 68:1229–1242. 186. Fisher RS, Rock E, Levin G, Malmud LS. Effects of somatostatin on gallbladder emptying. Gastroenterology 1987; 92:885–889 187. Neri M, Cuccurullo F, Marzio L. Effect of somatostatin on gallbladder Volume and small intestinal motor activity in humans. Gastroenterology 1990; 98:316– 320. 188. Catnach SM, Anderson JV, Fairclough PD, Trembath RC, Wilson PAJ, Parker E, Besser GM, Wass JAH. Effect of octrotide on gallstone prevalence and gallbladder motility in acromegaly. Gut 1993; 34:270–273. 189. Apstein MD, DaleckiChipperfield K. Spinal cord injury is a risk factor for cholesterol gallstone disease. Gastroenterology 1987: 92:966–968. 190. Sapala MA, Sapala JA, RestoSoto AD, Bouwman DL. Cholelithiasis following subtotal gastric resection with truncal vagotomy. Surg Gynecol Obstet 1979; 148:36–38. 191. Nino Murcia M, Burton D, Chang P, Stone L, Perkash I. Gallbladder contractility in patients with spinal cord injuries: a sonographic investigation. AJR 1990; 154:521–524. 192. Katoka S, Syoji K. Elevated cholecystokininlike activity in the duodenal mucosa in patients with cholelithiasis. Tohoku J Exp Med 1985; 145:395–402. 193. Pauletzki J, Cicala M, Holl J, Sauerbruch T, Schafmayer A, Paumgartner G. Correlation between gall bladder fasting volume and postprandial emptying in patients with gallstones and healthy controls. Gut 1993; 34:1443–1447. 194. Bailey IS, Walsh TN, Hill ADK, Jazrawi S, Hennessy TPS. Effect of cholecystectomy on plasma CCK. Br J Surg 1992; 79:456–460. 195. Behar J, Lee KY, Thompson WR, Biancani P. Gallbladder contraction in patients with pigment and cholesterol gallstones. Gastroenterology 1989; 97:1479– 1484.
Page 294
196. Thompson JC, Fried GM, Ogden MD, Fagan CJ, Inoue K, Wiener J, Watson LC. Correlation between release of CCK and contraction of the gallbladder in patients with gallstones. Ann Surg 1982; 195:670–676. 197. O'Leary DP, LaMorte WW, Scott TE, Booker ML, Stevenson J. Inhibition of prostaglandin synthesis fails to prevent gallbladder mucin hypersecretion in the cholesterolfed prairie dog. Gastroenterology 1991; 101:812–820. 198. Kuckenbecker SL, Doty JE, Pitt HA, DenBesten L. Salicylate prevents gallbladder stasis and cholesterol gallstones in the prairie dog. Surg Forum 1981; 32:154–155. 199. Neiderheiser DH, Harmon CK, Roth HP. Absorption of cholesterol by the gallbladder. J Lip Res 1976; 17:117–124. 200. Braghetto I, Antezana C, Hurtado C, Csendes A. Triglyceride and cholesterol content in bile, blood, and gallbladder wall. Am J Surg 1988; 156:26–28. 201. English M, Hopwood D. Lipid in the human gallbladder mucosa: a histochemical study by light and electron microscopy. J Pathol 1985; 146:333–336. 202. Jacyna MR, Ross PE, Bakar MA, Hopwood D, Bouchier IAD. Characteristics of cholesterol absorption by human gallbladder: relevance to cholesterolosis. J Clin Pathol 1987; 40:524–529. 203. Ross PE, Butt AN, Gallagher C. Cholesterol absorption by the gallbladder. J Clin Pathol 1990; 43:572–575. 204. Sahlin S, Stahlberg D, Einarsson K. Cholesterol metabolism in liver and gallbladder mucosa of patients with cholesterolosis. Hepatology 1995; 21:1269–1275. 205. Kaminski DL, Deshpande Y, Thomas L, Blank W. Evaluation of the role of prostaglandins E and F in human cholecystitis. Prostaglandins Leukot Med 1984; 16:109–120. 206. Kaminski DL, Deshpande YG, Westfall S, Herbold D. Evaluation of prostacyclin production by human gallbladder. Arch Surg 1989; 124:277–280. 207. Myers SI, Bartula L. Human cholecystitis is associated with increased gallbladder prostaglandin I2 and prostaglandin E2 synthesis. Hepatology 1992; 16:1176– 1179. 208. Kotwall CA, Clanachan AS, Baer HP, Scott GW. Effects of prostaglandins on motility of gallbladders removed from patients with gallstones. Arch Surg 1984; 119:709–712. 209. Hood KA, Gleeson D, Ruppin DC, Dowling RH. Gall stone recurrence and its prevention: the British/Belgian Gall Stone Study Group's postdissolution trial. Gut 1993; 34:1277–1288. 210. Marks JW, Bonorris GG, Albers G, Schoenfield LJ. The sequence of biliary events preceding the formation of gallstones in humans. Gastroenterology 1992; 103:566–570. 211. Marks JW, Uhler ML, Bonorris GG, Judd HL. Nucleation of biliary cholesterol, arachidonate, prostaglandin E2, and glycoproteins in postmenopausal women. Gastroenterology 1997; 112:1271–1276. 212. McKirdy ML, McKirdy HC, Johnson CD. Nonadrenergic noncholinergic inhibitory innervation shown by electrical field stimulation of isolated strips of human gallbladder muscle. Gut 1994; 35:412–416. 213. Yu P, Chen Q, Harnett KM, Amaral J, Biancani P, Behar J. Direct G protein activation reverses impaired CCK signalling in human gallbladders with cholesterol stones. Am J Physiol 1995; 269:G659–G665. 214. Yu P, Chen Q, Biancani P, Behar J. Membrane cholesterol alters gallbladder muscle contractility in prairie dogs. Am J Physiol 1996; 271:G56–G61. 215. Chert Q, Amaral J, Biancani P, Behar J. Excess membrane cholesterol alters human gallbladder muscle contractility and membrane fluidity. Gastroenterology 1999; 116:678–685. 216. Catnach SM, Fairclough PD, Trembath RC, O'Donnel LJ, McLean AM, Law PA, Wickham JE. Effect of oral erythromycin on gallbladder motility in normal subjects and subjects with gallstones. Gastroenterology 1992; 102:2071–2075. 217. Portincasa P, Di Ciaula A, Palmieri V, Baldassarre G, Palasciano G. Enhancement of gallbladder emptying in gallstone patients after oral cholestyramine. Am J Gastroenterol 1994; 89:909–914.
Page 295
218. Zoli G, Ballinger AB, Healy J, O'Donnell LJD, Clark ML, Farthing MJG. Promotion of gallbladder emptying by intravenous amino acids. Lancet 1993; 341:1240–1241. 219. Marzio L, Neri M, De Angelis C, Di Felice F, Celiberti V, Cuccurullo F. Effect of cisapride on gallbladder kinetics in normals and patients with decreased gallbladder response to a liquid meal. Curr Ther Res 1987; 42:895–900. 220. Marzio L, Di Felice F, Laico MG, Imbibo B, Lapenna D, Cuccurullo F. Gallbladder hypokinesia and normal gastric emptying of liquids in patients with dyspeptic symptoms: a double blind, placebo controlled trial with cisapride. Dig Dis Sci 1992; 37:262–267. 221. Zieganhagen DJ, Glimm E, Kruis W, Zehtner E. Oral cisapride increases gallbladder volume in volunteers. J Gastrointest Motil 1992; 4:119–123. 222. Ziegenhagen DJ, Heitz W, Kruis W, Pohl C, Zehtner E. Cisapride increases gallbladder volume in gallstone patients before and after extracorporeal shock wave lithotripsy. Aliment Pharmacol Ther 1993; 7:617–622.
Page 297
14— The Role of Intestinal Transit R. Hermon Dowling The Guy's, King's College and St. Thomas's Medical and Dental School, London, England I— Introduction The concept that slow intestinal transit, a deficiency of dietary fiber, and excess of refined carbohydrate intake might contribute to cholesterol gallstone formation is neither new (1–6) nor widely accepted. In part this is because over the past 30 years, thoughts about gallstone pathogenesis have been dominated by three different factors. First, there was an obsession about biliary cholesterol saturation (7,8). Second, it became clear that although supersaturation of bile with cholesterol was an essential prerequisite for cholesterol stone formation, supersaturation alone was not adequate (9); in addition, there had to be a nucleation defect due to an excess of promoters and/or a deficiency of inhibitors of cholesterol microcrystal precipitation (10–12). A third major factor—that of stasis of bile within the gallbladder (13–17) (secondary to impaired gallbladder motility and, in animals at least, to crystal trapping by excess mucus glycoprotein gel on the surface of the gallbladder mucosa)— also diverted attention away from the role of intestinal transit and other potentially important mechanisms, in cholesterol gallstone pathogenesis. The individual contributions to this socalled triple defect (18,19)—supersaturation, nucleation defects, and gallbladder stasis—are discussed fully elsewhere in this book. Their importance is not in question and this is recognized by the editor with multiple relevant chapters in this book. However, the brief for this chapter was to extend the limited evidence (4,20) incriminating intestinal transit in the development of cholesterolrich gallbladder stones. In fact, the different pathogenetic mechanisms discussed above are not mutually exclusive and may well be interrelated. It is just conceivable that all the various mechanisms are secondary to changes in intestinal transit. However, the argument is not about the relative importance of the ''lithogenic liver" versus the "guilty gallbladder" versus (now) the "indolent intestine," but rather about the attempt to develop a unifying hypothesis that might even harmonize the apparently divergent theories about gallstone formation. This hypothesis is based on the assumption that prolonged intestinal transit increases the formation and solubilization of deoxycholic acid (DCA) in the colon and absorption from it (21). It also depends on the validity of the evidence that increased proportions of DCA in bile promote biliary cholesterol hypersecretion (22,23), supersaturation, and stone formation. This too is controversial; therefore the evidence for and against this assumption is also reviewed. The story begins with the observation in the late 1980s (24,25) that some acromegalic patients treated long term with the somatostatin analogue octreotide developed gallbladder stones (26).
Page 298
II— Effect of Octreotide Treatment on the Prevalence/Incidence of Gallbladder Stones Patients with somatostatinsecreting tumors (somatostatinomas) are known to have a high prevalence of gallstones (27). Partly for this reason, clinicians treating patients with octreotide (OT; a relatively longacting analogue of native human somatostatin) were alert to the possibility that chronic OT treatment might induce gallstone formation. That it does so is now beyond dispute. Figure 1 summarizes the reported incidence of new gallstone formation in acromegalic patients treated long term with OT. As results in this figure indicate, in the different reports the dose and duration of OT treatment varied widely—as did the de novo development of gallstones. (In these 18 studies, all patients were gallstone free by ultrasound before the OT treatment was introduced.) Thus the incidence of stone development ranged from 10% in one study (28) to 60% in another (29). Despite this, most patients received 100 to 200 g of OT three times daily and, after 1 to 2 years of treatment, just under 30% of patients developed iatrogenic gallbladder stones (Fig. 1).
Figure 1 Reported incidence of gallbladder stones (GBS) in acromegalic patients treated with octreotide (OT) in doses ranging from 100 to 1500 g/day for periods ranging from 3 to 70 months. (Most patients received 100 to 200 g thrice daily by subcutaneous injection for approximately 1 to 2 years.) Since at the start of OT treatment these patients were free of GBS by ultrasound, the results of these 18 studies represent the frequency of OTinduced (rather than OTassociated) GBS. The vertical bars represent the percentage of patients developing cholelithiasis: the numbers at the top of the columns refer to the numbers of patients developing stones over the numbers of patients treated. The broken horizontal line represents the mean incidence of OTinduced stones (29%) in these 18 studies. (From Ref. 119.)
Page 299
A— Composition of OctreotideInduced Gallbladder Stones Based on rather scant evidence, it was initially assumed that all of these OTassociated stones were cholesterolrich. However, in common with "conventional" gallstones (developing spontaneously in the absence of acromegaly or octreotide treatment), most patients with OTinduced gallbladder stones (GBS) were symptom free and therefore did not warrant cholecystectomy (30). As a result, only a few patients with OTassociated gallstones came to surgery, when their stones could be retrieved and analyzed for their chemical composition. In these patients, the stones were indeed cholesterolrich (>70% cholesterol by weight) (31). But in those who did not undergo cholecystectomy, a number of indirect methods of estimating stone composition was used. These included (a) bile microscopy and studies of bile lipid composition, (b) the use of computed tomography (CT) scanning in vivo (gallstone attenuation scores, measured in Hounsfield units, predict stone composition) (32,33), and (c) the dissolution response of the gallstones to oral ursodeoxycholic acid therapy (34,35). Taking the results of these direct and indirect studies together, the consensus is that OTinduced stones are indeed cholesterolrich (31). But like most gallstones in industrialized societies, they were not pure cholesterol stones: rather, they were mixed in composition, containing, in addition to cholesterol, small amounts of bile pigments, calcium salts, and amorphous material. III— Mechanism for OctreotideInduced Gallbladder Stone Formation A— The Role of Gallbladder Emptying Acromegaly is characterized by high circulating levels of both growth hormone (GH) and insulinlike growth factorI (IGFI). OT is an effective treatment for acromegaly: it acts by suppressing GH and IGFI levels (36–38). However, this suppressive effect of OT is not specific: the somatostatin analogue also inhibits the mealstimulated release of many peptide hormones from the gastrointestinal tract, including cholecystokinin (CCK). In turn, suppression of CCK release is the principal but not the sole mechanism whereby both native somatostatin (39,40) and OT (41–45) inhibit postprandial gallbladder emptying. Thus, a single 50 g subcutaneous injection of OT virtually "paralyzes" the GB by abolishing mealstimulated gallbladder emptying for at least 4 h (44). Since longterm OT treatment is usually given thrice daily by subcutaneous injection, it was initially assumed that somatostatin analogueinduced gallstones were caused by gallbladder stasis alone. The author's team certainly confirmed that OT dramatically impairs GB emptying in response to a fatrich liquid test meal (45). However, given the fact that the pathogenesis of cholesterolrich gallstones is multifactorial, it seemed important to study not only gallbladder emptying but also bile composition and physical chemistry in acromegalic patients with OTassociated GBS. B— Bile Lipid Composition and Physical Chemistry Most but not all (46) investigators believe that to study the distribution of biliary cholesterol between vesicles and micelles and to measure the cholesterol microcrystal nucleation time (or, more correctly, the crystal detection or appearance time), one needs to study fresh gallbladder bile—rather than bilerich duodenal fluid or the mixture of hepatic and gallbladder bile that is obtained during endoscopic retrograde cholangiopancreatography (ERCP). Therefore the controversial approach of ultrasoundguided, percutaneous, transhepatic, fineneedle puncture of the gallbladder (47–49) was used to aspirate samples of fresh gallbladder bile from (a) stone free acromegalic patients untreated with OT (the closest approximation available to a control group), (b) acromegalics with OTassociated gallbladder stones, and (c) patients with conventional, cholesterolrich GBS unrelated to acromegaly or OT treatment. In these fresh samples, bile acid and bile lipid composition were measured and bile physical chemistry (the
Page 300
distribution of cholesterol between vesicles and micelles and the microcrystal nucleation/observation time) was studied (50). The results of these studies showed that all the acromegalic patients with OTassociated GBS had bile that was supersaturated with cholesterol. The excess biliary cholesterol was found mainly in the vesicular fraction, and the molar ratio of cholesterol:phospholipids (CH:PL) in these vesicles was high. High CH:PL molar ratios in biliary vesicles predict rapid nucleation of cholesterol microcrystals (51)—a prediction that was confirmed by direct measurement in the patients with OTGBS (50). In fact, the pattern of results in the patients with OTassociated gallstones was virtually identical to that in the "disease controls" with conventional gallstone disease. However, the changes in the bile lipid composition and physical chemistry were apparently due to the OT treatment rather than secondary to the presence of gallstones. Thus, when paired, before and during OT treatment studies were carried out in a small number of patients, the bile lipid changes were found to have developed within weeks of starting the OT, independent of stone formation (50). C— Biliary Bile Acid Composition The next chapter in the saga relates to induced changes in the bile acid composition and the concept that the bile lipid findings might be secondary to increases in the percentage of the hydrophobic DCA in bile. When biliary bile acid composition was measured by highperformance liquid chromatography (HPLC), the mean proportion of DCA (percentage of total bile acids) was approximately twice as high in the gallstone carriers as in the stonefree individuals (50). Furthermore, in the paired studies, the mean proportion of DCA increased significantly from approximately 12% before OT to around 24% during longterm treatment with the somatostatin analogue. At the same time, the biliary cholesterol saturation increased significantly and the bile became supersaturated in cholesterol, again independent of the presence or absence of iatrogenic stone formation. IV— Mechanism for the Increased Percentage of DCA in Bile during OT Treatment A— Small Bowel Transit Why OT should induce an increase in the percent DCA in bile was not immediately obvious. However, a possible explanation came from studies of small bowel transit, measured by the breath hydrogen technique, in nonacromegalic individuals (control subjects and patients with the irritable bowel syndrome) given a single subcutaneous injection (50 g) of OT 30 min before a liquid test meal "spiked" with lactulose (a substrate for breath hydrogen production). The results of four separate studies (41,52–54), in all of which a similar protocol was used, showed that OT markedly prolongs small bowel transit time—as measured by the breath hydrogen technique (55). However, when the Guy's studies began, there was little or no information about the effects of chronic (as opposed to the acute effect of a single injection) OT treatment in either control subjects or acromegalic patients, and no information about the effects of OT on large bowel transit—of importance, since the colon is the major intestinal site for DCA production (56,57). B— Large Bowel Transit The results of studies by the author's group (45) confirmed that a single 50 g injection of OT significantly prolonged small bowel transit both in control subjects and in acromegalic patients. More important (at least in the context of iatrogenic gallstone formation), chronic OT
Page 301
treatment also prolonged large bowel transit time (LBTT) as measured by the progress of ingested radiopaque marker shapes through the intestine (58). This method is simple and noninvasive, but it is a relatively crude technique that yields a considerable scatter of results (K.W. Heaton, personal communication). This may explain why, initially, no significant difference was found in the results for LBTT between acromegalic patients untreated and treated with OT (45). However, when LB. was measured before and during OT treatment, with each individual acting as his or her own control, it became clear that the somatostatin analogue did indeed significantly prolong LBTT by an average of approximately 15 h (59,60). C— Relationship between Large Bowel Transit Time and the Percentage of DCA in Serum/Bile For ethical reasons, we did not feel justified in extending our studies of bile composition in samples obtained by percutaneous fineneedle puncture of the gallbladder. Instead, as part of a study in which the size of the bile acid pool and the bile acid "synthesis" rates were measured (61) using serum sampling and stable isotopes (13C and 2Hlabeled bile acids) (62), we analyzed fasting serum bile acid composition by gas chromatographymass spectrometry (GCMS). In paired before and during treatment studies, we found that not only LBTT but also the percentage of DCA in fasting serum increased during the OT therapy (59,60). This raised an obvious question: Is there a relationship between colonic transit and the percent DCA in fasting serum (and, by implication, in gallbladder bile)? To answer this question, the study was extended, initially, to 64 individuals [control subjects and acromegalic patients ± longterm OT treatment (60)] in whom a highly significant (p < 0.001) linear relationship between the percentage of DCA in fasting serum and LBTT was found. This relationship has now been confirmed in over 200 individuals (Fig. 2) from five different studies (Veysey et al., unpublished observations): in other words, the longer the colonic transit time, the higher the proportion of DCA in serum and, again by inference, in bile.
Figure 2 Relationship between the proportion of deoxycholic acid (DCA) expressed as a percentage of total bile acids in fasting serum, measured by gas chromatographymass spectrometry, and large bowel transit time (LBTT), assessed by recording the progress of radioopaque marker shapes through the intestine. The data in these 207 individuals come from five separate studies (21,60,61,118,120) in which a similar pattern of results was found.
Page 302
D— Relationship between LBTT and DCA Pool Size/Formation Rate In some ways, the attempt to correlate LBTT and the percentage of DCA in fasting serum was a "chalk and cheese" comparison. The proportion of the individual bile acids in serum was measured in a "snapshot" by an exquisitely sensitive technique (63). In contrast, LBTT was measured over a 4day period using a relatively crude technique. The next step, therefore, was to measure the DCA pool size and the DCA formation or "synthesis" rate (input into the enterohepatic circulation) rate using stable isotope dilution and serum sampling (62). When this was done, the results of paired studies again showed that during OT treatment, there was a significant increase in both the DCA pool size and its formation rate with a corresponding decrease in the pool size of the parent bile acid, cholic acid (CA) (61,64). Indeed, by using dual isotopes, it was possible to measure the conversion rate of 13Clabeled CA into 13Clabeled DCA in serum (64). Both before and during OT treatment, there were highly significant linear relationships between (a) LBTT and the DCA pool size (61,64) and (b) LBTT and DCA formation (61,64), both variables now being measured over the same 4 to 5day period. Moreover, during OT treatment, there were significant increases in the conversion rate of 13CCA to 13CDCA on days 2, 3, and 4 of the study (64). V— Is the Percentage of DCA in Serum a Valid Marker for the Percentage of DCA in Bile? How valid is it to use the percent DCA in fasting serum as an indicator of the percent DCA in bile? To date, the evidence that it is limited: further studies are needed to confirm (or refute) that such a relationship exists. For two reasons, however, it seems that the bile acid composition of serum does, in fact, provide a valid surrogate marker for that in bile. First, although the total bile acid concentration in the peripheral circulation (approx 5 to 15 µM) is only onethousandth of that in the intestinal lumen (around 5 to 15 mM), which, in turn, is between onetenth and onetwentieth of that in concentrated gallbladder bile (100 to 200 mM); ultimately, the bile acid pools in these different "compartments" must be in equilibrium. Indeed, this is the justification for sampling serum, rather than bilerich duodenal fluid, when measuring pool sizes and turnover rates by isotope dilution (61,62). This does not imply that from minute to minute, or even from hour to hour, the bile acid composition in the different compartments will necessarily be the same. Indeed, the ability of the liver to take up bile acids from the portal blood (firstpass extraction efficiency) varies considerably from one bile acid to the next. This is explained, at least in part, by the avidity of the bile acid binding to albumin (and other proteins in the blood) (65,66). Thus, the firstpass extraction efficiency of the liver for the conjugates of cholic acid is high, while that for unconjugated secondary bile acids, such as DCA, is low (65–67). This results in a selective "spillover" of bile acids into the peripheral circulation, which inevitably leads to a temporary mismatch between the pattern of bile acids in serum and that in bile—particularly after a meal. Indeed, the turnover of bile acids in serum/plasma is very rapid, the t 1/2 being <10 min (68). However, over the course of a day (and certainly over the course of the 4 to 5 days required to measure bile acid kinetics by isotope dilution), these temporary inconsistencies are ironed out. These considerations were fully addressed by Stellaard and colleagues (62) in their validation of the serum sampling method when using stable isotopes to measure bile acid pool size and turnover. Second, the question has been addressed directly by Nagengast et al. (69), who compared the percentage of DCA in serum and the percentage of DCA in bile from 74 individuals. They showed that within an 8% variation, the molar percentage of DCA in bile could be predicted from the percentage of DCA in serum using a simple formula. It must be admitted, however, that for other bile acids, such as ursodeoxycholic acid (UDCA), the concordance between serum and biliary bile acids is variable. In some studies it is high (70,71), while in others it is
Page 303
indifferent (72,73). The reason for the variable concordance is unclear but may relate to the fact that in some of these studies, the serum bile acids were measured using suboptimal methods (gas chromatography) in disease states, such as primary biliary cirrhosis (73). VI— Evidence That DCA Influences Biliary Cholesterol Secretion, Saturation, and the Risk of Gallstone Formation A detailed discussion of this topic is beyond the scope of this chapter. However, since much of the argument advanced to date is based on the assumption that prolonged intestinal transit influences CH gallstone formation through its effect on DCA metabolism, the question about the central role of DCA in gallstone formation is not a trivial one. It demands at least a brief response. A— Percentage of DCA in Bile: Gallstone Carriers versus StoneFree Controls In 1988, Marcus and Heaton (4) reviewed the evidence incriminating DCA in the pathogenesis of CH gallstones. At that time, they cited 20 studies from the literature in which biliary bile acid composition had been compared in stonefree controls and patients with presumed CHrich gallstones. In 19 of the 20 comparisons, there was more DCA (percentage of total bile acids) in the bile of the gallstone carriers than in that of the controls. However, these differences were statistically significant in only 7 of the 19 comparisons—although in some of the nonsignificant comparisons, the numbers of control subjects and gallstone carriers were relatively small, with the attendant risk of a type II error. Marcus and Heaton (4) also noted that the percentage of DCA in bile was greater than normal in individuals with risk factors for developing GBS—namely increased age, female sex (gender), hypertriglyceridemia, and "not being a vegetarian." Since the 1988 review, there have been many subsequent publications in which similar comparisons of biliary bile acid composition have been made between stone carrying and stonefree groups. Most (21,50,74–77) but not all (78–82) confirmed that the proportion of DCA in bile is indeed significantly higher in gallstone patients than in matched controls. B— Effect of Increasing the Percentage of DCA in Bile on Biliary Cholesterol Secretion Two groups have shown that DCA enrichment of bile induces biliary cholesterol hypersecretion. Thus, Carulli et al. (22) studied postcholecystectomy Ttube patients in whom they had depleted the bile acid pool by external biliary drainage or "washout." They then replaced the endogenous bile acids with exogenous bile acids by intraduodenal infusion. This enriched the Ttube bile with the infused bile acid, and when DCA became the dominant bile acid (>90 to 95%), there was hypersecretion of biliary CH. Leiss and von Bergmann (23) used a different technique—that of steadystate secretionperfusion to measure the hourbyhour output of biliary lipids under the influence of different bile acids. Once again, in the presence of DCA, there was relative hypersecretion of biliary CH. The mechanism whereby DCArich bile induces biliary CH hypersecretion may involve the "leaching" of CH from selected domains of the outer hemileaflet of the canicular membrane as a function of bile acid hydrophobicity. Certainly the linear relationships between biliary CH and biliary bile acid secretion were much steeper when the bile was enriched with hydrophobic bile acids, such as DCA, as opposed to hydrophilic bile acids, such as chenodeoxycholic and ursodeoxycholic acids (22).
Page 304
C— The Relationship between the Percentage of DCA and the Moles Percent Cholesterol, the CH Saturation Index, and the Percentage of Arachidonic AcidRich Phospholipids in Bile A number of investigators have shown that there are significant linear relationships between the percentage of DCA in bile and (a) the moles percent cholesterol in GB bile (74,75,77,84–88), (b) the CH saturation index (CSI) (74,75,77,84–88), and (c) the CH mass in the gallbladder (89). Thus, in the American National Cooperative Gallstone Study, Hofmann et al. (85) showed that before treatment, men but not women with gallstones had a significant linear relationship between the percentage of DCA in bile, and both the moles percent CH, and the CSI. Similarly, Hussaini et al. (88) examined the relationship between these parameters in several groups of patients in whom the proportion of DCA in bile varied spontaneously rather than as a result of bile acid feeding (3,90), antibiotic therapy (90,91), or manipulation of the rates of intestinal transit (see below). Hussaini and colleagues also showed significant linear relationships between the percentage of DCA in bile and both the moles percent cholesterol and the CSI. This observation may explain why they also found that the percentage of DCA was significantly greater in individuals with abnormally rapid CH crystal nucleation times (<5 days) than in those with normal nucleation times (>10 days) (88). However, these findings have been challenged by others, who found no such relationship (82). Despite this, in model (artificial) biles, hydrophobic bile acids—such as DCA and its conjugates—induce faster and more extensive precipitation of various types of CH microcrystals, than hydrophilic bile acids, such as cholic acid and its conjugates (92). In humans, virtually all the biliary phospholipids are present as phosphatidylcholines. And when the percentage of DCA in bile is plotted against the percentage of phosphatidylcholines rich in arachidonic acid, both van Berge Henegouwen et al. (93) and Pereira et al. (94) showed, independently, that there are significant linear relationships between the two variables. In turn, high levels of arachidonic acidrich phosphatidylcholines (AAPCs) are risk factors for CH GBS formation (95,96). This may be due to the fact that after the AAPCs have been hydrolyzed (by phospholipases) and absorbed from the bile into the GB wall, the archidonic acid may serve as a precursor for prostaglandins. The prostaglandins so formed may influence the synthesis of mucus glycoprotein (MGP) and secretion by the gallbladder. (MGP is a potent promotor of CH crystal nucleation/precipitation and crystal trapping.) Prostaglandins may also affect GB motility (97). However appealing the logic, this theory contains many unproven assumptions. Thus, it is far from clear that the GB wall derives significant amounts of arachidonic acid from phospholipids in the GB lumen rather than from the circulation or as a result of synthesis in situ. D— Effect of Pharmacological Manipulation of Intestinal Transit on the Percentage of DCA and the Cholesterol Saturation Index in Bile A number of groups have shown that when intestinal transit is prolonged pharmacologically, the percentage of DCA and the CH saturation of bile both increase. This happens, for example, with loperamide (98) or octreotide (50). Conversely, when intestinal transit is accelerated with senna (98), lactulose (99), bran (100–102), or cisapride (103), the proportion of DCA and/or the cholesterol saturation of bile decrease. E— Effect of DCA Feeding on Biliary Cholesterol Saturation The fifth and weakest link in the chain of evidence incriminating DCA in the pathogenesis of gallstones comes from studies of bile acid feeding. Here the results are widely contradictory. Thus, when LowBeer and Pomare (3) fed normal people 100 to 150 mg of DCA per day for 2 weeks, there was a significant increase in the lithogenic (biliary CH saturation) index. Sim
Page 305
ilarly, when Carulli et al. (92) fed similar doses of DCA to gallstone patients, the mean CSI rose from 1.07 to 1.42. Conversely, when Ahlberg et al. (104), La Russo et al. (105), Carulli et al. (106) and Hillebrant et al. (78) fed DCA, they could not confirm the original observations. However, as is so often the case, the protocols used by different investigators varied considerably, and in these latter three studies the oral dose of DCA varied from 750 to 1000 mg/day. In an attempt to explain these discrepant results, the Bristol group suggested that the relationship between biliary CH saturation and the dose of DCA might conform to a "Starling curve." In other words, they suggested that the absence of effect of DCA feeding on the biliary CH saturation indices might be due to the fact that, in many of the subsequent studies, "pharmacological" (rather than physiological) doses of DCA were used. However, this explanation is not totally satisfactory. Ponz de Leon et al. (90) also fed large doses of DCA (800 to 1000 mg/day) and did find a significant rise in the lithogenic index, as did Di Donnato et al. (107), despite the fact that they fed small daily doses of DCA (180 to 250 mg/day), comparable to those used by LowBeer and Pomare in their original study (3). VII— Evidence That Prolonged Intestinal Transit and Altered DCA Metabolism Are Important in Conventional Cholesterol Gallstone Formation As indicated above, the author's interest in the role of intestinal transit in the pathogenesis of cholesterol gallstones began with studies of the unique human model of iatrogenic gallstone disease—that induced by OT. However interesting, the results of these studies would be of limited value if they applied only to this small group of patients. The question therefore arises: What is the evidence that prolonged colonic transit also plays a role in the genesis of "conventional" cholesterol gallstones? And if it does, how precisely does prolonged large bowel transit affect DCA metabolism? For example, does it influence DCA formation in the intestine? Does it favor the solubilization, and therefore the bioavailability, of the newly formed DCA? Or does the prolonged residence time in the colon simply allow more time for DCA absorption? A— Review of Published Literature To date, the number of published reports providing evidence in favor of a role for prolonged intestinal transit in conventional gallstone disease is limited (n = 5). However, the results of these studies all point in the same direction. For example, Heaton and colleagues from Bristol (108) compared wholegut transit time (measured directly in some and estimated indirectly in others) and fecal wet weight, in nonobese women with gallstones who had no known risk factors for cholelithiasis, and in matched stonefree controls. They showed that, on average, the wholegut transit time was almost 20 h longer in the gallstone patients than in the controls, while their mean fecal wet weight was approximately half that of the controls. To paraphrase their findings, the gallstone carriers seemed to suffer from slow transit constipation. Subsequently, the Bristol group traveled halfway round the world to carry out similar, although less detailed, studies in Ladakhi women from Northern India (109). The Ladakhis have a particularly high prevalence of gallstones and they also seem to suffer from constipation—allegedly because they suppress the urge to defecate outdoors in a primitive, lowtemperature mountainside environment. Then, in a study from Japan and Sweden, Shoda et al. (74) measured a large number of variables, again comparing gallstone patients with matched controls. These variables included measurements of intestinal transit, gallbladder emptying, and, in gallbladder bile, bile lipid and bile acid composition. It may well be unwise to "cherry pick" from the large number of results in this publication, but in support of the author's hypothesis, Shoda and colleagues (74) found that, when compared to controls, the gallstone patients had significantly prolonged small bowel
Page 306
transit times, approximately twice as much DCA (percentage of total bile acids), and supersaturated rather than unsaturated duodenal "bile." Azzaroli et al. from Bologna, Italy (76), also compared results in control subjects and gallstone patients. Despite the fact that DCA is thought to be formed in the colon (56,57) rather than in the small intestine, these authors also measured small rather than large bowel transit time by three different methods. Like the Japanese, they too found that oroileal, orocecal and duodenocolonic transit times were all significantly prolonged in the gallstone patients, with corresponding increases in the percentage of DCA and the CH saturation indices. Indeed, as noted above, these authors found significant linear relationships between small bowel transit and the percentage of DCA in bile. B— Roles of Cecal Anaerobes, Bile AcidMetabolizing Enzymes, and Intracolonic pH in the Formation and Solubilization of DCA in, and the Absorption of DCA from, the Colon At about the same time, Thomas et al. (21) conducted similar but independent studies in London. They, too, speculated that patients with conventional CHrich gallstones might have prolonged large bowel transit times. In their working hypothesis (21), they suggested that prolongation of LBTT might lead to increased numbers of total and Grampositive anaerobic bacteria in the proximal colon capable of bile acid deconjugation (removal of glycine or taurine by the intestinal bacterial enzyme cholyglycine hydrolase: CGH) and/or dehydroxylation (removal of the 7 hydroxyl group from unconjugated cholic acid, to form unconjugated deoxycholic acid by the enzyme 7 dehydroxylase: 7 DH). In other words, prolongation of large bowel transit could either increase the number of colonic anaerobes or the activity of the bile acidmetabolizing enzymes, either of which would favor increased DCA formation. Alternatively, the prolonged LBTT, if present, might increase the specific activities of the bile acidmetabolizing enzymes—that is, the amounts of enzyme per unit bacterial protein. The authors also hypothesized that transitinduced increases in fecal and/or colonic luminal pH (102,110) would favor the solubilization, and therefore the bioavailability, of newly formed unconjugated DCA for passive absorption from the colon. Finally, they suggested that any prolongation of colonic transit would allow more time for DCA absorption as the luminal contents traversed the colon. To test these working hypotheses, Thomas and colleagues (21) studied 20 individuals who were stonefree by cholecystosonography and 20 patients with GBS that were CHrich, as judged by maximum stoneattenuation scores of <100 Hounsfield units on in vivo CT scanning. All 40 individuals underwent clinically indicated colonoscopy, for which the contents of the left colon were washed out by instant enema, leaving those in the right side (cecum plus ascending colon) undisturbed and available for study. As before, LBTT was measured by the markershape technique (58). The pH profile throughout the gastrointestinal tract was measured by radiotelemetry (111–113) while the percentage of DCA in fasting serum was again quantitated by gas chromatographymass spectrometry (GCMS) (114). Then, in fresh homogenized cecal aspirates obtained at colonoscopy, the authors used serial dilution techniques to measure the number of total and Grampositive anaerobes and the activities of the two bile acidmetabolizing enzymes (CGH and 7 DH). The results of these studies confirmed that, compared to stonefree "controls," the gallstone carriers had significantly prolonged large bowel transit times. This was associated with two to threefold increases in the numbers of total and Grampositive anaerobes in the cecal aspirates. Assuming that the complement of bile acid metabolizing enzymes per bacterium did not change, it follows that the increase in anaerobic bacterial numbers leads to a corresponding increase in the mass (total amount) of enzymes present in the colon. In addition, however, the specific activity (units of enzyme per milligram of protein) of both CGH and 7 DH increased (significantly so only for 7 DH), allegedly because of substrateenzyme induction (115). The
Page 307
authors suggested that the combination of more time (prolonged LBTT) and more enzymes (as a result of more anaerobic bacteria and an increase in 7 DH specific activity), favored increased DCA formation. At the same time, the prolonged large bowel transit was associated with small but consistent and significant increases in colonic luminal pH—apparently because the slow transit allowed more time for shortchain fatty acid absorption (110,116,117). Parallel ex vivo studies showed that these subtle changes in pH markedly increased the solubilization, and therefore the bioavailability, of the newly formed DCA. The net result of these transitrelated increases in DCA formation and solubilization was enhanced DCA absorption (presumably by passive nonionic diffusion from the colon), as evidenced by significant increases in fasting serum DCA (percentage of total serum bile acids).
Figure 3 Flow diagram summarizing the results of several studies from the author's department (18,45,59,120–124) showing how prolongation of large bowel transit time (LBTT) might increase the proportion of deoxycholic acid (DCA) in the bile acid pool and in serum and bile. Thus, an increase in LBTT favors an increase in the numbers of Grampositive (gram + ve) anaerobes in the proximal colon. This increase in the numbers of anaerobes leads to increases in the total amounts (biomasses) of the intestinal bacterial bile acid metabolizing enzymes responsible for deconjugation [cholylglycine hydrolase (CGH)] and 7 dehydroxylation [7 dehydroxylase (7 DH)] of the conjugated bile acids in the cecum and colon. The resultant increase in deconjugation means that more cholic acid (CA) is formed in the proximal colon, and this, in turn, may increase the specific activity (spec act) of 7 DH. The combination of increased total amounts of 7 DH and increased 7 DHspecific activity favors enhanced DCA formation. At the same time, the prolongation in LBTT increases colonic luminal pH, which ensures that the newly formed unconjugated DCA is solubilized in the colon and is therefore made bioavailable for passive nonionic (n.i.) diffusion from the colon. This expands the proportion of DCA in the bile acid pool, which is in dynamic equilibrium with the percentage of DCA in serum and bile. The consequences of increased proportions of DCA in bile are summarized in Fig. 4. (From Ref. 119.)
Page 308
Figure 4 Schematic flow diagram explaining how an increase in the percentage of deoxycholic acid (%DCA) in bile might lead to an increased formation (Formn) of cholesterol gallbladder stones (CHGBS). Thus, enrichment of bile with DCA leads to a relative increase in biliary cholesterol secretion (secrn) (22). This may explain way we (88) and others (85,125) find that there is a significant linear relationship between the %DCA in bile and the molar percentage (moles %) cholesterol, and the cholesterol saturation index (SI) in gallbladder bile. It may also explain, at least in part, why we find that the mean %DCA in bile is approximately twice as high in patients with abnormally rapid (<5 days) nucleation or precipitation of cholesterol microcrystals, as in individuals with normal (>10 days) nucleation times (88). We (94) and others (83,93,126,127) also showed that the percentage of DCA in bile was linearly related to the percentage of arachidonic acidrich phospholipids (AAPL) in bile. In turn, this may explain (98) why mucus glycoprotein (MGP) synthesis by, and secretion into, the gallbladder are increased in gallstone patients (128–130). An increase in the proportion of AAPLs in GB bile could also contribute to the reduced mealstimulated gallbladder emptying (13–15,131) that characterizes cholesterol gallstone disease. (From Ref. 119.)
C— Unifying Hypothesis: Overall Summary A schematic flow diagram summarizing the results of these, and related, studies is shown in Fig. 3. This suggests in detail how prolongation of colonic transit might increase DCA formation, solubilization, and absorption, leading to increases in the DCA pool size and the percentage of DCA in serum and bile. In turn, the multiple pathways whereby increases in the percentage of DCA might favor cholesterol gallstone formation is illustrated in Fig. 4. VIII— Reversal of TransitInduced Abnormalities in DCA Metabolism with Intestinal Prokinetics If these hypotheses are correct, in theory it should be possible to reverse the multiple steps depicted in Figs. 3 and 4 by accelerating transit through the intestine. The results of several studies in the literature had already shown that the use of regimens that shorten intestinal transit time resulted in declines in the percentage of DCA and in the CH saturation index of gallbladder bile (98–102).
Page 309
In the authors' unit, this problem was addressed in acromegalic patients treated long term with OT. From a long list of possible therapeutic agents, the intestinal prokinetic drug cisapride was chosen for a prospective doubleblind controlled trial (118). The acromegalic patients were all taking longterm OT to which either cisapride or placebo was added, after which mouthtocecum and large bowel transit times were measured and the results related to the percent DCA in fasting serum. These findings were then compared with those in a historic disease control group of acromegalic patients untreated with octreotide. The study confirmed that longterm OT prolonged both small and large bowel transit times and increased the percent DCA in fasting serum. However, the use of the intestinal prokinetic cisapride, in a dose of 10 mg four times daily, completely reversed the prolongation in both small and large bowel transit and the increase in the percent in DCA serum induced by OT treatment. The implication of these results is that treatment of individuals who are at high risk of forming gallstones should prevent (a) the high percent DCA in bile, (b) supersaturated bile, and (c) cholesterol gallstone formation. This hypothesis has yet to be tested by prospective controlled trials but, for the first time, we now have soundly based scientific strategies on which to base such studies. Acknowledgments The author wishes to thank the following colleagues and collaborators whose work forms the basis of this review: Michael Besser, Gary French, Hyder Hussaini, Paul Jenkins, Tony Mallet, Gerry Murphy, Steve Pereira, Linzi Thomas, Martin Veysey, and John Wass. Thanks are also due to Ken Heaton, Phil Hylemon, and Gustav Paumgartner for their help in establishing laboratory methods, training junior colleagues, and stimulating original ideas and discussion. Financial support of the Special Trustees of Guy's Hospital, the John Ellerman Foundation, and the Sandoz/Novartis Companies (UK and Switzerland), is gratefully acknowledged. Last, but not least, the author wishes to thank Mrs. Ann Hollington for her invaluable help in preparing this manuscript. References 1. Cleave TL, Campbell GD, Painter NS. Diabetes, Coronary Thrombosis and the Saccharine Disease. Bristol, UK: John Wright, 1969. 2. Heaton KW. Effects of increased dietary fibre on intestinal transit. Clin Gastroenterol 1973; 2:67–83. 3. LowBeer TS, Pomare EW. Can colonic bacterial metabolites predispose to cholesterol gallstones? BMJ 1975; 1:438–440. 4. Marcus SN, Heaton KW. Deoxycholic acid and the pathogenesis of gall stones. Gut 1988; 29:522–533. 5. Misciagna G, Leoci C, Guerra V, et al. Epidemiology of cholelithiasis in southern Italy: Part II. Eur J Gastroenterol Hepatol 1996; 8:585–593. 6. LowBeer TS. How the colon begets gallstones. Lancet 1998; 351:612–613. 7. Isaksson B. On the dissolving power of lecithin and bile salts for cholesterol in human bladder bile. Acta Soc Med Upsalien 1954; 59:296–306. 8. Admirand WH, Small DM. The physicochemical basis of cholesterol gallstone formation in man. J Clin Invest 1968; 47:1043–1052. 9. Holzbach RT, Marsh M, Olszewski M, Holan K. Cholesterol solubility in bile: evidence that supersaturated bile is frequent in healthy man. J Clin Invest 1973; 52:1467–1479. 10. Holan KR, Holzbach RT, Hermann RE, et al. Nucleation time: a key factor in the pathogenesis of cholesterol gallstone disease. Gastroenterology 1979; 77:611– 617.
Page 310
11. Sedeghat A, Grundy SM. Cholesterol crystals and the formation of cholesterol gallstones. N Engl J Med 1980; 302:1274–1277. 12. Burnstein MJ, Ilson RG, Petrunka CN, Taylor RD, Strasberg SM. Evidence for a potent nucleating factor in the gallbladder bile of patients with cholesterol gallstones. Gastroenterology 1983; 85:801–807. 13. Fisher RS, Stelzer, F, Rock E, Melmud LS. Abnormal gallbladder emptying in patients with gallstones. Dig Dis Sci 1982; 27:1019–1024. 14. Forgacs IC, Maisey MN, Murphy GM, Dowling RH. Influence of gallstones and ursodeoxycholic acid therapy on gallbladder motor function. Gastroenterology 1984; 87:299–307. 15. Pomeranz IS, Shaffer EA. Abnormal gallbladder emptying in a subgroup of patients with gallstones. Gastroenterology 1985, 88; 787–791. 16. Patankar R, Ozmen MM, Bailey IS, Johnson CD. Gallbladder motility, gallstones, and the surgeon. Dig Dis Sci 1995; 40:2323–2335. 17. O'Donnell LJD, Fairclough PD. Gall stones and gall bladder motility. Gut 1993; 34:440–443. 18. Dowling RH, Gleeson D, Ruppin DC, Murphy GM and the British/Belgian Gallstone Study Group. Gallstone recurrence and postdissolution management. In: Paumgartner G, Stiehl A, Gerok W, eds. Enterohepatic Circulation of Bile Acids and Sterol Metabolism. Lancaster, UK: MTP Press, 1985, pp 361–369. 19. Carey MC. Formation of cholesterol gallstones: the new paradigms. In: Paumgartner G, Stiehl A, Gerok W, eds. Trends in Bile Acid Research. Dordrecht/Boston/London: Kluwer, 1989, pp 259–281. 20. Dowling RH, Veysey MJ, Pereira SP, Hussaini SH, Thomas LA, Wass JAH, Murphy GM. Role of intestinal transit in the pathogenesis of gallbladder stones. Can J Gastroenterol 1997; 11:57–64. 21. Thomas LA, Bathgate T, Veysey MJ, King A, French GL, Murphy GM, Dowling RH. Is cholelithiasis an intestinal disease? (abstr). Gut 1997; 41(suppl 3):A2. 22. Carulli N, Loria P, Bertolotti C, Ponz de Leon M, Menozzi D, Medici G, Piccagli I. Effects of acute changes in bile acid pool composition on biliary lipid secretion. J Clin Invest 1985; 74:616–624. 23. Leiss O, von Bergmann K. Comparison of biliary lipid secretion in nonobese cholesterol gallstone patients with normal, young, male volunteers. Klin Wochenschr 1985; 63:1163–1169. 24. McKnight JA, McCance DR, Crothers JG, Atkinson AB. Changes in glucose tolerance and development of gall stones during high dose treatment with octreotide for acromegaly. BMJ 1989; 299:604–605. 25. Wass JAH, Anderson JV, Besser GM, Dowling RH. Gall stones and treatment with octreotide for acromegaly (letter). BMJ 1989; 299:1162–1163. 26. Dowling RH, Hussaini SH, Murphy GM, Besser GM, Wass JAH. Gallstones during octreotide therapy—Metabolism: clinical and experimental. 1992; 41(suppl 2):22–33. 27. Krejs G, Orci L, Conlon JM, et al. Somatostatinoma syndrome. N Engl J Med 1979; 301:285–292. 28. Catnach SM, Anderson JV, Fairclough PD, et al. The effect of octreotide on gallstone prevalence and gallbladder motility in acromegaly. Gut 1993; 34:270–273. 29. McKnight JA, McCance DR, Sheridan B, et al. Four years' treatment of resistant acromegaly with octreotide. Eur J Endocrinol 1995; 132:429–432. 30. Attili A, Carulli N, Roda E, et al. Epidemiology of gallstone disease in Italy: prevalence data of the Multicenter Italian Study on Cholelithiasis (MICOL). Am J Epidemiol 1995; 141:158–165. 31. Hussaini SH, Pereira SP, Murphy GM, Kennedy C, Wass JAH, Besser GM, Dowling RH. Composition of gallbladder stones associated with octreotide: response to oral ursodeoxycholic acid. Gut 1995; 36:126–132.
Page 311
32. Rajagopal SU, Keightley A, Bills P, Walters JRF, Murphy GM, Dowling RH. Predictive value of pretreatment CT scanning vs conventional radiology in determining composition, dissolvability and fragmentability of gallbladder stones (GBS) (abstr). J Hepatol 1989; 9:S211. 33. Brakel K, Lameris JS, Nijs HGT, et al. Predicting gallstone composition with CT: in vivo and in vitro analysis. Radiology 1990; 174:337–341. 34. Petroni ML, Jazrawi RP, Grundy A, et al. Prospective, multicenter study on value of computerized tomography (CT) in gallstone disease in predicting response to bile acid therapy. Dig Dis Sci 1995; 40:1956–1962. 35. Pereira SP, Veysey MJ, Kennedy C, Hussaini SH, Dowling RH. Gallstone dissolution with oral bile acid therapy: importance of pretreatment CT scanning and reasons for nonresponse. Dig Dis Sci 1997; 42:1775–1782. 36. Plewe G, Beyer J, Krause U, Neufeld M, Del Pozo E. Longacting and selective suppression of growth hormone secretion by somatostatin analogue SMS 201 995 in acromegaly. Lancet 1984; 2:782–784. 37. Lamberts SWJ, Uiterlinden P, Verschoor L, Dongegen van KJ, Del Pozo E. Long acting treatment of acromegaly with the somatostatin analogue SMS 201995. N Engl J Med 1985; 313:1576–1580. 38. Melmed S. Acromegaly. N Engl J Med 1990; 322:966–977. 39. Reichlin S. Somatostatin. N Eng J Med 1983; 309:1495–1501, 1556–1563. 40. Fisher RS, Rock E. Levin G, et al. Effects of somatostatin on gallbladder emptying. Gastroentology 1987; 92:885–890. 41. Lembcke B, Creutzfeldt W, Schleser S, et al. Effect of somatostatin analogue Sandostatin (SMS 201995) on gastrointestinal, pancreatic and biliary function, and hormone release in normal men. Digestion 1987; 36:108–124. 42. van Liessum PA, Hopman WP, Pieters GF. Postprandial gallbladder motility during long term treatment with the longacting somatostatin analog SMS 201995 in acromegaly. J Clin Endocrinol Metab 1989; 69:557–562. 43. Mitsukawa T, Takemura J, Nishizono F, et al. Effects of atropine, proglumide and somatostatin analogue (SMS 201995) on bombesininduced gallbladder contraction and CCK secretion in humans. Am J Gastroenterol 1989; 84:1371–1374. 44. Stolk MFJ, van Erpecum KJ, Koppeschaar HPF, et al. Postprandial gallbladder motility and hormone release during intermittent and continuous subcutaneous octreotide therapy in acromegaly. Gut 1993; 34:808–813. 45. Hussaini SH, Pereira SP, Veysey MJ, Kennedy C, Jenkins P, Murphy GM, Wass JAH, Dowling RH. The roles of gallbladder emptying and intestinal transit in the pathogenesis of octreotide induced gallbladder stones. Gut 1996; 38:775–783. 46. Marks J, Broomfield P, Bonorris GG, Schoenfield LJ. Factors affecting the measurement of cholesterol nucleation in human gallbladder and duodenal bile. Gasteroenterology 1991; 101:214–219. 47. Swobodnik W, Hagert N, Janowitz P, Wenk H. Diagnostic fineneedle puncture of the gallbladder with US guidance. Radiology 1991; 178:755–758. 48. Herbetko J, Fache JS. Diagnostic fineneedle puncture of the gallbladder (letter). Radiology 1991; 180:586. 49. Hussaini SH, Kennedy C, Pereira SP, Wass JAH, Dowling RH. Ultrasoundguided percutaneous fine needle puncture of the gallbladder for studies of bile composition. Br J Radiol 1995; 68:271–276. 50. Hussaini SH, Murphy GM, Kennedy C, Besser GM, Wass JAH, Dowling RH. The role of bile composition and physical chemistry in the pathogenesis of octreotideassociated gallbladder stones. Gastroenterology 1994; 107:1503–1513. 51. Collins JJ, Phillips MC. The stability and structure of cholesterolrich codispersions of cholesterol and phosphatidylcholine. J Lipid Res 1982; 23:291–298. 52. Fuessl HS, Carolan G, Williams G, Bloom SR. Effect of a longacting somatostatin
Page 312
analogue (SMS 201995) on postprandial gastric emptying of 99mTctin colloid and mouthtocaecum transit time in man. Digestion 1987; 36:101–107. 53. Moller N, Petrany G, Cassidy D, et al. Effects of the somatostatin analogue SMS 201955 (sandostatin) on mouthtocaecum transit time and absorption of fat and carbohydrates in normal man. Clin Sci 1988; 75:345–350. 54. O'Donnell LJD, Watson AJM, Cameron D, Farthing MJG. Effect of octreotide on mouthtocaecum transit time in healthy subjects and in the irritable bowel syndrome. Aliment Pharmacol Ther 1990; 4:177–182. 55. Bond JH, Levitt MD. Investigation of small bowl transit time in man utilizing pulmonary hydrogen measurements. J Lab Clin Med 1974; 85:546–556. 56. Hill MJ, Drasar BS. The normal colonic bacterial flora. Gut 1974; 6:318–323. 57. Midtvedt T. Microbial bile acid transformation. Am J Clin Nutr 1974; 27:1341–1347. 58. Metcalf AM, Phillips SF, Zinsmeister AR, et al, Simplified assessment of segmental colonic transit. Gastroenterology 1987; 92:40–47. 59. Veysey MJ, Arraton SRD, Mallet A, Jenkins P, Murphy GM, Wass JAH, Dowling RH. Longterm octreotide treatment increases large bowel transit time (LBTT), the proportion of deoxycholic acid (%DCA) in serum and the risk of gallstone formation (abstr). Gut 1996; 39(suppl 3):A134. 60. Veysey MJ, Thomas LA, Mallet AI, Jenkins PJ, Besser GM, Wass JAH, Murphy GM, Dowling RH. Prolonged large bowel transit increases serum deoxycholic acid: a risk factor for octeotide induced gallstones. Gut 1999; 44:675–681. 61. Veysey MJ, Gathercole DJ, Mallet A, Jenkins P, Besser GM, Wass JAH, Murphy GM, Dowling RH. Large bowel transit time influences deoxycholic acid input rate and pool size—risk factors for octeotideinduced gallstones (abstr). Gastroenterology 1997; 112:A525. 62. Stellaard F, Paumgartner G, van Berge Henegouwen GP, et al. Determination of deoxycholic acid pool size and input rate using [24 13C] deoxycholic acid and serum sampling. J Lipid Res 1986; 27:1222–1225. 63. Setchell KDR, Matsui A. Serum bile acid analysis. Clin Chim Acta 1983; 127:1–17. 64. Veysey MJ, Mallett A, Jenkins P, Besser GM, Murphy GM, Dowling RH. Deoxycholic (DCA) and cholic acid (CA) kinetics in acromegalic patients treated with octreotide (OT) (abstr.). Gut 1998; 42(suppl 1):All. 65. Rudman D, Kendall FE. Bile acid content of human serum: II. The binding of cholanic acids by human plasma proteins. J Clin Invest 1957; 36:538–542. 66. Burke CW, Lewis B, Panveliwalla D, Tabaqchali S. The binding of cholic acid and its taurine conjugate to serum proteins. Clin Chim Acta 1971; 32:207–214. 67. Forker EL, Luxon B. Albumin helps mediate removal of taurocholate by rat liver. J Clin Invest 1981; 67:1517–1522. 68. LaRusso NF, Hoffman NE, Korman MG, Hofmann AF, Cowen AE. Determinants of fasting and postprandial serum bile acid levels in healthy man. Am J Dig Dis 1978; 23: 385–391. 69. Nagengast FM, van Munster IP, Salemans JMJI. Deoxycholic acid metabolism in patients with adenomas. Gastroenterology 1993; 105:955–956. 70. Bazzoli F, Roda A, Fromm H, Sarva RP, Roda E, Barbara L. Relationship between serum and biliary bile acids as an indicator of chenodeoxycholic and ursodeoxycholic acidinduced hepatotoxicity in the rhesus monkey. Dig Dis Sci 1982; 27:417–424. 71. Bazzoli F, Fromm H, Roda A, et al. Value of serum determinations for prediction of increased ursodeoxycholic and chenodeoxycholic levels in bile. Dig Dis Sci 1985; 30:650–654. 72. van de Meeberg PC, Wolfhagen FHJ, van Berge Henegouwen GP, et al. Single or multiple dose ursodeoxycholic acid for cholestatic liver disease: biliary enrichment and biochemical response. J Hepatol 1996; 25:887–894. 73. Lindor KD, Lacerda MA, Jorgensen RA, et al. Relationship between biliary and serum
Page 313
bile acids and response to ursodeoxycholic acid in patients with primary biliary cirrhosis. Am J Gastroenterol 1998; 93:1498–1504. 74. Shoda J, He BF, Tanaka N, et al. Increase of deoxycholate in supersaturated bile of patients with cholesterol gallstone disease and its correlation with de novo syntheses of cholesterol and bile acids in liver, gallbladder emptying, and small intestinal transit. Hepatology 1995; 21:1291–1302. 75. Galatola G, Jazrawi RP, Corrao G, Fracchia M. Biliary lipid determinants of bile lithogenicity and cholesterol gallstone formation (abstr). Gut 1996; 39:A104. 76. Azzaroli F, Mazzella G, De Vegori E, Festi D, Colcchia A, Villanova N, Simoni P, Mazzo C, et al. Sluggish gallbladder and small bowel motility are associated with cholesterol gallstones. Gastroenterology 1997; 112:A499. 77. Jazrawi RP, Northfield TC. Effects of a pharmacological dose of cholecystokinin on bile acid kinetics and biliary cholesterol saturation in man. Gut 1986; 27:355– 362. 78. Hillebrant CG. On the regulation of hepatic cholesterol and bile acid metabolism in gallstone patients. Thesis. Karolinska Institute at Huddinge University Hospital, Stockholm, Sweden, 1998. 79. Sahlin S, Stahlberg D, Einarsson K. Cholesterol metabolism in liver and gallbladder mucosa of patients with cholesterolosis. Hepatology 1995; 21:1269–1275. 80. Berr F, Pratschke E, Fischer S, Paumgartner G. Disorders of bile acid metabolism in cholesterol gallstone disease. J Clin Invest 1992; 90:859–868. 81. Berr F, KullakUblick GA, Paumgartner G, et al. 7alpha dehydroxylating bacteria enhance deoxycholic acid input and cholesterol saturation of bile in patients with gallstones. Gastroenterology 1996; 111:1611–1620. 82. Noshiro H, Chijiiwa K, Makino I, Nakano K, Hirota I. Deoxycholic acid in gall bladder bile does not account for the shortened nucleation time in patients with cholesterol gall stones. Gut 1995; 36:121–125. 83. Cantafora A, Di Biase A, Alvaro D, Angelico M, Marin M, Attili AF. High performance liquid chromatographic analysis of molecular species of phosphatidylcholine—development of quantitative assay and its application to human bile. Clin Chim Acta 1983; 134:281–295. 84. van der Linden W, Bergmann F. An analysis of data on human hepatic bile. Relationship between main bile components, serum cholesterol and serum triglycerides. Scand J Clin Lab Invest 1977; 37:741–747. 85. Hofmann AF, Grundy SM, Lachin JM, et al. Pretreatment lipid composition in white patients with gallstones in the National Cooperative Gallstone Study. Gastroenterology 1982; 83:738–752. 86. Alvaro D, Angelico F, Atilli AF, et al. Plasma lipid lipoproteins and biliary lipid composition in female gallstone patients. Biochim Biophys Acta 1986; 45:761– 768. 87. Rutgeerts P, Ghoos Y, Vantrappen J, Fevery J. Biliary lipid composition in patients with nonoperated Crohn's disease. Dig Dis Sci 1986; 31:27–32. 88. Hussaini SH, Pereira SP, Murphy GM, Dowling RH. Deoxycholic acid influences cholesterol solubilisation and microcrystal nucleation time in gallbladder bile. Hepatology 1995; 22:1735–1742. 89. Fracchia M, Jazrawi RP, Galatola G. Determination of gallbladder bile lithogenicity in patients with primary biliary cirrhosis. Ital J Gastroenterol 1996; 28:255– 260. 90. Ponz de Leon M, Carulli N, Iori R, Loria P, Romani M. Regulation of cholesterol absorption by bile acids: role of deoxycholic acid and cholic acid pool expansion on dietary cholesterol absorption. Ital J Gastroenterol 1993; 15:86–93. 91. LowBeer TS, Nutter S. Colonic bacterial activity, biliary cholesterol saturation, and pathogenesis of gallstones. Lancet 1978; 11:1063–1065. 92. Carulli N, Ponz de Leon M, Loria P, et al. Effect of selective expansion of the cholic acid pool on bile lipid composition: possible mechanism of bile acid induced biliary cholestrol desaturation. Gastroenterology 1981; 81:539–546.
Page 314
93. van Berge Henegouwen GP, van der Weft SDJ, Ruben AT. Fatty acid composition of phospholipids in bile in man: promoting effect of deoxycholate on arachidonate. Clin Chim Acta 1987; 165:27–37. 94. Pereira SP, Hussaini SH, Cassell TB, Murphy, GM, Wass JAH, Dowling RH. Biliary phospholipids and mucin glycoprotein are altered in octreofide induced gallstones (abstr). Gut 1995; 36(S1):A47. 95. Ahlberg J, Curstedt T, Einarsson K, et al. Molecular species of biliary phosphatidylcholine in gallstone patients. J Lipid Res 1981; 22:404–409. 96. Cantafora A, Angelico M, Di Blase A, et al. Structure of biliary phosphatidyl choline in cholesterol gallstone patients. Lipids 1981; 16:589–592. 97. Carey MC, Cahalane MJ. Whither biliary sludge? Gastroenterology 1993; 95:508–523. 98. Marcus SN, Heaton KW. Intestinal transit, deoxycholic acid and the cholesterol saturation of bile—three interrelated factors. Gut 1986; 27:550–558. 99. Thornton JR, Heaton KW. Do colonic bacteria contribute to cholesterol gallstone formation? Effects of lactulose on bile. BMJ 1981; 282:1018–1020. 100. Pomare EW, Heaton KW, LowBeer TS, Espiner HJ. The effect of wheat bran upon bile salt metabolism and upon the lipid composition of bile in gallstone patients. Am J Dig Dis 1976; 21:521–526. 101. Marcus SN, Heaton KW. Effects of a new, concentrated, wheat fibre preparation on intestinal transit, deoxycholic acid metabolism and the composition of bile. Gut 1986; 27:893–900. 102. Lewis SJ, Heaton KW. Increasing butyrate concentration in the distal colon by accelerating intestinal transit. Gut 1997; 41:245–251. 103. Veysey MJ, Jenkins P, Besser GM, Wass JAH, Dowling RH. The effect of cisapride on large bowel transit time: a randomised doubleblind placebocontrolled study in four groups of individuals. Gut 1997; 41(suppl 3):A123. 104. Ahlberg J, Angelin B, Einarsson K, Hellstrom L, Leijd B. Influence of deoxycholic acid on biliary lipids in man. Clin Sci Mol Med 1977; 53:249–256. 105. LaRusso NF, Szcepanik PA, Hofmann AF, Coffin SB. The effect of deoxycholic acid ingestion on bile acid metabolism and biliary lipid secretion in normal subjects. Gastroenterology 1977; 72:132–140. 106. Carulli N, Ponz de Leon M, Zironi F, Iori R, Loria P. Bile acid feeding and hepatic sterol metabolism: effect of deoxycholic acid. Gastroenterology 1980; 79:637–641. 107. DiDonato P, Carubbi F, Ponz de Leon M, Carulli N. Effect of small doses of deoxycholic acid on bile cholesterol saturation in patients with liver cirrhosis. Gut 1986; 27:23–26. 108. Heaton KW, Emmett PM, Symes CL, Braddon FEM. An explanation for gallstones in normalweight women: slow intestinal transit. Lancet 1993; 341:8–10. 109. Spathis A, Heaton KW, Emmett PM, Norboo T, Hunt L. Gallstones in a community free of obesity but prone to slow intestinal transit. Eur J Gastroenterol Hepatol 1997; 9:201–206. 110. El Oufir L, Flourie B, Bruley des Varannes S, et al. Relations between transit time, fermentation products, and hydrogen consuming flora in healthy humans. Gut 1996; 38:870–877. 111. Bown RL, Gibson JA, Sladen GE, Hicks B, Dawson AM. Effects of lactulose and other laxatives on ileal and colonic pH as measured by a radiotelemetry device. Gut 1974; 15:999–1004. 112. Evans DF, Pye G, Bramley R, et al. Measurement of gastrointestinal pH profiles in normal ambulant human subjects. Gut 1988; 29:1035–1041. 113. Fallingborg J, Christensen LA, IngemanNielson M, et al. pH profile and regional transit times of the normal gut measured by a radiotelemetry device. Aliment Pharmacol Ther 1989; 3:605–613. 114. Goto J, Watanabe K, Miura H, et al. Trace analysis of bile acids by gas chromatographymass spectrometry with negative ion and chemical ionization detection. J Chromatogr 1987; 388:379–387.
Page 315
115. White BA, Lipsky RH, Fricke RJ, Hylemon PB. Bile acid induction specificity of 7alphadehydroxylase activity in an intestinal Eubacterium sp. Steriods 1980; 35:103–109. 116. Newmark HL, Lupton JR. Determinants and consequences of colonic luminal pH: implications for colon cancer. Nutr Cancer 1990; 14:161–173. 117. Hond ED, Heile M, Evenepoel P, Peeters M, Ghoos Y, Rutgeerts P. In vivo butyrate metabolism and colonic permeability in extensive ulcerative colitis. Gastroenterology 1998; 115:584–590. 118. Veysey MJ, Arraton SRD, Mallet A, Jenkins P, Murphy GM, Wass JAH, Dowling RH. Does cisapride overcome the effects of octreotide on intestinal transit, thereby reducing the proportion of deoxycholic acid in bile and serum? In: Paumgartner G, Stiehl A, Gerok W, eds. Bile Acids in Hepatobiliary Disease. Dordrecht: Kluwer, 1997, pp 82–91. 119. Dowling RH. The gallstone story. Ulster Med J 1998; 67:20–24. 120. Veysey MJ, Arraton SRD, Gilani SS, et al. The relationship between large bowel transit time (LBTT) and the proportion of deoxycholic acid (%DCA) in serum (abstr). Gut 1996; 38(suppl 1):A53. 121. Veysey MJ, Mallet A, Murphy GM, Dowling RH. Deoxycholic acid pool size and input rate, measured by stable isotope dilution, are increased in patients with slow transit constipation (abstr). Clin Sci 1997; 92:3P. 122. Thomas LA, Veysey MJ, Murphy GM, Dowling RH, King A, French GL. Bile acid metabolising intestinal bacterial enzyme activity: a novel factor in cholesterol gallstone pathogenesis. Gut 1997: 40(suppl 1):A67. 123. Thomas LA, Bathgate T, Veysey MJ, King A, French GL, Murphy GM, Dowling RH. Do changes in colonic luminal pH explain the increased proportions of serum and biliary deoxycholic acid seen in patients with cholesterol gallbladder stones (GBS)? (abstr). Gut 1997; 41(suppl 3):A32. 124. Veysey MJ, Arraton SRD, Mallet A, Jenkins P, Murphy GM, Wass JAH, Dowling RH. Cisapride reverses the effects of octreotide (OT) on intestinal transit and the proportion of deoxycholic acid (%DCA) in bile and serum (abstr). Gut 1996; 39(suppl 3):A103. 125. Berr F, Schrieber E, Frick U. Interrelationships of bile acid and phospholipid fatty acid species with cholesterol saturation of duodenal bile in health and gallstone disease. Hepatology 1992; 16:71–81. 126. Angelico M, Corradini GS, Masella R, Alvaro D, Cantafora A, Capocaccia L. Molecular composition of biliary phosphatidylcholines, as related to cholesterol saturation, transport and nucleation in human gallbladder bile. J Hepatol 1992; 15:59–66. 127. Hatsushika S, Tazuma S, Kajiyama G. Nucleation time and fatty acid composition of lecithin in human gallbladder bile. Scand J Gastroenterol 1993; 28:131– 136. 128. Lee SP. Lessons from experimental cholelithiasis: gallbladder and mucosa, nonsteroidal antiinflammatory drugs, and gallstones. Gastroenterology 1991; 101:857–860. 129. Marks JW, Bonorris GG, Albers G, Schoenfield LJ. The sequence of biliary events preceding the formation of gallstones in humans. Gastroenterology 1992; 103:566–570. 130. Shiffman ML, Sugarman HJ, Kellum JM, Moore EW. Changes in gallbladder bile composition following gallstone formation and weight reduction. Gastroenterology, 1992; 103:214–221. 131. Thompson JC, Fried GM, Ogden DW, et al. Correlation between release of cholecystokinin and contraction of the gallbladder in patients with gallstones. Ann Surg 1982; 145:670–675.
Page 317
15— Calcium Salt Precipitation in Bile and Biomineralization of Gallstones Huguette Lafont INSERM U476, Marseilles, France J. Donald Ostrow University of Amsterdam, Amsterdam, The Netherlands I— Introduction: The Structure of Gallstones Gallstones are organized structures in which crystals of cholesterol and calcium salts are arrayed on a matrix composed of mucin polymer and small regulatory proteins (1–3). Virtually all the mineral components of human gallstones (4–10) and their localization within the various types of gallstones (11–16) have been characterized. Gallstones are usually classified into four types (Tables 1 and 2) according to the proportion of cholesterol and calcium salts (especially bilirubinates) and the gross arrangement of these components (4,6,17–22). In addition to the cholesterol, mixed, brown pigment, and black pigment stone types, a rarer fifth type has been described, called a salt and pepper stone (Table 1) (17). There is considerable overlap among these groups, however, and gallstones actually exhibit a wide spectrum of compositions, ranging from over 98% to less than 5% cholesterol; the remainder is composed mainly of calcium salts (especially bilirubinates) and the proteins and glycoproteins of the matrix (Table 2). All gallstones, including the core of ''pure" cholesterol gallstones, contain calcium salts (2,8,12,15,16,23–29) of one or more of the five "calciumsensitive" anions (which form poorly soluble calcium salts: bilirubinates, phosphates, carbonates, fatty acylates, and bile salts (9,30,31). Thus, gallstone formation is not simply a process of cholesterol precipitation. It has been proposed that calcium salt precipitation may initiate the formation of all types of gallstones (9). Each compound found in gallstones is derived by precipitation from bile. This may occur only if the bile is thermodynamically supersaturated with that component. Supersaturation in itself is not sufficient to cause precipitation; rather, precipitation of cholesterol from biliary vesicles and of calcium salts from the bulk fluid phase of bile, seems to be governed by regulatory proteins (kinetic factors) in bile (9,32,33). In gallstones, the cholesterol crystals and calcium salt precipitates are not simply clumped together, but are arrayed on a network of organic matrix (2,33–35), which consists mainly of polymerized mucin gel (1,34,36–38), to which are bound smaller proteins, including amphipathic anionic polypeptide/calciumbinding protein (APF/CBP) (3,39–42) and probably aminopeptidaseN (APN) (3,43) and others (43). Examining the cut surface of some stones, the cholesterol and calcium salts sometimes seem to be distributed diffusely and randomly, but there is evidence for sequential waves of deposition of crystals of different composition as
Page 318 Table 1 Characteristics of Major Types of Gallstonesa Stone type
Cholesterol
Mixed
Brown pigment
Black pigment
Main components
Cholesterol
Cholesterol pigments
Ca bilirubinates, Ca fatty acylates, cholesterol
Gross appearance
Round, shiny yellow white, pigmented center
Round or faceted, brown pigment in rings or specks
Ovoid or irregular, Faceted or spiky, brown to orange, soft, black, shiny or dull laminated
Number in GB
Usually single
Usually multiple
Single or multiple
Many
Usual site
Gallbladder
Gallbladder ± ducts
Bile ducts
Gallbladder ± ducts
Bile bacteria
Culture (), DNA (+)
Culture (), DNA (+)
Infected (E. coli)
Culture (), DNA (+)
Geography
Mainly in West
Mainly in West
Mainly in Orient
West and Orient
Disease/drug associations
Multiparity, obesity, diabetes, octreotide
Multiparity, obesity, diabetes, octreotide
Postcholecystectomy, Hemolysis, cirrhosis strictures, sutures, lowprotein diet
a
Pigment polymer, Ca phosphates and/or carbonates
A fifth type of gallstone, called a saltandpepper stone, has been described (17). It is rare in western countries and is composed of white crystals of calcium carbonate or yellowishwhite crystals of cholesterol compacted with black to brown spherules of pigment, which is predominantly polymerized.
Page 319 Table 2 Composition of Major Types of Gallstonesa,b Stone type
Cholesterol
Cholesterolc
60–99% d
Total pigment
Mixed
Brown pigment
Black pigment
0–47%
0g
46–86%
<2%
3–20%
12–61%
12–59%
N.D.
50–77%
>80%
Pigment polymere
N.D.
Ca phosphate
0
0–8%
0
0–56%
Ca carbonate
0
0–9%
0
0–37%
0
0
Ca fatty acylates Ca bile salts
<3%
<3%
5–20%
0
<7%
<7%
Residuef
1–35%
0–49%
a
27–76%
0–76%
Percentage of dry weight, based on infrared spectroscopic analysis Stone classification and data from Refs. 6 and 17. c In "pure" cholesterol stones, pigment is visible only in the center, and cholesterol constitutes >98% of mineral components. In mixed stones, laminae of cholesterol and pigment alternate, but cholesterol is >50% of total mineral. d Includes calcium bilirubinate plus black pigment polymer. e Percent of pigment that is polymer, based on loss of vinyl grups of bilirubin assessed by infrared spectroscopy (17,158). N.D., not determined, due to very low pigment content. f 100% (minus sum of measured components). g 0, below the limit of detection. b
gallstones grow (13,16,44). Often (e.g., the mixed gallstone in Fig. 1), the components are clearly arranged in alternating rings of cholesterol and pigment deposits (3). As in Fig. 1, virtually all cholesterol and mixed gallstones also show a pigmented core (8,24,25,45,46) with at least one central cavity, and any type of stone may be encased in an outer shell of calcium salts with or without pigment. The organized structure of gallstones, with crystals arrayed on an organic matrix, is typical of formation of mineralized structures (e.g., bone, teeth, shells, concretions) throughout the phylogenetic tree and in geological formations (47–50). Thus, gallstone formation may be viewed as a biomineralization process, in which precipitation from a supersaturated solution is regulated by soluble proteins and organized on a matrix composed of these proteins bound to the mucin gel (2,3,33,40,41). In vitro studies suggest that both the mucin (51–53) and the small proteins present in the matrices of gallstones (54–56), when unbound in bile, play important roles in controlling the onset and rates of precipitation of calcium salts (41,57), bile pigments (2,58), and cholesterol (54–56,59–61). This section summarizes the evidence for these concepts and reviews the roles of calcium, mucin, APF, CBP, other proteins and bile salts in the regulation of calcium and cholesterol precipitation and in the formation of the matrix and organization of the precipitates in gallstones. It is first necessary to describe the thermodynamic states and regulation of the concentrations of calcium salts and "calciumsensitive" anions in bile. The state of cholesterol in bile, and the regulation of cholesterol precipitation by biliary glycoproteins, are detailed elsewhere in this volume. II— Calcium and Calcium Salts in Bile A— Free and Bound Calcium Total calcium [CaT] in bile consists of two major fractions—the unbound (free) calcium ions [Ca2+], and the bound calcium (9,30,62) (Fig. 2). The bound calcium is, in turn, divided into
Page 320
Figure 1 Crosssection of a mixed gallstone. Note the pigmented core with a central cavity (arrow) and alternating rings of pigment (dark) and cholesterol (light) zones. Bar = 1 mm. (From Ref. 3.)
two fractions: (a) complexes with electrolyte anions, bile pigments (including unconjugated and conjugated bilirubins), and anionic bile salt amidates (monomers and oligomers) of molecular weight <3 kDa; and (b) larger bile salt aggregates (simple and mixed micelles) and anionic proteins (mainly albumin, soluble mucins, and CBP). The group of smaller complexes, together with the unbound Ca2+ cations, constitute ultrafilterable calcium, which can traverse the "pores" in the hepatobilary epithelium. In the complexes, the calcium is apparently bound bifunctionally, either chelated between two anionic groups, or between an anionic group and an —OH group (9,30,63). B— Determinants of Ionized and Total Calcium Concentrations in Bile In both hepatic and gallbladder bile, the concentrations of both unbound calcium, [Ca2+], and total calcium, [CaT] (and, therefore, also bound calcium) increase linearly with total bile salt concentrations, [BST], but the slope of the regression for [CaT] versus [BST] is much steeper than that for [Ca2+] versus [BST] (Fig. 3) (64–66). In normal human bile, [Ca2+] may range from as low as 0.8 mM, when [BST] is 10 mM, to 3.0 mM in concentrated gallbladder bile with [BST] of 300 mM or more; corresponding values for [CaT] are 1.5 and 18 mM respectively
Page 321
Figure 2 Fractions of calcium in bile. Those that may contribute to ultrafilterable calcium are shown in italics. The relative contributions of each fraction are only crudely represented since they vary markedly according to bile composition, and differ greatly between hepatic versus gallbladder bile and normal versus pathological bile. (Based on Ref. 9.)
(66–68). Patients with gallstones of any type, compared with subjects without gallstones, often have gallbladder bile containing significantly higher [Ca2+] and [CAT] in relation to [BST] (66,69), and black pigment stones are often associated with higher [BST] (up to 300 mM) (66). The fraction of [Ca2+]/[CaT], however, is similar for subjects with and without stones and is about 0.4 at the lowest [BST] and decreases curvilinearly to a plateau of about 0.2 at [BST] above 100 mM (66,70). The findings in normal subjects are the result of two processes: (a) Ca2+ ions freely and rapidly cross the tight junctions between the hepatocytes bounding the bile canalicular space, as well as the tight junctions between the biliary epithelium lining the biliary tree and gallbladder, by both passive diffusion and convection with fluid flow (65,71,72). Except at very
Page 322
Figure 3 Plot of concentrations of ionized and total calcium versus bile salts in canine bile. Note linear relationships, with higher values and steeper slope for total calcium. (From Ref. 64.)
high bile flows, bile [Ca2+] equilibrates with, and is determined by plasma [Ca2+] (73). (b) The equilibrium between [Ca2+] in plasma and bile is determined by the GibbsDonnan relationship (Fig. 4) (9,74). This law dictates that the presence of large, impermeant anions on one side of a semipermeable membrane will result in an increase in the concentration of unbound cations on that side of the membrane (9,75). The summed concentrations of the impermeant large bile salt micelles and anionic proteins (especially mucin and albumin) in bile usually exceed the concentrations of plasma albumin, the major impermeant anion in plasma. Thus, at bile salt concentrations in bile above 18 mM, the GibbsDonnan relationship dictates that the [Ca2+] in bile must be above the normal plasma [Ca2+] (mean 1.1 mM) (i.e., ratio of bile/plasma [Ca2+] > 1.0) (9). Two corollaries of these concepts are (9) (a) infusion of cholephiles that bind Ca2+ (i.e., bile salts) will not decrease the [Ca2+] in bile but increase both the ionized and bound calcium concentrations in bile and (b) the bile/plasma ratio of calcium concentrations must increase as the biliary concentrations of bile salts and proteins increase (74,76). In the gallbladder, where the bile is concentrated by absorption of NaCl and water (77), concentrations of [BST] and proteins increase by four to ninefold (78). This, along with secretion of mucin by the gallbladder epithelium, engenders greater GibbsDonnan forces and increased binding of calcium (9). As a consequence, in concentrated gallbladder bile, the GibbsDonnan ratio for [Ca2+] may be as high as 3.0, and the fraction of bound calcium increases (74). This increase in [Ca2+] and, consequently in [CaT], in the gallbladder is, however, mitigated somewhat by passive absorption of Ca2+ through diffusion down a concentration gradient and convection with the absorbed water (65,71,72), as well as by possible nonequilibrium conditions (76). C— Measurement of Calcium in Bile The activity of free Ca2+ ions can be determined with either calciumselective electrodes (30,62,76) or with indicator dyes whose color or fluorescence changes as they bind calcium
Page 323
Figure 4 Schematic for GibbsDonnan equilibrium.
(79). Both methods are subject to errors due to bile salts in bile. Calciumselective electrodes are poisoned by bile salts, leading to severe underestimation of [Ca2+]; this can be prevented by covering the active end of the electrode with a plastic membrane that has pores small enough to prevent access of bile salts to the sensor (9,80,81). The best way to show that an electrode is not poisoned by bile salts is to demonstrate that simultaneous measurements of bile and plasma [Ca2+] fulfill the GibbsDonnan relationship (30,80). The absorbance and fluorescence of most calciumindicator dyes may be altered by their interaction with bile salts (79), and bile pigments may interfere spectrally (82). Since the great variation in concentration and composition of bile salts among bile samples renders it difficult to correct for these effects with appropriate standards, use of calciumindicator dyes in native bile is problematic. It should be realized that both methods measure the activity of unbound, ionized calcium in bile (millimoles per kilogram), not the concentration of unbound calcium ion, [Ca2+] (millimoles per liter) (30,62). Most commercial electrodes are designed to convert the measured calcium ion activity to report [Ca2+] = Ca2+ activity/activity coefficient, using the mean activity coefficient for normal plasma of 0.304 at pH 7.4 and an ionic strength of 0.16 (62,81). The mean activity coefficient for bile is also 0.3 (30,83) and, as in serum (62), may vary with changes in pH and ionic strength. Some electrodes convert the [Ca2+] value to pH 7.4, adjusting for the decreasing activity of calcium ions, due to increasing concentrations of calciumcomplexing anions (especially carbonate) as pH increases (9,67,68,76). The reader is cautioned that many published papers on ionized calcium in bile do not make clear whether calcium ion activity or [Ca2+] is being reported or whether the reported values are at the actual pH of the bile or are adjusted to pH 7.4.
Page 324 2+
Other pitfalls in the use of Ca electrodes are (a) variable water contents of the "anhydrous" CaCl2, or CaCl2∙2H2O, used to prepare the standards; (b) the use of standards of differing pH, ionic strength or buffer composition than the samples under study; and (c) poor selectivity for sensing Ca2+ as compared with the other major cations in bile, Na+, K+, and Mg2+ (81). Comparison of four commercial electrodes available in 1983 revealed differences in reproducibility and in values reported with standards and plasma samples (84); similar rigorous comparisons have not been reported for bile, or for electrodes available today. Total calcium concentrations [CaT], may be measured with either the "Calcette," based on a dyebinding method (30), or by atomic absorption spectroscopy (85). The indicator dyes used are powerful chelators, which preferentially strip bound calcium from other complexes in bile. Bile salts and pigments (vide supra) seem to interfere less with these indicator dyes than those used to assay [Ca2+], since there is good agreement with the [CaT] obtained by atomic absorption spectroscopy (30). III— Thermodynamics of Calcium Salts in Bile A— The Solubility Product (K¢ sp) of Poorly Soluble Calcium Salts Ca2+ may form insoluble salts with the five "calciumsensitive" anions of bile, bilirubinates, carbonates, phosphates, fatty acylates and bile salts (9,30,31). For a calcium salt to precipitate, its net charge must be zero. The equilibrium solubility of each salt is determined by its solubility product ( ) (9):
Note that only the unbound ionized fractions of the calcium and the anion contribute to the ion product (the righthand side of each equation) and strictly should be based on the products of the ion activities, but the above approximations are satisfactory. If the ion product exceeds the empirically determined supersaturated for that salt. Calcium cannot form a salt with the fully protonated, neutral form of a weak acid (HAo).
, the system is
B— The Formation Constant (K'f) of Soluble Calcium Complexes When the ion product of a calcium salt is below the , the salt exists only as free ions and as soluble complexes; the latter, which are usually positively charged, contribute to the bound calcium in bile (9,30,62,75). Ca2+ ions also form soluble complexes with anions in bile that do not form poorly soluble calcium salts; the most important are and three polyanionic proteins in bile: mucin, albumin, and calciumbinding protein (CBP). The affinity of the calcium for the anion(s) in the complex is defined, at equilibrium, by the formation (association) constant, K f:
The terms in the denominator refer only to the unbound species. If the stoichiometry of the complex (i.e., m and n) and the pKa of HA are known, K f can be determined from measured values of [Ca2+] and the total concentrations of [AT] and [CaT]. Thus:
is derived from [AT] and pKa, using the HendersonHasselbalch equation, and values of [Cam An] and [A] from Eqs. (4 and 5) are substituted in Eq. (3).
Page 325
C— Factors Affecting Calcium Salt Saturation increases with increasing temperature and decreases with increasing ionic strength; these variables are homeostatically maintained relatively constant in vivo. Of far greater importance, as pH increases in relationship to the pKa values of a given acid (H2A or HA), the proportions of A= and A increase and the proportion of H2Ao or HAo decreases (9). Consequently, as pH increases, the ion product increases, and the system becomes more saturated with the given Ca2+ salt (2,9,76,86); a decrease in bile pH has the opposite effects (87). This is important, since (a) ductal bile is alkalinized by secretinstimulated bicarbonate secretion by biliary epithelial cells (88) and (b) in the gallbladder during fasting, as solutes from hepatic bile are concentrated by water and NaCl absorption, the increases in concentrations of A= and A are countered by the acidification of the bile (78,87,89), mediated by an amiloridesensitive Na+/H+ exchanger in the gallbladder epithelium (9093). In the postprandial state, however, this process is reversed, due to effects of secretin released from the duodenum, resulting in dilution and alkalinization of bile (88,94–97). Increased intracellular [Ca2+] and activation of protein kinase C may mediate these effects of secretin (98,99), whereas octreotide causes net absorption (100). Net secretion and alkalinization of bile by the gallbladder has also been demonstrated after crystallization but before stones appear in animal models of cholesterol gallstone formation (101–103), and in diseased human gallbladders (104). The protective effect of acidification in gallbladder bile is reinforced by two other factors: (a) enhanced binding of bilirubinates and fatty acylates by the increasing concentrations of bile salts (105–108) and (b) passive absorption of calcium by convection (71,72) and of the protonated (acid) forms of the organic anions (unconjugated bile acids, unconjugated bilirubin and fatty acids) by nonionic diffusion (109–112). Impaired acidification of bile should favor precipitation of calcium salts and gallstone formation, and acidification of gallbladder bile is reportedly defective in many patients with any type of gallstone (76,87), though some find this only in biles harboring gallstones which are radiopaque (i.e., calciumcontaining) (67,68). One must consider the duration of overnight fasting in the subjects studied, since the pH of gallbladder bile declines progressively with fasting and may attain levels as low as pH 6.5 (76,113). The pH of normal gallbladder bile is seldom above 7.2 (68,76). Whether or not there is defective acidification, the elevated [Ca2+] in gallbladder bile of many patients with gallstones contributes to increased saturation with all calcium salts (66,69). The increase in [Ca2+] in plasma in patients with hyperparathyroidism should engender an increase in [Ca2+] in bile (73), which might explain the possibly increased incidence of gallstones in this condition (114,115). D— Status of Calcium Salts of CalciumSensitive Anions in Bile The pKa,
, and K'f values of calcium salts are summarized in Table 3.
1— Bile Salts and Simple and Mixed Micelles Ca2+ ions in bile are bound mainly by bile salt anions, (BS). This involves highaffinity binding by premicellar (mainly monomeric) bile salts in the form of CaBS+ complexes and loweraffinity binding to bile salt micelles as C032501.GIF complexes (9). Compared with dihydroxy BS, anions of trihydroxy BS (cholates) have lower affinity for Ca2+ ions (9), and their Ca2+ salts have much higher
values (116,117), which renders them unlikely to precipitate in vivo. In human bile, taurine
conjugated BS constitute only about 25 to 40% of total BS (118,119), and their Ca2+ salts have high values (117), so they are unlikely to precipitate. By comparison, glycineconjugated bile salt anions have much higher affinities for Ca2+ (9), but their Ca2+ salts have only moderately lower solubilities (116,117). Thus, though bile may become supersaturated with Ca2+ salts of glycineconjugated BS, they seldom precipitate because of marked and prolonged metastability (116,117). Due to their higher pKa values (5.0 to 5.5) and their very low total concentrations in bile (except in patients with intestinal bacterial
Page 326 Table 3 pKa , Solubility Products (
), and Formation Constants (K f) of Calcium Salts of Calcium
Sensitive Anions of Bile (Mean ± S.D.)a pKa b
Calcium complex
Mon.
(Bile Salts)
<1.5
TC
3 c
(M )
Micelle
1.85
TDC
<1.5
1.95
TCDC
<1.5
1.95
K'f, monomer
K'f, micellar
(M1)d
(M1)d
>1.0 × 105
1.3 ± 0.5
3.9
7
47.7 ± 2.0
10.0
7
71.2 ± 3.5
13.6
7
1.2 × 10 3.5 × 10
GC
3.88
4.09
10.1 ± 0.6
6.1
GDC
3.88
4.50
1.0 × 10
8
53.6 ± 1.8
25.8
GCDC
3.87
4.54
2.0 × 109
96.0 ± 2.9
34.8
8
GUDC
3.86
5.12
C
5.00
5.50
6.0 × 10
N.D.
e
N.D.
7
N.D.
N.D.
9
4.0 × 10
6.4 ± 0.3 × 10
DC
5.02
6.30
N.D.
N.D.
CDC
4.98
6.53
8
1.0 ± 0.2 × 10
N.D.
N.D.
6.25
9.1 ± 3.0 × 108
N.D.
N.D.
1870
N.A.e
UDC
5.02
Carbonate
6.01
N.A.
Bicarbonate
9.80
N.A.
Phosphates =
Bilirubins B
HB Fatty acylates
e
7.21
N.A.
8.44
N.A.
8.12
N.A.
4.5–7.0
5.1–7.5k
1.5 ± 0.6 × 10
8f
3.76 ± 0.1 × 10
12.5 or 5.4 ± 0.5g
Soluble
i
46 ± 17
N.A.
13j
N.D.
N.A.
1.65 × 10 ca. 2 × 10
21j
ca. 2 × 10 N.D.
N.A.
6h
N.D.
N.A.
S.D. given if available in original article. b a pK values in H2O, 25°C, except bilirubinates at ionic strength 0.15. Mon., monomer. (Data for bile salts based on Refs. 280 and 281.) a
of for bile salts are the apparent solubility products, at 25°C based on an electroneutral Ca(BS)2 salt with an ion product = [Ca2+] x [BS]2∙[Ca2+] = free calcium ion activity, measured with the (Ca2+) ionselective electrode. Since residual dissolved [BST] was below the CMC, so that bile salts were mostly monomeric with a fugacity close to 1.0, and the pH was high enough for full ionization of the bile salts, free (BS) was assumed to be equal to [BST], ignoring the small proportion bound to Ca2+. The value given thus probably represents a maximum value for . (Based on Refs. 116 and 117.) d K'f for both premicellar and micellar bile salts is expressed as if the complex was CaBS+ (1:1 stoichiometry). where [Ca2+] = free Ca2+ ion activity, determined with the (Ca2+) ionselective electrode at 25°C and total sodium concentration of 0.15 M. Above the CMC, the total concentration of micellar bile salts ([BST] CMC) is used in place of [BST] in the equation. (Based on unpublished data from Moore EW, Mukerjee P, and Ostrow JD.) e N.D., not determined; N.A., not applicable. f
For calcium carbonate as calcite at 37°C; value at 24°C is 1.33 ± 0.04 M2. Data from Ref. 121. values for vaterite and aragonite have not been determined. g For calcium carbonate and bicarbonate at 37°C, ionic strength 0.15. First value obtained with calcium electrode, is from Ref. 122. Second value obtained by calcium electrode (282) and by gel filtration on Biogel P2 (283). h For CaHPO4 at 25°C, ionic strength 0.15 (123,284). i
For calcium phosphate at 37°C and ionic strength 0.15, at pH 7.4, determined by gel filtration on Biogel P2. K f thus reflects affinity mainly for anions are present also (283). j Approximate value for Ca(HB)2, obtained by 24h partition of UCB from chloroform into aqueous CaCl2 solutions at ionic strength = 0.15 and 25°C, over the pH range from 6.0 to 7.8. Though a stable value was attained for (Ca2+) × (HB)2, it was not proven that true equilibrium was attained. The value given thus probably represents a maximum value for (137). k pKa values for fatty acylates increase with increasing chain length, and average about 0.6 pH unit higher when dissolved in bile salt micelles (161,162).
Page 327
overgrowth syndromes) (119), the activity of unconjugated bile salt anions is relatively low at the normal pH of gallbladder bile (6.5 to 7.0). Calcium salts of unconjugated bile acids, therefore, rarely precipitate, even though they have very low values and do not display metastability. Collectively, these concepts explain why calcium complexes of glycineconjugated dihydroxy bile salts are major binders of calcium in bile and why calcium salts of bile acids constitute only a small proportion of the minerals in gallstones. In bile, bile salts are carried mainly in mixed micelles with lecithin and cholesterol (32,120). Several studies utilizing calciumselective electrodes have reported that addition of eggyolk lecithin to simple micelles decreased the activity of Ca2+ ions in a doserelated fashion, implying increased calcium binding to mixed micelles as compared with simple micelles (82,116). This binding was not affected by addition of cholesterol, even to the point of saturating the mixed micelles (82). Though reservations have been raised concerning the interpretation of these findings (80), the concordant results with two different calciumsensitive electrodes and two different bile salts support their validity. In contrast to this apparent binding of Ca2+ to mixed micelles, little calcium is bound to lecithincholesterol vesicles (9,82). 2— Carbonate/Bicarbonate At the normally acid pH of gallbladder bile (67), ion products of CaCO3, (as calcite) are usually below the solubility product ( = 3.8 × 108 M) (67,68,76,89,121). By contrast, both canine and human hepatic biles are always markedly supersaturated with CaCO3 because the pH is usually 7.1 (67,89). Calcium bicarbonate, by contrast, is a very soluble salt that is present as soluble complexes (122). 3— Phosphates In simple solutions containing only calcium and phosphate at pH > 6.0, CaHPO4 is the only phosphate salt that is found in the precipitates, with a of 1.65 × 106 M (123) but marked metastability (124). Initial precipitates are amorphous, but they metamorphose gradually through a variety of crystal forms, including CaHPO4, ultimately yielding the most stable form, hydroxyapatite, which has the lowest (125,126) and is the principal form in gallstones (6,24). Biles from patients with and without gallstones seldom exhibit supersaturation with CaHPO4 (127), since the concentrations of inorganic phosphate are low (mean 0.35 mM in hepatic bile and 0.6 mM in gallbladder bile) (128,129). This suggests that the precipitation of calcium phosphates, found in the centers of most cholesterol gallstones (2,8,15,27), might be triggered by a temporary increase in phosphate concentration due to hydrolysis of biliary lecithins. 4— Bilirubinates Bilirubin conjugates (mainly glucuronides), which constitute over 99% of the total bilirubin in normal bile (130,131), do not form insoluble calcium salts, but they bind calcium in soluble complexes (K f about 200 L/mol) (132,133). Though bilirubin conjugates are fully ionized throughout the pH range of bile, they bind avidly to simple and mixed bile salt micelles (134,135), so that their unbound concentration is relatively low at total concentrations that are rarely above 10 mM. Nonetheless, bilirubin conjugates are found in some gallstones (136), presumably by mechanisms other than precipitation as calcium salts—for example, by covalent binding to CBP (see below) (41). By contrast, calcium salts of unconjugated bilirubin (UCB) have extremely low aqueous solubility (137). The small proportion of UCB in bile is derived primarily from hydrolysis of secreted conjugates, mediated by endogenous b glucuronidases (138) secreted into bile by the hepatocytes and biliary epithelium (18). At any given fractional rate of hydrolysis, an increase in the concentration of total bilirubin in bile will result in a proportional increase in the concentration of UCB (139).
Page 328
Upon solvent partition of UCB from chloroform into buffered, aqueous CaCl2 solutions, the Ca(HB)2 (acidic) salt precipitates only above pH 6.2 and the CaB (neutral) salt only above pH 8.0 (137). The very low apparent for Ca(HB)2 of approximately 1 × 1021 M3 (137) guarantees that even normal bile is always supersaturated with Ca(HB)2, even when fully acidified in the gallbladder. This is because HB is the major unbound UCB anion over the entire pH range of bile (105,140). The small fraction of UCB dianion (B=) present below pH 8.0 also has a very high affinity for bile salts (105–108,141,142), so that the concentration of unbound B= should be extremely small. Therefore, except when pH and total [UCB] are considerably elevated, CaB is usually the minor calcium bilirubinate salt in gallstones (11,13,25,143–145). The K f of the soluble Ca2+ complexes of unconjugated HB and B= have not been determined. The above considerations indicate that factors which increase the concentration of unbound HB and B= will favor precipitation of calcium bilirubinates (2). These include the following: 1. An increase in bile pH (86) 2. A decrease in bile salt and/or increase in lecithin concentration, decreasing the binding of bilirubin anions (105) 3. An increase in the concentration of total UCB in the bile, due to either a. Increased hepatic secretion of conjugated bilirubins, as in hemolysis (131) b. Increased secretion of UCB photoisomers during phototherapy (146) c. Increased hydrolysis of secreted bilirubin conjugates to UCB (138,147,149). Enhanced formation of UCB by hydrolysis can result from (a) increased activity of glucuronidase in bile (138) due to a decrease in pH (150) or decreased concentrations of the endogenous inhibitors of this enzyme [bile salts (150,151), b glucuronolactone and glucaric acid (152,153)]; (b) increased release of endogenous b glucuronidases into bile (138), as with hepatocellular necrosis or biliary tract inflammation (18,147); (c) introduction of bacterial glucuronidases (147,148,154); or (d) an increased proportion of bilirubin monoglucuronides as compared with diglucuronides, as occurs with hemolysis or in Gilbert's syndrome (131). Gross infection of bile is almost universal in the formation of brown pigment gallstones, whereas black pigment gallstones are commonly associated with hemolysis (18,21). Recent studies indicate, however, that DNA of bacteria and/or bacterial b glucuronidase is detectable in most gallstones, even when the surrounding bile yields no growth on aerobic and anaerobic cultures (154–157). The pigment polymer network, which accounts for the dark color and resistance to chemical dissolution of the black pigment gallstones, is believed to form by solid state polymerization of precipitated calcium bilirubinates, mediated by oxidative mechanisms (17,158). Though an increased proportion of this polymer is associated with loss of the 990 cm1 peak of the vinyl groups of bilirubin in infrared spectroscopy (158), it is disputed whether polymer formation involves these vinyl groups (17) or the —CONH— groups in the two outer pyrrolenone rings of bilirubin (159,160). 5— Fatty Acylates Like bilirubinate anions but unlike carbonate or phosphate, amphiphilic free fatty acid anions (FA) bind to mixed micelles and proteins in bile (161). Thus, knowledge of the solubilities and pKa values for FA in simple, buffered aqueous solutions (161,162) is insufficient to describe the solubilities of FA or their calcium salts in bile. In simple aqueous systems, since the FA anion is more soluble than the protonated FA, the overall solubility of FFA decreases and the pKa increases with increasing hydrocarbon chain length. Thus, the pKa is 4.5 to 5.0 for myristic acid (C12:0), but 6.5 to 7.0 for palmitic acid (C16:0), the most abundant fatty acid found in gallstones (7,163). Free fatty acids in gallstones probably derive mainly from hydrolysis of lecithins by phospholipaseA2, so that the composition of the FFAs mimics the fatty
Page 329
acid composition of the phospholipids (7,164) and are present mainly as calcium palmitate and oleate (4,7,163,165). IV— Kinetics of Calcium Salt and Cholesterol Precipitation and Biomineralization Theory Calcium salt and cholesterol precipitation from supersaturated solutions is both regulated and organized. The persistent supersaturation of even normal human bile with cholesterol (166) and (in hepatic bile) carbonates (167), and probably with calcium bilirubinates (2), indicates that supersaturation is a necessary but not sufficient condition for precipitation of these components of bile. Calcium salts (125,168,169) and cholesterol (170–172) precipitate initially as unstable polymorphs (crystal forms) and then undergo transition to more stable forms, which may differ in composition and degree of hydration, as well as crystal structure. As with cholesterol, proteins and other factors in bile regulate calcium salt precipitation, but the mechanisms differ somewhat. Factors promoting cholesterol nucleation act mainly by influencing the aggregation, fusion and/or stability of the unilamellar vesicles which are the major carriers of cholesterol in bile, forming multilamellar vesicles from which cholesterol seems to crystallize (32,173–177). By contrast, the proteins and bile salts that inhibit calcium crystallization do so primarily by adhering to the growth centers on the surface of nascent crystals, ''poisoning" their ability to grow to a size sufficient for precipitation (178–180) or to undergo transition from less stable to more stable polymorphs (169,181). Recently, similar mechanisms have been demonstrated for four biliary glycoproteins that bind to the growing faces of cholesterol crystals and alter the morphology of the crystals (182). In gallstones, the calcium salts and cholesterol appear to be arrayed on a threedimensional network of polymerized mucin gel (1,34,35,45,183); thus, the pattern of crystallization is organized (Fig. 5) (3). A key difference, however, is that immunoreactive APF/CBP, which
Figure 5 Schematic of biomineralized structure of a mixed gallstone. (Ref. 10.)
Page 330
binds to mucin (51), appears to be interposed between the calcium salts and the mucin (3), whereas cholesterol may be bound directly to the hydrophobic, nonglycosylated domains of the mucin (184,185) without the intervention of APF/CBP (3). A— Biomineralization The apparent regulation and organization of calcium and cholesterol crystallization in gallstones, mediated by proteins that may be involved in both aspects, is characteristic of biomineralization processes throughout nature (Fig. 6) (33,41,47–50,168,186). This applies both to physiological biomineralization [e.g., formation of bones and teeth (47,49), and the shells of eggs and bivalves (178)] and pathological biomineralization [e.g., calculus formation in the saliva (187,188), urine (189,190), and pancreatic juice (191,192)]. In all cases, the mineralized structure consists of three classes of components (41): 1. The crystals themselves, equivalent to the bricks in a building. 2. A large structural protein with hydrophobic domains, which can polymerize to form the threedimensional structure, equivalent to the girders in a building. 3. Small (usually <20kda), amphiphilic, acidic regulatory proteins/polypeptides, which anchor the calcium salt to the structural protein (equivalent to the mortar). The equivalent components in bile and gallstones are (a) calcium salts and cholesterol, (b) mucin (2 × 106 kDa) and (c) calciumbinding APF/CBP (7 kDa) and the cholesterolbinding immunoglobulin fragments (28 to 74 kDa).
Figure 6 Schematic of steps in biomineralization of a mixed gallstone. (Modified from Ref. 2.)
Page 331
B— Biomineralizaton of Calcium Salts The key players are acidic regulatory proteins/polypeptides (33,41,49,178,179), which have both hydrophobic and polyanionic domains, either on opposite faces of a b pleated sheet (resembling a bile salt) or as the tail and head of a linear molecule (resembling a fatty acid anion). In their inactive form, the anionic groups are shielded (e.g., by amidation of —COOH groups on glutamate or aspartate), and the hydrophobic domains are inaccessible or not in proper conformation to bind to the structural protein. When activated by deamidation of —CONH2 groups, phosphorylation of —OH groups, and/or carboxylation of glutamate residues, the regulatory protein alters its conformation, so that its hydrophobic domain can bind to the structural protein, forming a matrix, to which Ca2+ is bound by the now exposed anionic groups (Fig. 6) (2). The anionic groups on the regulatory protein are set at intervals of about 6.9 Å, the typical distance between Ca2+ ions in the crystal lattice of the calcium salts (49). Thus, the matrix forms a template on which the Ca2+ ions are arrayed, and on which the crystal grows by accretion of the relevant anions and more Ca2+ ions (Fig. 6). Studies with components of molluscan shells (50,178,180) or bile/gallstones (51,182,193) have shown that the regulatory proteins not only regulate the rate of crystal formation; they also direct the stoichiometry and morphology of the growing crystal. Ultimately, growth of the crystal may be arrested by binding of the anionic groups of free regulatory protein molecules to the calcium cations at the growth face of the crystal (Fig. 6). The exposed hydrophobic domains of the bound regulatory protein act as a barrier to accretion of more ions but can bind more mucin, allowing the process to be repeated and generating multiple layers or cells of calcium salt crystals (33). In bile, alternatively, cholesterol can bind to the mucin, forming a layer of cholesterol crystals, which can, in turn, bind more mucin (Fig. 6). This is considered a likely mechanism for the presence of a core of calcium phosphates and bilirubinates in the center of all cholesterol gallstones (2,8,15,27); the nucleus of calcium salts and matrix forms first and then serves as a nidus for accretion of cholesterol monomers to form crystals. Formation and growth of a biomineralized structure usually involves accretion of monomers from the supersaturated solution rather than aggregation of preformed crystals (47,49,194). When not combined in a matrix to trigger and organize calcium salt crystallization, unbound CBP and mucin can bind ionically to the growing face of microcrystals or stones and arrest their further growth by blocking further accretion of calcium salts (51). It is not known how APF/CBP is activated, whether APF binds to mucin, or whether factors other than the molar ratios of APF/CBP to mucin determine whether crystal growth is promoted or arrested in the presence of both proteins. C— Biomineralization of Cholesterol As with calcium salts (125), cholesterol crystals may undergo transitions from early, spiral, and filamentous forms (believed to be anhydrous cholesterol) to mature, platelike crystals of cholesterol monohydrate (170–172). Four candidate regulatory glycoproteins have been isolated from bile by their properties of binding to cholesterol crystals and affinity for Helix pomatia lectin (182); the latter suggests the presence of Nacetyl a Dgalactosamine residues. Three of the four are apparently subunits of polymeric human IgA, with apparent molecular weights on SDSPAGE of 28 kDa (light chain) 63 kDa (heavy chain), and 74 kDa (secretory component). In model bile at pH within ± 1.0 U of their pI values (6.3 to 8.3) (182), each glycoprotein binds to cholesterol crystals, slowing the maximum growth rates, diminishing the quantity of precipitated cholesterol at equilibrium, and altering crystal morphology (182). Similar effects were obtained with other inhibitory biliary glycoproteins that bound to concanavalin A (Con A) or lentil lectins, but the H. pomatia proteins were up to fivefold more potent, in the order 63 > 16 > 74 > 28 kDa (182). Though immunoglobulins are present in human gallstones (3,54), IgA is distributed randomly throughout mixed gallstones, without specific
Page 332
localization (3), suggesting that this immunoglobulin does not act as a "mortar" to anchor cholesterol crystals to the mucin scaffolding; it is currently believed that the cholesterol crystals are bound directly to the mucin (3). To be determined are the effects on cholesterol crystallization in human bile samples of the specific removal from, or addition of, immunoglobulins. D— The Roles of Mucins 1— Structure Mucintype glycoproteins are heavily glycosylated macromolecules; they are the most abundant proteins in bile (53,195). The soluble mucin monomers have a very high molecular mass (2 × 106 kDa), over 80% of which consists of the Olinked oligosaccharide chains (53,196). Mucins from human bile are known to be quite heterogeneous in regard to the proportions of different glycan chains and sulfation (197–201). The core polypeptide chain contains at least two distinct regions (202– 205). One consists of 20amino acid tandem repeat sequences that are rich in serine, threonine, and proline; the glycan (carbohydrate ± sulfate) chains are covalently bound to the first two residues. The second nonglycosylated, hydrophobic region, possibly internally located, consists of 123amino acid tandem repeat units rich in serine, glutamine/glutamic acid, and glycine, and 127amino acid tandem repeats rich in cysteine and glycine. The uniquely high proportion of anionic sialic acid (16% of sugar residues) in gallbladder mucin is correlated with its ability to bind calcium (206,207), while the hydrophobic domains bind ligands such as bilirubin (37,58), lipids (185), and some proteins, including APF/CBP (51); both domains are believed to contribute to the pathogenesis of cholesterol lithiasis (53,185,208,209). 2— Function Mucins polymerize at concentrations above about 4 g/dL, mediated by concentrationdependent selfaggregation and possibly by formation of disulfide bridges, generating a highly hydrated and viscous polyanionic gel (196,210). A network polymer of this gel constitutes the supporting matrix of both sludge (210,211) and cholesterol and pigment gallstones (1,34,35,45,183), and is believed to serve as a nidus for gallstone formation (53,210). The viscous mucous gel layer overlying the gallbladder epithelium is believed to prolong the residence time of lithogenic bile and microcrystals in the gallbladder, granting time for the accretion of cholesterol crystals on to gallstones (210). Addition of soluble mucins to supersaturated model biles has been reported variously to promote (a) fusion and/or aggregation of vesicles; (b) instability/leakiness of vesicles and crystallization of cholesterol therefrom; and (c) growth rate and size of cholesterol crystals (52,174,185,208,212–217). Mucin also stimulates crystallization of calcium phosphates and bilirubinates (33,51,147) and diminishes the size of hydroxyapatite crystals (218). The presence of mucins in the core of cholesterol and mixed gallstones also supports their important role in the crystallization process (1,3,36–38). In animal models of gallstone formation, hypersecretion of mucins has seemed to occur before crystals and stones were formed (219–222), but inhibition of mucin secretion with nonsteroidal antiinflammatory agents or aspirin has not consistently prevented crystal and stone formation (223–226). Aspirin, however, has been shown to have a slight protective effect against the high frequency of cholesterol gallstone formation during rapid weight loss in morbidly obese humans (227). In human bile samples, contradictory results have been obtained regarding the relationships between the concentrations of mucins and the presence of gallstones (210,228,229). This suggests that differences in the structure of mucin, rather than its concentration, might be more important in the pathogenesis of gallstones (200,201,210,228), but no qualitative differences have been detected between mucins from gallbladder bile of humans with versus without gallstones, regarding either structure or in vitro effects on crystallization (201,228).
Page 333
Though it has been proposed that both calcium salts and cholesterol precipitate within the mucin gel layer covering the gallbladder epithelium (32,210,230), this has not been demonstrated directly, and virtually all studies of the effects of mucin on cholesterol and calcium crystallization have been performed with soluble mucins. Though phospholipids (231), cholesterol (232), and protonated organic acids [such as unconjugated bilirubin (110) and longchain free fatty acids (111,112)] are absorbed readily from the alkaline bile across the thin mucus gel layer in the guinea pig gallbladder, this is not true of organic anions with lower pKa values (109), including conjugated bile salts. This might be expected, despite the favorable 10mV lumentoplasma gradient across the gallbladder mucosa (233), since the large, negatively charged anions would undergo electrostatic repulsion by the polyanionic mucin gel [pI about 4.0 (233)]. Moreover, the increasing acidity in the deeper layers of the gel (86), generated by the Na+/H+exchanger in the gallbladder epithelium (91–93), would largely protonate those anions that did penetrate, converting them to acids that are incapable of forming calcium salts and are more readily absorbed across the gallbladder epithelium (234,235). For these reasons and since most gallstones in humans are not embedded in the mucin gel layer, it may well be that the precipitates form and are organized on mucin monomers in the bulk bile [as has been shown in model biles (51)]; these could then polymerize to form the gallstone and adhere it to the mucin gel layer covering the gallbladder epithelium. Clearly, the activities of unbound Ca2+ and HB need to be determined in the mucin gel. E— Roles of APF and CBP—The Small Regulatory Proteins of Bile APF (anionic peptide fraction) and CBP (calciumbinding protein) are two closely related, immunologically crossreacting, small (about 7 kDa), amphipathic polypeptides (236), which together constitute the third most abundant protein(s) in human bile (39,237). APF/CBP is abundant in both hepatic and gallbladder bile, from controls as well as patients with gallstones, in concentrations ranging between 0.6 and 1.2 mg/mL (237). It is also uniformly present in all types of gallstones (51). Immunostaining of the cut surface of gallstones reveals that immunoreactive APF/CBP can be demonstrated only in the zones that contain calcium salts and bile pigments (3,238). 1— Preparations of APF and CBP from Bile and Gallstones Early preparations of APF, isolated from bile by zonal ultracentrifugation (239,240), and of CBP, isolated from gallstones by delipidation and demineralization (40,42,51,241) or from bile by coprecipitation on calcium carbonate (42,236), were contaminated with bile salts, lipids, bile pigments, or sodium dodecyl sulfate (SDS) and contained both APF and CBP in various proportions (236). SDS contamination was later minimized by excluding it from electroelution solutions and by extraction with 50 mM KCl (242). Removal of pigments was facilitated by performing the SDSpolyacrylamide gel electrophoresis (PAGE) on 15% Laemmli rather than Schägger von Jagow gels (236). It was also realized that preparations from gallstones would include denatured proteins formed during the long sojourn of the stones in the gallbladder (236). Recently, virtually pure preparations of APF and CBP, with minimal denaturation, have been obtained by using fresh human hepatic bile collected into tubes containing protease inhibitors and eliminating the use of lipid solvents and SDSPAGE (236). As before, CBP was recovered by calcium precipitation and then demineralization with EDTA, whereas bile was simply centrifuged for the APF preparation. Larger proteins and aggregates were removed by serial ultrafiltration through 100, 30, and 10kDa membranes, and a final hydrophobic 3.5kDa membrane removed residual salts, lipids, and bile salts (236). Preparative hydrophobic highperformance liquid chromatography (HPLC) (Fig. 7) then cleanly separated APF and CBP from small polypeptide fragments and most of the residual pigment (236). Analytical HPLC revealed that the APF preparations isolated by serial ultracentrifugation or serial ultrafiltration
Page 334
Figure 7 Hydrophobic HPLC patterns in the Marseille system, with detection at 280 nm. (top) APF from bile. (middle) CBP from a cholesterol gallstone. (bottom) CBP from calcium precipitation of bile. "O.D. ELISA" refers to reactivity to polyclonal antibody against peak 4 from (A) by enzymelinked immunosorbent assay, as O.D./area under the curve of HPLC peak. (Modified from Ref. 236.)
Page 335 Table 4 Characteristics of Amphipathic Anionic Polypeptide (APF) and Calcium Binding Protein (CBP) Characteristic
APF
CBP
Present in
Bile (and stones)
Bile and gallstones
Molecular weights (SDSPAGE)
7 and 12 kDa
7 and 12 kDa
Amphipathic
Yes
Yes
Anionic (acidic)
Yes (pI < 5.0)
Yes (pI < 5.0)
Hydrophobic binding
Strong
Strong
Glycosylation
Not determined
None detected
Stain with Coomassie blue
No
No
Stain with silver
Orangebrown
Orangebrown
Bound pigments
+
+ + +
Amino acids
Closely similar compositions (see Table 5)
Binding to membranes
Bind poorly to artificial blotting membranes
Antibody binding
Complete immunological crossreactivity.
Binds calcium
Little
Strongly
If unbound, inhibits calcium salt precipitation
No
Yes
When added to mucin, promotes calcium salt precipitation
No
Yes
Stabilizes biliary vesicles
Yes
No
Inhibits cholesterol crystallization
Yes
No
contained small amounts of CBP, but that CBP preps, obtained by initial extraction of gallstones or calcium precipitation of bile, contained no APF (236). This is important to consider in evaluating results obtained with these preparations. 2— Characteristics of APF and CBP APF and CBP share many common characteristics (Table 4). Both (a) are highly amphipathic, leading to avid selfaggregation, strong binding of phospholipids (39,243) and pigments (33,41,51) as well as bile salts, and a tendency to adhere to hydrophobic surfaces, such as concanavalin A and C18 reversephase columns (41,51); (b) are highly acidic, revealing up to three bands on isoelectric focusing, with pI values of approximately 4.5, 4.7, and 4.9 (39,41,243); (c) show two bands on SDSPAGE in Schägger von Jagow gels with apparent molecular weights of about 6.5 to 7.0 and 12 kDa (51,236); (d) do not stain with Coomassie blue but stain orangebrown with silver (39); (e) are yellow, brown, or green in color due to tightly (? covalently) bound bile pigments (more for CBP than APF) (40,51); (f) have closely similar amino acid compositions (Table 5) [with about 24% diacidic and 10% dibasic amino acids, about 14% serine + threonine, about 38% neutral amino acids, low proportions of proline
Page 336 Table 5 Amino Acid Compositions of Amphipathic Anionic Polypeptide/CalciumBinding Protein Preparationsa,b,c Source Preparation
Hepatic bile APFmar
CBPnewd
Mixed stone
Xol stone
CBPmixd
CBPxol
Amino acid Taue
ND
ND
0.0
ND
Pro
3.3
4.7
5.4
5.2
Asp
10.0
9.7
13.2
12.5
Glu
12.3
13.8
13.8
14.8
Thr
6.1
5.1
5.1
6.6
Ser
7.9
8.9
7.2
7.6
Gly
19.8
11.4
13.6
14.0
Ala
7.1
7.2
8.4
6.0
Val
6.9
7.5
6.1
4.6
Ile
4.0
3.8
2.9
3.3
Leu
8.0
8.6
7.8
8.7
Cys/2
0.6
0.0
0.0
0.3
Met
0.0
1.9
0.0
0.0
Tyr
1.7
3.8
2.0
1.4
Phe
3.6
4.7
4.0
4.3
His
1.4
1.6
1.8
1.2
Lys
3.8
4.5
4.7
5.2
Arg
3.4
2.9
4.1
4.3
Mol % diacid
22.2
23.5
27.0
27.2
Mol % dibasic
10.7
8.7
9.0
10.6
Mol % Ser + Thr
14.0
14.0
12.3
14.2
Diaciddibasic
13.5
14.5
16.4
16.6
a
Percent of residues. Data from Ref. 236, where the nature of the above APF and CBP preparations is defined. b With CBPnew as reference, values in bold are significantly higher and values in italics are significantly lower. c Absence of tryptophane confirmed by independent nondestructive analysis. d Average of closely similar values for 7 and 12kDa bands. e
N.D., not determined.
and aromatic amino acids, and little or no cysteine or methionine (236)]; (g) adhere poorly to most artificial membranes used for immunoblotting and amino acid sequencing (236), with the possible exception of the positively charged Immobilon membrane (MilliporeWaters) (39). Most importantly, APF and CBP show complete immunological crossreactivity with all poly and monoclonal antibodies thus far developed against either polypeptide (39,236). This renders it impossible to distinguish APF from CBP in immunoassays of body fluids or immunostaining of tissues or gallstones; the polypeptide thus assayed or identified is referred to as APF/CBP. Some monoclonal antibodies prepared against earlier preparations of APF or CBP also showed cross immunoreactivity with apolipoprotein A1 and HDL3 (39,236), but a certain number of monoclonal and all polyclonal antibodies against APF, used for enzyme linked immunosorbent assay (ELISA) of body fluids, did not. Nonetheless, the reported amounts of APF/CBP in biological fluids [e.g., 0.6 to 1.2 mg/mL in human gallbladder biles (237)], based on ELISA, may represent different estimates due to changed exposure of APF/CBP epitopes
Page 337
in the presence of lipids and detergent bile salts than with the APF/CBP standards (244). Using the latest procedures, up to 42 mg of pure CBP has been isolated from 1 L of human hepatic bile, and as much as 5.2 mg CBP has been isolated per gram of dried black pigment gallstones and 1 to 2 mg/g from cholesterol and mixed gallstones (van den Berg AA, Ostrow JD; unpublished data). The same values, 1 mg/g, were estimated for APF in mixed gallstones (238). The 7 and 12kDa bands, seen on SDSPAGE of all preparations of APF/CBP, share many common properties: (a) closely similar amino acid patterns (236); (b) a blocked Nterminal residue; (c) closely similar patterns on hydrophobic HPLC (236); and (d) inhibit the precipitation of calcium phosphate and bind to gallbladder mucin with similar affinity, forming complexes which alter the kinetics and structure of the formation of calcium phosphate (51). Though these findings suggest that the 12kDa band is a dimer of the 7kDa band, each band eluted from a preparative gel ran true on second gel, with no evident dissociation of the 12kDa band to yield the 7kDa band, or association of the 7kDa band to yield the 12kDa band (236). This suggests that, if the 12kDa band is a dimer, the two 7kDa bands must be covalently linked. Both bands, however, were detected in the presence of mercaptoethanol, excluding crosslinkage by SS bond(s), but other linkers—for example covalently bound pigment—have not been excluded (236). The relationship between these two bands of APF/CBP remains unsolved. The most important differences between APF and CBP are their affinities for calcium and their effects on cholesterol crystallization (236). CBP, but not APF, avidly binds 45Ca2+ (236), adsorbs to calcium carbonate precipitates (42,236), inhibits the nucleation of crystals of calcium carbonate (40) or phosphate (51), and—when prebound to bovine gallbladder mucin—promotes the precipitation of calcium phosphate and alters its crystal structure (51). By contrast, APF but not CBP reportedly regulates the rates and amounts of cholesterol precipitation from model bile solutions, whether or not they contain calcium (59,216,245). These differences are discussed further in Secs. F. 2 and F. 3 below. Since repeated efforts by four labs have failed to sequence the APF/CBP polypeptide(s), the true structural relationships between APF and CBP are not known. It has been shown that CBP, isolated from gallstones, is not glycosylated and does not bind specifically to Con A, H. pomatia, or lentil lectins (41), but this has not been assessed for preparations of CBP from fresh bile or for APF. Our current hypotheses, influenced by biomineralization theory, are that APF and CBP have the same polypeptide structure and that CBP is a posttranslational modification of the APF synthesized in the rough endoplasmic reticulum of the hepatocyte (236). The binding of calcium by CBP but not APF suggests that CBP, like the activated form of most calciumregulatory proteins, is formed by deamidation and/or phosphorylation (2,47,49). 3— Sources of APF/CBP in Bile Immunomicroscopy reveals APF/CBP in the rough endoplasmic reticulum of rat and human hepatocytes (246) but not in nonparenchymal liver cells (Lechène de la Porte P, Lafont H; unpublished data). Radiolabeled leucine is incorporated into immunoreactive APF/CBP by isolated rat hepatocytes (246), and APF/CBP appears in the bile secreted by the isolated perfused rat liver (247). The synthesis of APF/CBP by isolated rat hepatocytes (247), and the biliary secretion of APF/CBP by the bile fistula rat (248,249) and the isolated perfused rat liver (247) increase in parallel with secretion of bile salts. Surprisingly, the output of APF/CBP is greater the less the hydrophobicity and micelleforming properties of the bile salts (GDC < TC << TDHC) and does not correlate with biliary phospholipid or cholesterol secretion (247–249), despite an apparent close association of APF/CBP with the bile pigmentlipoprotein complex in bile (243). It thus appears that APF/CBP is synthesized by hepatocytes and secreted into the bile. It has not been determined whether, like other proteins in bile (250,251), it is absorbed across the gallbladder mucosa.
Page 338
F— Functions of Calcium, APF, CBP, and Calcium Salt Precipitates 1— Functions of Calcium Binding of calcium, by addition of EDTA to human bile, blocked precipitation of calcium bilirubinates but did not affect the time of the appearance of cholesterol crystals (252). Note, however, that this study failed to first remove cholesterol microcrystals from the bile or to measure the growth rate or amount of crystals formed; in addition, EDTA binds bilirubin and other bile components besides calcium. In contrast, addition of calcium to biles from patients with gallstones increases the number of cholesterol crystals formed (253). In model biles, addition of physiological concentrations of calcium has been shown to enhance both the hydrophobic binding of bilirubin (254) and cholesterol (255) to mucin and the promotion of cholesterol precipitation and crystal growth engendered by Con Abinding glycoproteins (256). Each of our groups have recently demonstrated, using phasecontrast light microscopy, that addition of calcium to model biles at concentrations 10 mM promotes cholesterol precipitation under conditions in which insoluble calcium salts are not formed (no phosphate in buffer, CO2 excluded from the system) (59,216). In the FrenchIsraeli study (59), the model biles were prepared without buffer (pH was <7.0), taurocholate was the only bile salt used, supersaturation was initiated by 10 fold dilution with 0.15 M NaCl, and crystals and vesicular phases were quantitated also by sucrose density gradient ultracentrifugation. In the Amsterdam study (216), model biles were buffered with PIPES to pH 7.0, equimolar concentrations of taurocholate and taurodeoxycholate were added, the solutions were supersaturated from the time of dissolution, and nephelometry was used also to quantitate crystallization of cholesterol. Despite comparable total calcium and bile salt concentrations, the addition of taurodeoxycholate in the Amsterdam study results in much lower [Ca2+] compared with the use of taurocholate alone in the FrenchIsraeli study (9), probably accounting for effects at lower total calcium concentrations in the latter study (1 mM versus 5 mM). In both studies, addition of calcium led to an accelerated rate of cholesterol crystal growth, but only the IsraeliFrench study detected an increase in the cholesterol crystal mass at equilibrium (Fig. 8) (59). By contrast, only the Amsterdam study detected a hastening of the time to the first appearance of crystals and more rapid attainment of equilibrium in the presence of calcium (216). The IsraeliFrench study was able to show also that the effect of added calcium was mediated by promoting the aggregation of vesicles and destablizing the multilamellar vesicles from which cholesterol crystallizes (59). In the Amsterdam study, addition of 1 mg/mL human hepatic bile mucin even more potently accelerated crystallization than did 10 mM calcium, and also doubled the crystal mass, and the effects of mucin plus calcium were additive (216). That study also showed that rapid crystallization of calcium phosphates more potently accelerated and increased cholesterol precipitation than did the same concentrations of soluble calcium ions, and the influence of mucin was still additive (216). This demonstrated potentiation of cholesterol crystallization by physiological concentrations of calcium in model biles must be interpreted cautiously until similar results are obtained with human bile. The observation, in one study (216) of an even greater effect of precipitated calcium phosphate is compatible with the demonstrated presence of calcium phosphates in the centers of cholesterol gallstones (2,8,15,27) and favors the hypothesis that calcium precipitation might initiate cholesterol gallstone formation (9). The IsraeliFrench study also documented that the effect of soluble calcium anions was to increase vesicle size (fusion), aggregation and instability (59), confirming Moore's earlier proposals (9). 2— Functions of APF APF is found in higher concentrations in the vesicular than in the nonvesicular fractions of bile (59,245). By its phospholipidbinding properties, APF appears to stabilize aggregated unilamellar vesicles and the fused, large multilamellar vesicles (59,174–176), highly supersatu
Page 339
Figure 8 Effects of calcium (lower two pairs of panels) and APF (right panels) on distribution of cholesterol among multilamellar vesicles (gray bars), early crystals (white bars) and mature cholesterol monohydrate crystals (black bars) at various intervals after acute dilution of concentrated model bile. (From Ref. 59.)
Page 340
rated with cholesterol (59), hindering cholesterol crystallization from cholesterolrich domains within these structures (257). Thus, APF had effects on cholesterol precipitation exactly opposite to calcium, and could markedly suppress the enhancement of cholesterol precipitation by added calcium (Fig. 8) (59). With fluorescein labeled APF, it was shown that the APF was bound primarily to the large vesicles, presumably accounting its effect (59), but binding of bile salts by APF might have contributed also. The small proportions of CBP in the APF preparation used (236) were too small to attribute the observations to binding of calcium ions, and APF has little affinity for calcium (236). It thus appears that APF stabilizes cholesterol within supersaturated multilamellar and aggregated vesicles, markedly decreases the rate of crystal formation, and largely suppresses the effect of calcium (59). Several observations to this concept should be noted, however: (a) addition of APF preparations to human bile samples has not altered the distribution of cholesterol among various carriers or the time to first appearance of cholesterol crystals (245); (b) ELISA assay has revealed no differences in APF/CBP concentrations in gallbladder biles between patients with cholesterol gallstones and control subjects without gallstones (237); and (c) nucleation times of biles from patients with gallstones are unrelated to APF concentrations (237). APF was increased in biles from pigmentstones subjects (237). Though, as noted earlier, APF does not seem to determine phospholipid or cholesterol secretion into bile, it may play a role in regulation of the uptake of lipoproteins and metabolism of lipids by the liver (258,259). Addition of APF to liposomes containing 14Ccholesterol, administered intravenously to rats, enhances the uptake and storage of the cholesterol by the liver while decreasing its conversion to bile salts (259). 3— Functions of CBP In mixed gallstones, immunoreactive APF/CBP is clearly associated with the calciumcontaining pigmented zones (Figs. 4 and 6 in Ref. 3), where it is in contact with the mucin bands at the interface between the pigment deposits and cholesterol layers (3,238). No APF/CBP is detected in the cholesterol layers, where the cholesterol crystals are associated with mucin (3,238). In view of the known calciumbinding properties of CBP as opposed to APF (236), the immunostained APF/CBP associated with the calciumpigment zones is most probably CBP. In model systems in vitro, mucin and CBP alone each inhibit formation of calcium phosphate (51) and calcium carbonate (41) crystals, while the complex of the two proteins promotes calcium phosphate precipitation and alters the stoichiometry and form of the crystals (51). By contrast, addition of CBP (0.25 mg/mL) had no effects on cholesterol crystallization from model biles, whether or not calcium, calcium phosphate precipitates, or mucin was present (216). It should be noted, however, that the marked degree of supersaturation with CaHPO4 in that study may have precluded inhibition of precipitation of that salt despite the high concentration of CBP used. The properties of CBP on calcium salt crystallization in model systems thus fulfill the criteria for a protein that regulates biomineralization of calcium salts (33,178), but these effects must be demonstrated also in supersaturated human biles before the role of CBP can be confirmed. Interestingly, the content of immunoreactive APF/CBP in gallbladder bile is onethird higher in patients with black pigment gallstones than in controls, and the ratio of APF/CBP to phospholipid concentrations is significantly higher in patients with pigment gallstones compared to cholesterol gallstones or to controls (237). By contrast, biomineralization theory would predict that lower CBP levels would favor calcium salt precipitation, since the excess of unbound CBP, which inhibits crystallization, would diminish in relation to the mucinbound CBP, which promotes crystallization. The apparent conflict may relate to the failure of ELISA assays to distinguish between APF and CBP (236).
Page 341
G— Biomineralization Functions of Other Proteins and Biliary Lipids AminopeptidaseN has been identified as the 130kDa protein which promotes crystallization of cholesterol from model biles (260). Curiously, immunostaining of gallstones reveals that this protein is not detected in the cholesterolrich zones but is distributed alongside APF/CBP in the calciumpigment zones, interposed between mucin and the calciumpigment precipitates (3). Effects of aminopeptidaseN on calcium binding and calcium salt precipitation need to be determined with the techniques used for APF/CBP (51). Other bile proteins which inhibit cholesterol precipitation from model biles (261) are discussed in Sec. 2, Chaps. 3, 5, and 7. Their distribution in relation to mucins, calcium, pigments and cholesterol need to be studied by immunostaining the cut surface of gallstones, akin to published studies for mucin, APF/CBP, aminopeptidase N and albumin (3). Bile salts. During the precipitation of calcium phosphates, the initial amorphous calcium phosphate (ACP) precipitate metamorphoses through a series of intermediate forms, including CaHPO4, to the least soluble and most stable form, hydroxyapatite (Ca10(PO4)6(OH)2) (125). Anionic bile salts can bind avidly to precipitates of ACP (Fig. 9) (262,263), with moderate affinity to Ca3(PO4)2 and with low avidity to other intermediate crystalline forms and hydroxyapatite (Fig. 10) (263), as well as CaCO3 (263). The bound bile salts inhibit both the transition of ACP to hydroxyapatite and the growth of hydroxyapatite crystals (169,264) as well as crystallization of CaCO3 (181). Both types of binding are greatest with glycineamidated and then unconjugated bile salts but very low with taurineamidated bile salts (Figs. 9A and 10) (262–264). Within each category, there is essentially no binding of trihydroxy or triketobile salts; among the avidlybinding dihydroxy bile salts, adsorption of ursodeoxycholates (3a ,7b hydroxy) is somewhat less than chenodeoxycholates (3a ,7 a hydroxy) or deoxycholates (3a , 12a hydroxy) (Fig. 9A and B and Fig. 10) (262–264). These relative affinities of various bile salts for precipitated calcium salts are reminiscent of, but not identical to, their relative affinities for dissolved Ca2+ ions (Table 3). The reasons for these differences in binding among bile salts are unknown. The adsorption is mainly of premicellar bile salts (Figs. 9B and 10) and is not evident below a certain threshold concentration of bile salts, which is less than half the critical micellar concentration (CMC) (262–264). It probably involves binding of the —COO and —OH groups of the bile salt monomers to exposed calcium ions on the surface of the crystal lattice, akin to their binding to soluble Ca2+ ions (9), followed by hydrophobic binding of a second bile salt monomer, covering the exposed hydrophobic b face of the bound bile salt (264). Above the critical micellar concentration, additional bile salts may complex with the bound monomer, but with less avidity than premicellar binding (Fig. 10) (264). Thus, since [BST] in bile are far above the CMC, it is unlikely that changes in [BST] in bile would greatly influence the inhibition of crystallization by bile salts. By contrast, increases in the ratio of glycine to taurineamidated bile salts in bile, due to interruption of the enterohepatic circulation or taurinedeficient diets (119), might augment the inhibitory effects of bile salts on crystal growth. The major mechanism by which bile salt anions inhibit the growth of hydroxyapatite crystals appears to be competition with dissolved anions for attachment to Ca2+ ions in the crystal lattice (264). The mechanisms for the steep increase in binding of BS as pH increases from 5.5 to 6.2 (Fig. 9C) (262), and the decline in binding as pH increases above that range (262,264), are not wellunderstood. Phospholipids are found in pigment gallstones, are almost entirely phosphatidylcholine, and have a fatty acid composition similar to that of biliary lecithins (7). It has been shown that added phospholipid affects both the time of onset of cholesterol crystallization from lithogenic human bile (265) and the nature and transitions of the crystals that are formed in supersaturated model biles (266) or in films on the surface of water (267). Different phospholipids may have different effects. The mechanisms of these phenomena are unclear.
Page 342
Figure 9 Adsorption of bile salts to amorphous calcium phosphate precipitates. A. Dependence on bile salt amidation and ring hydroxylation. (From Ref. 263.) B. Dependence on concentrations of glycineamidated bile salt. (From Ref. 263.) C. Dependence on pH. (From Ref. 262.) CBC denotes the critical binding concentration of the bile salt below which no bile salts were bound.
Page 343
Figure 9 Continued.
V— Overview and Future Directions A— Summary of Current Concepts The pathogenesis of gallstone disease is multifactorial: gallstone formation in the gallbladder results from supersaturation of bile with calcium salts and cholesterol, impaired balance among kinetic factors which either promote or inhibit precipitation, and an enhanced residence time in the gallbladder (9,32). Supersaturation with cholesterol is determined by its interactions with bile salts and phospholipids, particularly its relative concentrations in mixed vesicles with lecithin (32). Supersaturation with calcium salts is determined by the activities of Ca2+ ions and the ''calciumsensitive" anions in bile; the former is mainly governed by the activity of Ca2+ ions in plasma and GibbsDonnan forces across the biliary tract epithelium, generated by the large, impermeant polyanions in bile [mucins, and the simple and mixed micelles of bile salts (9)]. Once thermodynamic supersaturation is attained, the nucleation of microcrystals is apparently determined by kinetic factors (proteins, glycoproteins, and bile salts) (9,32), and the
Page 344
Figure 10 Adsorption of bile salts to calcium hydroxyapatite crystals. Effects of bile salt structure, concentration and selfassociation in the presence of 8 mM calcium chloride in the supernatant. Left ordinates, bile salt bound, micromoles per milligram of hydroxyapatite ( ); right ordinates, bile salt selfassociation, assessed by fluorescence of bile saltassociated 1,6dephenyl1,3,5hexatriene (DPH). (Modified from Ref. 264.)
subsequent growth and organization of gallstones is governed by biomineralization processes (2,3,33,41,51,268). In the structure of gallstones, by analogy with that of a building, mucins are the girders, the regulatory proteins are the mortar, and the calciumpigment granules and cholesterol crystals are the bricks (41). Mucins and the bound regulatory proteins not only form the matrix on which the deposition of the mineral components is organized, but also act as a template which directs the stoichiometry and form of the crystal structures. When free in solution, moreover, mucins and regulatory proteins, along with bile salts and calcium ions, may control the rates of crystallization by altering the stability of cholesterol in biliary vesicles and/or by "poisoning" growth centers on nascent crystals. Lectinbinding glycoproteins and APF appear to be the key regulatory proteins for cholesterol crystallization, while CBP is currently the only candidate regulatory protein for calcium precipitation. Mucins seem to have regulatory functions in both systems. B— Limitations of Current Concepts The concepts outlined above are based mainly on in vitro studies of the effects of addition of individual components to supersaturated model biles. Conflicting results in such studies may be related to differences in the species of bile salts or mucin utilized. Due to financial considerations, most studies in model biles have used taurocholate, rather than a natural mixture of the bile salts present in human bile, which are much more hydrophobic (269) and bind calcium
Page 345
much more avidly (9). Most studies using purified mucins have been performed with soluble mucins rather than polymerized mucin gels and with bovine gallbladder mucins, which, though similar, are not identical to human or hepatic bile mucins (202–205). While studies with model biles may demonstrate specific effects of a given component, conflicting results are frequently obtained when the same component is added individually to natural bile samples and/or when several components are added together (270,271). There may, moreover, be discrepancies between interactions shown in vitro and the localization or the association of a component with calcium salts or cholesterol in the stones [e.g., aminopeptidaseN promotes cholesterol crystallization (260), but is associated only with the calcium salts in gallstones (3)]. It has been proposed (261,270) that a specific role for a given bile component in gallstone formation is best demonstrated by showing that (a) its selective removal from, and addition to, normal and lithic samples of human bile produce consistent and concordant alterations in the precipitation of calcium salts and/or cholesterol and/or the organization and growth of crystals and stones and (b) its structure and/or its concentration in bile are significantly altered in patients with a given type of gallstone, by comparison with patients with other types of gallstones or no gallstones. Further support could be obtained from the demonstration, in animal models of pigment or cholesterol gallstone formation, that changes in these bile components precede the appearance of crystals and gallstones. Note, however, that some effects of some regulatory proteins are manifest only in cooperation with other specific proteins. C— Initiation of Mineral Nucleation and Precipitation Possibly the most interesting and least understood phenomenon in gallstone pathogenesis is the process that triggers the initial nucleation of calcium salts and cholesterol and their organization into gallstones. In animal models, a large increase in mucin secretion and gel formation seems to precede gallstone formation (221,222), but it is unclear if this applies also to human gallstone disease (34,53). Another possible trigger is a rapid increase in the relative proportion of pronucleating versus antinucleating factors and/or of activated and bound CBP, but there is as yet no evidence for these proposals. Black pigment gallstones contain calcium bilirubinates and, phosphates, and often carbonates also (11,272), suggesting that an increase in the Ca2+ activity and/or pH of bile is the trigger for forming these stones. By contrast, the presence of calcium bilirubinates and phosphates but little or no carbonates in the cores of cholesterol and mixed gallstones (2,8,15,26,27) as well as in some black pigment stones (15,272) militates against an increase in Ca2+ activity or pH as the trigger for formation of these types of stones, since those should lead also to precipitation of CaCO3. Calcium palmitate and bilirubinate are enriched in the cores of brown pigment gallstones (11,21,273), suggesting that the trigger in this case is bacterial hydrolases (147). Recent evidence for the presence of bacteria and bacterial b glucuronidase in the cores of mixed gallstones (154–157,274) indicates that bacterially mediated hydrolysis of bilirubin conjugates and phospholipids may initiate formation of these stones as well. Note that addition of b glucuronidase to lithogenic human bile enhances precipitation of other components as well as bilirubin (147,275). The single or multiple cavities, found almost uniformly in the core of mature gallstones (Fig. 1), appear to involve dissolution of matrix as well as mineral. Bacterial hydrolases are logical candidates for this process also and the salts and digested fragments of matrix proteins might well diffuse out through the porous interstices of the gallstones (276). In model biles, APF inhibits cholesterol crystallization (59) and unbound CBP inhibits calcium salt crystallization (51), while CBP bound to mucin promotes accretion of calcium salts in gallstones (51). The possibility that CBP is an activated form of APF suggests the hypothesis that the balance between APF and CBP levels in bile might determine whether cholesterol or calcium/pigment salts will precipitate. A low APF/CBP ratio would favor cholesterol precipitation, whereas a high ratio, mediated by activation of APF to CBP, would promote calcium/pigment precipitation. The testing of this hypothesis must await development
Page 346
of a means to individually measure APF and CBP concentrations in bile and distinguish their presence in various regions of gallstones. All these hypotheses need to be evaluated by analysis of bile components, including regulatory proteins and hydrolytic enzymes, in the period immediately preceding crystallization in animal models (221,222) or in humans at high risk for gallstone formation [e.g., morbidly obese patients undergoing rapid weight reduction (277)]. D— Conclusion In a rich and diversified fluid like bile, it is difficult to assess which components are directly or indirectly implicated in the precipitation processes and how they function (270). Major efforts are needed in the next few years to perform the types of studies proposed in the preceding paragraphs, to sequence and clone all the putative regulatory proteins (most importantly, APF and CBP), and to study the genetic regulation of gallstone disease in humans and in knockout mice, a process already begun for cholesterol gallstone formation (278,279). Acknowledgments We thank Paulette Lechène de la Porte, Nicole Domingo, Bert Groen, and Andre van den Berg for their critical reviews of the manuscript. Dr. Ostrow especially remembers Edward W. Moore, whose creative insights, during three decades of collaboration, led to the development of most of the concepts relating to calcium in bile, and to Arthur Veis, who opened the door for application of biomineralization theory to gallstone formation. Dr. Lafont sends posthumous thanks to Jacques Hauton, her former mentor, who guided her to the discovery of APF and the understanding of its multiple key roles in lipid metabolism and gallstone formation. References 1. Womack NA, Zeppa R, Irvin G. The anatomy of gallstones. Ann Surg 1963; 157:670–686. 2. Ostrow JD. Unconjugated bilirubin and cholesterol gallstone formation. Hepatology 1990; 12:219S–226S. 3. Lechène de la Porte P, Domingo N, van Wijland M, Groen AK, Ostrow JD, Lafont H. Distinct immunolocalization of mucin and other biliary proteins in human cholesterol gallstones. J Hepatol 1996; 25:339–348. 4. Nakayama F. Quantitative microanalysis of gallstones. J Lab Clin Med 1969; 72:602–611. 5. Ananthakrishnan N, Rao BNB, Kapur BML. Studies on gallstone composition: IV. Analysis of gallstones by microchemical methods. Indian J Med Res 1975; 63:810–817. 6. Trotman BW, Morris TA III, Sanchez HM, Soloway RD, Ostrow JD. Pigment vs cholesterol cholelithiasis. Identification and quantification by infrared spectroscopy. Gastroenterology 1977; 72:495–498. 7. Robins SJ, Fasulo JM, Patton GM. Lipids of pigment gallstones. Biochim Biophys Acta 1982; 712:21–25. 8. Malet PF, Williamson CE, Trotman BW, Soloway RD. Composition of pigmented centers of cholesterol gallstones. Hepatology 1986; 6:477–481. 9. Moore EW. Biliary calcium and gallstone formation. Hepatology 1990; 12:206S–218S. 10. Grigorescu M, Suciu A, Acalovski M, Parian I, Duca S. The chemical composition of gallstones. Rom J Gastroenterol 1993; 2:195–197.
Page 347
11. Malet PF, Takabayashi A, Trotman BW, Soloway RD, Weston NE. Black and brown pigment gallstones differ in microstructure and microcomposition. Hepatology 1984; 2: 227–234. 12. Kienzle HF, Radtke J. Lokalisation und Verteilung von Calciumsalzen in Gallensteinen. Langenbecks Arch Chir 1980; 353:171–182. 13. Malet PF, Weston NE, Trotman BW, Soloway RD. Cyclic deposition of calcium salts during growth of cholesterol gallstones. In: Johari OM, ed. Scanning Electron Microscopy. Vol 2. Chicago: SEM, 1985, pp 775–779. 14. Kaufman HS, Lillemoe KD, Magnusson TH, Frasca P, Pitt HA. Backscattered electron imaging and windowless energy dispersive xray microanalysis: a new technique for gallstone analysis. Scanning Microsc 1990; 4:853–860. 15. Kaufman HS, Magnusson TH, Pitt HA, Frasca P, Lillemoe KD. The distribution of calcium salt precipitates in the core, peripery and shell of cholesterol, black pigment and brown pigment gallstones. Hepatology 1994; 19:1124–1132. 16. Taylor DR, Crowther RS, Cozart JC, Sharrock P, Wu J, Soloway RD. Calcium carbonate in cholesterol gallstones: polymorphism, distribution and hypothesis about pathogenesis. Hepatology 1995; 22:488–496. 17. Ohkubo H, Ostrow JD, Carr SH, Rege RV. Polymer networks in pigment and cholesterol gallstones assessed by equilibrium swelling and infrared spectroscopy. Gastroenterology 1984; 87:805–814. 18. Soloway RD, Trotman BW, Ostrow JD. Pigment gallstones. Gastroenterology 1977; 72: 167–182. 19. Trotman BW, Ostrow JD, Soloway RD. Pigment vs cholesterol cholelithiasis: comparison of stone and bile composition. Am J Dig Dis 1974; 19:585–590. 20. Cetta F, DeNisi A, Malet PF, Soloway RD. Black and brown pigment gallstones differ in microstructure and microcomposition (letter). Hepatology 1986; 6:153– 154. 21. Soloway RD, Trotman BW, Maddrey WC, Nakayama F. Pigment gallstone composition in patients with hemolysis or infection/stasis. Dig Dis Sci 1986; 31:454– 460. 22. Trotman BW, Soloway RD. Pigment gallstone disease: summary of the National Institutes of HealthInternational Workshop. Hepatology 1982; 2:879–884. 23. Wosiewitz U. Limy bile and radiopaque, calcified gallstones: a combined analytical, radiographic and micromorphologic examination. Pathol Res 1980; 167:273– 286. 24. Been JM, Bills PM, Lewis D. Microstructure of gallstones. Gastroenterology 1979; 76: 548–555. 25. Bogren H, Larsson K. On the pigment in biliary calculi. Scand J Clin Lab Invest 1963; 15:569–572. 26. Ruiz de Aguiar A, Medina Nunez JA, Lopez Domingo MI, Villacorta Patino J, Leguey Jimenez S. Calcium carbonate in cholesterol gallstones. J Hepatol 1988; 6:71–79. 27. Srivastava AK, Lal K, Tandon RK. A layerwise quantitation of human cholesterol gallstone constituents. Ind J Med Res 1983; 77:733–738. 28. Bogren HG, Mutvei H, Renberg G. Scanning electron microscope studies of human gallstones after plasma etching. Ultrastruct Pathol 1995; 19:447–452. 29. Kodaka T, Mori R, Debari K, Takiguchi R, Higashi S. Backscattered electron imaging and energydispersive xray microanalysis studies of evidence for calcium salt heterogeneity in fifteen gallstones from an elderly human. Scanning Microsc 1995; 9:907–920. 30. Moore EW. The role of calcium in the pathogenesis of gallstones: Ca2+ electrode studies of model bile salt solutions and other biologic systems. Hepatology 1984; 4:228S–243S. 31. Sutor DJ, Wilkie LI. Calcium in bile and calcium salts in gallstones. Clin Chim Acta 1977; 79:119–127. 32. Carey MC. Formation and growth of cholesterol gallstones: the new synthesis. In: Fromm H, Leuschner U, eds. Bile Acids—Cholestasis—Gallstones: Advances in Basic and Clinical Bile Acid Research. Dordrecht: Kluwer, 1996, pp 147–175.
Page 348
33. Ostrow JD. The roles of calcium salts and matrix in gallstone formation. In: Meyer zum Büschenfelde KH, Paumgartner G, Schölmerich J, eds. Perspectives in Gastroenterology. Munich: Urban & Schwarzenberg, 1995:143–152. 34. Lamont JT, Smith BF, Moore JRL. Role of gallbladder mucin in pathophysiology of gallstones. Hepatology 1984; 4:51S–56S. 35. Sutor DJ, Wooley SE. The organic matrix of gallstones. Gut 1974; 15:487–491. 36. Lamont JT, Ventola AS, Trotman BW, Soloway RD. Mucin glycoprotein content of human pigment gallstones. Hepatology 1983; 3:377–382. 37. Smith BF, Lamont JT. Identification of gallbladder mucinbilirubin complex in human cholesterol gallstone matrix: effects of reducing agents on in vitro dissolution of matrix and intact gallstones. J Clin Invest 1985; 76:439–445. 38. Pearson JP, Foster SNE. Mucus glycoprotein content of human cholesterol gallstones. Digestion 1987; 36:132–140. 39. Domingo N, Grosclaude J, Bekaert ED, Chapman MJ, Shimizu S, AyraultJarrier M, Ostrow JD, Lafont H. Epitope mapping of the human biliary amphipathic anionic polypeptide (APF): similarity with a calciumbinding protein (CBP) isolated from gallstones and bile and immunologic crossreactivity with apolipoprotein AI. J Lipid Res 1992; 33:1419–1430. 40. Shimizu S, Sabsay B, Veis A, Ostrow JD, Rege RV, Dawes LG. Isolation of an acidic protein from cholesterol gallstones, which inhibits the precipitation of calcium carbonate in vitro. J Clin Invest 1989; 84:1990–1996. 41. Ostrow JD. APF/CBP, an anionic polypeptide in bile and gallstones that may regulate calcium salt and cholesterol precipitation from bile. Hepatology 1992; 16:1493–1496. 42. Kestell MF, Sekijima J, Lee SP, Park HZ, Long M, Kaler EW. A calciumbinding protein in bile and gallstones. Hepatology 1992; 16:1315–1321. 43. Murray FE, Smith BF. Nonmucin glycoproteins in the matrix of human cholesterol gallstones. Scand J Gastroenterol 1991; 26:717–723. 44. Sutor DJ, Wooley SE. The sequential deposition of crystalline material in gallstones: evidence for changing gallbladder bile composition during the growth of some stones. Gut 1974; 15:130–131. 45. Womack NA. The development of gallstones. Surg Gynecol Obstet 1971; 133:937–945. 46. Bills PM, Lewis D. A structural study of gallstones. Gut 1975; 16:630–637. 47. Westbroek P, DeJong EW. Biomineralization and Biological Metal Accumulation: Biological and Geological. Dordrecht: Riedel, 1982, pp 1–533. 48. Lowenstam HA, Weiner S. Mineralization by organisms and the evolution of biomineralization. In: Westbroek P, DeJong EW, eds. Biomineralization and Biological Metal Accumulation. Dordrecht: Reidel, 1983, pp 191–203. 49. Veis A, Sabsay B. Bone and tooth formation: insights into biomineralization strategies. In: Westbroek P, DeJong EW, eds. Biomineralization and Biological Metal Accumulation: Biological and Geological. Dordrecht: Riedel, 1982, pp 273–284. 50. Berman A, Hanson J, Leiserowitz L, Koetzle TF, Weiner S, Addadi L. Biological control of crystal texture: a widespread strategy for adapting crystal properties to function. Science 1993; 259:776–779. 51. Afdhal NH, Ostrow JD, Koehler RK, Niu N, Groen AK, Veis A, Nunes DP, Offner GD. Interaction of bovine gallbladder mucin and calciumbinding protein: effects on calcium phosphate precipitation. Gastroenterology 1995; 109:1661–1672. 52. Levy PF, Smith BF, Lamont JT. Human gallbladder mucin accelerates in vitro nucleation of cholesterol in artificial bile. Gastroenterology 1984; 87:270–275. 53. Smith BF, Lamont JT. Gallbladder mucin and gallstone formation. In: Cohen S, Soloway RD, eds. Gallstones. New York: Churchill Livingstone, 1985:101–111. 54. Lipsett PA, Hildreth J, Kaufman HS, Lillemoe KD, Pitt HA. Human gallstones contain pronucleating nonmucin glycoproteins that are immunoglobulins. Ann Surg 1994; 219: 25–33.
Page 349
55. Kibe A, Holzbach RT, LaRusso NF, Mao SJ. Inhibition of cholesterol crystal formation by apolipoproteins in supersaturated model bile. Science 1984; 225:514– 516. 56. Gallinger S, Harvey PRC, Petrunka CN, Ilson RG, Strasberg SM. Biliary proteins and the nucleation defect in cholesterol cholelithiasis. Gastroenterology 1987; 92: 867–875. 57. Dawes LG, Moore EW, Rege RV, Shimizu S, Ostrow JD. Canine bile contains anticrystallization factors which inhibit precipitation of CaCO3. Hepatology 1991; 14:701–706. 58. Smith BF, Lamont JT. Bovine gallbladder mucin binds bilirubin in vitro. Gastroenterology 1983; 85:707–712. 59. Konikoff FM, Lechène de la Porte P, Laufer H, Domingo N, Lafont H, Gilat T. Calcium and the anionic polypeptide fracton (APF)have opposing effects on cholesterol crystallization in model bile. J Hepatol 1997; 27:707–715. 60. Groen AK, Ottenhoff R, Jansen PLM, van Marle J, Tytgat GNJ. Effect of cholesterol nucleationpromoting activity on cholesterol solubilization in model bile. J Lipid Res 1989; 30:51–58. 61. Groen AK, Noordam C, Drapers JAG, Egbers P, Jansen PLM, Tytgat GNJ. Isolation of a potent cholesterol nucleationpromoting activity from human gallbladder bile: role in the pathogenesis of gallstone disease. Hepatology 1990; 11:525–533. 62. SiggaardAndersen O, Thode J, FoghAndersen N. What is ionized calcium? Scand J Clin Lab Invest 1983; 43(suppl 165):11–16. 63. Moore EW. Role of calcium in the pathogenesis of gallstones with an hypothesis of structural requirements for Ca++binding to proteins and bile acids. In: Barbara L, Dowling RH, eds. Recent Advances in Bile Acid Research. New York: Raven Press, 1985, pp 109–143. 64. Rege RV, Moore EW, Nahrwold DL. Pathogenesis of calciumcontaining gallstones: relationship of total calcium and free ionized Ca++ in canine gallbladder and duct bile. Surg Forum 1985; 36:132–134. 65. Rege RV, Dawes LG, Moore EW. Biliary calcium secretion in the dog occurs primarily by passive convection and diffusion and is linked to bile flow. J Lab Clin Med 1990; 115:593–602. 66. Shiffman ML, Sugerman HJ, Kellum JM, Moore EW. Calcium in human gallbladder bile. J Lab Clin Med 1992; 120:875–884. 67. Gleeson D, Hood KA, Murphy GM, Dowling RH. Calcium and carbonate ion concentrations in gallbladder and hepatic bile. Gastroenterology 1992; 102:1707– 1716. 68. Knyrim K, Vakil N. Bile composition, microspheroliths, antinucleating activity, and gallstone calcification. Gastroenterology 1992; 103:552–559. 69. Rudnicki M, Jorgensen T, Thode J. Increased activity of ionized calcium in gall bladder bile in gallstone disease. Gut 1992; 33:1404–1407. 70. Sutor DJ, Wilkie LI, Jackson MJ. Ionised calcium in pathological human bile. J Clin Pathol 1980; 33:86–88. 71. Rege RV, Nahrwold DL, Moore EW. Absorption of biliary calcium from the canine gallbladder: protection against the formation of calciumcontaining gallstones. J Lab Clin Med 1987; 110:381–386. 72. Rege RV, Moore EW. Convective movement of Ca2+ ions across guinea pig gallbladder epithelium. Am J Physiol 1992; 262:G990–G995. 73. Ahrendt SA, Ahrendt GM, Pitt HA, Moore EW, Lillimoe KD. Hypercalcemia decreases bile flow and increases biliary calcium in the prairie dog. Surgery 1995; 117:435–442. 74. Rege RV, Lee MJ, Moore EW. The GibbsDonnan equilibrium: its effect on the distribution of calcium (Ca2+) across rabbit gallbladder epithelium (abstr). Gastroenterology 1986; 90:1761. 75. FoghAndersen N, Thode J, SiggaardAndersen O. Ionized calcium during dialysis and ultrafiltration. Scand J Clin Lab Invest 1983; 43(suppl 165):39–41. 76. Shiffman ML, Sugerman HJ, Moore EW. Human gallbladder mucosal function: effect
Page 350
of concentration and acidification of bile on cholesterol and calcium solubility. Gastroenterology 1990; 99:1452–1459. 77. O'Grady SM, Wolters PJ. Sodium and chloride transport across the isolated porcine gallbladder. Am J Physiol 1989; 257:C45–C51. 78. Svanvik J. Role of gallbladder in modifying hepatic bile composition. In: Tavoloni N, Berk PD, eds. Hepatic Transport and Bile Secretion: Physiology and Pathophysiology. New York: Raven Press, 1993:607–618. 79. Scarpa A. Measurements of cation transport with metallochromic indicators. Methods Enzymol 1979; 56:301–308. 80. Moore EW, Ostrow JD. Some Ca2+ electrodes misrepresent Ca2+ binding by bile salts (letter). Gastroenterology 1995; 109:662. 81. Thode J, Wandrup J, Aas F, SiggaardAndersen O. Evaluation of a new semiautomatic electrode system for simultaneous measurement of ionized calcium and pH. Scand J Clin Lab Invest 1982; 42:407–415. 82. Donovan JM, Leonard MR, Batta AK, Carey MC. Calcium affinity for biliary lipid aggregates in model biles: complementary importance of bile salts and lecithin. Gastroenterology 1994; 107:831–846. 83. Moore EW, Ross JW. NaCl and CaCl2, activity coefficients in mixed aqueous solutions. J Appl Physiol 1965; 20:1332–1336. 84. Boink ABTJ, Gimpel JA, Maas AHJ. A comparison of four Ca2+ analyzers. Scand J Clin Lab Invest 1983; 43(suppl 165):17–19. 85. Bowers GN Jr, Rains TC. Measurement of total calcium in biological fluids: flame atomic absorption spectrometry. Methods Enzymol 1988; 158:302–319. 86. O'Brien CB, Malet PF, Saul SH, Shiau YF, Soloway RD. Mucosal and bulk phase gallbladder pH influences the calcium bilirubin composition of the surface layer of cholesterol stones in man (abstr). Gastroenterology 1986; 90:1752. 87. Marteau C, Sastre B, Iconomidis N, Portugal H, Pauli AM, Gérolami A. pH regulation in human gallbladder bile: study in patients with and without gallstones. Hepatology 1990; 11:997–1002. 88. Buanes T, Grotmol T, Landsverk T, Raeder MG. Secretin empties bile duct cell cytoplasm of vesicles when it initiates ductular Gastroenterology 1988; 95:417–424.
secretion in the pig.
89. Rege RV, Moore EW. Pathogenesis of calciumcontaining gallstones: canine ductular bile, but not gallbladder bile, is supersaturated with calcium carbonate. J Clin Invest 1986; 77:21–26. 90. Petersen KU, Wehner F, Winterhager JM. Na/H exchange at the apical membrane of guineapig gallbladder epithelium: properties and inhibition by cyclic AMP. Pflugers Arch Eur J Physiol 1985; 405(suppl 1):S115–S120. 91. Heintze K, Petersen KU, Wood JR. Effects of bicarbonate on fluid and electrolyte transport by guinea pig and rabbit gallbladder: stimulation of absorption. J Membr Biol 1981; 62:175–181. 92. Silviani V, Colombiani V, Heyries L, Gerolami A, Cartouzou G, Marteau C. Role of NHE3 isoform of the Na+/H+ exchanger in sodium absorption by the rabbit gallbladder. Pflugers Arch Eur J Physiol 1996; 432:791–796. 93. Colombiani V, Silviani V, Marteau C, Lerique B, Cartouzou G, Gerolami A. Presence of the NHE3 isoform of the Na+/H+ exchanger in human gallbladder. Clin Sci 1996; 91:209–212. 94. Jacyna MR, Ross PE, Hopwood D, Bouchier IAD. The effect of secretin on sodium ion absorption by the isolated human gallbladder. Aliment Pharmacol Therap 1989; 3:293–297. 95. Jacyna MR. Interactions between gall bladder bile and mucosa: relevance to gall stone formation. Gut 1990; 31:568–570. 96. Svanvik J, Allen B, Pellegrini C, Bernhoft R, Way L. Variations in concentrating function of the gallbladder in the conscious monkey. Gastroenterology 1984; 86:919–925.
Page 351
97. Igimi H, Yamamoto F, Lee SP. Gallbladder mucosal function: Studies in absorption and secretion in humans and cultured dog gallbladder epithelium. Am J Physiol 1992; 263:G69–G74. 98. Moser AJ, Abedin MZ, Cates JA, Giurgiu DI, Karam JA, Roslyn JJ. Converting gallbladder absorption to secretion: the role of intracellular calcium. Surgery 1996; 119:410–416. 99. Cates JA, Abedin MZ, SaundersKirkwood KD, Moser AJ, Giurgiu DI, Roslyn JJ. Protein kinase C regulates prairie dog gallbladder ion transport. Surgery 1995; 117:206–212. 100. Moser AJ, Abedin MZ, Giurgiu DI, Roslyn JJ. Octreotide promotes gallbladder absorption in prairie dogs: a potential cause of gallstones. Gastroenterology 1995; 108:1547–1555. 101. Conter RL, Roslyn JJ, PorterFink V, DenBesten L. Gallbladder absorption increases during early cholesterol gallstone formation. Am J Surg 1986; 151:184– 191. 102. Roslyn JJ, Doty JE, Pitt HA, Conter RL, DenBesten L. Enhanced gallbladder absorption during gallstone formation: the roles of cholesterol saturated bile and gallbladder stasis. Am J Med Sci 1986; 292:75–80. 103. Giurgiu DI, SaundersKirkwood KD, Roslyn JJ, Abedin MZ. Sequential changes in biliary lipids and gallbladder ion transport during gallstone formation. Ann Surg 1997; 225:382–390. 104. Jacyna MR, Ross PE, Hopwood D, Bouchier IAD. Sodium transport in the diseased human gallbladder and the effects of indomethacin. Clin Sci 1988; 75:147– 149. 105. Ostrow JD, Mukerjee P, Tiribelli C. Structure and binding of unconjugated bilirubin: relevance for physiological and pathophysiological function. J Lipid Res 1994; 35:1715–1737. 106. Ostrow JD, Celic L, Mukerjee P. Solvent partition of unconjugated bilirubin (B) from CHCl3 into bile salt solutions: evidence that affinity of B for bile salts increases with ionization of B (abstr). Gastroenterology 1993; 104:A968. 107. Ostrow JD, Celic L, Webster CC, Mukerjee P. Binding of unconjugated bilirubin (UCB) to various bile salts at pH 9.0, assessed by solvent partition from CHCl3 (abstr). Gastroenterology 1994; 106:A957. 108. D'Alagni M, Galantini L, Giglio E, Gavuzzo E, Scaramuzza L. Micellar aggregates of sodium glycocholate and sodium taurocholate and their interaction complexes with bilirubin IX . J Chem Soc Faraday Trans 1994; 90:1523–1532. 109. Ostrow JD. Absorption by the gallbladder of bile salts, sulfobromophthalein, and iodipamide. J Lab Clin Med 1969; 74:482–494. 110. Ostrow JD. Absorption of bile pigments by the gallbladder. J Clin Invest 1967; 46:2035–2052. 111. Neiderhiser DH, Pineda FM, Hejduk LJ, Roth HP. Absorption of oleic acid by the guinea pig gallbladder. J Lab Clin Med 1971; 77:985–992. 112. Rege RV, Wallner EI, Neri KM, Moore EW. Absorption of free fatty acids (FFA) from guinea pig gallbladder: effects of the unstirred water layer (UWL). Gastroenterology 1991; 100:A336. 113. Sutor DJ, Wilkie LI. Diurnal variations in pH of pathological gallbladder bile. Gut 1976; 17:971–974. 114. Selle JG, Altemeier WA, Fullen WD, Goldsmith RE. Cholelithiasis in hyperparathyroidism. Arch Surg 1972; 105:369–374. 115. Werner S, Hjern B, Sjoberg HE. Primary hyperparathyroidism. Acta Chir Scand 1974; 140:618–625. 116. Jones CA, Hofmann AF, Mysels KJ, Roda A. The effect of calcium and sodium ion concentration on the properties of dilute aqueous solutions of glycine conjugated bile salts: phase behavior and solubility products of the calcium salts of the common glycine conjugated bile acids. J Colloid Interface Sci 1986; 114:452– 470.
Page 352
117. Gu JJ, Hofmann AF, TonNu HT, Schteingart CD, Mysels KJ. Solubility of calcium salts of unconjugated and conjugated natural bile acids. J Lipid Res 1992; 33:635–646. 118. Rossi SS, Converse JL, Hofmann AF. High pressure liquid chromatographic analysis of conjugated bile acids in human bile: simultaneous resolution of sulfated and unsulfated lithocholyl amidates and the common conjugated bile acids. J Lipid Res 1987; 28:589–595. 119. Ostrow JD. Metabolism of bile salts in cholestasis in humans. In: Tavaloni N, Berk PD, eds. Hepatic Transport and Bile Secretion; Physiology and Pathophysiology. New York: Raven Press, 1993, pp 673–712. 120. Carey MC, Lamont JT. Cholesterol gallstone formation: 1. Physical chemistry of bile and biliary lipid secretion. In: Boyer JL, Ockner RK, eds. Progress in Liver Diseases. Philadelphia: Saunders, 1992, pp 139–163. 121. Moore EW, Verine HJ. Pathogenesis of pancreatic and biliary CaCO3, lithiasis: the solubility product (K sp) of calcite determined with the Ca+ + electrode. J Lab Clin Med 1985; 106:611–618. 122. Moore EW, Verine HJ. Pancreatic calcification: Formation constants of CaHCO3 and 241:G182–G190.
complexes determined with Ca2+ electrode. Am J Physiol 1981;
123. Moore EW, Kelley EH, Keith FB, Krell H. Pathogenesis of phosphatecontaining gallstones: II. CaHPO4, precipitation and solubility product constant (K'sp) (abstr). Gastroenterology 1989; 96:A632. 124. Moore EW, Kelley EH, Keith FB, Krell H. Pathogenesis of phosphatecontaining gallstones: IV. Kinetics of CaHPO4 precipitation: the metastable phase— crystallization times (abstr). Gastroenterology 1989; 96:A633. 125. Nancollas GH. Crystallization in bile. Hepatology 1984; 4:169S–172S. 126. Moore EW, Keith FB, Krell H, Kelley EH. Pathogenesis of phosphatecontaining gallstones: III. Kinetics of CaHPO4 precipitation—nucleation and crystallization (abstr). Gastroenterology 1989; 96:A632. 127. Moore EW, Keith FB, Krell H, Kelley EH. Pathogenesis of phosphatecontaining gallstones: VI. Gallbladder bile is unsaturated with CaHPO4 in patients with gallstones or morbid obesity (abstr). Gastroenterology 1989; 96:A633. 128. Krell H, Ritzmann A, Feger R, Moore EW. Inorganic phosphate in bile: concentrations in bile of rat, hamster, and man (abstr). Hepatology 1989; 10:738. 129. Sutor DJ, Wilkie LI. Inorganic phosphorus in human bile. Clin Chim Acta 1977; 77:31–36. 130. Boonyapisit ST, Trotman BW, Ostrow JD. Unconjugated bilirubin and the hydrolysis of conjugated bilirubin in gallbladder bile of patients with cholelithiasis. Gastroenterology 1978; 74:70–74. 131. Fevery J, Blanckaert N, Leroy P, Michiels R, Heirwegh KPM. Analysis of bilirubins in biological fluids by extraction and thinlayer chromatography of the intact tetrapyrroles: application to bile of patients with Gilbert's syndrome, hemolysis, or cholelithiasis. Hepatology 1983; 3:177–183. 132. Shimizu S, Rege RV, Webster CC, Ostrow JD, Celic L, Moore EW. Highaffinity binding of ionized calcium (Ca2+) by conjugated bilirubin (abstr). Hepatology 1987; 7:1110. 133. Ostrow JD, Murphy NH. Isolation and properties of conjugated bilirubin from bile. Biochem J 1970; 120:311–327. 134. Reuben A, Howell KE, Boyer JL. Effects of taurocholate on the size of mixed lipid micelles and their associations with pigment and proteins in bile. J Lipid Res 1982; 23:1039–1052. 135. Verkade HJ, Havinga R, Gerding A, Vonk RJ, Kuipers F. Mechanism of bile acidinduced biliary lipid secretion in the rat: effect of conjugated bilirubin. Am J Physiol 1993; 264:G462–G469. 136. Trotman BW, Nair CR, Bernstein SE. Monoconjugated bilirubin is a major component of hemolysisinduced gallstones in mice. Hepatology 1988; 8:919–924.
Page 353
137. Vadali M, Ostrow JD, Mukerjee P, Celic L. Aqueous solubility products of calcium bilirubinate salts determined by solvent partition from chloroform into buffer solutions of CaCl2, (abstr). Proceedings of the 12th International Bile Acid Meeting, 1992. 138. Ho KJ, Hsu SC, Chen JS, Ho LHC. Human biliary b glucuronidase: correlation of its activity with deconjugation of bilirubin in the bile. Eur J Clin Invest 1986; 16:361–367. 139. Shull SD, Wagner CI, Trotman BW, Soloway RD. Factors affecting bilirubin excretion in patients with cholesterol or pigment gallstones. Gastroenterology 1977; 72:625–629. 140. Hahm JS, Ostrow JD, Mukerjee P, Celic L. Ionization and selfassociation of unconjugated bilirubin, determined by rapid solvent partition from chloroform, with further studies of bilirubin solubility. J Lip Res 1992; 33:1123–1137. 141. Carey MC, Koretsky AP. Self association of unconjugated bilirubin IXa in aqueous solution at pH 10 and physicochemical interactions with bile salt monomers and micelles. Biochem J 1979; 179:675–689. 142. Ostrow JD, Celic L, Mukerjee P. Unconjugated bilirubin (B) dianions ''comicellize" with taurocholate (TC): evidence from partition of B from CHCl3 into aqueous TC solutions (abstr). Hepatology 1991; 14:150A. 143. Sutor DJ, Wilkie LI. The crystalline salts of calcium bilirubinate in human gallstones. Clin Sci Mol Med 1977; 53:101–103. 144. Malet PF, Dabezies MA, Huang G, Long WB, Gadacz TR, Soloway RD. Quantitative infrared spectroscopy of common bile duct gallstones. Gastroenterology 1988; 94:1217–1221. 145. Crowther RS, Soloway RD. Pigment gallstone pathogenesis from man to molecules. Semin Liver Dis 1990; 10:171–180. 146. McDonagh AF, Lightner DA. Phototherapy and photobiology of bilirubin. Semin Liver Dis 1988; 8:272–281. 147. Maki T. Pathogenesis of calcium bilirubinate gallstones: Role of E. coli, b glucuronidase and coagulation by inorganic ions, polyelectrolytes and agitation. Ann Surg 1966; 164:90–100. 148. Maki T, Sato T, Saitoh T. A study on the activity of b glucuronidase in bile in connection with precipitation of calcium bilirubinate. Tohoku J Exp Med 1962; 77:179–186. 149. Wosiewitz U, Althoff J, Langhans P. Betaglucuronidase activity in human cholesterol and pigment stone bile. Z Gastroenterol 1982; 12:237–239. 150. Ho KJ. Human b glucuronidase: Studies on the effects of pH and bile acids in regard to its role in the pathogenesis of cholelithiasis. Biochim Biophys Acta 1985; 827:197–206. 151. Ho KJ, Ho LHC. Inhibitory effect of bile acids on human b glucuronidase activity at its optimal pH. Proc Soc Exp Biol Med 1981; 167:304–309. 152. Ho YD, Ho KJ. Biliary Dglucaric acid: Its quantitation and preventive role in gallstone formation. Scand J Gastroenterol 1990; 25:631–640. 153. Matsushiro T, Suzuki N, Sato T, Maki T. Effects of diet on glucaric acid concentraton in bile and the formation of calcium bilirubinate gallstones. Gastroenterology 1977; 72:630–633. 154. Liu YL, Lam K, Tang YJ, Gumerlock PH, Lee SP, Silva J Jr, Leung JW. Detection of bacterial b glucuronidase gene in human intrahepatic stone by the polymerase chain reaction (abstr). Gastroenterology 1997; 112:A515. 155. Swidsinski A, Khilkin M, Pahlig H, Swidsinski S, Priem F. Time dependent changes in the concentration and type of bacterial sequences found in cholesterol gallstones. Hepatology 1998; 27:662–665. 156. Liu YL, Lam K, Shi XB, Gumerlock PH, Tang YJ, Lee SP, Leung JW. Analysis of Clostridium sp. in human intrahepatic stones by SSCP method (abstr). Gastroenterology 1997; 112:A1322. 157. Lee DK, Tarr PI, Haigh WG, Lee SP. Bacterial DNA in mixed cholesterol gallstones. Am J Gastroenterol 1999; 94:3502–3506.
Page 354
158. Rege RV, Webster CC, Ostrow JD, Carr SH, Ohkubo H. Validation of infrared spectroscopy for assessment of vinyl polymers of bilepigment gallstones. Biochem J 1984; 224:871–876. 159. Zilm KW, Grant DM, Englert E Jr, Straight RC. The use of 13C nuclear magnetic resonance for the characterization of cholesterol and bilirubin pigment composition of human gallstones. Biochem Biophys Res Commun 1980; 93:857–866. 160. Woolfenden WR, Grant DM, Straight RC, Englert E Jr. Chemical characterization of pigment gallstones, using 13Cnuclear magnetic resonance analysis. Biochem Biophys Res Commun 1982; 107:684–694. 161. Shankland W. The ionic behavior of fatty acids solubilized by bile salts. J Colloid Interface Sci 1970; 34:9–25. 162. Sallee VL. Apparent monomer activity of saturated fatty acids in micellar bile salt solutions measured by a polyethylene partitioning system. J Lipid Res 1974; 15:56–64. 163. Sutor DJ. Calcium palmitate and palmitic acid in gallstones. Gut 1970; 11:618–619. 164. Ahlberg J, Curstedt T, Einarsson K, Sjövall J. Molecular species of biliary phosphatidylcholine in gallstone patients: the influence of treatment with cholic acid and chenodeoxycholic acid. J Lipid Res 1981; 22:404–409. 165. Wosiewitz U, Schenk J, Sabinski F, Schmack B. Investigations on common bile duct stones. Digestion 1983; 26:43–52. 166. Holan KR, Holzbach RT, Hermann RE, Cooperman WJ, Claffey WJ. Nucleation time: a key factor in the pathogenesis of cholesterol gallstone disease. Gastroenterology 1979; 77:611–617. 167. Shiffman ML, Carithers RCJ, MendezPicon G, Moore EW. Duct bile is supersaturated with CaCO3 in man: implications for the pathogenesis of gallstone formation (abstr). Hepatology 1987; 7:110. 168. Nancollas GH, Sawada K, Schuttringer E. Mineralization reactions involving calcium carbonates and phosphates. In: Westbroek P, DeJong EW, eds. Biomineralization and Biological Metal Accumulation. Dordrecht: Reidel, 1983, pp 155–169. 169. Qiu SM, Wen G, Hirakawa N, Soloway RD, Hong NK, Crowther RS. Glycochenodeoxycholic acid inhibits calcium phosphate precipitation in vitro by preventing the transformation of amorphous calcium phosphate to calcium hydroxyapatite. J Clin Invest 1991; 88:1265–1271. 170. Kaplun A, Talmon Y, Konikoff FM. Rubin M, Eitan A, Tadmor M, Lichtenberg D. Direct visualization of lipid aggregates in native human bile by light and cryo transmission electronmicroscopy. FEBS Lett 1994; 340:78–82. 171. Wang DQH, Carey MC. Complete mapping of crystallization pathways during cholesterol precipitation from model bile: influence of physicalchemical variables of pathophysiologic relevance and identification of a stable liquid crystalline state in cold, dilute and hydrophobic bilesalt containing systems. J Lipid Res 1996; 37:606–630. 172. Wang DQH, Carey MC. Characterization of crystallization pathways during cholesterol precipitation from human gallbladder biles: identical pathways to corresponding model biles with three predominating sequences. J Lipid Res 1996; 37:2539–2549. 173. Holzbach RT. Recent progress in understanding cholesterol crystal nucleation as a precursor to human gallstone formation. Hepatology 1986; 6:1403–1406. 174. Afdhal NH, Niu N, Nunes DP, Bansil R, Cao X, Gantz D, Small DM, Offner GD. Mucinvesicle interactions in model bile: evidence for vesicle aggregation and fusion before cholesterol crystal formation. Hepatology 1995; 22:856–865. 175. Halpern Z, Dudley MA, Lynn MP, Nader JM, Breuer AC, Holzbach RT. Vesicle aggregation in model systems of supersaturated bile: relation to crystal nucleation and lipid composition of the vesicular phase. J Lipid Res 1986; 27:295–306. 176. Halpern Z, Dudley MA, Kibe A, Lynn MP, Breuer AC, Holzbach RT. Rapid vesicle formation and aggregation in abnormal human bile: a timelapse video enhanced contrast microscopy study. Gastroenterology 1986; 90:875–885.
Page 355
177. Konikoff FM, Laufer H, Messer G, Gilat T. Monitoring cholesterol crystallization from lithogenic model bile by timelapse density gradient ultracentrifugation. J Hepatol 1997; 26:703–710. 178. Weiner S, Addadi L. Acidic macromolecules of mineralized tissues: the controllers of crystal formation. Trends Biochem Sci 1991; 16:252–256 179. MoradianOldak J, Frolow F, Addadi L, Weiner S. Interactions between acidic matrix macromolecules and calcium phosphate ester crystals: relevance to carbonate apatite formation in biomineralization. Proc R Soc Lond B 1992; 247:47–55. 180. Addadi L, Weiner S. Interactions between acidic proteins and crystals: stereochemical requirements for biomineralization. Proc Natl Acad Sci USA 1985; 82:4110–4114. 181. Marteau C, Portugal H, Pauli AM, Gerolami A. Effect of glycoursodeoxycholate on precipitation of calcium carbonate. Hepatology 1985; 5:1209–1212. 182. Busch N, Lammert F, Marschall HU, Matern S. A new subgroup of lectinbound biliary proteins binds to cholesterol crystals, modifies crystal morphology, and inhibits cholesterol crystallization. J Clin Invest 1995; 96:3009–3015. 183. Maki T, Matsushiro T, Suzuki N, Nakamura N. Role of sulfated glycoprotein in gallstone formation. Surg Gynecol Obstet 1971; 132:846–851. 184. Smith BF, Lamont JT. Hydrophobic binding properties of bovine gallbladder mucin. J Biol Chem 1984; 259:12170–12177. 185. Smith BF. Human gallbladder mucin binds biliary lipids and promotes cholesterol crystal nucleation in model bile. J Lipid Res 1987; 28:1088–1097. 186. Nancollas GH, Tomson MB. The precipitation of biological materials. Faraday Discuss 1976; 61:175–183. 187. Moreno EC, Varughese K, Hay DI. Effect of human salivary proteins on the precipitation kinetics of calcium phosphate. Calcif Tissue Int 1979; 28:7–16. 188. Schlesinger DH, Hay DI. Complete covalent structure of statherin, a tyrosinerich acidic peptide which inhibits calcium phosphate precipitation from human parotid saliva. J Biol Chem 1977; 252:1689–1695. 189. Nakagawa Y, Abram V, Kezdy FJ, Kaiser ET. Purification and characterization of the principal inhibitor of calcium oxalate monhydrate crystal growth in human urine. J Biol Chem 1983; 258:12594–12600. 190. Nakagawa Y, Ahmed M, Hall SL, DeGanello S, Coe FL. Isolation from human calcium oxalate renal stones of a glycoprotein inhibitor of calcium oxalate crystal growth: evidence that nephrocalcin from patients with calcium oxalate nephrolithiasis is deficient in carboxyglutamic acid. J Clin Invest 1987; 79:1782–1787. 191. DeCaro A, Multigner L, Lafont H, Lombardo D, Sarles H. The molecular characteristics of a human pancreatic acidic phosphoprotein that inhibits calcium carbonate crystal growth. Biochem J 1984; 222:669–677. 192. Bernard JP, Adrich Z, Montalto G, DeCaro A, De Reggi M, Sarles H, Dagorn JC. Inhibition of nucleation and crystal growth of calcium carbonate by human lithostatine. Gastroenterology 1992; 103:1277–1284. 193. Okido M, Soloway RD, Crowther RS. Inhibition of vaterite (CaCO3) formation by a calciumbinding protein from black pigment gallstones (abstr). Gastroenterology 1993; 104:A968. 194. van den Berg AA, van Buul JD, Tytgat GNJ, Ostrow JD, Groen AK. Cholesterol gallstone growth in model biles is inversely related to cholesterol crystal formation. Gastroenterology 1998; 114:G2237. 195. Strous GJ, Dekker J. Mucintype glycoproteins. In: Critical Reviews in Biochemistry and Molecular Biology. Boca Raton, FL: CRC Press, 1992, pp 57–92. 196. Smith BF, Peetermans JA, Tanaka T, Lamont JT. Subunit interactions and physical properties of bovine gallbladder mucin. Gastroenterology 1989; 97:179– 187. 197. Matsushiro T, Nemoto T, Endo M, Yosizawa Z. Glycoproteins and sulfated glycoproteins isolated from human bile. Clin Chim Acta 1970; 30:645–655.
Page 356
198. Lee SP, Lim TH, Scott AJ. Carbohydrate moieties of glycoproteins in human hepatic and gallbladder bile, gallbladder mucosa and gallstones. Clin Sci Mol Med 1979; 56:533–538. 199. Stanley RA, Lee SP, Roberton AM. Heterogeneity in gastrointestinal mucins. Biochim Biophys Acta 1983; 760:262–269. 200. van Wijland MJA, Klinkspoor JH, de Wit LT, Oude Elferink RPJ, Tytgat GNJ, Groen AK. Heterogeneity of human gallbladder mucin in bile. Clin Sci 1994; 86:67–74. 201. Klinkspoor JH, van Wijland MJA, Koeleman CAM, van Dijk W, Tytgat GNJ, Groen AK. Heterogeneity of human biliary mucin: functional implications. Clin Sci 1994; 86:75–82. 202. Afdhal NH, Offner GD, Smith BF. Characterization of bovine gallbladder mucin: amino acid sequences of tryptic peptides from the glycosylated domain of the protein core. Gastroenterology 1990; 99:1493–1501. 203. Afdhal NH, Offner GD, Murray FE, Troxler RF, Smith BF. Isolation and characterization of peptides from the protein core of bovine gallbladder mucin. Gastroenterology 1990; 98:1633–1641. 204. Nunes DP, Keates AC, Afdhal NH, Offner GD. Bovine gallbladder mucin contains two distinct tandem repeating sequences: evidence for scavenger receptor cysteinerich repeats. Biochem J 1995; 310(pt 1):41–48. 205. Keates AC, Nunes DP, Afdhal NH, Troxler RF, Offner GD. Molecular cloning of a major human gallbladder mucin: complete Cterminal sequence and genomic organization of MUC5B. Biochem J 1997; 324(pt 1):295–303. 206. Shiffman ML, Smith BF, Hirsch JI, Moore EW. Ca++binding to bovine gallbladder mucin: high affinity but low capacity (abstr). Hepatology 1989; 10:601. 207. Sterling RK, Shiffman ML, Smith BF, Moore EW. Ca2+binding by gallbladder mucin (GBM) is related to sialic acid content (abstr). Hepatology 1992; 16:155A. 208. Smith BF, Lamont JT. The central issue of cholesterol gallstones. Hepatology 1986; 6:529–531. 209. Malet PF, Deng SQ, Soloway RD. Gallbladder mucin and cholesterol and pigment gallstone formation in hamsters. Scand J Gastroenterol 1989; 24:1055– 1060. 210. Lee SP, Nicholls JF. Nature and composition of biliary sludge. Gastroenterology 1986; 90:677–686. 211. Carey MC, Cahalane MJ. Whither biliary sludge? Gastroenterology 1988; 95:508–523. 212. Gallinger S, Taylor RD, Harvey PRC, Petrunka CN, Strasberg SM. The effect of mucous glycoprotein on nucleation time of human bile. Gastroenterology 1985; 89:648–658. 213. Lee TJ, Smith BF. Bovine gallbladder mucin promotes cholesterol crystal nucleation from cholesteroltransporting vesicles in supersaturated model bile. J Lipid Res 1989; 30:491–498. 214. Afdhal NH, Niu N, Nunes DP, Bansil R, Caoxx, Gantz D, Small DM, Offner GD. Mucinvesicle interaction in model bile: evidence for vesicle aggregation and fusion before cholesterol crystal formation. Hepatology 1995; 22:856–865. 215. Afdhal NH, Niu N, Gantz D, Small DM, Smith BF. Bovine gallbladder mucin accelerates cholesterol monohydrate crystal growth in model bile. Gastroenterology 1993; 104:1515–1523. 216. van den Berg AA, van Buul JD, Tytgat GNJ, Groen AK, Ostrow JD. Mucins and calcium phosphate precipitates additively stimulate cholesterol crystallization. J Lipid Res 1998; 39:1744–1751. 217. de Bruijn MAC, Goldhoorn BG, Zijlstra AIM, Tytgat GNJ, Groen AK. Interaction of cholesterolcrystallization promoting proteins with vesicles. Biochem J 1995; 305:93–96. 218. Qiu SM, Wen G, Wen J, Soloway RD, Crowther RS. Interaction of human gallbladder mucin with calcium hydroxyapatite: binding studies and the effect on hydroxyapatite formation. Hepatology 1995; 21:1618–1624.
Page 357
219. Lee SP. Hypersecretion of mucus glycoprotein by the gallbladder epithelium in experimental cholelithiasis. J Pathol 1981; 134:199–207. 220. Rege RV, Nahrwold DL. Animal models of pigment gallstone disease. J Surg Res 1987; 43:196–203. 221. Rege RV, Dawes LG, Ostrow JD. Animal models of pigment gallstones. In: Cornelius CE, ed. Animal Models in Liver Research. Orlando, FL: Academic Press, 1993, pp 257–287. 222. Rege RV, Ostrow JD. Animal models of pigment and cholesterol gallstone disease. In: Muraca M, ed. Methods in Hepatobiliary Research. Boca Raton, FL: CRC Press, 1995, pp 203–243. 223. Lee SP, Lamont JT, Carey MC. Role of gallbladder mucus hypersecretion in the evolution of cholesterol gallstones: studies in the prairie dog. J Clin Invest 1981; 67:1712–1723. 224. Lee SP, Carey MC, Lamont JT. Aspirin prevention of cholesterol gallstone formation in prairie dogs. Science 1981; 211:1429–1431. 225. O'Leary DP, LaMorte WW, Scott TE, Booker ML, Stevenson J. Inhibition of prostaglandin synthesis fails to prevent gallbladder mucin hypersecretion in the cholesterolfed prairie dog. Gastroenterology 1991; 101:812–820. 226. Cohen BI, Mosbach EH, Ayyad N, Yashii M, McSherry CK. Aspirin does not inhibit cholesterol cholelithiasis in two established animal models. Gastroenterology 1991; 101:1109–1116. 227. Broomfield PH, Chopra R, Sheinbaum RC, Bonorris GG, Silverman A, Schoenfield LJ, Marks JW. Effects of ursodeoxycholic acid and aspirin on the formation of lithogenic bile and gallstones during loss of weight. N Engl J Med 1988; 319:1567–1572. 228. Harvey PRC, Rupar CA, Gallinger S, Petrunka CN, Strasberg SM. Quantitative and qualitative comparison of gallbladder mucus glycoprotein from patients with and without gall stones. Gut 1986; 27:374–381. 229. Shiffman ML, Shamburek RD, Schwartz CC, Sugerman HJ, Kellum JM, Moore EW. Gallbladder mucin, arachidonic acid, and bile lipids in patients who develop gallstones during weight reduction. Gastroenterology 1993; 105:1200–1208. 230. Trotman BW, Bongiovanni MB, Kahn MJ, Bernstein SE. A morphologic study of the liver and gallbladder in hemolysisinduced gallstone disease in mice. Hepatology 1982; 2:863–869. 231. Neiderhiser DH, Morningstar WA, Roth HP. Absorption of lecithin and lysolecithin by the gallbladder. J Lab Clin Med 1973; 82:891–897. 232. Neiderhiser DH, Harmon CK, Roth HP. Absorption of cholesterol by the gallbladder. J Lipid Res 1976; 17:117–124. 233. Gerlarden RT, Rose RC. Electrical properties and diffusion potentials in the gallbladder of man, monkey, dog, goose and rabbit. J Membrane Biol 1974; 19:37– 54. 234. Shiau YF, Kelemen RJ, Reed MA. Acidic mucin layer facilitates micelle dissociation and fatty acid diffusion. Am J Physiol 1990; 259:G671–G675. 235. Wallner EI, Moore EW, Rege RV. Amiloride decreases butyrate permeability across guinea pig gallbladder (abstr). Hepatology 1991; 14:268A. 236. Lafont H, Domingo N, Groen AK, Kaler EW, Lee SP, Koehler RK, Ostrow JD, Veis A. APF/CBP, the small, amphipathic, anionic protein(s) in bile and gallstones, consists of lipidbinding and calciumbinding forms. Hepatology 1997; 25:1054–1063. 237. Domingo N, Lafont H, Halpern Z, Peled Y, Grosclaude J, Gilat T. Anionic polypeptide fraction (APF) in bile of patients with and without gallstones. Hepatology 1993; 17:778–780. 238. Martigne M, Domingo N, Lechène de la Porte PL, Lafont H, Hauton JC. Identification and localization of the apoprotein fraction of the bile lipoprotein complex in human gallstones. Scand J Gastroenterol 1988; 23:731–737. 239. Hauton JC. The apoprotein of the bile lipoprotein complex (apoBLC). In: Fisher MM,
Page 358
Goresky CA, Shaffer EA, Strasberg SM, eds. Gallstones. New York: Plenum Press, 1979, pp 71–87. 240. Martigne M, Domingo N, Lafont H, Nalbone G, Hauton JC. Purification of the human anionic polypeptide fraction of the bile lipoprotein complex by zonal ultracentrifugation. Lipids 1985; 20:884–889. 241. Okido M, Shimizu S, Ostrow JD, Nakayama F. Isolation of a calcium regulatory protein from black pigment gallstones: similarity with a protein from cholesterol gallstones. Hepatology 1992; 15:1079–1085. 242. Suzuki H, Terada T. Removal of dodecyl sulfate from protein solution. Anal Biochem 1988; 172:259–263. 243. Nalbone G, Lafont H, Vigne JL, Domingo N, Lairon D, Chabert C, Lechène de la Porte P, Hauton JC. The apoprotein fraction of the bile lipoprotein complex: isolation, partial characterization and phospholipid binding properties. Biochimie 1979; 61:1029–1041. 244. Nalbone G, Lafont H, Lairon D, Vigne JL, Domingo N, Leonardi J, Hauton JC. Immunogenicity of the apoprotein of the bile lipoprotein complex. Biochimie 1978; 60:691–694. 245. Halpern Z, Lafont H, Arad J, Domingo N, Peled Y, Konikoff FM, Gilat T. The distribution of the biliary anionic polypeptide fraction between cholesterol carriers in bile and its effects on nucleation. J Hepatol 1994; 21:979–983. 246. Domingo N, Botta D, MartigneCros M, Lechène de la Porte P, PakLeung P, Hauton JC, Lafont H. Evidence for the synthesis and secretion of APF—a bile lipid associated protein—by isolated rat hepatocytes. Biochim Biophys Acta 1990; 1044:243–248. 247. Domingo N, Chanussot F, Botta D, Reynier MO, Crotte C, Hauton JC, Lafont H. Modulating effects of bile salt hydrophobicity on bile secretion of the major protein of the bile lipoprotein complex. Lipids 1993; 28:883–887. 248. Chanussot F, Domingo N. Tuchweber B, Lafont H, Yousef IB. Influence of dehydrocholic and cholic acid infusions on the biliary secretion of anionic polypeptide fraction, the major apoprotein of the biliary lipoprotein complex. Scand J Gastroenterol 1992; 27:238–242. 249. Verkade HJ, Kuipers F, Domingo N, Havinga R, Leonardi J, Vonk RJ, Lafont H. Biliary secretion of anionic polypeptide fraction is not coupled to that of phospholipids and cholesterol in rats. Hepatology 1997; 25:38–47. 250. Toth JL, Harvey PRC, Upadyha GA, Strasberg SM. Albumin absorption and protein secretion by the gallbladder in man and in the pig. Hepatology 1990; 12:729–737. 251. Keulemans YCA, Mok KS, de Wit LT, Gouma DJ, Groen AK. Hepatic bile versus gallbladder bile; a comparison of protein and lipid concentration and composition in cholesterol gallstone patients. Hepatology 1998; 28:11–16. 252. Gallinger S, Harvey PRC, Petrunka CN, Strasberg SM. Effect of binding of ionized calcium on the in vitro nucleation of cholesterol and calcium bilirubinate in human gallbladder bile. Gut 1986; 28:1382–1386. 253. Neithercut WD. Effect of calcium, magnesium and sodium ions on in vitro nucleation of human gall bladder bile. Gut 1989; 30:665–670. 254. Gourdin TG, Afdhal NH, Niu N, Smith BF. Calcium augments binding of bilirubin to bovine gallbladder mucin (abstr). Hepatology 1990; 12:899. 255. Niu N, Gourdin TG, Smith BF. Calcium (Ca++) augments hydrophobic binding properties of bovine gallbladder mucin (BGM) (abstr). Gastroenterology 1990; 98:A615. 256. Teramen K. Tazuma S, Ohya T, Kajiyama G. Comparative effects of biliary concanavalin Abound glycoproteins and calcium ion on cholesterol crystal nucleation and growth in model bile. J Gastroenterol 1995; 30:500–507. 257. Carey MC, Cohen DE. Biliary transport of cholesterol in vesicles, micelles and liquid crystals. In: Paumgartner G, Stiehl A, Gerok W, eds. Bile Acids and the Liver. Lancaster, UK: MTP Press, 1986, pp 287–300. 258. Martigne M, Meli B, Mahlberg F, Domingo N, Chanussot F, Lafont H, Hauton JC. Detection and characterization of anionic polypeptidic fraction binding sites in rat liver
Page 359
plasma membranes and cultured hepatocytes. Biochim Biophys Acta 1989; 979:341–346. 259. Martigne M, Domingo N, Chanussot F, Nalbone G, Lafont H, Hauton JC. Effect of bile anionic polypeptidic fraction on the fate of cholesterol carried by liposomes in the rat. Proc Soc Exp Biol Med 1988; 187:229–234. 260. Offner GD, Gong D, Afdhal NH. Identification of a 130kilodalton human biliary concanavalin A binding protein as aminopeptidase N. Gastroenterology 1994; 106:755–762. 261. Harvey PRC, Strasberg SM. Will the real cholesterolnucleating and antinucleating proteins please stand up? (editorial). Gastroenterology 1993; 104:646–650. 262. Van der Meer R, De Vries HT. Differential binding of glycine and taurineconjugated bile acids to insoluble calcium phosphate. Biochem J 1985; 22:265–268. 263. Govers MJAP, Termont DSML, Van Aken GA, Van der Meer R. Characterization of the adsorption of conjugated and unconjugated bile acids to insoluble, amorphous calcium phosphate. J Lipid Res 1994; 35:741–748. 264. Qiu SM, Soloway RD, Crowther RS. Interaction of bile salts with calcium hydroxyapatite: inhibitors of apatite formation exhibit highaffinity premicellar binding. Hepatology 1992; 16:1280–1289. 265. Jüngst D, Lang T, Huber P, Lange V, Paumgartner G. Effect of phospholipids and bile acids on cholesterol nucleation time and vesicular/micellar cholesterol in gallbladder bile of patients with cholesterol stones. J Lipid Res 1993; 34:1457–1464. 266. Konikoff FM, Cohen DE, Carey MC. Phospholipid molecular species influence crystal habits and transition sequences of metastable intermediates during cholesterol crystallization from bile saltrich model bile. J Lipid Res 1994; 35:60–70. 267. Lafont S, Rapaport H, Sömjen GJ, Renault A, Howes PB, Kjaer K, AlsNielsen J, Lieserowitz L, Lahav M. Monitoring the nucleation of crystalline films of cholesterol on water and in the presence of phospholipids. J Phys Chem B 1998; 102:761–765. 268. Verder JM. Les inhibiteurs macromoleculaires de cristallisation dans la salive et dans le bile. Nephrologie 1993; 14:251–255. 269. Heuman DM. Quantitative estimation of the hydrophobichydrophilic balance of mixed bile 2salt solutions. J Lipid Res 1989; 30:719–730. 270. de Bruijn MAC, Mok KS, Out T, Tytgat GNJ, Groen AK. Immunoglobulins and 1 acid glycoprotein do not contribute to the cholesterol crystallization promoting effect of concanavalin Abinding biliary protein. Hepatology 1994; 20:626–632. 271. Keulemans YCA, Mok KS, Gouma DJ, Groen AK. The role of concanavalin Abinding fraction in cholesterol crystallization in native human bile. J Hepatol 1997; 27:1041–1050. 272. Trotman BW, Morris TA III, Cheney HM, Ostrow JD, Sanchez HM, Soloway RD, Conn HO. Pigment gallstone composition in cirrhotic and noncirrhotic subjects. Am J Dig Dis 1978; 230:872–876. 273. Liu CH, Chen GH, Soloway RD, Wu JG. Comparing core, rim and peripheral composition of intraductal gallstones with pathogenesis by infrared spectroscopy (abstr). Gastroenterology 1997; 112:A1322. 274. Stewart L, Oesterle A, Erden I, Way LW. Infectious and sterile gallstones: morphology, chemical composition, and bacterial betaglucuronidase production (abstr). Gastroenterology 1997; 112:A1477. 275. Higashijima H, Ichimiya H, Nakano T, Yamashita H, Kuroki S, Satoh H, Chijiiwa K, Tanaka M. Deconjugation of bilirubin accelerates coprecipitation of cholesterol, fatty acids, and mucin in human bile—in vitro study. J Gastroenterol 1996; 31:828–835. 276. Sanabria JR, Upadhya GA, Harvey PRC, Strasberg SM. Diffusion of substances into human cholesterol gallstones. Gastroenterology 1994; 106:749–754. 277. Shiffman ML, Sugerman HJ, Kellum JM, Brewer WH, Moore EW. Gallstones in patients with morbid obesity: relationship to body weight, weight loss, and gallbladder bile cholesterol solubility. Int J Obesity 1993; 17:153–158.
Page 360
278. Khanuja B, Cheah YC, Hunt M, Nishina PM, Wang DQH, Chen HW, Billheimer JT, Carey MC, Paigen B. Lithl, a major gene affecting cholesterol gallstone formation among inbred strains of mice. Proc Natl Acad Sci USA 1995; 92:7729–7733. 279. Wang DQH, Paigen B, Carey MC. Phenotypic characterization of Lith genes that determine susceptibility to cholesterol cholelithiasis in inbred mice: physical chemistry of gallbladder bile. J Lipid Res 1997; 38:1395–1411. 280. Cabral DJ, Small DM. Physical chemistry of bile. In: Schultz SG, Forte JG, Rauner BB, eds. Handbook of Physiology. The Gastrointestinal System III, Section 6. New York: Waverly Press, 1989:621–662. 281. Fini A, Roda A. Chemical properties of bile acids: IV. Acidity constants of glycineconjugated bile acids. J Lipid Res 1987; 28:755–759. 282. Hughes WS, Aurbach GD, Sharp ME, Marx SE. The effect of bicarbonate anion on serum ionized calcium concentration in vitro. J Lab Clin Med 1984; 103:93–103. 283. Toffaletti J, Gitelman HJ, Savory J. Separation and quantitation of serum constituents associated with calcium by gel filtration. Clin Chem 1976; 22:1968–1972. 284. Moore EW, Kelley EH, Keith FB, Krell H. Pathogenesis of phosphatecontaining gallstones: V. Saturation limits for total inorganic phosphate in bile as functions of [Ca2+] and pH (abstr). Gastroenterology 1989; 96:A633.
Page 361
16— Prevention of Gallstones Mitchell L. Shiffman Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia I— Introduction Gallstone disease is a major health problem throughout the western world. Approximately 10 to 15% of adults, or more than 20 million persons in the United States, have gallstones. These are found nearly twice as frequently in women than in men—18 versus 9% (1,2). Approximately 80% of all patients with gallstones are asymptomatic. However, several large prospective studies have demonstrated that 1 to 3% of patients with silent gallstones become symptomatic each year (3–5). It is the appearance of symptoms that leads to a new diagnosis of gallstones in approximately 1 million patients annually. Severe complications of gallstones include acute cholelithiasis, cholangitis with sepsis, acute pancreatitis, and gallbladder carcinoma. Of these, acute cholecystitis is the most common complication, occurring in nearly 10% of symptomatic patients (6,7). Cholecystectomy is a very safe and effective therapy for symptomatic gallstone disease. Biliary pain is relieved in 99% of cases (8,9). Mortality in very large series remains under 1% (10–12). The most common cause of death following cholecystectomy results from unsuspected cardiac disease (10). Morbidity following cholecystectomy, both long and short term, appears to develop in 5% of patients or less (10–12). A variable percentage of patients develop postcholecystectomy sequellae, which include bilious gastroesphageal reflux and postprandial diarrhea or dumping (13,14). Cholecystectomy reduces the bile salt pool, increases bile salt recycling, and increases the content of hydrophobic bile acids, which may be toxic and/or precarcinogenic to colonic mucosa (15). More than a dozen epidemiological studies from seven different countries have suggested that cholecystectomy may increase the longterm risk of developing colonic carcinoma (16–27), although the methodology utilized in many of these studies remains controversial. Cholecystectomy remains one of the most common operative procedures performed in the United States. In 1991, shortly after the introduction of laparoscopic cholecystectomy to this country, approximately 600,000 patients underwent cholecystectomy (28). Today, cholecystectomy is performed via laparoscopy in over 90% of cases. This technique has significantly reduced morbidity, inpatient hospital days, and recovery following cholecystectomy compared with the open surgical approach (29). However, laparoscopic cholecystectomy has not reduced I would like to dedicate this chapter to my mentor, colleague, and friend Edward W. Moore, MD, who taught me how to think scientifically and critically and shared with me his love for medicine, science, and life.
Page 362
the overall cost of caring for patients with gallstone disease, as originally predicted (29,30). Rather, the widespread acceptance of this technique by patients and surgeons alike has led to a dramatic increase in the number of cholecystectomies performed to over 800,000 annually (28). As a result, treatment of gallstone disease has become the most costly of all digestive disorders. In 1994 this cost exceeded $2 billion (31). Preventing gallstones from forming may therefore be the only manner by which to reduce the morbidity, mortality, and the cost of caring for patients with gallstone disease. This chapter focuses on ways to prevent the development of gallstones and/or to prevent gallstones from becoming symptomatic and requiring surgical treatment. The pathogenesis and risk factors associated with the development of cholesterol and pigment gallstones were discussed in previous chapters. In this chapter, those pathogenic factors thought to be important for development of gallstones in select highrisk populations are reviewed. Special emphasis is placed on those factors that could be modified by diet, exercise, or pharmacological therapy to reduce gallstone formation. Several studies have already demonstrated that ursodeoxycholic acid is very effective therapy for prevention of cholesterol gallstones in patients undergoing rapid weight reduction (32–35). These data are reviewed and the applicability of this treatment to other populations at high risk for cholesterol gallstone formation is discussed. Data regarding other pharmacological agents or treatments that have been hypothesized but not proven to reduce the risk for gallstone formation are also summarized. II— Cholesterol Gallstones Approximately 80% of gallstones found in patients living in the western world are classified as cholesterol (36). These stones are made up of more than 50% cholesterol but also contain variable amounts of calcium, bilirubin, and mucin glycoprotein (37–39). The pathogenesis of cholesterol gallstones was discussed in previous chapters. This is reviewed briefly here, with special emphasis on those factors thought to be important for the prevention of gallstones. Four factors are considered essential for cholesterol gallstones to form: (a) bile must be supersaturated with cholesterol, (b) precipitation of cholesterol crystals must occur, (c) gallbladder mucosa must secrete excess mucin, and (d) gallbladder motility must be reduced. Each of these is considered below. A— Cholesterol Solubility Cholesterol is virtually insoluble in an aqueous environment (40). To solubilize this biliary lipid, the liver secretes bile salts and phospholipids. These amphipathic molecules form both micelles and vesicles, which incorporate cholesterol and thereby markedly increase cholesterol solubility within bile (36,37,41). The proportion of bile salts and phospholipids necessary to maintain cholesterol in solution was originally described by Amirand and Small (42). Additional studies on model solutions composed of bile salts, lecithin, and cholesterol by Carey and Small led to the concept and definition of the cholesterol saturation index (CSI) (43). The CSI has since been utilized to assess cholesterol solubility in human bile under various conditions. Whenever the amount of cholesterol in bile exceeds the solubilizing capacity of bile salts and phospholipids, CSI exceeds 1 and bile is referred to as being either lithogenic or supersaturated with cholesterol. In this state, solid crystals of cholesterol monohydrate may precipitate out of solution and begin the process of gallstone formation. Gallbladder bile is supersaturated with cholesterol in most patients with cholesterol gallstones (44). In contrast, when gallbladder bile is unsaturated, cholesterol cannot precipitate and gallstone formation is impossible. Any factor that increases biliary cholesterol secretion to a greater extent than the secretion of bile salts and/or phospholipids will render bile supersaturated. Genetic alterations in the relative proportions of cholesterol, bile salts, and phospholipids secreted into bile are thought
Page 363
to account for the increase in CSI and increased incidence of gallstones observed in some American Indians and the Scandinavian population (45,46). Certain drugs utilized to treat hyperlipidemia, such as clofibrate, also enhance secretion of cholesterol into bile and increase CSI (47). A reduction in the secretion of bile salts relative to cholesterol also reduces cholesterol solubility within bile. The increased risk for gallstone formation observed in some nonobese women has been attributed to such an alteration in biliary lipid secretion (48,49). Sex steroids are an important factor affecting biliary cholesterol secretion. Estrogens increase the secretion of cholesterol into bile and CSI (50–52). This effect of estrogens is observed not only during pregnancy but also in women taking oral contraceptive steroids. In contrast, biliary cholesterol secretion appears to be unaffected by the normal ovulatory cycle (51). This effect of estrogens on biliary cholesterol secretion accounts in part for the increased incidence of gallstone formation observed during and following pregnancy and in multiparous women (2,45,53). Of the various factors that enhance cholesterol secretion into bile, alterations in body weight appear to have the most profound effect. Both biliary cholesterol secretion and CSI increase with increasing body weight (54–57). This may account for the stepwise increase in gallstone risk observed with increasing body weight (2,58–62). Cholesterol secretion into bile is also increased during rapid weight loss (56,57), such as that which occurs in patients participating in a very low calorie diet program (32) and following gastric bypass surgery (63,64). Both of these populations are at high risk for gallstone formation (32–35, 65–68). B— Nucleation and Cholesterol Crystal Growth Crystals of cholesterol do not always form and precipitate out of solution when bile is supersaturated with this lipid (69,70). Rather, it appears that bile contains both pro and antinucleating proteins, which regulate the formation, growth, and precipitation of cholesterol crystals from supersaturated bile (71–73). Candidate proteins that appear to have strong nucleating effects include immunoglobulins (73,74) and gallbladder mucin (75–78). This later glycoprotein is discussed in detail below. The balance between pro and antinucleating proteins may be altered in patients who develop gallstones. For example, cholesterol crystals develop within days from supersaturated gallbladder bile obtained from patients with gallstones (69). In contrast, crystals may not develop for weeks to months, if at all, when gallbladder bile with an equal CSI is obtained from patients without gallstones. Cholesterol crystals will also precipitate from supersaturated model bile solutions when gallbladder bile from patients with gallstones is added, but not after the addition of bile from patients without gallstones (79). Despite extensive work in this area little is known regarding the mechanism by which many of these proteins function and how they may be affected by body weight, weight loss, and other factors thought to be associated with gallstone formation. C— Gallbladder Mucin Several lines of evidence strongly support the role of gallbladder mucin in gallstone pathogenesis. In animal models of cholesterol gallstone disease hypersecretion of mucin by gallbladder mucosa precedes the development of gallstones (80–82), and inhibition of mucin secretion abolishes gallstone formation (83). Gallbladder mucin is a potent stimulus for nucleation and growth of cholesterol crystals (75–78). Mucin also binds biliary lipids and is found within the central nidus region of cholesterol gallstones (78,84,85). Secretion of gallbladder mucin appears to be stimulated by prostaglandins (86–88), which, in turn, are synthesized from arachidonic acid within gallbladder mucosa (88,89). Inhibitors of prostaglandin synthesis, such as aspirin and other nonsteroidal antiinflammatory drugs (NSAIDs), reduce the secretion of gallbladder mucin and prevent gallstone formation in animal models (83,88). It has been hypothesized that patients who develop gallstones secrete bile that is rich in the arachidonic acid
Page 364
precursor phosphatidylcholine (90). Alternatively, synthesis of prostaglandins by gallbladder mucosa may occur in response to inflammation, which is stimulated by either an increase in the concentration of hydrophobic bile acids within bile (91,92) and/or by the presence of cholesterol crystals (93). Hypersecretion of mucin creates a thick gel that lines the gallbladder mucosa, traps cholesterol, and provides an environment for crystal fusion and subsequent gallstone growth (36). During this process, mucin is incorporated within the gallstone matrix (85). The viscous mucus gel prevents cholesterol crystals and small gallstones from being expelled from the gallbladder during contractions. Studies in persons at high risk for gallstone formation also suggest that mucin may play an important role in gallstone formation. Gallbladder mucin increases in patients on rapidweightloss diets (32,94) and in patients who develop gallstones following gastric bypass surgery (63). One study has also suggested that both arachidonic acid and prostaglandin E2 increase in bile prior to the increase in mucin concentration (94). However, this observation has not been confirmed by other studies (64). The effect of other wellknown risk factors of gallstone formation on gallbladder mucin secretion remain unexplored. D— Gallbladder Motility Numerous studies have demonstrated that gallbladder motility is frequently abnormal in patients with gallstones and in persons at increased risk for gallstone formation (95). Gallbladder contractility is reduced in patients with gallstones following a fatty meal (96,97) and in some patients following direct stimulation with intravenous cholecystokinin (98,99). Patients with gallstones have an increase in both gallbladder resting and residual volumes (95,100), which leads to stasis of bile within the gallbladder. These abnormalities in gallbladder volume and contractility appear to be intrinsic defects. Placement of synthetic stones within the gallbladder does not reduce motility (101), and gallbladder contractility does not appear to improve following complete gallstone dissolution (96,98,102). Animal studies have demonstrated that a reduction in gallbladder motility precedes the development of cholesterol gallstone formation (103–105). Supersaturation of bile with cholesterol may be the primary event leading to a reduction in gallbladder motility. Cholesterol is absorbed from supersaturated bile across gallbladder mucosa (106,107). In the submucosa, cholesterol appears to alter the fluidity of smooth muscle cell membranes and to reduce the contractility of these cells (108). Gallbladder smooth muscle contractility is reduced both in patients with gallstones and in patients with cholesterolosis and no gallstones. The contractility of gallbladder smooth muscle cells derived from patients without gallstones is also reduced following incubation with supersaturated model bile solutions (109). A decline in gallbladder contractility has also been demonstrated to be present in several populations at high risk to develop gallstones (95). The most common of these populations includes those on a very low calorie weightreduction diet and patients following gastric surgery. The increased risk for gallstone formation during the third trimester of pregnancy may also be secondary to a reduction in gallbladder contractility that has been documented to develop during this time. III— Pigment Gallstones Pigment gallstones are found in nearly onethird of patients who undergo cholecystectomy (2,110). They are composed of calcium salts of bilirubin, phosphates and fatty acids, mucin glycoprotein, and small amounts of cholesterol (111). Several studies have demonstrated that the incidence of black pigment gallstones increases with advancing age. Black pigment gallstones also develop in patients with cirrhosis or chronic hemolytic disorders and those on longterm parenteral alimentation (110–116).
Page 365
No current treatments are available to either dissolve pigment gallstones once they have formed or to prevent these stones from developing. Prevention of pigment gallstones in these highrisk populations is not considered in this chapter. IV— Gallbladder Sludge Gallbladder sludge is an echogenic substance that layers within the dependent portion of the gallbladder and does not cast an acoustic shadow (117,118). It is composed of gallbladder mucin, calcium, bilirubin, and cholesterol crystals (117,119). Gallbladder sludge may appear, dissolve spontaneously, and then reappear; this is primarily a function of gallbladder motility. Factors that promote stasis of gallbladder bile are all associated with the appearance of biliary sludge. This includes the third trimester of pregnancy, parenteral alimentation, prolonged fasting, a reduction in the fat content of the diet, and gastric bypass surgery (32,68,114,115,121–123). Although sludge may resolve spontaneously in many cases, many other patients with sludge will eventually develop gallstones. Indeed, prospective studies have clearly demonstrated that gallbladder sludge is an essential event in the development of gallstones (114,118,124,125). As such, any treatment that either prevents gallbladder sludge from forming or removes sludge from the gallbladder may prevent the development of gallstones. V— HighRisk Populations Major risk factors associated with the development of cholesterol gallstones and gallbladder sludge are listed in Table 1. Each of these factors alters biliary cholesterol and bile salt secretion in a manner that reduces cholesterol solubility within bile and/or reduces gallbladder motility. Each of these highrisk populations is discussed below. Emphasis is placed on how these factors might be altered to prevent gallstone formation. Table 1 Populations at High Risk for Gallstone Formation in Whom Prevention Has Been Proven Effective or May Be Effective Cholesterol gallstones Obesity Rapid weight lossa Acute spinal cord injury Pregnancy and the immediate postpartum state Longterm somatostatin (octreotide) therapy Increasing age Diabetes mellitus Intestinal disease Biliary sludge Total parenteral nutritionb UDCA proven to reduce incidence of gallstones. CCK proven to reduce incidence of biliary sludge.
a
b
Page 366
A— Obesity The incidence of gallstones increases stepwise with increased body weight (58,61,62). As illustrated in Fig. 1, patients whose body mass index is as little as 1.5 above ideal have a threeto fivefold increased risk for developing gallstones. The risk for developing gallstones increases to a maximum of eightfold in patients who are morbidly obese and weigh in excess of 100 lb over ideal. Since dieting is common among obese patients, some authors have suggested that simply being overweight may not be as strong a risk factor for gallstones as originally thought. However, this hypothesis was shown to be incorrect. When controlled for the effects of dieting, obesity was still found to be a strong risk factor for developing gallstones (126). The risk for developing symptomatic gallstones also appears to increase with increasing body weight (127). Approximately 30% of patients with morbid obesity have either undergone prior cholecystectomy or have been found to have gallstones at the time of bariatric surgery (2,59,60,68,128–130). Obesity increases the risk for gallstone formation by increasing cholesterol secretion into bile (54–57). Obesity may also enhance nucleation of cholesterol from supersaturated bile (131). Several studies have evaluated the effects of obesity on gallbladder motility. Gallbladder resting volume appears to increase with body size, but this appears to be true for large nonobese persons as well (132). Gallbladder contraction either in response to a fatty meal or to cholecystokinin (CCK) appears to be unaffected by body weight (132,133). It is therefore unlikely that obesity in and of itself alters gallbladder motility in a manner that could increase the risk of gallstone formation. B— Rapid Weight Loss Rapid weight loss is one of the most studied risk factors associated with gallstone formation. Several prospective studies have demonstrated that approximately 25 to 30% of persons develop gallstones while participating in a very low calorie weightloss program (32,35,65,66) or following gastric bypass surgery (33,34,67,68). Two approaches have been taken to obtain gallbladder bile during and following weight reduction and to study the mechanisms by which rapid weight reduction may contribute to gallstone formation. In one set of studies, bile was obtained by aspirating duodenal contents following CCK administration at periodic intervals while patients participated in a very low
Figure 1 Relationship between body weight and the risk of developing gallstones. (Redrawn from Ref. 58.)
Page 367
calorie weightreduction diet (94,134). In another set of studies, bile was aspirated directly from the gallbladder at the time of gastric bypass surgery (prior to gallstone formation) and at cholecystectomy after gallstones had formed (63,64). These studies demonstrated that rapid weight loss was associated with an increase in the secretion of cholesterol into bile, a rise in CSI, and a marked increase in the concentration of mucin within gallbladder bile (63,94). No alteration in the bile salt pool (i.e., no increase in the percentage of deoxycholic acid) during weight reduction was observed in either of these studies (64,134). In one of these studies, both arachidonic acid and prostaglandin E2 were noted to increase with weight reduction (94). In the other set of studies, no alteration in the phospholipid content of gallbladder bile was observed following gastric bypass surgery (64). Weight loss in and of itself does not appear to alter gallbladder motility. However, the mechanism by which weight is lost does appear to be a factor. Both gastric bypass surgery and very low calorie diets are associated with a reduction in gallbladder motility. The absence of fat in most very low calorie diets provides no stimulus for gallbladder contraction and thereby promotes stasis of gallbladder bile (135). Patients who have undergone Billroth II gastrectomy with RouxenY gastroenterotomy have an increase in gallbladder resting volume and a reduction in gallbladder contractility following a fatty meal (136). The decline in gallbladder emptying associated with gastric bypass surgery may result from an alteration in the secretion of several gastrointestinal hormones affecting gallbladder motility (137). However, this decline in gallbladder contractility appears to resolve and gallbladder motility returns back to its presurgical baseline within 6 to 12 months (138). C— Spinal Cord Injury Patients who have sustained spinal cord injury were found to have a nearly threefold increased risk for gallstones compared to a controlled population (139). In another study, 30% of patients with spinal cord injury were found either to have gallstones on screening ultrasound examination or to have undergone prior cholecystectomy (140). It has been proposed that the increased incidence of gallstones in this population results from a reduction in gallbladder contractility following injury to the spinal cord. However, the only study to assess gallbladder motility following spinal cord injury could not confirm that a motility defect existed in these patients (140). Most of these patients had conventional risk factors associated with cholelithiasis. D— Pregnancy and Sex Steroid Hormones Pregnancy has long been thought to be a risk factor for cholesterol gallstone disease. The risk of developing gallstones appears to increase with the number of pregnancies (2,59,141). The risk of gallstone disease is increased in women who become pregnant before the age of 30 years (142). Alternatively, the risk for developing gallstones appears to be reduced if the first pregnancy occurs at an older age (53). Women who take contraceptives steroids (53,95,141) and men treated with estrogens (143) are also at increased risk for gallstone formation. Pregnancy and sex steroid hormones affect both bile lipid composition and gallbladder motility in ways that promote gallstone formation. During the later stages of pregnancy, biliary cholesterol secretion into bile and the cholesterol saturation index of gallbladder bile are increased (51). This effect appears to result from the effects of estrogens. Estrogen therapy in males with prostate cancer was associated with a marked increase in biliary cholesterol secretion and cholesterol saturation index compared to a control group of men who underwent orchidectomy (143). Biliary lipid secretion in women taking oral contraceptives and estrogen replacement therapy is also increased (50,52). Pregnancy is also associated with a marked reduction in gallbladder motility and an increase in resting gallbladder volume (144,145). These effects lead to stasis of bile within the gallbladder, are especially pronounced during the third trimester, and resolve within weeks of
Page 368
delivery (146). In contrast, studies that have examined the effect of oral contraceptive hormones on gallbladder motility have yielding mixed results. Some of these studies have demonstrated that gallbladder contractility is reduced in response to a test meal and that gallbladder residual and fasting volumes are increased (145). Other studies have been unable to confirm these observations (144). The high concentration of progestins appears to be directly responsible for this reduction in gallbladder contractility during the later phase of pregnancy. Progesterone inhibits the contractility of gastrointestinal smooth muscle strips in vitro (147). Additional studies have demonstrated that progestins but not estrogens inhibit the contractility of human gallbladder muscle strips obtained from either men or women (148). Progesterone reduces cholecystokinin and mealstimulated gallbladder contraction in animal studies (149) and treatment of women with progestins reduces cholecystokininmediated gallbladder contraction (150). Despite the overwhelming epidemiological evidence relating pregnancy and gallstone formation, it is actually uncommon for women to develop symptomatic gallstone disease during the pregnancy (151–153). Rather, most gallstones that either develop or are present during pregnancy become symptomatic during the peripartum period. This most likely results from the rapid decline in serum progestins and restoration of gallbladder contractility following delivery. E— Somatostatin The relationship between somatostatin and gallstone formation was first recognized in patients with somatostatinsecreting tumors. It appears that gallstones are identified in approximately 65 to 80% of these patients (154–156). Similarly, the synthetic somatostatin analogue octreotide is also associated with gallstone formation. Numerous studies have demonstrated that gallstones develop in approximately 20% of patients treated with longterm administration of octreotide for acromegaly, carcinoid tumors, secretory diarrhea, and other conditions (154,157–161). The risk for gallstone formation increases with the duration of octreotide therapy but plateaus after 6 to 12 months of treatment (161). Octreotide leads to the development of gallstones by inhibiting the postprandial release of cholecystokinin and other gastrointestinal hormones that stimulate gallbladder contraction (154,161–163). This, in turns, leads to an increase in the resting volume of the gallbladder and stasis of gallbladder bile. Administration of octreotide does not appear to alter the relative proportion of bile salts, phospholipids, or cholesterol in bile or the CSI (164). However, both cholesterol crystals and gallstones have been shown to develop in patients who had supersaturated bile prior to the initiation of octreotide (164,165). F— Increasing Age The incidence of gallstones increases with age. The reasons for this have not been clearly defined. In one study, the ratio of biliary cholesterol to bile salt secretion was noted to increase with age (166). In contrast, both gallbladder contractility and CCK release in response to a fatty meal appear unaffected by advancing age (167). G— Diabetes Mellitus Many patients with obesity also have type II diabetes. However, most studies have demonstrated that it is obesity, not diabetes, that increases the risk for gallstone disease in this population (59,60,141,168). Despite this, patients with diabetes have been shown to have a reduction in gallbladder contractility, and this appears to be pronounced in patients with autonomic neuropathy (169). Diabetes may therefore contribute to the increased incidence of gallstones in some obese patients.
Page 369
H— Small Bowel Disease Bile acids are reabsorbed in the terminal ileum and return to liver via portal blood to complete the enterohepatic circulation. This process is necessary to maintain a steadystate pool of bile acids. Disruption of the enterohepatic circulation leads to a marked reduction in the size of the bile acid pool. In some patients with Crohn's disease of the terminal ileum and patients who have had surgical resection of the terminal ileum, the enterohepatic circulation is disrupted (170,171). This increases the relative proportion of cholesterol to bile acids secreted into bile and leads to supersaturation of bile with cholesterol. These observations explain the increased incidence of gallstones in these populations. VI— Prevention of Gallstones Gallstone disease is the most costly of all digestive disorders. Given the staggering health care costs associated with treating gallstones—over $2 billion annually (31)—preventing gallstones from forming is of paramount importance. At this time, studies to prevent gallstone formation have been performed only in those patients undergoing rapid weight reduction. However, it is quite possible that lifestyle factors could be altered and/or pharmacological agents could be utilized to reduce the risk of gallstone formation in many other highrisk populations (Table 2). A— Participation in a Regular Exercise Program Several epidemiological studies have examined the relationship between symptomatic gallstones disease and exercise. Although some studies did not find a relationship between exercise and symptomatic gallstones (2,60,62), others have demonstrated that such a relationship may indeed exist (172–174). The most recent and extensive of these studies evaluated over 45,000 men between the ages of 45 and 75 years (174). In men younger than 65 years of age, 2 to 3 h of moderate physical activity per week seemed to reduce the risk of gallstone disease by 20 to 40%. This degree of activity was achieved by either running, jogging, brisk walking, or participation in racquet sports. The protective effective of moderate exercise was still evident even when adjusted for body weight and body mass index. In contrast, sedentary behavior, defined as watching more than 40 h of television per week and failure to participate in regular exercise, strongly correlated with the development of symptomatic gallstones. Table 2 Lifestyle Factors or Pharmacological Agents That May Reduce the Risk of Cholesterol Gallstone Formation Intervention
Proposed mechanism of action
Exercise
Increases gallbladder motility
Slowing of the rate of weight lossa
Reduces biliary cholesterol secretion
Increasing fat within the low calorie diet
Increases gallbladder motility
Increasing dietary vitamin C
Reduces biliary cholesterol secretion
Ursodeoxycholic acid
a
Reduces biliary cholesterol secretion Increases cholesterol solubility
b
NSAIDs
Reduce biliary mucin secretion
Inhibitors of hepatic cholesterol synthesis
Reduce cholesterol secretion into bile
Cholecystokinin
Increases gallbladder motility
Proven to be effective in large clinical trials. b Nonsteroidal antiinflammatory drugs. a
Page 370
The mechanism(s) by which regular exercise may contribute to this decreased risk of gallstone disease remains unexplored. Exercise affects several metabolic pathways that could reduce the risk associated with gallstone formation. This includes an increase in serum highdensitylipoprotein (HDL) cholesterol (175) and reduction in serum triglycerides (176). HDL cholesterol is preferentially utilized for bile salt synthesis (177), and patients with elevated serum HDL secrete bile with a lower CSI (178) and have a reduced risk for gallstone disease (179). Triglycerides have been shown to be a stimulus for mucin secretion (180). Regular exercise is also associated with an increased secretion of cholecystokinin, which may increase gallbladder motility (181). B— Lowering of the Rate of Weight Reduction Rate of weight loss appears to be an important factor leading to gallstone formation. In the largest epidemiological study to evaluate this relationship, nearly 90,000 women were evaluated over a 2year period (104). Compared to women who lost less than 4 kg, those who lost 4 to 10 kg had a 44% increase in the risk for symptomatic gallstone disease. Women who lost more than 10 kg over the 2year observation period had a 94% increase in the risk of developing symptomatic gallstones. Several prospective studies have examined the relationship between weight loss and gallstone formation in the placebo arm of randomized controlled trials designed to prevent gallstone formation during rapid weight loss. In the study by Yang et al. (66), patients who lost more than 25% of their baseline body weight during a 16week very low calorie rapidweightloss diet had an incidence of new gallstone formation more than twice that observed for patients with a lower rate of weight loss. In two other studies (34,35), the rate of new gallstone formation increased stepwise with the rate of weight loss. Gallstones developed in only 8% of patients who lost less than 10 lb per month. This increased to 31% in patients who lost 10 to 15 lb per month and 49% in patients who lost 15 to 20 lb per month (Fig. 2). The relationship between rate of weight loss and gallstone formation appeared to be stronger in women than men. These data suggests that the risk of gallstone formation during weight reduction could be reduced to less than 10% if the rate of weight loss is limited to only 1 to 2 lb per week.
Figure 2 Relationship between the rate of weight loss and the incidence of new gallstone formation. Data was compiled from the placebo arms of two randomized, doubleblinded, placebocontrolled clinical trials designed to prevent gallstone formation during rapid weight reduction. (From Refs. 34 and 35.)
Page 371
C— Changes in the Composition of LowCalorie Diets Gallstones develop in approximately 25 to 30% of patients who consume very low calorie diets that supply only 420 calories per day and 0 to 2 g of fat (32,35,65). It is believed that the low fat content of these diets provides an insufficient stimulus for gallbladder contraction (135). This, coupled with the enhanced biliary cholesterol secretion that occurs during weight loss (56,57) leads to the increased incidence of gallstones observed in these patients. An uncontrolled trial has suggested that diets that supply a slightly higher caloric content, 800 kcal/day, and 15 to 25 g of fat could reduce the incidence of new gallstone formation during dieting (182). Only 4% (2 of 53) of patients consuming this diet over 10 weeks developed gallstones. Interestingly, the mean rate of weight loss in the two patients who developed gallstones in this study (2.5 kg/week) was far greater than that observed in the remaining individuals (only 0.9 kg/week) who did not develop gallstones. Thus, rate of weight loss, as discussed above, may be a more important factor in gallstone formation than the specific diet and/or its fat content. D— Vitamin C Several small casecontrolled studies have suggested that increased dietary consumption of ascorbic acid is associated with a reduction in gallstones (183–185). This benefit appears to be isolated to women, where a 50 to 65% reduction in gallstone disease and cholecystectomy were observed. In contrast, vitamin C did not appear to reduce gallstone disease in men. In the second National Health and Nutrition Examination Survey (NHANES II) the incidence of gallstone disease was also found to be inversely related to serum ascorbic acid concentration in women but not in men (185). Previous studies have demonstrated that animals fed an ascorbic acid—deficient diet develop gallstones (186). However, the manner in which vitamin C reduces the incidence of gallstones remains unclear. Animal studies have demonstrated that ascorbic acid enhances hepatic metabolism of cholesterol to bile acids (187). This process could lead to an increase in bile salt secretion relative to cholesterol secretion and thereby improve the solubility of biliary cholesterol. Why this only occurs in women is speculative. It is also unknown what effects if any ascorbic acid may have upon cholesterol nucleation from supersaturated bile, secretion of gallbladder mucin, and gallbladder motility. E— Ursodeoxycholic Acid Ursodeoxycholic acid (UDCA) is the major bile acid in the polar bear and has long been employed in Asian folk medicine as a treatment for liver disorders. UDCA is the 7b epimer of chenodeoxycholic acid (CDCA). It is relatively hydrophilic and a poor detergent (188). UDCA affects biliary cholesterol synthesis, secretion, solubility, and lithogenicity in a manner that reduces the risk for gallstone formation. UDCA appears to reduce hepatic cholesterol synthesis by inhibiting b hydroxy b methylglutarylcoenzyme A (HMGCoA) reductase (189,190), although this is not a universal finding (191,192). UDCA has also been shown to directly reduce canalicular cholesterol secretion (193) and intestinal cholesterol absorption (194). These effects lead to a reduction in hepatic cholesterol secretion into bile (195). In contrast, UDCA has little effect on the synthesis and secretion of bile salts (194,196). As a result, treatment with UDCA leads to a marked reduction in CSI. UDCA also increases the cholesterol content of both mixed micelles and vesicles (197) and directly inhibits nucleation of cholesterol from supersaturated bile (198). UDCA may also effect gallbladder motility in a manner that reduces the risk for gallstone formation. Some studies have demonstrated that treatment with UDCA increases fasting gallbladder volume in patients with gallstones as well as in control patients without gallstones (199). In addition, both meal and CCKstimulated gallbladder contraction appear to be increased in patients treated with UDCA. Other studies have been unable to confirm these ob
Page 372
servations (200,201). The varying results observed in these studies may reflect differences in the manner in which gallbladder volume and contraction have been assessed and the specific stimulus utilized to stimulate gallbladder contraction. UDCA was originally developed as a nonsurgical treatment for symptomatic cholesterol gallstones. Approximately, 30% of patients treated with this bile acid achieved complete gallstone dissolution within 12 months (190,202,203). UDCA treatment has also been reported to reduce symptoms associated with cholelithiasis (203,204). Unfortunately, gallstones appear to reform in nearly 50% of patients within 5 years following successful stone dissolution and discontinuation of treatment (205). UDCA is well tolerated. The most common side effect is diarrhea, and this develops in only 2% of treated patients (190,202,203). During treatment, the percentage of UDCA in bile increases from a baseline value of 2 to 5% to nearly 50 to 60% of the total bile salt pool (188,206). Several studies have now demonstrated that UDCA is very effective for prevention of cholesterol gallstone formation in patients undergoing rapid weight reduction (32–35). In the largest of these studies, over 1000 patients participating in a 16week very low calorie rapidweightloss program were randomly assigned to receive one of three doses of UDCA or a placebo. UDCA reduced gallstone formation from nearly 28% in placebotreated patients to 8% in patients treated with 300 mg/day and to only 3% in patients treated with 600 mg of UDCA daily (34). In another large study, 310 patients who underwent proximal gastric bypass surgery as a treatment for morbid obesity were also randomized to receive either UDCA or placebo for 16 weeks. As in the dietary weightloss study, UDCA significantly reduced the incidence of new gallstone formation following gastric bypass surgery from 32% in placebo treated patients to only 2% in patients treated with 600 mg of UDCA daily (35). Based upon the results of these two large studies, the Food and Drug Administration approved UDCA for prevention of gallstone formation in persons undergoing rapid weight reduction via either dietary or surgical means. UDCA remains the only treatment proven to be effective for prevention of gallstone formation in highrisk populations (Fig. 3). Although no formal safety and efficacy studies of UDCA treatment have been performed during pregnancy, at least one report has demonstrated that this agent may be utilized during pregnancy without adverse effects to the developing infant (207). While this observation in no way supports the routine use of UDCA for prevention of gallstone formation during the third trimester of pregnancy, it does suggest that UDCA might be safely utilized in this population.
Figure 3 Prevention of gallstone formation with ursodeoxycholic acid in patients undergoing rapid weight loss either while participating in a very low calorie diet program or following gastric bypass surgery. (Redrawn from Refs. 34 and 35.)
Page 373
F— Aspirin and Other NSAIDs Both aspirin and NSAIDs have been evaluated as potential agents to prevent gallstone formation. As noted above, gallbladder mucin plays an essential role in gallstone pathogenesis. Animal studies have demonstrated that hypersecretion of gallbladder mucin occurs prior to gallstone formation (80–82), and human studies have demonstrated that the mucin content of gallbladder bile increases in patients who develop gallstones during rapid weight loss (32,63,64,94). Mucin secretion is stimulated by prostaglandins, which, in turn, are synthesized from arachidonic acid (86–89). Aspirin and NSAIDs inhibit prostaglandin synthesis (86–88), and these agents have been shown to both inhibit gallbladder mucin secretion and gallstone formation in animal models of gallstone disease (83,88). NSAIDs have been shown to improve gallbladder motility in an animal model of gallstone disease (208), although this has not been confirmed by subsequent studies (209,210). The effect of aspirin on bile composition has been evaluated in patients participating in a very low calorie rapidweightloss diet (32). In this randomized, doubleblind, placebocontrolled trial, an increase in biliary prostaglandins, mucin, and cholesterol saturation was observed in placebotreated patients. In contrast, treatment with 600 mg/day of aspirin prevented these changes. In another study, gallbladder bile was sampled from patients with and without gallstones at the time of bariatric surgery (211). Those patients who utilized NSAIDs on a regular basis had less gallbladder mucin and a lower cholesterol/phospholipid ratio than patients who did not use NSAIDs. NSAIDs have also been shown to have a positive effect on gallbladder motility. Use of indomethacin in patients with symptomatic gallstones was associated with an increase in postprandial gallbladder emptying and a reduction in residual volume (212). Each of these observations lends support to the hypothesis that NSAIDs could be utilized for the prevention of gallstones. Several epidemiological studies have also examined the effect of NSAIDs on gallstone disease. The most promising of these studies evaluated gallstone recurrence following oral bile salt dissolution therapy (213,214). Gallstones did not develop in those patients who utilized NSAIDs on a regular basis, whereas 32% of non NSAID users developed gallstone recurrence within 5 years followup. In contrast, two other studies could not demonstrate that regular NSAID use reduced the incidence of either gallstones or symptomatic gallstone disease. The prevalence of gallstones was found to be nearly identical in patients who were prescribed NSAIDs for various rheumatological conditions compared to a population matched for age, gender, and body weight who did not take NSAIDs (215). In the Myocardial Infarction Study, regular aspirin use did not reduce the hospitalization rate for symptomatic gallstone disease (216). Finally, the incidence of new gallstone formation observed in a randomized, doubleblind, placebocontrolled trial was not significantly reduced by treating patients with 600 mg of aspirin daily as compared with placebo while they were participating in a very low calorie rapidweightloss diet (32). In summary, although NSAID use appears to affect bile composition in a manner that could reduce gallstone formation, epidemiological studies have been unable to provide definitive evidence that aspirin or other NSAIDs could be utilized as an effective prophylactic agent in patients at high risk for gallstone formation (217). G— Inhibitors of Cholesterol Synthesis Cholesterol biosynthesis occurs within hepatic microsomes and is controlled by the ratelimiting enzyme HMGCoA reductase (218). This enzyme is blocked by the family of drugs known as HMGCoA reductase inhibitors or ''statins." These agents are potent inhibitors of cholesterol biosynthesis and widely utilized for the treatment of hypercholesterolemia. Several studies have demonstrated that HMGCoA reductase inhibitors reduce biliary cholesterol secretion and the percentage of cholesterol in bile without affecting bile salt synthesis (219–222). This reduces the biliary CSI. Combining HMGCoA reductase inhibitors
Page 374
with UDCA leads to an even greater decline in cholesterol saturation than is observed with either of these agents alone (221,222). At least one report has documented dissolution of cholesterol gallstones in a patient treated with an HMGCoA reductase inhibitor (223). These observations suggest that these agents may be effective for the dissolution and/or prevention of gallstones. However, these observations were made in patients taking HMGCoA reductase inhibitors for the treatment of hypercholesterolemia. One study has suggested that HMGCoA reductase inhibitors do not have beneficial effects on biliary cholesterol secretion and solubility in patients with normal plasma lipids (224). Although not yet proven to be effective, HMGCoA reductase inhibitors remain a potential therapy for gallstone prophylaxis, at least in patients with hypercholesterolemia. H— Cholecystokinin A reduction in gallbladder motility is believed to play an important role in the formation of both biliary sludge and gallstones (95–99). Several studies have documented that both mealstimulated and CCKinduced gallbladder contractility is reduced in gallstone patients (96–99). A reduction in gallbladder contractility is also observed during pregnancy, in response to oral contraceptive steroids, and following gastric bypass surgery (138,144–150). The lack of dietary fat in many very low calorie diets and in patients on longterm parenteral nutrition does not provide a stimulus for gallbladder contraction (135). The increased risk of gallstones and biliary sludge observed in these patients may result, at least in part, from a reduction in gallbladder motility. One possible treatment for such patients is to contract the gallbladder via pharmacological stimulation at periodic intervals. Indeed, in a randomized, blinded, placebo controlled trial, daily administration of an intravenous CCK bolus to patients receiving longterm parenteral nutrition led to a significant reduction in gallbladder resting volume (225) and reduced the incidence of gallbladder sludge from 60% to less than 5% (Fig. 4). Although not specifically studied, it is possible that periodic treatment with CCK to induce gallbladder contraction could reduce the risk of gallstone formation in other populations in whom a reduction in gallbladder motility appears to be the primary defect. This includes patients with acute spinal cord injury,
Figure 4 Effect of cholecystokinin on the incidence of gallbladder sludge in patients who were unable to eat for a prolonged period and received total parenteral nutrition. (Redrawn from Ref. 225.)
Page 375
patients on lowfat weightloss diets, and those who have undergone gastric bypass surgery or gastrectomy. Pharmacological agents that stimulate gastrointestinal motility might also enhance gallbladder contractility; if so, they could be utilized to reduce the risk of gallstone formation in certain highrisk populations. Cisapride increases bile salt recycling, probably by reducing intestinal transit time; in some studies, this was associated with a reduction in CSI (226,227). However, cisapride appears to offer no beneficial effect on gallbladder contractility (227,228). The effect of erythromycin on gallbladder contractility and CSI has been evaluated only in an animal model of gallstone formation. In this ground squirrel model of cholesterol gallstone disease, treatment with erythromycin increased bile salt recycling, reduced the bile salt pool, reduced CSI, and improved gallbladder motility (229). It remains to be seen if erythromycin could provide similar benefits in humans and develop into an effective agent for gallstone prevention. VII— Summary Gallstone disease is one of the most common and costly of all digestive disorders. Preventing gallstones and biliary sludge from forming has the potential for reducing morbidity and health care costs associated with gallbladder disease. This chapter has reviewed data clearly demonstrating that prevention of gallstones and biliary sludge is indeed possible in certain highrisk populations. Daily administration of intravenous CCK is highly effective for prevention of biliary sludge in patients receiving parenteral nutrition. The applicability of this treatment to other highrisk populations has yet to be explored. The most effective agent for prophylaxis against cholesterol gallstones appears to be UDCA. This has been demonstrated to be highly effective in patients undergoing rapid weight loss via either dietary means or following gastric bypass surgery. It is quite possible that UDCA could be utilized to prevent cholesterol gallstone formation in many other highrisk populations as well. References 1. Diehl AK. Epidemiology and natural history of gallstone disease. Gastroenterol Clin North Am 1991; 20:1–19. 2. Friedman GD, Kannel WB, Dawber TR. The epidemiology of gallbladder disease: observations in the Framingham study. J Chronic Dis 1966; 19:273–292. 3. Gracie WA, Ransohoff DF. The natural history of silent gallstones: the innocent gallstone is not a myth. N Eng J Med 1982; 307:798–800. 4. McSherry CK, Ferstenberg H, Calhoun WF, Lahman E, Virshup M. The natural history of diagnosed gallstone disease in symptomatic and asymptomatic patients. Ann Surg 1987; 202:59–63. 5. Friedman GD, Raviola CA, Fireman B. Prognosis of gallstones with mild or no symptoms: 25 years of followup in a health maintenance organization. J Clin Epidemiol 1989; 42:127–136. 6. Wenckert A, Robertson B. The natural course of gallstone disease: eleven year review of 781 nonoperated cases. Gastroenterology 1966; 50:376–381. 7. Newman HF, Northup JD, Rosenblum M, Abrams H. Complications of cholelithiasis. Am J Gastroenterol 1968; 50:476–496. 8. Wetter LA, Way LW. Surgical therapy for gallstone disease. Gastroenterol Clin North Am 1991; 20:157–169. 9. Lee SP, Kuver R. Gallstones. In: T Yamada, DH Alpers, C Owyang, DW Powell, FE Silverstein, eds. Textbook of Gastroenterology. Philadelphia: Lippincott, 1995, pp 2187–2212.
Page 376
10. McSherry CK, Glen F. The incidence and causes of death following surgery for nonmalignant biliary tract disease. Ann Surg 1980; 191:271–275. 11. Kelley JE, Burrus RG, Graham LD, Chandler KE. Safety, efficacy, cost and morbidity of laparoscopic versus open cholecystectomy: a prospective analysis of 228 consecutive patients. Am Surg 1993; 59:23–27. 12. Stoker ME, Vose J, O'Mara P, Maini BS. Laparoscopic cholecystectomy: a clinical and financial analysis of 280 operations. Arch Surg 1992; 127:589–594. 13. Glinatsis MT, Griffith JP, McMahon MJ. Open vs laparoscopic cholecystectomy: a retrospective comparative study. J Laparoendosc Surg 1992; 2:81–86. 14. Gagic N, Frey CF. The results of cholecystectomy for the treatment of acute cholecystitis. Surg Gynecol Obstet 1975; 140:255–257. 15. Almond HR, Vlahcevic ZR, Bell CC Jr, Gregory DH, Swell L. Bile acid pools, kinetics and biliary lipid composition before and after cholecystectomy. N Engl J Med 1973; 289:1213–1216. 16. Lee SS, Cha S, Lee RL. The relationship between cholecystectomy and colon cancer: an Iowa study. J Surg Oncol 1989; 41:81–85. 17. Werner B, De Heer K, Mitschke H. Cholecystectomy and carcinoma of the colon: An experimental study. Z Krebforsch 1977; 88:223–230. 18. Capron JP, Delamarre J, Canarelli JP, Brousse N, Dupas JL. Does cholecystectomy predispose to the development of colorectal cancer? Gastroenterol Clin Biol 1978; 2:383–389. 19. Turunen MJ, Kivilaakso EO. Increased risk of colorectal cancer after cholecystectomy. Ann Surg 1981; 194:639–641. 20. Linos DA, Beard CM, O'Fallon WH, Dockerty MB, Beart RW Jr, Kurland LT. Cholecystectomy and carcinoma of the colon. Lancet 1981; 2:379–381. 21. Vernick LJ, Kuller LH. Cholecystectomy and rightsided colon cancer: an epidemiological study. Lancet 1981; 2:381–383. 22. Alley PG, Lee SP. The increased risk of proximal colon cancer after cholecystectomy. Dis Colon Rectum 1983; 26:522–524. 23. Turnbull PRG, Smith AH, Isbister WH. Cholecystectomy and cancer after cholecystectomy. Dis Colon Rectum 1983; 26:522–524. 24. Weiss NS, Daling JR, Chow WH. Cholecystectomy and the incidence of large bowel cancer. Cancer 1982; 49:1713–1715. 25. Kwai AH. Cholecystectomy and large bowel cancer. Mt Sinai J Med 1983; 50:359–363. 26. Narisawa T, Sano M, Sato M, Takahashi T, Arakawa H. Relationship between cholecystectomy and colon cancer in lowrisk Japanese population. Dis Colon Rectum 1983; 26:512–515. 27. Lowenfels AB, Domellof L, Lindstrom CG, Bergman F, Monk MA, Sternby NH. Cholelithiasis, cholecystectomy and cancer: a case control study in Sweden. Gastroenterology 1982; 83:672–676. 28. Digestive disease statistics. NIH publication no. 95–3873. Bethesda, MD: National Institute of Diabetes and Digestive and Kidney Diseases, US Dept of Health and Human Services, 1995. 29. The Southern surgeons club: a prospective analysis of 1518 laproscopic cholecystectomies. N Engl J Med 1991; 324:1073–1078. 30. Legorreta AP, Silber JH, Costantino GN, Kobylinski RW, Zatz SL. Increased cholecystectomy rate after the introduction of laparoscopic cholecystectomy. JAMA 1993; 270:1429–1432. 31. National Center for Health Statistics. National hospital discharge survey. Advance data from vital and health statistics of the national center for health statistics. No 278. Rockville, MD: US Dept of Health, Education and Welfare, Public Health Service, Health Resources Administration, 1994.
Page 377
32. Broomfied PH, Chopra R, Sheinbaum RC, Bonnorris GG, Silverman A, Schoenfield LJ, Marks JW. Effects of ursodeoxycholic acid and aspirin on the formation of lithogenic bile and gallstones during loss of weight. N Engl J Med 1988; 319:1567–1572. 33. Worobetz LJ, Inglis FG, Shaffer EA. The effect of ursodeoxycholic acid therapy on gallstone formation in the morbidly obese during rapid weight loss. Am J Gastroenterol 1993; 88:1705–1710. 34. Sugerman HJ, Brewer WH, Shiffman ML, Brolin RE, Fobi MAL, Linner JH, MacDonald KG, MacGregor AM, Martin LF, OramSmith JC, Popoola D, Schirmer BD, Vickers FF. Prophylactic ursodeoxycholic acid prevents gallstone formation following gastric bypass surgery induced rapid weight loss: a multicenter, placebo controlled, randomized, doubleblind prospective trail. Am J Surg 1995; 169:91–97. 35. Shiffman ML, Kaplan GD, BrinkmanKaplan V, Vickers FF. Prevention of gallstone formation with ursodeoxycholic acid in patients participating in a very low calorie diet program. Ann Intern Med 1995; 122:899–905. 36. Heuman DM, Shiffman ML. Prevention and medical treatment of gallstones. In: Friedman G et al., eds. Gastrointestinal Pharmacology and Therapeutics. Philadelphia: LippincottRaven, 1997. 37. Donovan JM, Carey MC. Physicalchemical basis of gallstone formation. Gastroenterol Clin North Am 1991; 20:47–66. 38. Heuman DM, Moore EW, Vlahcevic ZR. Pathogenesis and dissolution of gallstones. In: Zakim D, Boyer JD, eds. Hepatology: A Textbook of Liver Disease. Philadelphia: Saunders, 1996, pp 1833–1876. 39. Carey MC. Pathogenesis of gallstones. Am J Surg 1993; 165:410–419. 40. Renshaw PE, Janoff AS, Miller KW. On the nature of dilute aqueous cholesterol suspensions. J Lipid Res 1987; 28:673–683. 41. Carey MC, LaMont JT. Cholesterol gallstone formation: 1. Physical chemistry of bile and biliary lipid secretion. Prog Liver Dis 1993; 10:139–163. 42. Admirand WH, Small DM. The physical chemical basis of cholesterol solubility in bile: relationship to gallstone formation and dissolution in man. J Clin Invest 1968; 47:1043–1052. 43. Carey MC, Small DM. The physical chemistry of cholesterol solubility in bile: relationship to gallstone formation and dissolution in man. J Clin Invest 1978; 61:998–1026. 44. Mackay C, Crook JN, Smith DC, McAllister RA. The composition of hepatic and gallbladder bile in patients with gallstones. Gut 1972; 13:759–762. 45. Bennion L, Grundy SM. Risk factors for the development of cholelithiasis in man. N Engl J Med 1978; 299:1161–1167. 46. Grundy SM, Metzger AL, Adler RD. Mechanism of lithogenic bile formation in American Indian women with cholesterol gallstones. J Clin Invest 1972; 51:3026– 3042. 47. Grundy SM, Ahrens EHJ, Salen G, Schreibman PH, Nestel PJ. Mechanisms of action of clofibrate on cholesterol metabolism in patients with hyperlipidemia. J Lipid Res 1972; 13:531–551. 48. Vlahcevic ZR, Bell CCJ, Buhac I, Farrar JT, Swell L. Diminished bile acid pool size in patients with gallstones. Gastroenterology 1970; 59:165–173. 49. Reuben A, Maton PN, Murphy GM, Dowling RH. Bile lipid secretion in obese and nonobese individuals with and without gallstones. Clin Sci 1985; 69:71–79. 50. Kern FJ, Everson GT. Contraceptive steroids increase cholesterol secretion in bile: mechanism of action. J Lipid Res 1987; 28:828–839. 51. Kern FJ, Everson GT, DeMark B, McKinley C, Showalter R, Erfling W, Braverman DZ, SzczepanikVan Leeuwen P, Klein PD. Biliary lipids, bile acids and gallbladder function in the human female: effects of pregnancy and the ovulatory cycle. J Clin Invest 1981; 68:1229–1242. 52. Kern FJ, Everson GT, DeMark B, McKinley C, Showalter R, Erfling W, Braverman DZ, SzczepanikVan Leeuwen P, Klein PD. Biliary lipids, bile acids and gallbladder function
Page 378
in the human female: effects of contraceptive steroids. J Lab Clin Med 1982; 99:798–805. 53. Scragg RKR, McMichael AJ, Seamark RF. Oral contraceptives, pregnancy and endogenous oestrogen in gallstone disease: a case controlled study. BMJ 1984; 2:1795. 54. Mabee TM, Meyer P, DenBesten L, Mason EE. The mechanism of increased gallstone formation in obese human subjects. Surgery 1976; 79:460–468. 55. Madura JA, Loomis RC, Harris RA, Grosfield J, Tompkins RK. Relationship of obesity to bile lithogenicity in man. Ann Surg 1979; 189:106–111. 56. Mok HYI, Von Bergmann K, Crouse JR, Grundy SM. Biliary lipid metabolism in obesity. Effects of bile acid feeding before and during weight reduction. Gastroenterology 1979; 76:556–567. 57. Bennion LJ, Grundy SM. Effects of obesity and caloric intake on biliary lipid metabolism in man. J Clin Invest 1975; 56:996–1011. 58. Shiffman ML, Sugerman HJ, Kellum JH, Brewer WH, Moore EW. Gallstones in patients with morbid obesity: relationship to body weight, weight loss and gallbladder bile cholesterol solubility. Int J Obesity 1993; 17:153–158. 59. Barbara L, Sama C, Labate AMM, Taroni F, Rusticali AG, Festi D, Sapio C, Roda E, Banterle C, Puci A, Formentini F, Colasanti S, Nardin F. A population study on the prevalence of gallstone disease: the Sirmione study. Hepatology 1987; 7:913–917. 60. Capocaccia L, Giunchi G, Pocchiari F and GREPCO. The epidemiology of gallstone disease in Rome, Italy: Part II. Factors associated with the disease. Hepatology 1988; 8:907–913. 61. Maclure KM, Hayes KC, Colditz GA, Stampfer MJ, Speizer FE, Willett WC. Weight, diet and the risk of symptomatic gallstones in middleaged women. N Engl J Med 1989; 321:563–569. 62. Jorgensen T. Gallstones in a Danish population: relation to weight, physical activity, smoking, coffee consumption and diabetes mellitus. Gut 1989; 30:528–534. 63. Shiffman ML, Sugerman HJ, Kellum JM, Moore EW. Changes in gallbladder bile composition following gallstone formation and weight reduction. Gastroenterology 1992; 103:214–221. 64. Shiffman ML, Shamburek RD, Schwartz CC, Sugerman HJ, Kellum JM, Moore EW. Effect of gallstone formation on the phospholipid, bile salt and mucin content of gallbladder bile in persons undergoing rapid weight reduction. Gastroenterology 1993; 105:1200–1208. 65. Liddle RA, Goldstein RB, Saxton J. Gallstone formation during weight reduction dieting. Arch Intern Med 1989; 149:1750–1753. 66. Yang H, Petersen GM, Roth MP, Schoenfield LJ, Marks JW. Risk factors for gallstone formation during rapid loss of weight. Dig Dis Sci 1992; 37:912–918. 67. Wattchow DA, Hall JC, Whiting MJ, Bradley B, Iannos J, Watts JM. Prevalence and treatment of gall stones after gastric bypass surgery for morbid obesity. BMJ 1993; 286:763. 68. Shiffman ML, Sugerman HJ, Kellum JM, Brewer WH, Moore EW. Gallstone formation following rapid weight loss: a prospective study in patients undergoing gastric bypass surgery for treatment of morbid obesity. Am J Gastroenterol 1991; 86:1000–1005. 69. Holan KR, Holzbach RT, Hermann RE, Cooperman AM, Claffey WJ. Nucleation time: a key factor in the pathogenesis of cholesterol gallstone disease. Gastroenterology 1979; 77:611–617. 70. Gollish SH, Burnstein MJ, Ilson RG, Petrunka CN, Strasberg SM. Nucleation of cholesterol monohydrate crystals from hepatic and gallbladder bile of patients with cholesterol gall stones. Gut 1983; 24:836–844. 71. Groen AK, Drapers JAG, Tytgat GNJ. Cholesterol nucleation and gallstone formation. J Hepatol 1988; 6:383–387. 72. Harvey PRC, Strasberg SM. Will the real cholesterol nucleating and antinucleating proteins please stand up. Gastroenterology 1979; 77:611–617.
Page 379
73. Abei M, Schwarzendrube J, Nuutinen H, Broughan TA, Kawczak P, Williams C, Holzbach RT. Cholesterol crystalizationpromoters in human bile: comparative potencies of immunoglobulins, a 1acid glycoprotein, phospholipase C and aminopeptidase N. J Lipid Res 1993; 34:1141–1148. 74. Aravinda Upadhya G, Harvey PRC, Strasberg SM. Effect of human biliary immunoglobulins on the nucleation of cholesterol. J Biol Chem 1993; 268:5193–5200. 75. Afdhal NH, Niu N, Gantz D, Small DM, Smith BF. Bovine gallbladder mucin accelerates cholesterol monohydrate crystal growth in model bile. Gastroenterology 1993; 104:1515–1523. 76. Levy PF, Smith BF, LaMont JT. Human gallbladder mucin accelerates in vitro nucleation of cholesterol in artificial bile. Gastroenterology 1984; 87:270–275. 77. Gallinger S, Taylor RD, Harvey PRC, Petrunka CN, Strasberg SM. Effect of mucous glycoprotein on the nucleation time of human gallbladder bile. Gastroenterology 1985; 89:648–658. 78. Smith BF. Human gallbladder mucin binds biliary lipids and promotes cholesterol crystal nucleation in model biles. J Lipid Res 1987; 28:1088–1097. 79. Sedaghat A, Grundy SM. Cholesterol crystals and the formation of cholesterol gallstones. N Engl J Med 1980; 302:1274–1277. 80. Lee SP, LaMont JT, Carey MC. The role of gallbladder mucus hypersecretion in the evolution of cholesterol gallstones: studies in the prairie dog. J Clin Invest 1981; 67:1712–1723. 81. Pemsingh RS, MacPherson BR, Scott GW. Mucus hypersecretion in the gallbladder epithelium of ground squirrels fed a lithogenic diet for the induction of cholesterol gallstones. Hepatology 1987; 7:1267–1271. 82. Lee SP, Scott AF. The evolution of morphologic changes in the gallbladder before stone formation in mice fed a cholesterolcholic acid diet. Am J Pathol 1982; 108:1–8. 83. Lee SP, Carey MC, LaMont JT. Aspirin prevention of cholesterol gallstone formation in prairie dogs. Science 1985; 98:445–451. 84. Smith BF, LaMont JT. Bovine gallbladder mucin binds bilirubin in vitro. Gastroenterology 1983; 85:707–712. 85. Smith BF, LaMont JT. Identification of gallbladder mucinbilirubin complex in human cholesterol gallstone matrix: effects of reducing agents on in vitro dissolution of matrix and intact gallstones. J Clin Invest 1985; 76:439–445. 86. LaMorte WW, LaMont JT, Hale W, Booker ML, Scott TE, Turner B. Gallbladder prostaglandins and lysophospholipids as mediators of mucin secretion during cholelithiasis. Am J Physiol 1986; 14:G701–G709. 87. LaMore WW, Booker ML, Scott TE, Williams LF. Increases in gallbladder prostaglandin synthesis before the formation of cholesterol gallstones. Surgery 1985; 98:445–451. 88. Lee SP. Lessons from experimental cholelithiasis: gallbladder and mucosa, nonsteroidal inflammatory drugs and gallstones. Gastroenterology 1991; 101:857–860. 89. LaMont JT, Turner BS, DiBenedetto D, Handlin R, Schafer Al. Arachidonic acid stimulates mucin secretion in prairie dog gallbladder. Am J Physiol 1983; 8:G92–G98. 90. Carey MC, Cahalane MJ. Whither biliary sludge? Gastroenterology 1988; 95:508–523. 91. O'Leary DP, Murray FE, Turner BS, LaMont JT. Bile salts stimulate glycoprotein release by guinea pig gallbladder in vitro. Hepatology 1991; 13:957–961. 92. Van Berge Henegouwen GP, van der Werf SDJ, Ruben AT. Fatty acid composition of phospholipids in bile in man: promoting effect of deoxycholate on arachidonate. Clin Chim Acta 1987; 165:27–37. 93. Rege RV, Prystowsky JB. Crystalline cholesterol reduces sodium transport in guinea pig gallbladder. Gastroenterology 1993; 104:1515–1523. 94. Marks JW, Bonnorris GG, Albers G, Schoenfield LJ. The sequence of biliary events preceeding the formation of gallstones in humans. Gastroenterology 1992; 103:566–570.
Page 380
95. Everson GT. Gallbladder function in gallstone disease. Gastroenterol Clin North Am 1991; 20:85–110. 96. Fisher RS, Stelzer F, Rock E, Malmud LS. Abnormal gallbladder emptying in patients with gallstones. Dig Dis Sci 1982; 27:1019–1024. 97. Forgacs IC, Maisey MN, Murphy GM, Dowling RH. Influence of gallstones and ursodeoxycholic acid therapy on gallbladder emptying. Gastroenterology 1984; 87:299–307. 98. Pomeranz IS, Shaffer EA. Abnormal gallbladder emptying in a subgroup of patients with gallstones. Gastroenterology 1985; 88:787–791. 99. Sylwestrowicz TA, Shaffer EA. Gallbladder function during gallstone disolution: effect of bile acid therapy in patients with gallstones. Gastroenterology 1988; 95:740–748. 100. Pauletzki J, Cicala M, Holl J, Sauerbruch T, Schafmayer A, Paumgartner G. Correlation between gall bladder fasting volume and postprandial emptying in patients with gall stones and healthy controls. Gut 1993; 34:1443–1447. 101. Pomeranz IS, Davison JS, Shaffer EA. The effects of prosthetic gallstones on gallbladder function and bile composition. J Surg Res 1986; 41:47–52. 102. Pellegrini C, Ryan T, Broderick W, Way LW. Gallbladder filling and emptying during cholesterol gallstone formation in the prairie dog: a cholescintigraphic study. Gastroenterology 1986; 90:143–149. 103. Fridhandler TM, Davison JS, Shaffer EA. Defective gallbladder contractility in the ground squirrel and prairie dog during the early stages of cholesterol gallstone formation. Gastroenterology 1983; 85:830–836. 104. Doty JE, Pitt HA, Kuchenbecker SL, DenBesten L. Impaired gallbladder emptying before gallstone formation in the prairie dog. Gastroenterology 1983; 85:168–174. 105. Li YF, Moody FG, Weisbrodt NW, Zalewsky CA, Coelho JCU, Senninger N. Decrease in contractility of prairie dog gallbladder muscle strips following cholesterol feeding. Surg Forum 1984; 35:224–226. 106. Shiffman ML, Sugerman HJ, Moore, EW. Human gallbladder mucosal function: effects of concentration and acidification of bile on biliary cholesterol and calcium solubility. Gastroenterology 1990; 99:1452–1459. 107. Neiderhiser DH, Harmon CK, Roth HP. Absorption of cholesterol by the gallbladder. J Lipid Res 1976; 17:117–124. 108. Behar J, Lee KY, Thompson WR, Biancani P. Gallbladder contraction in patients with pigment and cholesterol gallstones. Gastroenterology 1989; 97:1479– 1484. 109. Yu P, Chen Q, Biancani P, Behar J. Membrane cholesterol alters gallbladder muscle contractility in prairie dogs. Am J Physiol 1996; 271:G56–G61. 110. Trotman BW, Soloway RD. Pigment vs cholesterol cholelithiasis: clinical and epidemiological aspects. Am J Dig Dis 1975; 20:735–740. 111. Trotman BW. Pigment gallstone disease. Gastroenterol Clin North Am 1991; 20:111–126. 112. Nicholas P, Rinaudo PA, Conn HO. Increased incidence of cholelithiasis in Laennec's cirrhosis: a postmortem evaluation of pathogenesis. Gastroenterology 1972; 63:112–118. 113. Fornari F, Civardi G, Buscarini E, Cavanna L, Imberti D, Rossi S, Sbolli G, Di Stasi M, Buscarini L. Cirrhosis of the liver: a risk factor for development of cholelithiasis in males. Dig Dis Sci 1990; 35:1403–1408. 114. Messing B, Bories C, Kunstlinger F, Bernier JJ. Does total parenteral nutrition induce gallbladder sludge formation and lithiasis. Gastroenterology 1983; 84:1012–1019. 115. Levinson DE, Fromm H. Medical management of gallbladder disease. In: Zakim D, Boyer JD, eds. Hepatology: A Textbook of Liver Disease. Philadelphia: Saunders, 1996, pp 1877–1897. 116. Roslyn JL, Pitt HA, Mann LL, Ament ME, DenBesten L. Gallbladder disease in patients on long term parenteral nutrition. Gastroenterology 1983; 84:148–154. 117. Soloway RD, Trotman BW, Maddrey WC, Nakayama F. Pigment gallstone composition in patients with hemolysis of infection/stasis. Dig Dis Sci 1986; 31:454–460.
Page 381
118. Lee SP. Pathogenesis of biliary sludge. Hepatology 1990; 12:200S–205S. 119. Lee SP, Maher K, Nicholls JF. Origin and fate of biliary sludge. Gastroenterology 1988; 94:170–178. 120. Lee SP, Nicholls JF. Nature and composition of biliary sludge. Gastroenterology 1986; 90:677–686. 121. Bolondi L, Gaini S, Testa S, Labo G. Gallbladder sludge formation during prolonged fasting after gastrointestinal surgery. Gut 1985; 26:734–738. 122. Cohen S. The sluggish gallbladder of pregnancy. N Engl J Med 1980; 302:397–399. 123. Maringhini A, Ciambra M, Baccelliere P, Raimondo M, Pagliaro L. Sludge, stones and pregnancy. Gastroenterology 1988; 95:1160–1161. 124. Gafa M, Sarli L, Miselli A, Pietra N, Carreras F, Peracchia A. Sludge and microlithiasis of the biliary tract after total gastrectomy and postoperative total parenteral nutrition. Surg Gynecol Obstet 1987; 165:413–418. 125. Janowitz P, Kratzer W, Zemmler T, Tudyka J, Wechsler JG. Gallbladder sludge: spontaneous course and incidence of complications in patients without stones. Hepatology 1994; 20:291–294. 126. Thijs C, Knipschild P, Leffers P. Is gallstone disease caused by obesity or by dieting? Am J Epidemiol 1992; 135:274–280. 127. Stampfer MJ, Maclure KM, Colditz GA, Manson JE, Willett WC. Risk of symptomatic gallstones in women with severe obesity. Am J Clin Nutr 1992; 55:652–658. 128. Sugerman HJ, Starkey JV, Birkenhauer R. A randomized prospective trial of gastric bypass versus vertical banded gastroplasty for morbid obesity and their effects on sweets versus nonsweets eaters. Ann Surg 1987; 205:613–624. 129. Thiet MD, Mittelstaedt CA, Herbst CA, Buckwalter JA. Cholelithiasis in morbid obesity. South Med J 1984; 77:415–417. 130. Amaral JF, Thompson WR. Gallbladder disease in the morbidly obese. Am J Surg 1985; 149:551–557. 131. GinanniCorradini S, Yamashita G, Nuutinen H, Chernosky A, Williams C, Hays L, Shiffman ML, Walsh RM, Svanvik J, Della Guardia P, Capocaccia L, Holzbach RT. Variations in pigment and carbohydrate content of gallbladder bile affect accurate quantitation of total protein when using the fluorescamine method. Scand J Gastroenterol 1997; 32:340–349. 132. Vezina WC, Paradis RL, Grace DM, Zimmer RA, Lamont DD, Rycroft KM, King ME, Hutton LC, Chey WY. Increased volume and decreased emptying of the gallbladder in large (morbidly obese, tall normal and muscular normal) people. Gastroenterology 1990; 98:1000–1007. 133. Marzio L, Capone F, Neri M, Mezzetti A, De Angelis C, Cuccurullo F. Gallbladder kinetics in obese patients: effect of a regular meal and a lowcalorie meal. Dig Dis Dis 1988; 33:4–9. 134. Marks JW, Bonorris GG, Schoenfield LJ. Roles of deoxycholate and arachidonate in pathogenesis of cholesterol gallstones in obese patients during rapid loss of weight. Dig Dis Sci 1991; 36:957–960. 135. Stone BG, Ansel HJ, Peterson FJ, Gebhard RL. Gallbladder emptying stimuli in obese and normal weight subjects. Hepatology 1992; 15:795–798. 136. Vassilakis JS, Pechlivanides G, Fountos A, Zoras OJ, Xynos E. RouxenY gastroenterostomy severely disturbs emptying of the gallbladder. J Am Coll Surg 1994; 179:313–317. 137. Kellum JH, Kuemmerle J, O'Dorisio T, Rayford P, Martin D, Engle K, Wolf N, Sugerman H. Gastrointestinal hormone responses to meals before and after bypass and vertical banded gastroplasty. Ann Surg 1990; 211:763–771. 138. Inoue K, Fuchigami A, Higashide S, Sumi S, Kogire M, Suzuki T, Tobe T. Gallbladder sludge and stone formation in relation to contractile function after gastrectomy. Ann Surg 1992; 215:19–26.
Page 382
139. Apstein MD, DaleckiChipperfield K. Spinal cord injury is a risk factor for gallstone disease. Gastroenterology 1987; 92:966–968. 140. Ketover SR, Ansel HJ, Goldish G, Roche B, Gebhard RL. Gallstones in chronic spinal cord injury: is impaired gallbladder emptying a risk factor. Arch Phys Med Rehabil 1996; 77:1136–1138. 141. GREPCORome group for the epidemiology and prevention of cholelithiasis: prevention of gallstone disease in an Italian adult female population. Am J Epidemiol 1984; 119: 796–805. 142. Buiumsohn A, Albu E, Gerst PH, Subbara O. Cholelithiasis and teenage mothers. J Adolesc Health Care 1990; 11:339–342. 143. Henriksson P, Einarsson K, Eriksson A, Kelter U, Angelin B. Estrogeninduced gallstone formation in males. J Clin Invest 1989; 84:811–816. 144. Braverman DZ, Johnson ML, Kern F Jr. Effects of pregnancy and contraceptive steroids on gallbladder function. N Engl J Med 1980; 302:362–364. 145. Everson GT, McKinley C, Lawson M, Johnson M, Kern F Jr. Gallbladder function in the human female: effect of the ovulatory cycle, pregnancy and contraceptive steroids. Gastroenterology 1982; 82:711–719. 146. Braverman DZ, Herbert D, Goldstein R, Persitz E, Eylath U, Jacobsohn WZ. Postpartum restoration of pregnancyinduced cholecystoparesis and prolonged intestinal transit time. J Clin Gastroenterol 1988; 10:642–646. 147. Fisher RS, Roberts GS, Grabowski CJ, Cohen S. Inhibition of lower esophageal sphincter circular muscle by female sex hormones. Am J Physiol 1978; 234:E243–E247. 148. Keane P, Colwell D, Baer HP, Clanachan AS. Effects of age, gender and female sex hormones upon contractility of the human gallbladder in vitro. Surg Gynecol Obstet 1986; 163:555–560. 149. Ryan JP, Pellecchia D. Effect of progesterone pretreatment on guinea pig gallbladder motility in vitro. Gastroenterology 1982; 83:81–83. 150. Shaffer EA, Taylor PJ, Logan K, Gadomski S, Corenblum B. The effect of progestin on gallbladder function in young women. Am J Obstet Gynecol 1984; 148:504–507. 151. Friley MD, Douglas G. Acute cholecystitis in pregnancy and the puerperium. Am Surg 1972; 38:314–317. 152. Glen F, McSherry CK. Gallstones and pregnancy among 300 young women treated by cholecystectomy. Surg Gynecol Obstet 1968; 127:1067–1072. 153. Hill LM, Johnson CE, Lee RA. Cholecystectomy in pregnancy. Obstet Gynecol 1975; 46:291–293. 154. Dowling RH, Hussaini SH, Murphy GM, Besser GM, Was JAH. Gallstones during octreotide therapy. Metabolism 1992; 41(suppl 2):22–33. 155. Boden G, Shimoyama R. Somatostatinoma. Contemp Issues Gastroenterol 1985; 5:95–99. 156. Fisher RS, Rock E, Levin G, Malmud L. Effects of somatostatin on gallbladder emptying. Gastroenterology 1987; 92:885–890. 157. Page MD, Millward ME, Taylor A, Preece M, Hourihan M, Hall R. Long term treatment of acromegally with a longacting analogue of somatostatin, octreotide. Q J Med 1990; 74:189–201. 158. Ho KY, Weissberger AJ, Marbach P, Lazarus L. Therapeutic efficacy of the somatostatin analog SMS 201–995 (octreotide) in acromegaly. Ann Intern Med 1990; 112:173–181. 159. Plockinger V, Dienemann D, Quabbe HJ. Gastrointestinal sideeffects of octreotide during long term treatment of acromegaly. J Clin Endocrinol Metab 1990; 71:1658–1662. 160. Roti E, Minelli R, Gardini E, Salvi M, Bianconi L, Balducci L, Manfredi A, Braverman LE. Chronic treatment with a longacting somatostatin analogue in a patient with intestinal carcinoid tumor: occurrence of cholelithiasis. J Endocrinol Invest 1990; 13:69–72. 161. Redfern JS, Fortuner WJ II. Octreotide associated biliary tract dysfunction and gallstone formation: pathophysiology and management. Am J Gastroenterol 1995; 90:1042–1052.
Page 383
162. Hussaini SH, Pereira SP, Veysey MJ, Kennedy C, Jenkins P, Murphy GM, Wass JAH, Dowling RH. Roles of gall bladder emptying and intestinal transit in the pathogenesis of octreotide induced gall bladder stones. Gut 1996; 38:775–783. 163. Stolk MFJ, van Erpecum KJ, Koppeschaar HPF, Samsom M, Smout AJPM, Akkermans LMA, Peeters TL, vanBergeHenegouwen GP. Effect of octreotide on fasting gall bladder emptying, antroduodenal motility and motilin release in acromegally. Gut 1995; 36: 755–760. 164. Erlinger S, Chanson P, Dumont M, Ponsot P, Warnet A, Harris AG. Effects of octreotide on biliary lipid composition and occurrence of cholesterol crystals in patients with acromegaly: a prospective study. Dig Dis Sci 1994; 39:2384–2388. 165. Buscail L, Tauber JP, Escourrou J, et al. Gallstone formation and occurrence of cholesterol monohydrate crystals in gallbladder bile of patients with longterm sandostatin treatment. Gastroenterol Clin Biol 1991; 15:800–804. 166. Einarsson K, Nilsell K, Leijd B, Angelin B. Influence of age on secretion of cholesterol and synthesis of bile acids by the liver. N Engl J Med 1985; 313:277– 282. 167. Khalil T, Walker JP, Wiener I, Fagan CJ, Townsend CM Jr, Greeley GH Jr, Thompson JC. Effect of aging on gallbladder contraction and release of cholecystokinin33 in humans. Surgery 1985; 98:423–429. 168. Capocaccia L, Giunchi G, Pocchiari F and GREPCO. The epidemiology of gallstone disease in Rome, Italy: Part I. Prevalence data in men. Hepatology 1988; 8:904–906. 169. Stone BG, Gavaler JS, Belle SH, Shreiner DP, Peleman RR, Sarva RP, Yingvorapant N, Van Thiel DH. Impairment of gallbladder emptying in diabetes mellitus. Gastroenterology 1988; 95:170–176. 170. Marks JW, Conley DR, Capretta TL, Bonorris GG, Chung A, Coyne MJ, Schoenfield LJ. Gallstone prevalence and biliary lipid composition in inflammatory bowel disease. Am J Dis Sci 1977; 22:1097–1100. 171. Heaton KW, Read AE. Gall stones in patients with disorders of the terminal ileum and disturbed bile acid metabolism. BMJ 1969; 3:494–496. 172. Kato I, Nomura A, Stemmermann GN, Chyou PH. Prospective study of clinical gallbladder disease and its association with obesity, physical activity, and other factors. Dig Dis Sci 1992; 37:784–790. 173. Williams CN, Johnson JL. Prevalence of gallstones and risk factors in Caucasian women in a rural Canadian community. Can Med Assoc J 1980; 122:664– 668. 174. Leitzmann MF, Giovannucci EL, Rimm, EB, Stampfer MJ, Spiegelman D, Wing AL, Willett WC. The relation of physical activity to risk for symptomatic gallstone disease in men. Ann Intern Med 1998; 128:417–425. 175. Baker TT, Allen D, Lei KY, Willcox KK. Aterations in lipid and protein profiles of plasma lipoproteins in middleaged men consequent to an aerobic exercise program. Metabolism 1986; 35:1037–1043. 176. Tran ZV, Weltman A, Glass GV, Mood DP. The effects of exercise on blood lipids and lipoproteins: a metaanalysis of studies. Med Sci Sports Exerc 1983; 15:393–402. 177. Halloran LG, Schwartz CC, Vlahcevic ZR, Nisman RM, Swell L. Evidence for highdensity lipoproteinfree cholesterol as the primary precursor for bile acid synthesis in man. Surgery 1978; 84:1–7. 178. Thornton JR, Heaton KW, Macfarlane DG. A relation between highdensitylipoprotein cholesterol and bile cholesterol saturation. BMJ 1981; 283:1352–1354. 179. Petitti DB, Friedman GD, Klatsky AL. Association of a history of gallbladder disease with a reduced concentration of highdensitylipoprotein cholesterol. N Engl J Med 1981; 304:1396–1398. 180. Mingrone G, Greco AV, Finotti E, Passi S. Free fatty acids: a stimulus for mucin hypersecretion in cholesterol gallstone biles. Biochim Biophys Acta 1988; 958:52–59. 181. Philipp E, Wilckens T, Friess E, Platte P, Pirke KM. Cholecystokinin, gastrin and stress hormone responses in marathon runners. Peptides 1992; 13:125–128. 182. Hoy MK, Heshka S, Allison DB, Grassett E, Blank R, Abiri M, Heymsfield SB. Reduced
Page 384
risk of liverfunctiontest abnormalities and new gallstone formation with weight loss on 3350J (800kcal) formula diets. Am J Clin Nutr 1994; 60:249–254. 183. Ortega RM, FernandezAzuela M, EncinasSolillos A, Andres P, LopezSobaler AM. Differences in diet and food habits between patients with gallstones and controls. J Am Coll Nutr 1997; 16:88–95. 184. Simon JA, Grady D, Snabes MC, Fong J, Hunninghake DB. Ascorbic acid supplement use and the prevalence of gallbladder disease. J Clin Epidemiol 1998; 51:257–265. 185. Simon JA, Hudes ES. Serum ascorbic acid and other correlates of gallbladder disease among US adults. Am J Public Health 1998; 88:1208–1212. 186. Jenkins SA. Biliary lipids, bile acids and gallstone formation in hypovitaminotic C guineapigs. Br J Nutr 1978; 40:317–322. 187. Ginter E. Cholesterol: vitamin C controls its transformation to bile acids. Science 1973; 179:702–704. 188. Salen G, Tint GS, Shefer S. Treatment of cholesterol gallstones with litholytic bile acids. Gastroenterol Clin North Am 1991; 20:171–182. 189. Maton PN, Ellis HJ, Higgins MJ, Dowling RH. Hepatic HMG CoA reductase in human cholelithiasis: effects of chenodeoxycholic and ursodeoxycholic acids. Eur J Clin Invest 1980; 10:325–332. 190. Salen G, Colalillo A, Verga D, Bagan E, Tint GS, Shefer S. Effect of high and low doses of ursodeoxycholic acid on gallstone dissolution in humans. Gastroenterology 1980; 78:1412–1418. 191. Angelin B, Ewerth S, Einarsson K. Ursodeoxycholic acid treatment in cholesterol gallstone disease: effect on hepatic 3hydroxy3methylglutaryl coenzyme A reductase activity, biliary lipid composition and plasma lipid levels. J Lipid Res 1983; 24:461–468. 192. Carulli N, Ponz De Leon M, Zironi F, Pinetti A, Smerieri A, Iori R, Loria P. Hepatic cholesterol and bile acid metabolism in subjects with gallstones: comparative effects of short term feeding of chenodeoxycholic and ursodeoxycholic acids. J Lipid Res 1980; 21:35–43. 193. Carulli N, Loria P, Bertolotti M, Ponz de Leon M, Menozzi D, Medici G, Piccagli I. Effects of acute changes of bile acid pool composition on biliary lipid secretion. J Clin Invest 1984; 74:614–624. 194. Hardison WGM, Grundy SM. Effect of ursodeoxycholic acid and its taurine conjugate on bile acid synthesis and cholesterol absorption. Gastroenterology 1984; 87:130–135. 195. Stiehl A, Raedsch R, Rudolph G, Walker S. Effect of ursodeoxycholic acid on biliary bile acid and bile lipid composition in gallstone patients. Hepatology 1984; 4(1):107–111. 196. Von Bergmann K, EppleGutsfeld M, Leiss O. Differences in the effects of chenodeoxycholic and ursodeoxycholic acid on biliary lipid secretion and bile acid synthesis in patients with gallstones. Gastroenterology 1984; 87:136–143. 197. Igimi H, Carey MC. Cholesterol gallstone dissolution in bile: dissolution kinetics of crystalline (anhydrate and monohydrate) cholesterol with chenodeoxycholate, ursodeoxycholate and their glycine and taurine conjugates. J Lipid Res 1981; 22:254–270. 198. Tazuma S, Sasaki H, Mizuno S, Sagawa H, Hashiba S, Horiuchi I, Kajiyama G. Effect of ursodeoxycholic acid administration on nucleation time in human gallbladder bile. Gastroenterology 1989; 97:173–178. 199. Van Erpecum KJ, Van Berge Henegouwen GP, Stolk MFJ, Hopman WPM, Jansen JBMJ, Lamers CBHW. Effects of ursodeoxycholic acid on gallbladder contraction and cholecystokinin release in gallstone patients and normal subjects. Gastroenterology 1990; 99: 836–842. 200. Forgacs IC, Maisey MN, Murphy GM, Dowling RH. Influence of gallstones and ursodeoxycholic acid therapy on gallbladder emptying. Gastroenterology 1984; 87:299–307. 201. Sylwestrowicz T, Shaffer EA. Gallbladder function during gallstone dissolution: effect of bile acid therapy in patients with gallstones. Gastroenterology 1988; 95:740–748.
Page 385
202. Kutz C, Schulte A. Effectiveness of ursodeoxycholic acid in gallstone dissolution. Gastroenterology 1977; 73:632–633. 203. Maton PN, Murphy GM, Dowling RH. Ursodeoxycholic acid treatment of gallstones. Lancet 1977; 2:1297–1301. 204. Bachrach WJ, Hofmann AF. Ursodeoxycholic acid in the treatment of cholesterol cholelithiasis: part II. Dig Dis Sci 1982; 27:833–856. 205. O'Donnell LDJ, Heaton KW. Recurrence and rerecurrence of gallstones after medical dissolution: a longterm followup. Gut 1988; 29:655–658. 206. Tint GS, Salen G, Shefer S. Effect of ursodeoxycholic acid and chenodeoxycholic acid on cholesterol and bile acid metabolism. Gastroenterology 1986; 91:1007–1018. 207. Scott LD. Gallstone disease and pancreatitis in pregnancy. Gastroenterol Clin North Am 1992; 21:803–815. 208. Brotschi EA, LaMorte WW, Williams LF. Effect of dietary cholesterol and indomethacin in cholelithiasis and gallbladder motility in guinea pig. Dig Dis Sci 1984; 29:1050–1056. 209. Li YF, Russell DH, Myers SI, Weisbrodt NW, Moody FG. Gallbladder contractility in aspirin and cholesterolfed prairie dogs. Gastroenterology 1994; 106:1662–1667. 210. Borch K, Chu M, Carlsson B, Rehfeld JF. Endogenous hypercholecystokininemia but not aspirin reduces the gallstone incidence in the hamster model. Scand J Gastroenterol 1994; 29:740–743. 211. Sterling RK, Shiffman ML, Sugerman HJ, Moore EW. Effect of NSAIDs on gallbladder bile composition. Dig Dis Sci 1995; 40:2220–226. 212. O'Donnell LJD, Wilson P, Guest P, Catnach SM, McLean A, Wickham JEA, Fairclough PD. Indomethacin and postprandial gallbladder emptying. Lancet 1992; 339:269–271. 213. Hood KA, Ruppin DC, Gleeson D, Dowling RH. Prevention of gallstone recurrence by nonsteroidal antiinflammatory drugs. Lancet 1988; 26:1223–1225. 214. Hood KA, Gleeson D, Ruppin RH, Dowling RH. BritishBelgian gall stone study group: gall stone recurrence and its prevention: the BritishBelgian gall stone study group's postdissolution trial. Gut 1993; 34:1277–1288. 215. Pazzi P, Scagliarini R, Sighinolfi D, Govoni M, La Corte R, Gullini S. Nonsteroidal antiinflammatory drug use and gallstone disease prevalence: a case control study. Am J Gastroenterol 1998; 93(9):1420–1424. 216. Kurata JH, Marks J, Abbey D. One gram of aspirin per day does not reduce the risk of hospitalization for gallstone disease. Dig Dis Sci 1991; 36:1110–1115. 217. Sterling RK, Shiffman ML. Nonsteroidal antiinflammatory drugs and gallstone disease: will an aspirin a day keep the gallstones away? Am J Gastroenterol 1998; 93(9):1405–1407. 218. Grundy SM. HMGCoA reductase inhibitors for treatment of hypercholesterolemia. N Engl J Med 1988; 319:24–33. 219. Reihner E, Rudling M, Stahlberg D, Berglund L, Ewerth S, Bjorkhem I, Einarsson K, Angelin B. Influence of pravastatin, a specific inhibitor of HMGCoA reductase, on hepatic metabolism of cholesterol. N Engl J Med 1990; 323:224–228. 220. Hillebrant CG, Axelson M, Bjorkhem I, Wang FH, Nyberg B, Einarsson C. Effects of short term treatment with pravastatin on the hepatic synthesis of cholesterol and bile acids in gallstone patients. Eur J Clin Invest 1998; 28:324–328. 221. Duane WC, Hunninghake DB, Freeman ML, Pooler PA, Schlasner LA, Gebhard RL. Simvistatin, a competitive inhibitor of HMGCoA reductase, lowers cholesterol saturation index of gallbladder bile. Hepatology 1988; 8:1147–1150. 222. Hoogerbrugge van der Linden N, de Rooy FWM, van Blankenstein M. The effect of pravastatin on biliary lipid composition and bile acid synthesis in familial hypercholesterolemia. Gut 1990; 31:348–350. 223. Smit JWA, Van Erpecum KJ, Stolk MFJ, Geerdink RA, Cluysenaer OJJ, Erkelens DW, Van Berge Henegouwen GP. Successful dissolution of cholesterol gallstone during treatment with pravastatin. Gastroenterology 1992; 103:1068–1070.
Page 386
224. Sharma BC, Agarwal DK, Baijal SS, Saraswat VA. Pravastatin has no effect on bile lipid composition, nucleation time and gallbladder motility in persons with normal levels of cholesterol. J Clin Gastroenterol 1997; 25:433–436. 225. Sitzmann JV, Pitt HA, Steinborn PA, Pasha ZR, Sanders RC. Cholecystokinin prevents parenteral nutrition induced biliary sludge in humans. Surg Gynecol Obstet 1990; 170: 25–31. 226. Patankar R, Ozen MM, Sanderson A, Johnson CD. Effect of cisapride on gallbladder emptying and plasma CCK in normal and vagotomized human subjects. Dig Dis Sci 1996; 41:543–548. 227. Takaoka M, Kubota Y, Fujimura K, Ogura M, Kin H, Yamamoto S, Inoue K. Effect of single and multiple administrations of cisapride on postprandial gallbladder emptying in healthy humans. Intern Med 1994; 33:381–386. 228. Thorens J, Schnegg JF, Brignoli R, Froehlich F, Jansen JB, Dorta G, Blum AL, Gonvers JJ, Fried M. Effect of cisapride on gallbladder motility after extracorporeal shockwave lithotripsy. J Hepatol 1995; 22:333–337. 229. Xu QW, Scott RB, Tan DTM, Shaffer EA. Effect of prokinetic agent, erythromycin, in the Richardson round squirrel model of cholesterol gallstone disease. Hepatology 1998; 28:613–619.
Page 387
17— The Gallbladder and Biliary Tree in Cystic Fibrosis Michael P. Curry and John E. Hegarty St. Vincent's University Hospital, Dublin, Ireland I— Introduction Cystic fibrosis (CF) is the most common severe autosomal recessive disorder in the Caucasian population, with a frequency of 1 in 2000 births and a calculated carrier frequency of 5%. The first comprehensive report of cystic fibrosis was published in 1938 and highlighted the variability in clinical presentation, pathological findings, and outcome of patients with the disease. The term mucoviscidosis was coined in 1944 to reflect the importance of exocrine gland obstruction by inspissated mucus throughout multiple organ systems. Subsequently, in 1985, the gene for CF was mapped to chromosome 7. The gene and its product are termed the cystic fibrosis transmembrane conductance regulator (CFTR). The basic defect in the CFTR involves the abnormal control of ion and water transport across epithelial cells, resulting in the production of an abnormally viscous luminal secretion and reduced hydration of ductal mucus in the airways, pancreas, liver, and reproductive systems, leading to progressive obstructive damage. Until recent years, the incidence of subclinical liver disease has been overshadowed by respiratory and pancreatic complications. However, with increasingly effective management of such complications, resulting in better survival, management of liver disease and its complications has become important to a larger number of patients. Some 60% of CF patients now survive to adulthood. Liver disease is the second leading cause of death in CF and affects approximately 20 to 30% of adolescents and adults. Focal biliary cirrhosis is the pathognomonic hepatic lesion, which can progress to multilobular biliary cirrhosis, with signs and symptoms of cirrhosis and portal hypertension. Approximately 10% of this group will require liver transplantation for complications of their liver disease. Liver disease in CF may also occur as a consequence of malnutrition, bacterial or viral infection, druginduced hepatotoxocity, or right heart failure. II— Prevalence Since 1938, when Anderson first reported the hepatobiliary manifestations associated with CF, there has been considerable controversy in the literature regarding the prevalence of liver disease (1). The wide variation in the reported series probably relates to factors such as the different age groups studied and the sensitivity of methods used to detect liver disease, ranging from physical examination to diagnosis at autopsy. Autopsy studies have shown abnormal
Page 388
histological appearances of the liver of 10 to 30% of patients with CF. These studies suggest an agerelated increase in the incidence of histologically diagnosed CF liver disease from 10.8% in infants less than 3 months to 26.8% in those over the age of 1 year. This is further supported by an incidence of 72% in those patients who died over the age of 24 years (2,3). However, necropsy studies may not reflect the true agerelated prevalence, as liver disease itself may contribute to death. A number of reports have attempted to assess the clinical prevalence of CFrelated liver disease. In a review of 2500 patients over a 25year period, it was noted that 2% of children had portal hypertension, and this incidence increased tenfold in adolescent patients. Furthermore, a study of 1100 patients has shown a prevalence of clinically overt liver disease of 4.4%, with a progressive rise in prevalence from 0.3% in the 0 to 5 age group to a peak of 8.7% in those aged 16 to 20 years. Thereafter, instead of the expected rise, there was a fall to 4.1% in those over 20 years of age, which may reflect an increased mortality from respiratory or liver failure in those with liver disease during teenage years. The mean age at presentation was 9.8 years, with a male preponderance (4). The male preponderance has been supported in two further studies (5,6). One prospective study has been performed to assess the incidence and outcome of liver disease. Hepatic status was assessed at yearly intervals using a combination of clinical, biochemical, and ultrasound examinations. A cumulative incidence of 17% was reported with a male predominance. In contrast to the reported retrospective data, liver disease developed in the first decade of life and no incidence peak was observed in any age group (7). Like liver disease, the reported incidence of gallbladder and biliary tract disease among patients with CF is variable. Nonfunctioning gallbladder is the commonest abnormality, and microgallbladder has been observed in up to onethird of cases. Cholelithiasis has been a recognized complication of CF since 1973 and has an incidence of 14 to 24% (2,3,8). Cholangiographic features of sclerosing cholangitis have been reported in CF patients with liver disease (9–11). Strictures of the distal common bile duct (CBD) have been demonstrated in 90% of patients with CF liver disease and implicated as an etiological factor (11). Two further studies have failed to confirm this finding; therefore the role of distal CBD stenosis in the pathogenesis of CFrelated liver disease is controversial (9,10). III— Genetics CF is the most significant autosomal recessive disorder in the Caucasian population and has been reported in all racial groups with varying frequencies (12). The frequency of homozygote carrier status varies from 1:2500 in the United Kingdom to 1:16,000 in the AfricanAmerican population. This frequency has been determined by case finding from large population studies rather than widespread neonatal screening and thus may underestimate the true incidence of CF. The genetic defect responsible for CF was localized to the long arm of chromosome 7 in 1985 and the gene was cloned in 1989. Since then the precise location of the gene has been established and sequenced; it was found to encode for a 1480amino acid protein termed the cystic fibrosis transmembrane conductance regulator (CFTR) (13–17). Definitive evidence that this is the correct gene comes from gene transfer experiments resulting in correction of the defective chloride channels. The amino acid sequence indicates that the CFTR belongs to the ABC (ATPbinding cassette) family of proteins with transmembrane transport function. The protein has two groups of six membranespanning regions, two intracellular nucleotidebinding folds (NBFs), and a highly charged ''R domain" containing multiple phosphorylation sites. Activation of the chloride channel requires protein kinase Amediated phosphorylation of the R domain and continuous presence of ATP in the NBF. The CFTR functions as a cyclic adenine monophosphate (cAMP)/protein kinase Aactivated chloride (Cl) channel. The cAMPstimulated secretion of Cl generates an osmotic gradient that drives sodium (Na+) and water into the lumen, leading to dilution of the bile. As in the pancreatic ducts, the change in the apical
Page 389 Table 1 Classification of the CFTR Mutations Mutations
Description of mutation
Class I
Defective protein production—This mutation results in premature termination on the mRNA and complete absence of the CFTR protein.
Class II
Defective protein processing—This mutation prevents the CFTR protein trafficking to the correct cellular location.
Class III
Defective regulation—This mutation leads to diminished channel activity in response to ATP.
Class IV
Defective conduction—This mutation results in correct production, localization, and cAMPmediated stimulation of the CFTR, with reduced rate of ion flow and duration of channel opening.
Source: From Ref. 21.
Cl gradient facilitates bicarbonate (
) excretion from the biliary epithelium, resulting in biliary alkalinization.
The most common defect of the CFTR is caused by a 3base pair deletion at position 508 with loss of a phenylalanine residue. This F508 defect accounts for approximately 70% of CF mutations, but its frequency varies considerably among ethnic groups (18,19). The F508 mutation is associated with classic severe CF with a high risk of lung disease, pancreatic insufficiency, and meconium ileus (20). Over 500 other genetic mutations have been identified. Heterozygotes for the CF are entirely asymptomatic. The F508 mutation results in disease because the mutated CFTR fails to reach the apical membrane of the epithelium; this is a result of incomplete glycosylation and failure to leave the endoplasmic reticulum or Golgi apparatus, where it becomes degraded (class II mutation). Other mutations may allow the CFTR to reach the apical membrane, but it fails to function (Table 1) (21). IV— Pathogenesis It has been clearly established by immunohistochemical techniques that the CFTR is expressed on the apical membrane of epithelial cells lining the intra and extrahepatic biliary tree (22). While the pathogenesis of CFassociated liver disease is not completely understood, it is thought that a mutation in the CFTR results in failure of the apical Cl channel function, resulting in reduced hydration and abnormally viscous ductal secretions. The resultant inspissation of bile ductules with tenacious eosinophilic secretions may lead to the characteristic histological features of duct obstruction, dilatation, hyperplasia, proliferation, and subsequent periductular inflammation and fibrosis. While focal fibrosis increases with advancing age, mucous obstruction of bile ducts is rarely seen beyond the neonatal period and in some cases is reversible. Furthermore, cholestasis is not a prominent feature of CFrelated liver disease beyond the neonatal period (23). These data would suggest that intrahepatic bile duct mucus, while important, is not the only determinant of the pathogenesis or progression of liver disease in CF. There is some evidence to suggest that necrosis of biliary epithelial cells occurs in the absence of bile duct obstruction and dilatation, implicating toxic injury to biliary epithelial cell death by cytotoxic compounds as a primary event in CFrelated liver disease (24). Glycineconjugated bile acids prevalent in those patients with CF as a result of bile acid malabsorption may be responsible for cytotoxic damage to cholangiocytes. Retention of hydrophobic bile acids within the hepatic parenchyma has been implicated as a major cause of liver disease, since it was shown that lithocholic acid induced cirrhosis in animal models (25). The association of certain HLA antigens with the development of liver disease and the presence of
Page 390
antibodies to liverspecific lipoproteins suggests that lymphocytemediated immune responses may either contribute to the primary damage or further exacerbate secondary progressive liver disease (26,27). Gallbladder abnormalities are present in approximately onethird of patients with CF, the majority of which are due to microgallbladder. The role of cholecystokinin (CCK) (hormonal stimulus promoting gallbladder contraction) in the pathogenesis of microgallbladder and gallbladder disease has been extensively investigated. High resting and postprandial CCK levels are observed in CF patients, consistent with microgallbladder and the negative feedback loop between reduced luminal bile acids and CCK release (28). Administration of a CCK antagonist (Loxiglumide) does not produce the expected relaxation of the gallbladder in CF patients. In view of this and the frequently observed association of cholelithiasis with microgallbladder, it is speculated that gallstone disease results in chronic inflammation and the development of microgallbladder. While the mechanism of stone formation is not completely understood, fat malabsorption has been incriminated because of its contribution to increased fecal bile acid loss and subsequent development of lithogenic glycineconjugated bile. The clinical practice of pancreatic enzyme supplementation and dietary measures has not, however, been associated with a reduced frequency of gallstones. The proposed hypothesis of cholesterol supersaturation of bile promoting the formation of radiolucent cholesterol gallstones has been refuted by many studies showing normal or reduced cholesterol saturation in CF (29). Furthermore, there is no difference in cholesterol saturation between CF patients with and without gallstones (30). These studies would suggest that bile acid depletion alone is not sufficient for the observed increased incidence of gallstone disease. It has therefore been hypothesized that bile stasis resulting from cystic duct and biliary tract disease may impair gallbladder emptying, promote nucleation, and allow gallstone formation. V— Clinical Variability The large clinical variability of hepatobiliary manifestations and the familial clustering of CF patients with liver disease has led some authors to investigate the correlation between genotype and phenotype. Mutation analysis of large series of patients with CF has shown clear associations between genotype and pancreatic function; however, there is no association between CF genotype and the development or severity of liver disease (31,32). A familial concordance for liver disease has suggested that genes outside the CF locus may be involved in the pathogenesis of liver disease. Duthie et al. (26) have demonstrated that the HLA class III antigen DQw6 was present in 66.7% of a British population of CF patients with liver disease, compared with 32.9% of those without liver disease and 28.8% of controls. This and weaker association with HLADR2 (DR15), B7, B16, B21, and B22 are for male patients only, suggesting that a genetic susceptibility to chronic liver disease in male CF patients is associated with genes at or close to the DQ locus. The association between chronic liver disease in male patients with CF and the B7 DR2DQ6 haplotype is similar to that observed in a subgroup of patients with primary sclerosing cholangitis, which has similar biliary features to CF and also has a male predominance (33). This may have therapeutic implications for establishing early treatment with ursodeoxycholic acid (UDCA) in male patients with the HLA DQw6 genotype. The ability to predict the development of liver disease on the basis of genetic testing is hampered by the lack of association with genotype and the association of HLA class II molecules only with the male sex. The possibility that a mutation not as yet identified may be responsible for CFrelated liver disease is an attractive but unlikely concept. Autopsy studies have demonstrated that there is a strong association with meconium ileus or meconium ileus equivalent and the development of liver disease (34). These studies are strongly supported by the findings of Colombo et al., showing that CF patients who have a history of neonatal meconium ileus are six times more likely to develop liver disease than those who do not (6,7).
Page 391 Table 2 Clinical Liver Problems in Cystic Fibrosis Neonatal cholestasis with or without meconium ileus Fatty liver causing transient hepatomegaly associated with malnutrition Asymptomatic hepato(spleno)megaly Abdominal pain Variceal hemorrhage Hypersplenism Decompensated cirrhosis Asymptomatic abnormalities of liver biochemistry or ultrasound Sources: From Refs. 1, 4, 7, 34, and 35.
VI— Clinical Features Hepatic disease in CF may be asymptomatic and usually follows a slowly progressive course characterized by focal bile duct damage, portal hypertension, and preservation of hepatic architecture and liver cell integrity. As a result of the characteristic pathological process, the major clinical features of CFassociated liver disease relate to the development of portal hypertension and its complications (Table 2). Some asymptomatic patients will be diagnosed as having liver disease on routine screening in the CF clinic on the basis of abnormal clinical findings with or without associated biochemical or radiological abnormalities. Rarely, the first clinical manifestations of CF can be liverrelated and present to the gastroenterologist/hepatologist. Neonatal cholestasis (35,36) or vitamin Kdeficient hemorrhagic diseases are rare presentations of CF (37). Neonatal jaundice occurs more frequently in association with meconium ileus and is sometimes due to obstruction of the extrahepatic biliary tree with tenacious bile; laparotomy and biliary lavage may be required to clear the obstruction (38). Hepatomegaly can be detected in isolation in infancy due to massive hepatic steatosis and malnutrition, but it is more commonly a presenting sign in combination with splenomegaly due to cirrhosis in childhood or adolescence (4,39). It is therefore worthwhile to determine the sweat electrolyte concentration in the evaluation of obscure liver disease in children and adolescents (39). Progressive enlargement of liver and spleen may give rise to abdominal distention and pain, which must be differentiated from intestinal obstruction secondary to meconium ileus equivalent. Abdominal pain may also result from symptomatic gallbladder disease, common bile duct obstruction, and intrahepatic microlithiasis in a small number of patients (11,40–42). Encephalopathy and ascites are rare and late clinical features and cutaneous stigmata are usually absent (43). The first clinical presentation of liver disease may be an acute variceal hemorrhage and usually occurs in those with splenomegaly. The percentage of CF patients with portal hypertensive varices is unknown, but in one cohort of 1100 patients, only 6 patients had a variceal bleed (4). Massive splenomegaly may occur as a consequence of portal hypertension and result in the hematological features of hypersplenism. This rarely requires intervention, as the response to bleeding and infection is not significantly impaired. Leftupperquadrant pain may occur as a direct result of splenic enlargement or as a consequence of infarction; if severe, it may require splenectomy in selected cases. VII— Diagnosis The traditional diagnostic criteria for CF of persistently elevated concentration of sweat electrolytes plus characteristic clinical findings or a family history still apply. When these criteria
Page 392
are fulfilled, the diagnosis of CF can be made; however, if the criteria are not met, CF cannot be ruled out. These atypical patients usually present as adolescents or adults with complications of pancreatitis and/or infertility. Liver disease has long been a recognized feature of CF; however, early detection of liver involvement remains difficult. This is confounded by the fact that commencement of prophylactic therapy in the form of ursodeoxycholic acid (UCDA) may serve to prevent progression to biliary cirrhosis. Early detection and diagnosis of liver disease in CF is therefore essential and should be based on a combination of clinical assessment, biochemical analysis, and diagnostic imaging. A— Clinical Assessment While clinical assessment of the patient offers the best available method for the detection of cirrhosis, it is neither sensitive nor specific enough to confirm the diagnosis of early CFrelated liver disease. The presence of an enlarged liver alone may reflect depression of a normalsized liver below the costal margin by hyperinflated lungs. Reliance on cutaneous stigmata as diagnostic aids is problematic as they are usually absent in CFrelated liver disease (43). B— Laboratory Investigations Conventional biochemical tests of liver function are abnormal in a high proportion of patients with a predominantly cholestatic pattern, but the degree of abnormality does not correspond with the extent of the fibrotic process. The correlation with clinical evidence of liver disease is also poor, as 13% of patients with clinical liver disease reported in a series by ScottJupp et al. had normal liver function tests and 12.9% with elevated liver enzymes had no clinical liver disease (4). Elevation of the alkaline phosphatase, seen in 38% of patients, is the commonest abnormality and must be differentiated from the skeletal isoenzyme, which may be elevated in association with growth or metabolic bone disease. Serum glutathione Stransferase B1 may be a more sensitive method of detecting liver impairment, as elevated levels were observed in 16 of 18 patients with clinically apparent CFrelated liver disease and none of 12 patients with abnormal liver function tests in the absence of clinical liver disease (44). Further, larger followup studies will be required to verify the usefulness of this assay, as elevations of liver function tests are sometimes only transient events secondary to selflimiting coexistent disease or medication. Hepatic synthetic function as reflected by the prothrombin time is usually normal, but it may be prolonged in cases of vitamin K deficiency and advanced liver disease. Noninvasive markers of liver fibrosis for the diagnosis and followup of CF liver disease are urgently required. Circulating collagen VI levels, which are elevated in adults with liver fibrosis and cirrhosis, have been found to be significantly elevated in children with CFrelated liver fibrosis or cirrhosis and normal in all those with no evidence of liver disease and normal controls. This may provide a sensitive and specific serum marker for the detection of liver fibrosis in children with CF (Fig. 1) (45). C— Ultrasound Ultrasound examination is a noninvasive method of assessing liver echotexture and morphology and therefore is useful in determining the presence of absence of hepatobiliary disease, portal hypertension, and architectural distortion. Abnormal echotexture may be due to either steatosis or fibrosis. The differentiation can be reliably made only by histological analysis. Duplex Doppler ultrasound has been used to assess the diameter of the portal vein so as to determine the presence of liver disease; however, these measurements serve only as a more sensitive method for the detection of portal hypertension reflecting already established liver disease (46,47). The application of a systematic scoring system to assess the echotexture of the hepatic
Page 393
Figure 1 Serum collagen VI levels in 224 healthy children and 54 children with liver fibrosis (left panel) and 133 CF children with no overt liver disease and 30 CF children with liver disease (right panel). (From Ref. 44.)
parenchyma, contour of the liver edge, and presence of portal fibrosis has resulted in a reproducible method of assessing CF patients for the presence of liver disease. This technique may be of value in identifying precirrhotic liver disease and therefore allow for the early detection and subsequent prophylactic treatment of CFrelated liver disease (48). D— Hepatobiliary Scintigraphy Hepatobiliary scintigraphy is a useful investigation because it provides both functional and structural information about the liver. Scintigraphy can demonstrate a spectrum of abnormal findings in CFrelated liver disease, including delayed clearance of isotope from the liver parenchyma, nonvisualization of the gallbladder, dilated intrahepatic ducts, and intrahepatic duct strictures (49). Beading and stricturing of the intrahepatic biliary tree, which is found most frequently in those patients with established liver disease, probably represents the strictures found during cholangiography (50). Delayed uptake and retention of isotope within the parenchyma is significantly more common in those patients with established liver disease (Fig. 2) (10). Intrahepatic retention is also seen in patients with abnormal liver enzymes and no clinical evidence of advanced liver disease and may represent focal cholestasis. This is supported by the observation that significant improvement in isotope clearance is noted following treatment with UDCA (50,51). These studies suggest that hepatobiliary scintigraphy has a role in the diagnosis of CFrelated liver disease. It is sufficiently sensitive to detect liver abnormalities in asymptomatic individuals and provides a mechanism to monitor the response to UDCA. E— Liver Histology Liver biopsy is not routinely used to establish a diagnosis of CF because of the focal nature of the pathological process and the possibility of sampling error. Percutaneous needle biopsy
Page 394
Figure 2 Maximum time (Tmax) for hepatic uptake of 99Tc DISIDA and percentage clearance of 99Tc DISIDA from the liver and biliary tree at 45 (E45) and 60 (E60) minutes in CF patients with and without liver disease. (From Ref. 10.)
can underestimate the pathological changes in CFrelated liver disease as compared with perioperative wedge biopsy (52). This contrasts with studies from other liver diseases. F— Endoscopic Retrograde Cholangiopancreatography Endoscopic retrograde cholangiopancreatography (ERCP) is an invasive investigation and is therefore not useful as a screening test for liver disease. In clinical studies, it has identified intrahepatic bile duct strictures similar to those of primary sclerosing cholangitis and extrahepatic biliary obstruction, which may be important in determining the progression of the disease (Fig. 3) (10,11). Despite the possible associated morbidity, it is an invaluable tool for the diagnosis and treatment of biliary obstruction secondary to cholelithiasis (10). G— Magnetic Resonance Imaging Magnetic resonance cholangiography (MRC) has rapidly become an important noninvasive technique for the evaluating intra and extrahepatic ducts. Unlike conventional cholangiography, MRC does not require the introduction of contrast material into the ductal system and so avoids the associated morbidity. MRC provides diagnostic cholangiograms in 90 to 100% of patients with a dilated system and can identify the larger intrahepatic ducts in 83 to 100% of patients with a normalcaliber biliary tree. As a result, MRC may not be sufficiently sensitive to evaluate the second and thirdorder intrahepatic ducts (53). MRC is currently being evaluated in CFrelated liver disease and may provide the necessary noninvasive tool for its early detection (54).
Page 395
Figure 3 Endoscopic retrograde cholangiogram in a 15yearold girl with cystic fibrosis related liver disease, demonstrating beading and stricturing of the intrahepatic biliary tree. (From Ref. 10.)
VIII— Pathology The biliary cirrhosis of cystic fibrosis has distinctive histopathology. Large sections of hepatic parenchyma are spared, but the liver architecture may be sufficiently distorted to produce portal hypertension, hypersplenism, and gastrointestinal hemorrhage. The pathological features of fatty liver, mild portal fibrosis, and biliary cirrhosis were first described in Andersen's original paper on CF in 1938 (1). Subsequently Farber et al. described small bile ducts blocked by inspissated eosinophilic material resembling that found in the pancreatic ducts and acini (55). The focal nature of the fibrotic lesion with associated eosinophilic concretions was termed focal biliary cirrhosis, and the term multilobular biliary cirrhosis was used to describe the large, irregular nodules typical of cirrhosis in those patients with CF. Hepatic changes may be
Page 396
present at any age. Neonatal cholestasis is present in 30% of cases and may be associated with neonatal hepatitis or biliary atresia. Excessive biliary mucus associated with mild periportal inflammation and early fibrosis is common in infants, while focal biliary fibrosis with associated eosinophilic PASpositive material in ductules, bile duct reduplication, and chronic inflammatory infiltrates is present in >20% of adolescents (Fig. 4). In some patients, focal lesions coalesce and progress to multilobular biliary cirrhosis characterized by irregular nodules, large bands of fibrosis, regenerative microscopic nodules, and bile duct reduplication adjacent to areas of preserved hepatic lobular architecture (Fig. 5). Bile plugs and cholestasis may be present, but even in advanced disease they are rare beyond the neonatal period (56). Fatty liver, which occurs in approximately 30% of CF patients, was previously attributed to malnutrition with specific deficiencies of fatty acids or carnitine; however, increased amounts of fat in hepatocytes has been documented in wellnourished patients with high Scwachman scores (Fig. 6). The relationship between steatosis and the other hepatic lesions has not been defined, but there is no correlation between fatty infiltration and the amount of collagen deposition (24,57). Electron microscopy of CF liver tissue has identified disrupted and necrotic cholangiocytes in the absence of biliary dilatation or bile plugs. Bile duct reduplication is a prominent feature. Inflammatory cells—including lymphocytes, occasional neutrophils, and macrophages—are present in the portal tracts and are more numerous in those with extensive fibrosis. Activated fatstoring cells, known to produce collagen, are present in fibrotic portal tracts and are possibly engaged in collagen production (24). IX— Bile Acid Metabolism and Fat Malabsorption Bile acids represent the predominant component of bile and are synthesized de novo in the liver from cholesterol. The primary bile acids, cholic and chenodeoxycholic acids, are conjugated with glycine and taurine normally in a ratio of 3:1 to increase solubility. Further conjugation may occur with the addition of a sulfate or a glucuronide to one or more hydroxyl groups of the cholesterol nucleus. These conjugates are more soluble and less readily absorbed from the gut. In the intestine, bacteria convert primary bile acids into secondary forms (deoxycholic and lithocholic acids) through a process of dehydroxylation. Approximately 95% of the bile acids are reabsorbed in the terminal ileum, enter the enterohepatic circulation, and are reexcreted in the bile. The absorbed fraction of lithocholic acid is conjugated with sulfate, thus reducing the amount of this potentially harmful hydrophobic bile acid that is reabsorbed. Bile acids act as detergents, participating in the solubilization and absorption of lipids. The more hydrophobic bile acids are better detergents and are therefore potentially more destructive to cell membranes. Hydrophobic bile acids, in contrast to the hydrophilic bile acids, decrease bile flow and may further promote liver injury (58). Fecal bile acid (FBA) excretion is markedly increased in CF patients with and without pancreatic insufficiency, leading to a reduced total bile acid pool. Fecal fat loss as a consequence of pancreatic insufficiency is also increased in CF, resulting in nutritional deficiencies. This association of fecal fat and bile acid excretion may result from inhibition of bile acid absorption by intraluminal unhydrolyzed triglycerides. However, although the addition of pancreatic enzymes and H2 receptor antagonists and substitution of dietary fat for mediumchain triglycerides reduces fecal fat excretion, the same does not always hold true for bile acid excretion (59). The administration of sodium bicarbonate also reduces fecal fat excretion but has no effect on FBA loss, and patients with near normal fat excretion continue to lose large amounts of FBA (59). These data would suggest that mechanisms other than inhibition of bile acid absorption by intraluminal unhydrolyzed triglycerides plays a role in excessive bile acid excretion. Decreased intestinal motility due to viscid intraluminal mucus, or altered intestinal hormones with resulting bacterial overgrowth, deconjugation, and dehydroxylation of bile acids has been suggested to reduce bile acid absorption and contribute to fat malabsorption (60). It is unlikely that bacterial overgrowth alone contributes to FBA and fecal fat loss, as the mal
Page 397
Figure 4 Histological section demonstrating proliferating ductules within fibrous bands in a patient with cystic fibrosis. The ductules are distended by eosinophilic granular material (H&E, ×20). (Courtesy of Dr. Niamh Nolan.)
Figure 5 Biliary cirrhosis with hepatocyte nodules and broad fibrous bands (H&E, ×20). (Courtesy of Dr. Niamh Nolan.)
Page 398
Figure 6 H&E section demonstrating extensive hepatic steatosis. This is the most common finding in cystic fibrosis, present in onethird of patients. (Courtesy of Dr. Niamh Nolan.)
absorption of fat and bile acids is similar in patients who have evidence of bacterial overgrowth and those that do not. In vitro studies have suggested that dysfunction of ileal transport processes may contribute to excessive FBA loss. The presence of a primary defect in ileal mucosal transport selective for cholic acid is further supported by the observations of reduced postprandial serum cholic acid level and increased fecal loss of cholic acid in CF patients with and without pancreatic insufficiency. Absorption of chenodeoxycholic acid is reduced in CF patients with pancreatic insufficiency as compared to those with normal pancreatic function, suggesting that cholic acid absorption is impaired by a defect in mucosal transport and that absorption of both cholic and chenodeoxycholic acid is reduced by pancreatic insufficiency (61). The malabsorption of these primary bile acids results in the production of hydrophobic secondary bile acids as a consequence of dehydroxylation by colonic flora. Secondary bile acids are reported to be cytotoxic and their increased absorption, circulation, and accumulation within the liver may contribute to the pathogenesis of CFrelated liver disease (62). Serum levels of several potentially toxic secondary bile acids are increased in CFrelated liver disease. Fasting levels of taurochenodeoxycholic acid are increased 30fold in patients with liver disease and correlate closely with biochemical cholestasis. Administration of UDCA results in reduction in the serum and biliary level of taurochenodeoxycholic acid, with associated improvement in liver biochemistry (63). X— Management of CFRelated Hepatobiliary Disease A— Nutrition Malnutrition in cystic fibrosis has been described for over a half a century and results from malabsorption and increased energy expenditure as a consequence of chronic disease. Severe
Page 399
liver disease with an associated reduction in the bile acid pool and poor protein synthesis may contribute to the patient's poor nutritional status. Energy intake in CF patients on a free diet has been calculated to be 80.3 ± 4.6% of the recommended daily allowance (RDA), which falls well below the recommended total energy intake of 120 to 150% (64). A dietary fat intake of 40% of total daily energy intake along with adequate pancreatic enzyme supplementation is now advocated for all patients (65). Deficiencies of fatsoluble vitamins D, E, and K occur in CFrelated liver disease and require supplementation with adequate nontoxic amounts. Judicious use of vitamin A supplementation is recommended, as serum levels do not reflect hepatic stores and there are reports of toxicity (66). B— Ursodeoxycholic Acid The use of UDCA for the treatment of a wide range of cholestatic liver diseases has been increasing rapidly since the first reports of its beneficial effects on serum bilirubin (67). The mechanism of action of UDCA is not fully understood; however, experimental and clinical evidence currently available suggests that the effects of UDCA are due to the protection of cell membranes at the level of the biliary tree and stimulation of impaired hepatocellular secretion of toxic hydrophobic bile acids. UDCA conjugates have a stabilizing effect on biomembrane exposed to hydrophobic bile acids, possibly by the formation structure of mixed micelles. Taurine conjugated UDCA also promotes secretion from cholestatic hepatocytes through a complex of signals mediated by Ca2+ and protein kinase C (68). The accumulation of inspissated bile within the intrahepatic biliary tree and toxic hydrophobic bile acids in the liver parenchyma are two possible mechanisms involved in the pathogenesis of CF liver disease. The use of UDCA, a hydrophilic bile acid with choleretic properties, is therefore reasonable in the treatment of this condition. A randomized, double blind, placebocontrolled trial has demonstrated that UDCA improves biochemical cholestasis, nutritional status in compromized individuals, pulmonary function, and general wellbeing in patients treated for 1 year (69,70). The optimal dose of 20 mg/kg body weight to achieve biliary enrichment is higher than that for other cholestatic diseases because of impaired intestinal absorption due to pancreatic insufficiency. UDCA at this dose has been shown to increase in the duodenal bile to an average of 42% of bile acids (71). That UDCA stimulates biliary excretion and improves hepatic morphology has been confirmed by hepatobiliary scintigraphy (51). In one uncontrolled study, UDCA administered for 2 years was shown to improve the histological features of CFrelated liver disease with a reduction in portal based inflammation, fibrosis, bile duct proliferation, and infiltration (72). UDCA is a promising and welltolerated treatment that confers a beneficial effect on biochemical cholestasis, nutrition, and perhaps liver morphology. Longterm followup studies are required to assess its effectiveness in preventing progression to liver cirrhosis resulting in the need for liver transplantation or death. C— Biliary Complications Up to 4% of the CF population will develop symptomatic gallbladder disease, and some of these will require surgical management (40). A decision to perform a cholecystectomy in such patients is difficult due to the possible pulmonary complications and the risk of hepatic decompensation in those with advanced cirrhosis. Biliary surgery in patients whose respiratory function has been optimized with the pre and postoperative use of mucolytic aerosols, intravenous antibiotics, and intensive physiotherapy is complicated by a morbidity and mortality of 10 and 5% respectively (73). Laparoscopic cholecystectomy under general or continuous epidural anaesthesia may have advantages in those patients with poor respiratory function (74,75). Choledocholithiasis complicated by cholangitis or distal common bile duct obstruction or stricture causing abdominal pain can be treated by endoscopic sphincterotomy, stone extraction, and stent insertion with less respiratory complications than may be involved in abdominal
Page 400
surgery. In those patients unsuitable for surgery with mild symptoms that can be controlled with analgesia who have a functioning gallbladder and a small number of radiolucent stones, medical therapy with oral bile salts and/or extracorporeal shockwave lithotripsy may succeed in dissolving the gallstones. D— Complications of Portal Hypertension Acute variceal hemorrhage should be managed by direct admission to hospital. If there is hemodynamic compromise, blood volume should be replaced and coagulation abnormalities corrected with caution to prevent right heart failure, pulmonary edema, and further respiratory compromise. Once the patient is hemodynamically stable, an endoscopy by a skilled endoscopist should be performed to establish the site of bleeding and proceed with injection sclerotherapy or variceal banding to control hemorrhage. A course of sclerotherapy or variceal ligation should be embarked upon until varices have been obliterated, followed by a surveillance program to maintain obliteration. A metanalysis comparing injection sclerotherapy with variceal banding revealed a reduced rate of rebleeding, esophageal stricture, and death due to rebleeding with variceal banding in patients with portal hypertension. It was also noted that the number of session of treatment was lower with the banding technique (76). Vasoactive drugs such as octreotide, somatostatin, vasopressin, or terlipressin—which are potent splanchnic vasoconstrictors— lower portal pressure and can be used as adjuvants to endoscopic therapy (77). Balloon tamponade, first used in 1930, is a highly effective method of arresting variceal hemorrhage but can cause significant complications (78). Patients are usually sedated and may require ventilation to protect them from aspiration pneumonia. Therefore balloon tamponade should be reserved for cases of uncontrolled bleeding and used only by experienced personnel adhering to a strict protocol. Portosystemic shunt surgery for the control variceal bleeding has largely been replaced by endoscopic methods and also by the emergence of the transjugular intrahepatic portosystemic shunt (TIPS). Shunt surgery involves a major operation with significant morbidity and mortality. It is usually complicated by significant deterioration in pulmonary function and also complicates any subsequent attempt at liver transplantation. The use of TIPS in the management of uncontrolled bleeding in CFrelated disease remains to be evaluated (79). Prophylactic injection sclerotherapy for the obliteration of esophageal varices is not recommended as the risk of rebleeding is increased until the varices are obliterated. Similarly, the use of beta blockers is not recommended for prophylaxis because of the risk of bronchospasm. The role of prophylactic banding of varices remains to be evaluated. E— Liver Transplantation Liver transplantation offers the only potentially curative treatment for portal hypertension and its complications. Until recent years, liver transplantation was not considered a suitable therapy for patients with CFrelated liver disease because of the high incidence of pulmonary complications and because it was thought that the lifelong immunosuppression required for recipients would predisposse to further pulmonary infection. A number of series have been published that have documented good survival rates following liver transplantation with no increase in the incidence of respiratory compromise (80–82). The optimal time for liver transplantation in CF is difficult to judge, as standard liver biochemistry is sometimes normal and so may underestimate the severity of liver disease. Liver transplantation should be reserved for those patients in whom there is evidence of hepatocellular failure, such as impaired protein synthesis, prolonged prothrombin time unresponsive to vitamin K, raised bilirubin in the absence of biliary obstruction, and hepatic encephalopathy. A helpful scoring system has been devised to indicate the need for referral for transplantation based on the severity of portal hypertension, impaired hepatic function, nutritional status, and hypersplenism (83). Prior to transplantation, malnutrition should be aggressively managed with supplemental feeding, preferably by the enteral route.
Page 401
Pulmonary function should be optimized by intense chest physiotherapy, the use of nebulized DNase to loosen sticky secretions, and the treatment of active infection with intravenous antibiotics. Patients with poor lung function (as evidenced by an FEV1 of 30 to 50%) can now undergo successful liver transplantation with appropriate pre and postoperative pulmonary care. An improvement in postoperative respiratory function may result from the use of corticosteroids, which reduce production of the mucus, and inhibit both leukocyte activation and the synthesis of inflammatory cytokines. Furthermore, the removal of a large liver, the reduction in portal hypertension, and consequent reduction in ascites will result in abdominal decompression and improved respiratory function. In the intraoperative period, there is a dramatic improvement in oxygenation and CO2 exchange when the abdomen is open and diaphragmatic splinting is relieved (83). There is controversy as to whether the biliary anastomosis should be directly from bile duct to bile duct or whether a RouxenY loop should be fashioned (84). The high incidence of biliary complications reported by NobleJamieson et al. (83) in patients who had direct duct to duct biliary anastomosis, would seem to suggest that a RouxenY loop is preferable. Splenectomy may be necessary in some cases due to the large size of the organ, and therefore prophylactic vaccination against Haemophilus influenzae and Streptococcus pneumoniae is recommended in children. Postoperatively, chest physiotherapy, nebulized DNase, and intravenous antibiotics are commenced early to prevent serious respiratory tract infections. Antifungal agents are used in those patients with positive Aspergillus serology and culture. The use of bronchoalveolar lavage through the endotracheal tube both preoperatively and at the end of the transplant has been advocated to reduce postoperative respiratory dysfunction (82). Malabsorption may prevent adequate levels of immunosuppression; therefore larger doses, increased dosing frequency, or intravenous immunosuppression may be required to achieve therapeutic levels. The newer immunosuppressive agents, neoral and tacrolimus, are better absorbed than conventional cyclosporine. Successful liver transplantation with relief of portal hypertension results in an improvement in absorption, nutrition, and respiratory function. Combined heartlungliver or lungliver transplantation can be performed successfully in patients with hepatic cirrhosis and severe, irreversible pulmonary disease with significant functional improvement (85). However, the increasing shortage of organ donors means that patients requiring multiple organ transplantation will wait longer, and it may be beneficial to transplant these patients early, before they develop severe respiratory compromise. XI— Gene Therapy Gene therapy remains the ultimate treatment for CF. Although many organ systems are involved in CF, efforts to develop gene therapy have focused almost entirely upon curing pulmonary disease, as it is the cause of greater than 90% of the mortality from CF. Recombinant adenoviruses have been extensively studied as vectors to deliver CFTR genes. The adenoviruses used for gene transfer are modified to replace adenoviral genome with the CFTR, resulting in a nonreplicating virus that is capable of CFTR expression. Clinical trials have indicated that recombinant adenoviruses will infect human airway cells with limited efficiency, and some but not all of the clinical trials report the generation of CFTR chloride channel activity (86,87). CFTR gene expression is shortlived, usually lasting less than a few weeks. This may result from virusinduced innate immune responses and tissue injury, as antiviral antibodies prevent epithelial cell infection and T cellmediated killing of transfected cells. The use of nonviral vectors has failed to overcome these problems, as inflammation has been reported when high doses of liposomes are delivered and the duration of expression of CFTR is short. Human biliary epithelial cell lines have been successfully infected by adenovirus vector with expression of functioning CFTR (88). In transgenic animal studies, adenovirus vectors
Page 402
have been infused into the biliary tract, with resultant expression of CFTR in almost all cells for a period of 21 days (89). An efficient method for the delivery of recombinant CFTR that can be sustained within the transfected epithelial cell is required for a successful outcome to gene therapy. XII— Conclusions Liver disease is an important complication of CF, with significant morbidity and mortality in the everincreasing number of CF survivors. CFassociated hepatobiliary disease is clearly the result of genetic expression of a basic defect in the biliary epithelium. Defective chloride secretion results in reduced hydration and increased viscosity of the intraluminal bile and plugging of small bile ductules, which may directly or by some other means cause periductular inflammation and fibrosis. The inflammatory and fibrotic process may be perpetuated by an increase in the ratio of toxic hydrophobic to hydrophilic bile acids, which accumulate within the liver and cause damage to cholangiocytes. The early detection of CFrelated liver disease—using a combination of clinical, biochemical, and imaging techniques—might allow for the introduction of UDCA prior to the development of irreversible liver damage. The patient with mild to moderate impairment of respiratory function and severe lifethreatening liver disease and portal hypertension should be referred for liver transplant assessment, as survival following a liver graft is good. For those patients with severe respiratory impairment, multiple organ transplantation remains an option. References 1. Anderson DH. Cystic fibrosis of pancreas and its relation to coeliac disease. Am J Dis Child 56:344–349, 1938. 2. Oppeinheimer EH, Esterly JR. Hepatic changes in young infants with cystic fibrosis: possible relation to focal biliary cirrhosis. J Pediatr 86:683–689, 1975. 3. Vawter GF, Shwachman H. Cystic fibrosis in adults: an autopsy study. Pathol Ann 14:357–382, 1979. 4. ScottJupp R, Lama M, Tanner MS. Prevalence of liver disease in cystic fibrosis. Arch Dis Child 66:698–701, 1991. 5. Feigelson J, Anagnostopoulos C, Poquet M, Pecau Y, Munck A, Navarro J. Liver cirrhosis in cystic fibrosistherapeutic implications and long term follow up. Arch Dis Child 68:653–657, 1993. 6. Colombo C, Apostolo MG, Ferrari M, Seia M, Genoni S, Giunta A, Sereni LP. Analysis of risk factors for the development of liver disease associated with cystic fibrosis. J Pediatr 124:393–399, 1994. 7. Colombo C, Battezzati PM, Strazzabosco M, Podda M. Liver and biliary problems in cystic fibrosis. Semin Liver Dis 18:227–235, 1998. 8. Rovsing H, Sloth K. Microgallbladder and biliary calculi in mucuviscidosis. Acta Radiol (Diagn) 14:588–592, 1973. 9. Nagel RA, Westaby D, Javaid A, Kavani J, Meire HB, Lombard MG, Wise A, Williams R. Liver disease and bile duct abnormalities in adults with cystic fibrosis. Lancet 11:1422–1425, 1989. 10. O'Brien S, Keogan M, Casey M, McErlean D, Fitzgerald MX, Hegarty JE. Biliary complications of cystic fibrosis. Gut 33:387–391, 1992. 11. Gaskin KJ, Waters DLM, HowmanGiles R, De Silva M, Earl JW, Martin HCO, Kan AE, Brown JM, Dorney SFA. Liver disease and common bile duct stenosis in cystic fibrosis. N Engl J Med 318:340–346, 1988.
Page 403
12. Marino CR, Gorelick FS. Scientific advances in cystic fibrosis. Gastroenterology 103:681–693, 1992. 13. Tsui LC, Buchwald M, Barker D, Braman JC, Knowlton R, Schumm JW, Eiberg H, Mohr J, Kennedy D, Plavisc N. Cystic fibrosis locus defined by a genetically linked polymorphic DNA marker. Science 230:1054–1057, 1985. 14. Knowlton RG, CohenHaguenaur O, Van Cong NV, Frezal L, Brown VA, Barker D, Braman JC, Schumm JW, Tsui LC, Buchwald M. A polymorphic DNA marker linked to cystic fibrosis is located on chromosome 7. Nature 318:380–382, 1985. 15. Riordan JR, Rommens JM, Kerem B, Alon N, Rozmahel R, Grelczak Z, Zielenski J, Lok S, Plavsic N, Chou JL, Drumm ML, Iannuzzi MC, Collins F, Tsui LC. Identification of the cystic fibrosis gene: cloning and characterisation of complementary DNA. Science 245:1066–1073, 1989. 16. Kerem B, Rommens JM, Buchanan JA, Markiewicz D, Cox TK, Chakravarti A, Buchwald M, Tsui LC. Identification of the cystic fibrosis gene: genetic analysis. Science 245:1073–1077, 1989. 17. Rommens JM, Iannuzzi MC, Kerem B, Drumm ML, Melmer G, Dean M, Rozmahel R, Cole JL, Kennedy D, Hidaka N, Zsiga M, Buchwald M, Riordan JR, Tsui LC, Collins FS. Identification of the cystic fibrosis gene: chromosome walking and jumping. Science 245:1059–1065, 1989. 18. Kerem E, Corey M, Kerem B, Rommens J, Markiewicz D, Levison H, Tsui LC, Durie P. The relationship between genotype and phenotype in cystic fibrosis analysis of the most common mutation ( F508). N Engl J Med 323:1517–1522, 1990. 19. Knight RA. Genetics of cystic fibrosis. Br J Hosp Med 47:502–506, 1992. 20. Santis G, Osborne L, Knight RA, Hodson M. Independent genetic determinants of pancreatic and pulmonary status in cystic fibrosis. Lancet 336:1081–1084, 1990. 21. Davis PB, Drumm M, Konstan MW. Cystic fibrosis. Am J Respir Crit Care Med 154:1229–1256, 1996. 22. Cohn JA, Strong TV, Picciotto MR, Nairn AC, Collins FS, Fitz JG. Localisation of the cystic fibrosis transmembrane conductance regulator in human bile duct epithelial cells. Gastroenterology 105:1857–1864, 1993. 23. Hultcrantz R, Mengarelli S, Strandvik B. Morphological findings in the liver of children with cystic fibrosis: a light and electron microscopical study. Hepatology 6:811–889, 1986. 24. Lindbald A, Hultcrantz R, Strandvik B. Bileduct destruction and collagen deposition: a prominent ultrastructural feature of the liver in cystic fibrosis. Hepatology 16:372–381, 1992. 25. Hosti P. Cirrhosis of the liver induced in rabbits by gastric instillation of 3–monohydroxycholynic acid. Nature 186:250–252, 1960. 26. Duthie A, Doherty DG, Donaldson PT, ScottJupp R, Tanner MS, Eddleston A, Mowat AP. The major histocompatibly complex influences the development of chronic liver disease in male children and young adults with cystic fibrosis. J Hepatol 23:532–537, 1995. 27. MieliVergani G, Psacharopoulous HT, Nicholson AM, Eddleston ALWF, Mowat AP, Williams R. Immune responses to liver membrane antigens in patients with cystic fibrosis and liver disease. Arch Dis Child 55:696–701, 1980. 28. Van Haren EH, Hopman WPM, Jansen JBMJ, Rosenbusch G, Lamers CBHW, Van Herwaarden CLA. Increased plasma cholecystokinin levels and small gallbladders in adult patients with cystic fibrosis. Clin Sci 81:85–89, 1991. 29. Roy CC, Weber AM, Morin CL, Combes JC, Nussle D, Megevand A, Lasalle R. Abnormal biliary lipid composition in cystic fibrosis: effect of pancreatic enzymes. N Engl J Med 297:1301–1305, 1977. 30. Angelico M, Gandin C, Cannuzzi P, Bertasi S, Cantafora A, De Santis A, Quattrucci S, Antonelli M. Gallstones in cystic fibrosis: a critical appraisal. Hepatology 14:768–775, 1991.
Page 404
31. De Arce M, O'Brien S, Hegarty J, O'Mahoney SM, Cashman SM, Delgado M, Fitzgerald MX. Deletion delta F508 and clinical expression of cystic fibrosis— related liver disease. Clin Genet 42:271–272, 1993. 32. Duthie A, Doherty DG, Williams C, ScottJupp R, Warner JO, Tanner MS, Williamson R, Mowat AP. Genotype analysis for F508, G551D and R553X mutations in children and young adults with cystic fibrosis with and without chronic liver disease. Hepatology 15:660–664, 1992. 33. Donaldson PT, Farrant JM, Wilkinson ML, Hayllar KM, Portmann BC, Williams R. Dual association of HLA DR2 and DR3 with primary sclerosing cholangitis. Hepatology 13:129–133, 1991. 34. Maurage C, Lenaerts C, Weber A, Brochu P, Yousef I, Roy CC. Meconium ileus and its equivalent as a risk factor for the development of cirrhosis: an autopsy study in cystic fibrosis. J Pediatr 9:17–20, 1989. 35. Smith FEM, Doughty IM, David TJ, Miller V, Patel L. Severe jaundice in two infants with cystic fibrosis. J R Soc Med 89:289–290, 1996. 36. Valman HB, France NE, Wallis PG. Prolonged neonatal jaundice in cystic fibrosis. Arch Dis Child 46:805–809, 1971. 37. Torstenson OL, Humphrey GB, Edson JR, Warwick WJ. Cystic fibrosis presenting with severe haemorrhage due to vitamin K malabsorption: a report of three cases. Paediatrics 45:857–861, 1970. 38. Evans JS, George DE, Mollit D. Biliary infusion therapy in the inspissated bile syndrome of cystic fibrosis. J Pediatr Gastroenterol Nutr 12:131–135, 1991. 39. Wilroy RS, Crawford SE, Johnson WW. Cystic fibrosis with extensive fat replacement of the liver. J Pediatr 68:67–73, 1966. 40. Stern RC, Rothstein FC, Doershuk CF. Treatment and prognosis of symptomatic gallbladder disease in patients with cystic fibrosis. J Pediatr Gastroenterol Nutr 5:35–40, 1986. 41. Patrick MK, HowanGiles R, De Silva M, Van Asperen P, Pitkin J, Gaskin KJ. Common bile duct obstruction causing right upper abdominal pain in cystic fibrosis. J Pediatr 108:101–102, 1986. 42. Magrude MJ, Munden MM. Intrahepatic microlithiasis: another gastrointestinal complication of cystic fibrosis. J Ultrasound Med 16:763–765, 1997. 43. Tanner MS, Taylor CJ. Liver disease in cystic fibrosis. Arch Dis Child 72:281–284, 1995. 44. Rattenbury JM, Taylor CJ, Haeth PK, Howie AF, Beckett GJ. Serum glutathione Stransferase B1 activity as an index of liver function in cystic fibrosis. J Clin Pathol 48:771–774, 1995. 45. Gerling B, Becker M, Staab D, Schuppan D. Prediction of liver fibrosis according to serum collagen VI levels in children with cystic fibrosis. N Engl J Med 336:1611–1612, 1997. 46. Vergesslich KA, Gotz M, Mostbeck G, Sommer G, Ponhold W. Portal venous blood flow in cystic fibrosis: assessment by duplex Doppler sonography. Pediatr Radiol 19:371–374, 1989. 47. KumariSubaiya S, Gorvoy J, Phillips G, Ross P, Riddelsberger MM. Portal vein measurement by ultrasonography in patients with long standing cystic fibrosis: preliminary observations. J Pediatr Gastroenterol Nutr 6:71–78, 1987. 48. Williams SGJ, Evanson JE, Barret N, Hodson ME, Boultbee JE, Westaby D. An ultrasound scoring system for the diagnosis of liver disease in cystic fibrosis. J Hepatol 22:513–521, 1995. 49. Dogan AS, Conway JJ, LloydStill JD. Hepatobiliary scintigraphy in children with cystic fibrosis and liver disease. J Nucl Med 35:432–435, 1994. 50. O'Connor PJ, Southern KW, Bowler IM, Irving HC, Robinson PJ, Littlewood JM. The role of hepatobiliary scintigraphy in cystic fibrosis. Hepatology 23:281– 287, 1996.
Page 405
51. Colombo C, Castellani MR, Balistreri WF, Seregni E, Assaisso ML, Giunta A. Scintigraphic documentation of an improvement in hepatobiliary excretory function after treatment with ursodeoxycholic acid in patients with cystic fibrosis and associated liver disease. Hepatology 4:677–684, 1992. 52. Picciotto A, Ciravegna G, Lapertosa G, Celle G. Percutaneous of laparoscopic needle biopsy on cystic fibrosis in the evaluation of chronic liver disease? Am J Gastroenterol 79:567–68, 1984. 53. Hintze RE, Adler A, Veltzke W, AbouRebyeh H, Hammerstring R, Vogl T, Felix R. Clinical significance of magnetic resonance cholangiopancreatography (MRCP) compared to endoscopic retrograde cholangiopancreatography (ERCP). Endoscopy 29:182–187, 1997. 54. Durieu I, Pellet O, Gaillard C, Bellon G, Vital Durand D. Magnetic resonance cholangiography in cystic fibrosis (abstr). Paediatr Pulmonol 14(suppl):307, 1997. 55. Farber S. Pancreatic function and disease in early life: pathological changes associated with pancreatic insufficiency in early life. Arch Pathol 37:238–250, 1944. 56. Park RW, Grand RJ. Gastrointestinal manifestations of cystic fibrosis: a review. Gastroenterology 81:1143–1161, 1981. 57. Colombo C, Apostolo M, Assaisso M, Roman B, Bottani P. Liver disease in cystic fibrosis. Neth J Med 41:119–122, 1992. 58. Palmer RH. Bile acids, liver injury and liver disease. Arch Intern Med 130:606–617, 1972. 59. Weber AM, Roy CC, Chartrand L, Lepage G, Dufour OL, Morin CL, Lasalle R. Relationship between bile acid malabsorption and pancreatic insufficiency in cystic fibrosis. Gut 17:295–299, 1976. 60. Allen JM, Penketh ARL, Adrian TE. Adult cystic fibrosis: postpramdial responses of gut regulatory peptides. Gastroenterology 85:1379–1383, 1983. 61. Colombo C, Aldo R, Roda E, Sereni LP, Brega A, Fugazza R, Giunta A. Bile acid malabsorption in cystic fibrosis with and without pancreatic insufficiency. J Pediatr Gastroenterol Nutr 3:556–562, 1984. 62. Scholmerich J, Becher MS, Schmidt K. Influence of hydroxylation and conjugation of bile acids on their membrane damaging properties: studies on isolated hepatocytes and lipid membrane vesicles. Hepatology 2:187–201, 1982. 63. Nakagawa M, Colombo C, Setchell KDR. Comprehensive study of the biliary bile acid composition of patients with cystic fibrosis and associated liver disease before and after UDCA administration. Hepatology 12:322–334, 1990. 64. Chase HP, Long MA, Lavin MH. Cystic fibrosis and malnutrition. J Pediatr 95:337–347, 1979. 65. Heymans HS. Gastrointestinal dysfunction and its effects on nutrition in cystic fibrosis. Acta Paeditrica Scandinavia; 363(Suppl):74–78, 1989. 66. Eid NS, Shoemaker LR, Saminec TD. Vitamin A in cystic fibrosis: case reports and review of the literature. J Ped Gastroenterol Nutr 10:265–269, 1990. 67. Leuschner U, Fischer H, Kurtz W, Guldutuna S, Hubner K, Hellstern A, Gatzen M. Ursodeoxycholic acid in primary biliary cirrhosis: results of a controlled double blind trial. Gastroenterology 97:1268–1274, 1989. 68. Beuers U, Boyer JL, Paumgartner G. Ursodeoxycholic acid in cholestasis: potential mechanisms of action and therapeutic applications. Hepatology 28:1449– 1453, 1998. 69. Colombo C, Battezzati PM, Podda M, Bettinardi N, Giunta A. Ursodeoxycholic acid for liver disease associated with cystic fibrosis: a doubleblind, multicentre trial—the Italian group for the study of ursodeoxycholic acid in cystic fibrosis. Hepatology 23:1484–1490, 1996. 70. O'Brien SM, Campbell GR, Burke AF, Maguire OC, Rowlands BJ, Fitzgerald MX, Hegarty JE. Serum bile acids and ursodeoxycholic acid treatment in cystic fibrosisrelated liver disease. Eur J Gastroenterol Hepatol 5:477–483, 1996.
Page 406
71. Colombo C, Crosignani A, Assaiaao ML. Ursodeoxycholic acid therapy in cystic fibrosis associated liver disease: a dose response study. Hepatology 16:924– 930, 1992. 72. Lindbald A, Glaumann H, Strandvik B. A twoyear prospective study of the effect of ursodeoxycholic acid on urinary bile acid excretion and liver morphology in cystic fibrosisassociated liver disease. Hepatology 27:166–174, 1998. 73. Snyder CL, Ferrell K, Saltzmann DA, Warwick WJ, Leonard AS. Operative therapy of gallbladder disease in patients with cystic fibrosis. Am J Surg 157:557– 561, 1989. 74. McGrath DS, Short C, Bredin CP, Kirwan WO, Rooney E, Meeke R. Laparoscopic cholecystectomy in adult cystic fibrosis. Irish J Med Sci 166:70–71, 1997. 75. Edelman DS. Laparoscopic cholecystectomy under continuous epidural anaesthesia in patients with cystic fibrosis. Am J Dis Child 145:723–724, 1991. 76. Laine L, Cook D. Endoscopic ligation compared with sclerotherapy for treatment of oesophageal variceal bleed. Ann Intern Med 123:280–287, 1995. 77. Tanner MS. Current clinical management of hepatic problems in cystic fibrosis. J R Soc Med 12(suppl):38–43, 1986. 78. Conn HO. Hazards attending the use of esophageal tamponade. N Engl J Med 259:701–707, 1958. 79. Sergent G, Gottrand F, Delemazure O, Ernst O, Bonvarlet P, Mizrahi D, L'Hermine C. Transjugular intrahepatic portosystemic shunt in an infant. PaediatrRadiol 27:588–590, 1997. 80. Mieles LA, Orenstein D, Teperman L, Podesta L, Koneru B, Starzl TE. Liver transplant in cystic fibrosis. Lancet 1:1073, 1989. 81. Mack DR, Traystman MD, Colombo JL. Clinical denouement and mutation analysis of patients with cystic fibrosis undergoing liver transplantation for biliary cirrhosis. J Pediatr 127:881–887, 1995. 82. Rocca GD, Pompei L, Pugliese F, Coccia C, Ruberto F, Montecchi C, Antonini M, Rossi M, Alfani D, Cortesini R, Gasparetto A. Anaesthesia for liver transplantation in cystic fibrosis patients. Eur J Surg 8:278–281, 1998. 83. NobleJamieson G, Barnes N, Jamieson N, Friend P, Calne R. Liver transplantation for hepatic cirrhosis in cystic fibrosis. J R Soc Med 89(suppl):31–37, 1996. 84. Sharp HL. Cystic fibrosis liver disease and transplantation. J Pediatr 127:944–946, 1995. 85. Couetil JP, Houssin DP, Sourbane O, Chevalier PG, Dousset BE, Loulmet D, Achkar A, Tolan MJ, Amrein CI, Guinvarch A. Combined lung and liver transplantation in patients with cystic fibrosis: a 41/2 year experience. J Thorac Cardiovasc Surg 110:1415–1422, 1995. 86. Zabner J, Couture LA, Gregory RJ, Graham SM, Smith AE, Welsh MJ. Adenovirusmediated gene transfer transiently corrects the chloride transport defect in nasal epithelia of patients with cystic fibrosis. Cell 75:207–216, 1993. 87. Grubb BR, Pickles RJ, He H, Yankaskas JR, Vick RN, Engelhardt JF, Wilson JM, Johnson LG, Boucher RC. Inefficient gene transfer by adenovirus vector to cystic fibrosis airway epithelia of mice and humans. Nature 371:802–806, 1994. 88. Grubman SA, Fang SL, Mulberg AE, Perrone RD, Rogers LC, Lee DW, Armentano D, Murray SL, Dorkin HL, Cheng SH. Correction of the cystic fibrosis defect by gene complementation in human intrahepatobiliary cell lines. Gastroenterology 108:584–592, 1995. 89. Yang Y, Raper SE, Cohn JA, Engelhardt JR, Wilson LM. An approach for treating the hepatobiliary disease of cystic fibrosis by somatic gene transfer. Proc Natl Acad Sci USA 90:4601–4605, 1993.
Page 407
18— Selected Advances in Imaging of the Gallbladder and Bile Ducts Matthew Barish and Joseph T. Ferrucci Boston Medical Center, Boston, Massachusetts Michael Blake Beth Israel Deaconess Medical Center, Boston, Massachusetts I— Introduction Imaging of the gallbladder and biliary tree has undergone major changes in the last 5 to 10 years predominantly due to advances in ultrasound (US), computed tomography (CT), and magnetic resonance imaging (MRI). These techniques are now capable of imaging the entire biliary tract noninvasively with accuracy rates approaching or equaling those of invasive techniques such as endoscopic retrograde cholangiopancreatography (ERCP) and percutaneous transhepatic cholangiography (PTC), dramatically changing the approach to the diagnosis of diseases of the biliary system. This chapter reviews the recent advances in imaging of the gallbladder and biliary tract, focusing on the techniques of US, helical CT, and MRI. The role of these various techniques in specific disease entities is discussed. II— Imaging Advances A— Ultrasonography Ultrasound (US) remains one of the primary modalities for imaging the biliary system and is particularly successful in examining the gallbladder. Recent advances in US equipment have improved both spatial and contrast resolution, which have, in turn, increased the radiologist's sensitivity and specificity in the detection and characterization of diseases of the biliary tract. The major innovation in US over the past 10 years has been the introduction of color and power Doppler vascular imaging techniques. Their application has expanded the role of US to permit evaluation of vessel patency—flow direction as well as tissue perfusion—without the need for contrast administration. The use of Doppler imaging in the evaluation of the biliary tree and cholecystitis is discussed in subsequent sections. B— Helical Computed Tomography The introduction of helical CT has improved our ability to image the biliary tract by allowing the rapid acquisition of a volume of data capable of imaging all portions of the ductal system. Previously, nonhelical CT scanners acquired data in separate slices during repetitive breathhold acquisitions, which led to the risk of portions of the biliary tract being excluded from the images. The use of helical CT permits the acquisition of a larger volume of tissue in which
Page 408
the entire biliary tract can be included in a single breathhold period. In addition, the use of a helical technique allows retrospective reconstruction of images with variable slice overlap. This postprocessing of data has led to an increase in the diagnostic accuracy of CT in the evaluation of diseases of the biliary tract. Current techniques for imaging of the biliary tract require the use of spiral acquisition with thin collimation (3 to 5 mm), 20 to 40% reconstruction overlap, pitch of 10 to 1.5, and use of both conventional and narrow imagedisplay window widths. The use of oral and intravenous contrast material depends on the suspected disease entity under study. However, dedicated biliary CT is improved with the use of negative (lowattenuation) contrast agents such as a water or lipidcontaining fluids such as milk. The use of highdensity oral contrast material can create undesired artifacts as well as obscure stones of the distal common bile duct. In addition, scans should be obtained both prior to and following the injection of intravenous contrast. Noncontrast scans are useful to detect both cholelithiasis and choledocholithiasis, which can be obscured by enhancement of the walls of the gallbladder or bile ducts. C— Magnetic Resonance Imaging MRI of the biliary system has undergone a major change due to the recent introduction of noninvasive methods, which provide projectional images of the biliary tree simulating those obtained at ERCP. This technique has been termed magnetic resonance cholangiopancreatography (MRCP), and because of its recent introduction, a review of the technique is warranted. MRCP uses the inherent tissue contrast between fluidfilled structures and the surrounding solid organs to generate images of only the fluidcontaining structures (gallbladder, bile duct, and pancreatic ducts). No exogenous contrast (either by intravenous or direct injection) is necessary and the technique is completely noninvasive (1). The images rendered by MRCP resemble those provided by direct cholangiography obtained through ERCP or PTC. They are projectional images and display ducts as bright against a dark background, which makes them easily understandable (Fig. 1). This has allowed MRCP to become the primary diagnostic imaging modality in several clinical situations. It competes successfully with invasive tests, such as ERCP and PTC, and with more traditional and widely accepted noninvasive imaging techniques, such as CT and US. Besides the wellknown advantages of MRI—such as noninvasiveness and multiplanar capabilities—other elements that have led to the widespread use of MRCP in biliary tract disease include faster sequences with shorter total exam duration, use of highresolution parameters without compromising signaltonoise ratio (SNR), and improved threedimensional rendering techniques. The reasons for the great interest that MRCP has generated among radiologists, gastroenterologists, and surgeons are many. First, highquality images can be obtained with nearly all clinical scanners. Second, images are easy to interpret and agreement between different observers interpreting the same images is moderate to substantial in most cases. Third, the available data demonstrate that MRCP has replaced diagnostic ERCP for many indications, with major implications for patient management (1). Various pulse sequences and different approaches for MRCP have been described. Regardless of the specific sequence used, the principle underlying the technique is the same: on heavily T2weighted images, structures with long T2 relaxation times, such as those containing stationary fluids, exhibit very high signal intensity, while the surrounding organs and tissues show marked decreases in signal intensity. The signal arising from fat can be further decreased with the use of chemically selective fat suppression. The optimal sequence for routine clinical practice varies with different scanners and depends upon the availability of software applications. Most or all techniques for MRCP described acquire images in a coronal or oblique coronal plane (following the usual anatomic arrangement of the extrahepatic bile ducts). Imaging times depend on the sequences used with highresolution imaging, which allow for threedimensional, rotational viewing requiring approximately 6 to 15 min. Alternatively, one can obtain twodimensional projectional images or lowresolution imaging in 2 to 30 s. These rapid
Page 409
Figure 1 Normal MR cholangiogram demonstrating the projectional overview of the entire biliary system provided by the technique. gb, gallbladder; curved arrow, common hepatic duct; arrowhead and arrow, pancreatic duct.
imaging techniques have shown relatively high accuracy in the detection of cholecystolithiasis, choledocholithiasis, and biliary obstruction. III— Disease Entities A— Cholelithiasis 1— Ultrasound The normal appearance of the gallbladder on US is that of an anechoic structure with a hyperechoic, welldefined wall measuring 3 mm or less in thickness. Minor angulation of the transducer or decentering can cause pseudothickening of the gallbladder wall. The shape of the gallbladder can vary because of infolding of its wall. A ''Phrygian cap" deformity is caused by folding of the fundus over its body; occasionally, prominent folds may cause acoustic shadowing mimicking a polyp or a stone. The prone view is useful for demonstrating the mobility of stones, especially those hidden in the neck, and for avoiding shadowing from bowel gas and reverberation artifacts. Occasionally, stones or sludge in the gallbladder can be demonstrated exclusively on prone views (2). Classically, a gallstone appears as an echogenic structure within the gallbladder lumen that causes distal acoustic shadowing and moves with gravity (Fig. 2). Accuracy of up to 96% of sonography in the diagnosis of gallstones has been reported, results improving with advances in technology. (3) Gallstone size and number, however, cannot be estimated accurately by US (4). When calculi completely fill the gallbladder, the demonstration of a wall echo shadow (WES) or doublearc shadow sign, which consists of two parallel hyperechoic lines separated by a thin hypoechoic space and distal acoustic shadowing, is the key to the diagnosis (5). The proximal arc represents the gallbladder wall, the distal arc represents the gallstones' reflections, and the hypoechoic space in between represents either a hypoechoic portion of the wall or a small sliver of bile. Shadowing from the gallbladder fossa may also be caused by porcelain
Page 410
Figure 2 Gallstone demonstrated by sonography. Transverse image of the gallbladder demonstrates echogenic focus (arrow) with distal acoustic shadowing (arrowheads).
gallbladder or by acute emphysematous cholecystitis due to gas in the gallbladder. Focal intramural gas in acute emphysematous cholecystitis is usually associated with reverberations or a comettail artifact. Gascontaining stones are rarely recognized on sonographic examination, even though gas has been noted by computed tomography in 4% of patients with visible stones. The fissures in these calculi are usually arranged in a triradiate pattern. Gascontaining stones may also appear as two parallel linear echoes (doubleecho sign) or have a large, rounded gas bubble surrounded by a rim of solid material (6). Gallstones may be mimicked by intraluminal gas bubbles that cause shadowing. Gas bubbles are usually associated with reverberation or comettail artifacts but may appear simply as echogenic foci. Cholesterol polyps causing subtle shadowing and small gallstones not producing any acoustic shadow can also sometimes cause confusion. Sonographic findings of milk of calcium bile include layering echogenic material with a flat fluidfluid level or a convex meniscus usually associated with acoustic shadowing. Milk of calcium bile may also produce a weak reverberation artifact (7). 2— Computed Tomography CT detects approximately 75% of gallstones due to the differential attenuation between the bile and the stone. Approximately 25% of gallstones are not detected, since they are either
Page 411
isodense to bile or too small to visualize adequately (8,9). Screening for gallstones with CT is not a routine technique because of its high cost, radiation exposure, and low sensitivity. Although not a screening method, the widespread use of CT scanning of the upper abdomen for a variety of indications frequently reveals the incidental presence of gallstones. Recognition and appreciation of the varied appearances of gallstones on CT images (Fig. 3) is therefore necessary and can avoid further imaging. Gallstones can appear as homogeneously hyperdense to bile and are then readily detected (Figure 3c and d). Soft tissue—density stones are of only slightly greater density than bile and can be difficult to visualize without narrow windowwidth settings (Fig. 4). In addition, the variable density of bile in the gallbladder can change the relative densities between calculi and concentrated bile, rendering them isodense. Less frequently, a stone may appear homogeneously hypodense to bile; these stones are felt to represent cholesterol calculi. Stones can have a more complex pattern, with hyperdense rims, varying layers of hypodensity and hyperdensity (laminated) (Fig. 3c and d), central punctate hyperdensity, and even a central lucency containing gas. A proportion of routine intravenously administered iodinated contrast is excreted in bile and can render bile within the gallbladder variably hyperdense. Specific biliary contrast agents that are preferentially excreted in bile exist in both oral and intravenous formulations. After administration of these agents, bile in the gallbladder will become homogeneously hyperdense. This will improve detection of lower density stones (Fig. 5) but may rarely obscure homogeneously hyperdense calculi. These CT biliary contrast agents are discussed in greater detail further on in this chapter. 3— Magnetic Resonance Imaging Gallbladder calculi generally show low signal on T1 and T2weighted MR images due to the lack of mobile protons (Fig. 6). However, most stones show at least small areas of signal on T1 or T2weighted images typically in a central location. The size, distribution, and configuration of these areas of signal result in gallstones with a variety of patterns (Fig. 7) (10). The ability to detect gallstones on MRI depends on the difference in signal between the bile and the stone. On T1weighted sequences, the bile within the gallbladder can vary in signal intensity from uniformly low (watersignal intensity) to uniformly bright (fatsignal intensity) due to variations in lipid and bile salt concentrations in the concentrated bile of the gallbladder. This results in a variable detection rate of gallstones on T1weighted sequences. On T2 weighted sequences, bile will be of high signal intensity, similar to fluid elsewhere in the body. Rarely, sufficiently concentrated bile may appear to be of low signal even on heavily T2weighted sequences. MRCP or similar bright bile sequences are particularly effective in detecting calculi due to the high contrast between bile and stones. MR exceeds CT and US in stone visualization because of this high contrast. B— Cholecystitis 1— Ultrasound of Cholecystitis Findings on grayscale US supportive of a diagnosis of acute cholecystitis but lacking specificity include gallstones, sludge, gallbladder distention, a thickened wall with hypoechoic zones, pericholecystic fluid, and intraluminal membranes (Fig. 8) (11). Gallbladder wall thickening may be seen in other disorders such as anasarca, cirrhosis, pancreatitis, hepatitis, AIDS cholangiopathy, gallbladder carcinoma, and hyperplastic cholecystopathy (Fig. 9). A positive sonographic Murphy's sign, defined as the presence of maximal tenderness elicited by direct pressure of the ultrasound transducer over the visualized gallbladder, has been reported to have a positive predictive value for acute cholecystitis up to 73% (12). The positive predictive value of sonographic Murphy's sign combined with the presence of gallstones is higher and reported to be 77 to 92% (12,13). Sonographic Murphy's sign may be negative in up to 70% of patients with gangrenous cholecystitis or on ventilator support.
Page 412
Figure 3 Various appearances of gallstones on CT. (a) Noncontrast CT showing several calculi with faint central and peripheral calcifications (arrows). (b) Noncontrast CT shows innumerable small calculi of mixed attenuation with punctate central calcification (arrows). Oral dilute barium contrast is seen in the stomach and duodenal bulb. St, stomach; d, duodenum. (c) and (d) Noncontrast CT demonstrating large, laminated gallstone (arrows) on normal windows (c) and wide bone windows (d).
Page 413
Figure 4 Demonstration of calculi at different window settings on CT. (a) Stone with calcified rim in gallbladder (arrow) is barely visible at window level 40 and width 370. d, duodenum. (b) Stone becomes more conspicuous (arrow) at window level 50 and narrower width of 129. d, duodenum.
In recent years the main area of research in the diagnosis of cholecystitis has been related to vascular imaging innovations such as power Doppler imaging. Schiller et al. showed the hyperemic changes in a thickened gallbladder wall to be more sensitive than a sonographic Murphy's sign and abnormal laboratory values (14). A subsequent study showed the sensitivity of power Doppler sonography to be 95%, compared with 33% for color Doppler sonography, in revealing a hypervascularized gallbladder wall. Power Doppler sonography revealed hyperemia within a nonthickened gallbladder wall in some patients with surgically proven acute cholecystitis. Although the sensitivity of power Doppler sonography in diagnosing acute cholecystitis was similar to that of grayscale sonography, the specificity of power Doppler sonography was significantly higher, which substantially improved diagnostic confidence (15). Color Doppler sonography (CDS) of the cystic artery flow in the anterior wall of the gallbladder has also been used to diagnose acute cholecystitis by demonstrating increased color flow in patients with acute cholecystitis (16,17). With normal CDS, the color flow is visible only in the proximal body and neck of the gallbladder and can be seen in up to 40% of healthy persons (16,17). Jeffrey et al. reported increased color flow with visualization of cystic artery length greater than half of the anterior gallbladder wall length in 26% of patients with acute cholecystitis, compared with only 2% of normal gallbladders (18). Even though increased color flow is suggestive of acute cholecystitis, the usefulness of CDS in the diagnosis of acute cholecystitis is limited by its low sensitivity (40%) and low positive predictive value (24%) (16). It is important to be aware that power Doppler sonography is significantly more sensitive than CDS for revealing cystic artery flow in patients with normal gallbladders (19). In this study, power Doppler sonography revealed flow in 73% of patients with normal gallbladders, compared with 53% revealed by CDS. The flow patterns in patients with normal gallbladders
Page 414
Figure 5 Gallbladder calculi seen on CT following intravenously administered cholangiographic contrast. (Courtesy of Jorge Soto.) Stones are seen as multiple filling defects (arrows) within the opacified gallbladder. Note opacification of several intrahepatic bile ducts (arrowheads).
obtained with power Doppler sonography overlapped the flow patterns previously reported as fairly specific criteria for diagnosing acute cholecystitis using CDS (i.e., flow within the distal fundal quartile and flow spanning greater than 50% of the anterior wall). Therefore, different power Doppler sonography criteria are necessary, as CDS criteria are not applicable to the diagnosis of acute cholecystitis with power Doppler sonography (19). Researchers from Paris have reported on color velocity imaging, a color sonographic technique that uses data contained in grayscale Bmode image scan lines, to determine blood flow velocity. Sensitivity, specificity, accuracy, positive predictive value, and negative predictive value of color velocity imaging and power Doppler sonography for revealing acute cholecystitis were 95, 100, 99, 100, and 99%, respectively. The accuracy of color velocity imaging and power Doppler sonography in revealing acute cholecystitis was significantly greater than the accuracy of grayscale sonography (20). If these results are confirmed on further studies, color velocity imaging would be a major breakthrough. Although advances in vascular imaging using Doppler have been reported to improve the accuracy of ultrasound in the diagnosis of cholecystitis, further study is needed before these techniques are fully accepted. The many complications of acute cholecystitis can have varied appearances on US. Empyema with pus in a distended gallbladder may simply resemble echogenic sludge. Several sonographic features can suggest gangrenous cholecystitis, including striated thickening of the wall, intraluminal membranes, marked wall asymmetry, and pericholecystic debrisfilled collections (Fig. 9) (21,22). Sonographic findings in hemorrhagic cholecystitis are similar to those in gangrenous cholecystitis, with which it is often associated. Following gallbladder perforation, pericholecystic fluid collections may appear anechoic or complex. The gallbladder frequently has an irregular indistinct outline; sometimes a focal interruption of the gallbladder wall signifies the site of perforation (23). If acute emphysematous cholecystitis develops, gas in the
Page 415
Figure 6 Cholelithiasis on MRI. (a) Axial image from T1weighted sequence shows foci of reduced signal within the gallbladder representing gallstones (arrow). (b) Axial image from heavily T2weighted sequence show foci of reduced signal within the gallbladder representing gallstones (arrow). d, duodenum; open arrow, common bile duct.
gallbladder lumen typically produces a hyperechoic reflection with reverberations or comettail artifacts within its acoustic shadow. Similarly gas within the wall of the gallbladder is usually seen as a hyperechoic ring around the fluidfilled gallbladder, often with associated reverberations (24). Acute acalculous cholecystitis (AAC) accounts for 5 to 15% of all cases of acute cholecystitis, but clinical diagnosis is difficult. Sludge within a distended gallbladder is nonspecific, but abnormalities more indicative of AAC include diffuse intraluminal echogenicity secondary to pus or hemorrhage, hypoechoic regions within a thickened wall, and a positive sonographic Murphy's sign (25). Chronic cholecystitis is most commonly (90%) associated with gallstones. The longstanding chronic inflammation may cause muscular hypertrophy of the gallbladder wall and a fibrotic reaction leading to dilatation of the RokitanskyAschoff sinuses and hyperplastic cholecystopathy. In appropriate clinical circumstances, sonographic findings of gallbladder wall thickening and gallstones are suggestive even though nonspecific. Chronic acalculous cholecystitis is a poorly understood entity and no single test provides a specific diagnostic sign. Diffuse gallbladder wall thickening and intraluminal nonshadowing echoes may be seen. Biliary scintigraphy may be the most sensitive technique for the diagnosis (26). Sonographic findings of xanthogranulomatous cholecystitis may mimic carcinoma and include marked thickening of the gallbladder wall, which may be irregular and lobulated with hypoechoic nodules, and an indistinct interface between the gallbladder and liver.
Page 416
Figure 7 Variable appearance of gallstones on MRCP. (a) Four consecutive source coronal images from MRCP demonstrate a large filling defect in the gallbladder representing a stone (arrow). (b) Oblique coronal maximumintensity projection (MIP) MRCP demonstrates several rounded filling defects representing gallstones in the dependent portion of the gallbladder (open arrow). (c) Oblique coronal MIP MRCP demonstrates several filling defects in the gallbladder representing gallstones (black arrow). The central stellate high signal represents the MR equivalent of a "Mercedes Benz" sign (white arrow).
Page 417
Figure 8 Acute cholecystitis on US. Longitudinal (a) and transverse (b) sonograms show marked circumferential thickening of the gallbladder wall (arrows) and a pericholecystic complex collection (curved arrows) in a patient with acute cholecystitis and a positive sonographic Murphy's sign.
Page 418
Figure 9 Gallbladder wall thickening in the absence of acute cholecystitis in a 35yearold HIVpositive male. Longitudinal (a) and transverse (b) sonograms of the gallbladder show asymmetrical circumferential thickening of the gallbladder wall (arrows) secondary to HIV infection and a lowalbumin state.
Page 419
2— Computed Tomography of Cholecystitis As in the evaluation of cholecystolithiasis, CT is not the primary method of evaluation of acute cholecystitis. CT is used, however, in the evaluation of patients with unexplained abdominal pain where cholecystitis may be the etiology. Since complicated cholecystitis may present confusing signs, symptoms, and clinical presentations, CT scanning may be the initial imaging modality chosen in these patients with unsuspected gallbladder disease. In one series of complicated cholecystitis by Terrier et al., gallbladder disease was suspected in only 7 of 23 patients (27). It is important, therefore, to recognize and appreciate the appearances of acute cholecystitis on CT images. The CT appearance of acute cholecystitis mimics the ultrasound findings and consists of gallbladder wall thickening (>3 mm) and cholelithiasis (Fig. 10) (28). These findings are nonspecific and gallbladder wall thickening may be seen in multiple other disorders. The lack of an equivalent CT diagnostic criterion to the sonographic Murphy's sign may decrease specificity. CT can visualize the infiltration and inflammation of the pericholecystic fat, however, which is not visible on ultrasound (Fig. 11). Additional CT findings suggestive of cholecystitis include increased density of bile, loss of definition of the gallbladder wall, and dilatation of the gallbladder lumen. As in US, complicated cholecystitis can present an array of findings which, on CT, include hypodense regions in the thickened gallbladder wall, pericholecystic fluid collections, intrahepatic parencymal low attenuation from edema, direct spread of the inflammatory process, or intrahepatic abscesses. Emphysematous cholecystitis is demonstrated on CT as air within the gallbladder wall or lumen (Fig. 12). Air within the gallbladder lumen can also be a sign
Figure 10 Acute cholecystitis demonstrated on CT. (a) Gallstones (arrows) are of almost the same attenuation as the bile in the gallbladder, which has a thickened wall. (b) Low attenuation within the liver superior to gallbladder fundus represents adjacent hepatic inflammation (arrowheads).
Page 420
Figure 11 Acute cholecystitis demonstrated on contrastenhanced CT. There is circumferential thickening of the wall with muscosal enhancement (arrows) and extensive stranding of the pericholecystic fat (curved arrow).
Figure 12 Emphysematous cholecystitis on CT. Consecutive axial CT images show multiple calcified gallstones within the gallbladder (black arrows; a and b), air in the gallbladder lumen (curved arrows; a and b), and air in the gallbladder wall (white arrow; b).
Page 421
of emphysematous cholecystitis; however, intraluminal gas can also be due to prior ERCP, sphincterotomy, biliaryenteric anastomoses, or biliaryenteric fistulae. 3— Magnetic Resonance Imaging of Cholecystitis As in the CT diagnosis of acute cholecystitis, the MR hallmark of the disease is gallbladder wall thickening in the presence of gallstones. In addition, the added tissue and fluid characterization of MR has been used in an attempt to increase the diagnostic accuracy of MR by evaluating the functional capability of the gallbladder to concentrate bile. This technique of evaluating the signal intensity of bile on MR was used to diagnose cholecystitis as early as 1986 (29). The diseased gallbladder is unable to concentrate bile and therefore, on T1weighted images, the gallbladder bile in patients with cholecystitis is isointense or hypointense to hepatic parenchyma, while in normal gallbladders the bile is hyperintense (29,30). Other authors, however, reported no significant difference in T1 and T2 relaxation times between extracted bile (in vitro) from normal gallbladders and those with acute cholecystitis (31). The morphological changes of acute cholecystitis are also well demonstrated by MR imaging. Gallbladder wall thickening is well demonstrated on both T1 and T2 weighted images, while intramural fluid, pericholecystic fluid, and edema are better seen on T2weighted imaged (Fig. 13) (32). Recent advances in MRI have led to more rapid imaging techniques capable of imaging the upper abdomen with T2weighted images in a single breathhold. These
Figure 13 Acute cholecystitis on MRI. T2weighted axial MR image depicting several lowsignal foci representing gallstones (black arrows) and pericholecystic high signal representing surrounding inflammation within the pericholecystic fat of Morrison's pouch (white arrows) and edema within the liver parenchyma (curved arrows).
Page 422
techniques have been applied to the evaluation of cholecystitis by Regan and coworkers, who have reported a sensitivity of 91% and a specificity of 79%. The diagnosis of cholecystitis was based on the presence of pericholecystic high signal and gallbladder calculi (33). In this study, HASTE MRI detected seven patients with choledocholithiasis, while ultrasound visualized the calculi in only five. Since this examination can be performed in less than 10 min, it is possible that MR may compete with US in the evaluation of patients with atypical rightupperquadrant pain in whom either acute cholecystitis or choledocholithiasis with acute cholangitis is suspected. 4— Nuclear Medicine Imaging of Cholecystitis Radionuclide cholescintigraphy was one of the major radiological advances of the 1970s and is of proven value in the diagnosis of acute cholecystitis (34). The radiopharmaceutical is injected intravenously, metabolized by hepatocytes, excreted in bile, and concentrated by the gallbladder. With gamma camera, images of the biliary tree can be obtained by recording the photons emitted from the radionuclide within the bile. Multiple images are obtained over a prolonged period of time (up to 6 h), which demonstrate the distribution of the ''tagged" bile. The principle underlying interpretation of radionuclide cholescintigraphy is that the lack of gallbladder visualization indicates occlusion of the cystic duct, while lack of visualization of the duodenum suggests obstruction of the bile duct. Recent progress has centered on technical and pharmacological developments. The evolution of biliary imaging compounds such as 99mTc disofenin and mebrofenin now allow the depiction of the biliary tree even in patients with a high level of bilirubin (35). Administration of morphine reduces the examination duration and gives functional gallbladder information. Nonvisualization of the gallbladder by 90 min with morphine in an appropriate clinical setting is diagnostic of acute cholecystitis. When the gallbladder is not observed by 60 min but then appears within 60 min following morphine administration, a positive diagnosis of abnormal gallbladder function can be made (36). Morphineenhanced radionuclide cholescintigraphy has also been shown to be useful in the evaluation of critically ill patients for suspected acute cholecystitis, particularly in patients with known risk factors or documented gallstones (37). Administration of morphine thus has many advantages, including a shortened examination time and a reduction in the incidence of falsepositive results. False positives still occur, however, due to chronic cholecystitis, hyperalimentation, or excessive fasting (38). When the gallbladder is observed in a clinical setting of biliary pain, cholecystokinin8 (CCK8) may be infused intravenously for the measurement of the ejection fraction. An ejection fraction value of less than 35% is indicative of calculous or acalculous chronic cholecystitis (36). The excellent direct sonographic visualization of the gallbladder along with the generally greater availability of US has seen scintigraphy supplanted in many centers, but it remains a good technique for the diagnosis of acute cholecystitis. Cystic duct calculi are sometimes difficult to visualize ultrasonographically, so hepatobiliary scintigraphy may be particularly useful to evaluate the functional patency of the cystic duct in patients whose clinical presentation strongly supports stone disease (39). C— Choledocholithiasis 1— Ultrasound of Choledocholithiasis Choledocholithiasis occurs in approximately 15% of patients with gallstones, with the majority of common duct stones originating from the gallbladder (40). In Asia, parasitic infection can be a major cause of intraductal formation of calculi (primary CBD stones). Sonography is usually considered to be the screening method of choice due to its low cost, noninvasiveness, availability, and lack of ionizing radiation. Sensitivity for sonographic detection of CBD stones has improved with the newer higherresolution US machines and has approached 75% in certain studies (41). This high sensitivity depends on high operator skill, meticulous technique, and cooperative patients and is therefore difficult to achieve routinely in clinical practice (Fig. 14a).
Page 423
Figure 14 Choledocholithiasis on US. (a) Sonogram. reveals dilated common bile duct (arrow) measuring 11.2 mm. (cursors), but the obstructing stone was not visualized. (b) Sagittal oblique sonography shows a dilated common bile duct (CBD) with an intraluminal focus of increased echogenicity representing a CBD stone (arrow) (no associated demonstrable shadowing). Portal vein (V) lies posterior to CBD.
Page 424
Calculi are often impacted in the distal CBD and detection may be optimized by using a gastric window after the patient ingests water or scanning transversely with the patient in an erect position. CBD stones appear as hyperechoic structures with distal acoustic shadowing, similar to stones within the gallbladder. Occasionally, distal acoustic shadowing may be difficult to demonstrate (Fig. 14b). It is easier to demonstrate stones in a dilated duct, but in about onethird of cases of CBD calculi, no duct dilatation is seen due to the intermittent nature of stone impaction and obstruction. Several possible interpretative pitfalls exist in the diagnosis of CBD stones. Pancreatic calcification, pneumobilia, surgical clips, and folds in tortuous ducts may all be associated with distal shadowing and can mimic stones. The distal cystic duct insertion or right hepatic artery indentation may also produce an echo within the CBD. The true cause of these mimics of stone disease can usually be recognized by a careful scanning technique and by assessing the patient in different positions. Sludge in the CBD appears as echogenic material without shadowing and occasionally demonstrates layering or a pluglike appearance. Biliary sludge is seen in 10 to 29% of patients following liver transplantation and may lead to biliary obstruction and ascending cholangitis (42). There is considerable interest in new applications of US in assessing the biliary tree. In a study of 65 patients intraoperative US compared favorably with intraoperative cholangiography in terms of untility in exploring bile ducts for stones and with respect to its safety, shorter examination period, and ease of administration. The authors found intraoperative US to be an effective procedure for biliary exploration during laparoscopic cholecystectomy (43). In a larger study of 216 patients with symptomatic cholelithiasis, the main intra and extrahepatic ducts were well documented by laparoscopic US in all patients, but in eight cases the distal tract of the CBD was not well visualized. The authors concluded that laparoscopic ultrasonographic examination may be a real alternative to cholangiography during laparoscopic cholecystectomy, but considerable ultrasonographic experience is required for the examination to be performed successfully (44). 2— Computed Tomography of Choledocholithiasis A— Noncontrast Computed Tomography In patients in whom the diagnosis of choledocholithiasis is the main concern, ultrasonography is the usual screening technique. Direct referral to ERCP may be indicated where there is a high clinical suspicion of ductal stones owing to the high accuracy of ERCP and the ability to extract the stone during the same procedure. However, patients often present with biliary obstructive signs and symptoms without a clear etiology. In these patients, US, CT, and recently MRI have become acceptable screening modalities for the evaluation of the site and etiology of the biliary obstruction. The reported sensitivity of noncontrast CT for the detection of biliary stones varies from 45 to 90% (45–49). More recent reports with thinsection helical CT have shown improved performance. Neitlich et al. demonstrated a sensitivity of 88% with an overall accuracy of 94% for the detection of choledocholithiasis using thinsection breathhold helical CT. Neither hyperdense oral contrast nor intravenous contrast was used, as both of these agents can obscure biliary calculi. In addition, detection of calculi was improved by the use of narrow "bile windows" in which the displaywindow width was set to 150 HU and the level was set to the mean attenuation of bile (50). The appearance of a duct stone is variable and may resemble (a) a calcific density surrounded by a crescent of hypointense biles, (b) a hyperattenuating ring surrounded by hypoattenuating bile, or (c) a soft tissue attenuation focus either surrounded by hypoattenuating bile or with hypoattenuating bile above and below (50). Stones within the biliary tract show the same variable appearance as stones within the gallbladder. Ductal calculi may appear hyperintense or hypointense to bile. Calcific stones are easily seen as hyperintense foci along the course of the bile duct (Fig. 15). When stones have a peripheral rim of calcium, the rim can be difficult to differentiate from the duct wall. This differentiation is even more difficult after
Page 425
Figure 15 Choledocholithiasis evaluated with CT and MRI. a to c. Axial CT (a), T1weighted MRI (b), and T2weighted MRI at the same level. (a) Axial CT showing calcified calculus (arrow) within the gallbladder. Small stone in the common bile duct (CBD) is difficult to visualize (curved arrow). (b) T1 weighted axial MR image shows a high signal gallbladder calculus (arrow) and a stone in the common bile duct (curved arrow). (c) T2weighted axial MR images shows a lowsignal filling defect in the dependent portion of the gallbladder representing the calculus (arrow) and the CBD calculus (curved arrow). (d) T2weighted axial MR image shows a CBD calculus (arrow). (e) Source coronal image from MR cholangiopancreatogram (MRCP) demonstrates a filling defect in the CBD representing the stone (curved arrow). (f) Oblique coronal maximumintensity projection (MIP) MRCP clearly demonstrates the CBD calculus (curved arrow). (g) CT demonstrates a calcified calculus (arrow) in the CBD. (h) T1weighted axial MR image shows the stone in the distal CBD (curved arrow). (i) Source coronal image from MRCP demonstrates the calculus impacted in the distal CBD (curved arrow).
the administration of intravenous contrast, which produces enhancement of the bile duct wall simulating a hyperdense calcified rim of an intraductal stone. Cholesterol stones can appear isointense to bile and therefore may be difficult or impossible to detect with noncontrast helical CT (50). The bile in the ducts is not as concentrated as the bile in the gallbladder; and therefore. hypointense CBD stones are less common. B— Computed Tomography Following Cholangiographic Contrast Agents In an effort to improve the accuracy of helical CT in the detection of common duct stones, multiple investigators have proposed the use of exogenously administered contrast agents that are preferentially excreted in bile (51,52). Several intravenous agents exist that include meglumine salts, such as iodoxamate, iotroxamate, ioglycamate, and iododipamide. These agents are slowly infused over 45 min, conjugated by the liver, and excreted in bile, resulting in the opacification of the biliary tract approximately 30 min after the end of infusion. This technique, known as CT cholangiography, attempts to improve the visualization of calculi by increasing the attenuation of bile to a level greater than that of biliary stones. The resultant images would have
Page 426
Figure 15 Continued.
Page 427
Figure 15 Continued.
the greatest contrast differential for hypoattenuating stones such as cholesterol stones (Fig. 16). The resultant axial images can be reviewed as in conventional CT interpretation or the helical CT data set can be subjected to a variety of postprocessing tools such as maximumintensity pixel projection (MIP) or shaded surface displays (SSDs). These techniques yield projectional images similar to ERCP or threedimensional images capable of fully demonstrating the complex biliary anatomy. In a study by Stockberger et al., bile duct stones were detected with a
Page 428
Figure 16 Calculi on CT following an orally administered cholangiographic contrast agent. (Courtesy of Jorge Soto.) Stones are seen as multiple filling defects within the opacified gallbladder (arrows) and CBD (curved arrow).
sensitivity of 86%, a specificity of 100%, and an accuracy of 94% (51). There was one stone that was isodense to the enhanced bile and was not detected by the technique. The intravenous agent produced poor opacification of the biliary tree in 75% of patients, in whom the serum bilirubin levels were elevated above 2 mg/dL (34 mmol/L). Intravenous CT cholangiography is not likely to obtain widespread use for many reasons, which include the poor biliary excretion of the contrast agent in patients with elevated bilirubin levels, lack of FDA approval of these agents in the United States, and the reported incidence of severe reactions, including death (53). Orally administered cholangiographic contrast agents are also being used to opacify the biliary tract for CT cholangiography. Soto used 6 g of orally administered iopanoate ingested 12 h prior to helical CT scanning (54). This agent is generally considered safe and is well tolerated by most patients. Adequate opacification of the biliary tract was achieved in 27 of 29 patients. Sensitivity, specificity, and accuracy for choledocholithiasis were 93, 100, and 97% respectively. Common duct opacification was further improved by administering a fatty meal to evacuate the gallbladder (54). The use of orally administered cholangiographic contrast agents as part of the routine preparation for abdominal CT in patients with suspected biliary disease still awaits further evaluation. Because of the ease of administration, lack of significant adverse reactions, and wide availability of these agents, their routine use may become standard practice in the future. The major limitations of the technique are the lack of sufficient biliary excretion of the agent with elevated bilirubin levels and the theoretical risk of masking hyperdense calculi by increasing the attenuation of bile to equal that of the stone. 3— Magnetic Resonance Imaging/Magnetic Resonance Cholangiopancreatography of Choledocholithiasis The high contrast resolution of MRI allows the detection of CBD stones without the need for injection of contrast material or invasive techniques such as ERCP or PTC. MRI techniques
Page 429
for the detection of biliary stone disease capitalize on the inherent contrast between highsignal bile and the low signal of nearly all types of stones. Routine axial T2 weighted images are capable of demonstrating intraductal stones as lowsignal filling defects usually seen on the dependent wall (Figs. 17 to 20). The major limitation of routine axial imaging is the large number of slices necessary to cover the entire biliary tree. This results in prolonged scan times, possible gaps between slices, degradation of image quality by motion artifact, and loss of spatial resolution. Recent use of coronal acquisitions and the addition of fast imaging techniques have allowed MRI to produce highcontrast images capable of demonstrating the entire pancreaticobiliary tree. The resultant coronal images can be reviewed as individual source images, or the volumetric data can be subjected to a variety of postprocessing tools such as maximumintensity pixel projection (MIP) or shaded surface displays (SSD). These techniques yield projectional images similar to ERCP or threedimensional images that better demonstrate the complex biliary anatomy. These dedicated MR sequences and the postprocessing techniques are termed MR cholangiopancreatography. MRCP has allowed the detection of bile duct stones with high accuracy, nearing or equaling ERCP. Recent experience (55–58) has demonstrated sensitivities for the diagnosis of choledocholithiasis approaching 98%, when stones 4 mm or greater in size are considered. The higher SNR afforded by phasedarray coils allows imaging with highresolution parameters, thereby improving detection of small stones (59). Sensitivity for the diagnosis of choledocholithiasis by MRCP is certainly superior to that of US (20 to 75%) (41) and CT (45 to 85%) (60,61). The interpretation of MRCP studies for detecting intraductal stones demands a meticulous review of the source images, since in the MIP reconstructions the foci of low signal (the typical appearance of stones on MRCP) can be obscured by the very high signal intensity of surrounding bile. The importance of careful review of these source images in confirming the presence of stones cannot be overemphasized. Another potential error is misdiagnosing a stone for another type of intraluminal filling defect, such as an intraductal tumor. The recent acceptance of laparoscopic cholecystectomy as the procedure of choice for the management of patients with cholelithiasis has resulted in a decrease in postoperative
Figure 17 Choledocholithiasis on MRI. Signal voids throughout the CBD representing multiple stones (arrow) on oblique coronal maximumintensity projection (MIP) MR cholangiopancreatogram. GB, gallbladder; Du, duodenum.
Page 430
Figure 18 Choledocholithiasis on MRI. a and b. Signal void in distal common bile duct (CBD) representing a CBD stone (arrow) on oblique coronal maximumintensity projection MR cholangiopancreatogram (MRCP) and coronal source image from MRCP.
recovery time and hospital stay (62). The role of MRCP in the preoperative diagnosis of choledocholithiasis is complicated, especially since minimally invasive laparoscopic methods to remove bile duct calculi have also emerged. Approximately 10 to 15% of patients who undergo cholecystectomy have CBD calculi (63), and currently the preoperative evaluation of patients almost always includes US and measurement of serum liver function tests. These studies however lack sufficient sensitivity and the incidence of CBD stones unsuspected on preoperative investigations but discovered at time of intraoperative cholangiography (IOC) ranges from 3 to 5% (64). Liver function tests also lack specificity, since only 33% of patients with abnormal results have choledocholithiasis (65). This lack of specificity results in a 40 to 70% incidence of negative ERCP examinations for CBD stones (66). Since MRCP has been shown to have high sensitivity and specificity for choledocholithiasis, MRCP could limit preoperative ERCP to therapy for those patients with stones. ERCP is a technically challenging procedure. Reported rates of failure of ductal cannulation vary between 3 and 10% (67–69), the frequency clearly being determined by the experience and expertise of the endoscopist. Although many failures are due to inexperience, several anatomical factors such as periampullary diverticula and duodenal stenosis (68,69) may be responsible for difficulties in duct cannulation. When the ducts are not cannulated with conventional ERCP, a precut papillotomy with a needleknife is often required. The incidence and severity of complications following precut papillotomy are much higher than those after conventional diagnostic ERCP. The reported rate of postprocedure pancreatitis and other complications for this needleknife procedure (6 to 15%) (67,70) is substantially higher than the 1 to 5% for conventional diagnostic ERCP (67,71,72). Other available options, such as PTC and surgery, are also associated with significant morbidity. In a study that specifically addressed
Page 431
Figure 19 Choledocholithiasis on MRI. (a) Two consecutive source coronal images from MRCP demonstrate multiple filling defects in the common bile duct (CBD) representing calculi (arrows and curved arrow). (b) Corresponding oblique coronal maximumintensity projection MR cholangiopancreatogram again demonstrates filling defects in the CBD representing calculi (arrow and curved arrow).
Figure 20 T2weighted axial MR images shows a lowsignal filling defect in the dependent portion of the gallbladder (GB), representing the gallbladder calculus (arrow), and a further common bile duct (CBD) lowsignal focus representing the CBD calculus (curved arrow).
Page 432
this issue, MRCP was shown to be useful for evaluating patients who have had a failed ERCP attempt, regardless of the reason for failure, and that it can be helpful in guiding patient management (73). The failure rate of ERCP in patients who have had a previous biliaryenteric anastomosis or gastroenteric drainage procedure such as Bilroth II has been shown to be considerably higher than in intact patients, with reported figures varying between 10 and 48% (74–76). In patients with Bilroth II anatomy, the length of the afferent limb is the most important factor determining the ability to reach the periampullary region (76). In this group of patients, MRCP will demonstrate the site of the biliary enteric anastomosis as well as the status of the intrahepatic ducts and can therefore help determine which patients may benefit from antegrade duct cannulation with a drainage procedure or a possible balloon dilatation of a stenotic anastomosis (73,77). Even though the high sensitivity and specificity for MRCP in the diagnosis of CBD stones is established, the precise role of MRCP in the workup of patients with suspected choledocholithiasis has not yet been defined. Many patients are referred for ERCP because of high suspicion for stones on the basis of clinical history, symptoms, results of liver function tests, or finding on US or CT. Routine use of MRCP for confirmation of the presence of stones prior to ERCP is difficult to justify in this era of cost awareness (78,79), since most of these patients will need to be treated endoscopically with stone extraction immediately following cholangiographic diagnosis. MRCP is clearly indicated when the diagnosis is being entertained and the CBD cannot be cannulated on ERCP, since definitive therapy usually requires more invasive intervention, such as precut papillotomy, antegrade cholangiography with stone extraction, or surgical exploration. In these cases, MRCP will increase diagnostic certainty, thereby limiting these therapeutic procedures to those patients who will truly benefit from them (73). MRCP may also be indicated when clinical suspicion is only moderate and a primary diagnostic ERCP is difficult to justify. IV— Conclusion Recent advances in ultrasound, CT, and MRI have changed and will continue to change the diagnostic evaluation of patients with biliary stone disease. As these noninvasive methods are refined and improved, they will compete with more invasive diagnostic examinations such as PTC and ERCP. Currently, MRCP has demonstrated accuracy rates equivalent to ERCP for the detection of common duct stones and accuracy rates surpassing ultrasound for the detection of gallstones. It seems prudent to surmise that in the future, invasive diagnostic techniques with their higher rates of complications, higher costs, and lower availability will be replaced by noninvasive imaging techniques. Therapy will remain, for now at least, the domain of invasive techniques. References 1. Barish MA, Soto JA. MR Cholangiopancreatography: techniques and clinical applications. AJR 1997; 5:1295–1303. 2. Parulekar SG. Evaluation of the prone view for cholecystosonography. J Ultrasound Med 1986; 5:617. 3. Zeman R. Cholelithiasis and cholecystitis. In: Gore RM, Levine MS, Laufer I, eds. Gastrointestinal Radiology. Philadelphia: Saunders, 1994, p 1657. 4. Brakel K, Lameris JS, Nijs HG, et al. Accuracy of ultrasound and oral cholecystography in assessing the number and size of gallstones: implications for non surgical therapy. Br J Radiol 1992; 65:779. 5. MacDonald FR, Cooperberg, PL, Cohen MM. The WES triad: a specific sonographic sign of gallstones in the contracted gallbladder. Gastrointest Radiol 1981; 6:39.
Page 433
6. Mitchell DG, Needleman L, Frauenhoffer S, et al. Gas containing gallstones: the sonographic ''double echo sign." J Ultrasound Med 1988; 7:39. 7. Parulekar SG. Ultrasonic detection of calcification in gallstones: "the reverberation shadow." J Ultrasound Med 1984; 3:123. 8. Baron RL, Rohrmann CA Jr, Le SP, Shuman WP, Teefey SA. CT evaluation of gallstones in vitro: correlation with chemical analysis. AJR 1988; 151:1123–1128. 9. Barakos JA, Ralis PW, Lapin SA, et al. Cholelithiasis: evaluation with CT. Radiology 1987; 162:415–418. 10. Baron RL, Shuman WP, Lee SP, Rohrman CA, Golden RN, Richards TL, Richardson L, Nelson JA. MR appearance of gallstones in vitro at 1.5 T: correlation with chemical composition. AJR 1989; 153:497–502. 11. Laing FC, Federle MP, Jeffrey RB, et al. Ultrasonic evaluation of patients with acute right upper quadrant pain. Radiology 1981; 140:449. 12. Bree RL. Further observations on the usefulness of the sonographic Murphy sign in the evaluation of suspected acute cholecystitis. JCU J Clin Ultrasound 1995; 23:169. 13. Ralls PW, Colletti PM, Lapin SA, et al. Realtime sonography in suspected acute cholecystitis. Radiology 1985; 155:767. 14. Schiller VL, Turner RR, Sarti DA. Color Doppler imaging of the gallbladder wall in acute cholecystitis. Abdom Imaging 1996 May–Jun; (3):233–237. 15. Uggowitzer M, Kugler C, Schramayer G, et al. Sonography of acute cholecystitis: comparison of color and power Doppler sonography in detecting a hypervascularized gallbladder wall. AJR 1997; 168(3):707–712. 16. Parulekar SG, Hillier SA, Adell JA, et al. Color Doppler sonography of the gallbladder wall. J Ultrasound Med 1995; 14(suppl):S21. 17. Paulson EK, Kliewer MA, Hertzberg BS, et al. Diagnosis of acute cholecystitis with color Doppler sonography: significance of arterial flow in thickened gallbladder wall. AJR 1994; 162:1105. 18. Jeffrey RB Jr, NinoMurcia M, Ralls PW, et al. Color Doppler sonography of the cystic artery: comparison of normal controls and patients with acute cholecystitis. J Ultrasound Med 1995; 14:33. 19. Olcott EW, Brooke Jeffrey R, Jain KA. Power versus color Doppler sonography of the normal cystic artery: implications for patients with acute cholecystitis. AJR 1997; 168:703–705. 20. Soyer P, Brouland JP, Boudiaf M, et al. Color velocity imaging and power Doppler sonography of the gallbladder wall: a new look at sonographic diagnosis of acute cholecystitis. AJR 1998; 171:183–188. 21. Teefey SA, Baron RL, Radke HM, et al. Gangrenous cholecystitis: new observations on sonography. J Ultrasound Med 1991; 10:603. 22. Teefey SA, Baron RL, Bigler SA. Sonography of the gallbladder: the significance of striated (layered) thickening of the gallbladder wall. AJR 1991; 156:945. 23. Chau WK, Wong KB, Chan SC, et al. Ultrasonic hole sign: a reliable sign of perforation of the gallbladder. J Clin Ultrasound 1992; 20:294. 24. Bloom RA, Libson E, Lebensart PD, et al. The ultrasound spectrum of emphysematous cholecystitis. J Clin Ultrasound 1989; 17:251. 25. Cornwell EE III, Rodriquez A, Mirvis SE, et al. Acute acalculous cholecystitis in critically injured patients: preoperative diagnostic imaging. Ann Surg 1989; 210:52. 26. Raptopoulos V, Compton CC, Doherty P, et al. Chronic acalculous gallbladder disease: multiimaging evaluation with clinicalpathologic correlation. AJR 1986; 147:721. 27. Terrier F, Becker CD, Stoller C, Triller JK. Computed tomography in complicated cholecystitis. J Comput Assist Tomogr 1984; 8:58–62. 28. Kane RA, Costello P, Duszlak E. Computed tomography in acute cholecystitis: new observations. AJR 1983; 141:697–701. 29. McCarthy S, Hricak H, Cohen M, Fisher MR, Winkler ML, Filly RA, Margulis AR. Cholecystitis: detection with MR imaging. Radiology 1986; 158:333–336.
Page 434
30. Pu Y, Yamamoto F, Igimi H, et al. A comparative study of usefulness of magnetic resonance imaging in the diagnosis of acute cholecystitis. J Gastroenterol 1994; 14:192–198. 31. Loflin TG, Simeone JF, Mueller PR, et al. Gallbladder bile in cholecystitis: in vitro MR evaluation. Radiology 1985; 157:457–459. 32. Weissleder R, Stark D, Compton CC, Simeone JF, Ferrucci JT. Cholecystitis: diagnosis by MR imaging. Magn Reson Imaging 1988; 6:345–348. 33. Regan F, Schaefer DC, Smith DP, Petronis JD, Bohlman ME, Magnuson TH. The diagnostic utility of HASTE MRI in the evaluation of acute cholecystitis; half Fourier acquisition single shot turbo SE. J Comput Assist Tomogr 1998; 22(4):638–642. 34. Ralls PW, Colletti PM, Halls JM, Siemsen JK. Prospective evaluation of 99mTcIDA cholescintigraphy and gray scale US in the diagnosis of acute cholecystitis. Radiology 1982; 144:369–371. 35. Krishnamurthy S, Krishnamurthy K. Quantitative assessment of hepatobiliary diseases with 99mTcIDA scintigraphy. In: Nuclear Medicine Annual. New York: Raven Press, 1988, pp 309–313. 36. Krishnamurthy S, Krishnamurthy K. Cholecystokinin and morphine pharmacological intervention during 99mTcHIDA cholescintigraphy: a rational approach. Semin Nucl Med 1996; 26(1):16–24. 37. Flancbaum L, Choban PS. Use of morphine cholescintigraphy in the diagnosis of acute cholecystitis in critically ill patients. Intens Care Med 1995; 21(2):120– 124. 38. Kim EE, Moon TY, Delpassand ES, Podoloff DA, Haynie TP. Nuclear hepatobiliary imaging. Radiol Clin North Am 1993; 31:923–33. 39. Zemen RK, Garra BS. Gallbladder imaging, the state of the art. Gastroenterol Clin North Am 1991; 2:127–156. 40. Laing FC. Ultrasound diagnosis of choledocholithiasis. Semin Ultrasound CT MR 1987; 8:103. 41. Dong B, Chen M. Improved sonographic visualization of choledocholithiasis. J Clin Ultrasound 1987; 15:185. 42. Barton P, Maier A, Steininger R, et al. Biliary sludge after liver transplantation: 1. Imaging findings and efficacy of various imaging procedures. AJR 1995; 164:859. 43. Ohtani T, Kawai C, Shirai Y, Kawakami, Yoshida K, Hatakeyama K. Intraoperative ultrasonography versus cholangiography during laparoscopic cholecystectomy: a prospective comparative study. J Am Coll Surg 1997; 185(3):274–282. 44. Santambrogio R, Montorosi M, Bianchi P, et al. Common bile duct exploration and laparoscopic cholecystectomy: role of intraoperative ultrasonography. J Am Coll Surg 1997; 185(1):40–48. 45. Baron RL, Stanley RJ, Lee JKT, Koehler RE, Melson GL, Balfe DM, Weyman PJ. A prospective comparison of the evaluation of biliary obstruction using computed tomography and ultrasonography. Radiology 1982; 145:91–98. 46. Baron RL, Stanley RJ, Lee JKT, Koehler RE, Leavitt RG. Computed tomographic features of biliary obstruction. AJR 1983; 140:1173–1178. 47. Baron RL. Common bile duct stones: reassessment of criteria for CT diagnosis. Radiology 1987; 162:419–424. 48. Jeffrey RB Jr, Federle MP, Laing FC, Wall S, Rego J, et al. Computed tomography of choledocholithiasis. AJR 1983; 140:1179–183. 49. Mitchell SE, Clark RA. Comparison of computed tomography and sonography in choledocholithiasis. AJR 1984; 142:729–732. 50. Neitlich JD, Topazian M, Smith R, Gupta A, Burrell MI, Rosenfield AT. Detection of choledocholithiasis: comparison of unenhanced helical CT and endoscopic retrograde cholangiopancreatography. Radiology 1997; 203:753–757. 51. Stockberger SM, Waas JL, Sherman S, Lehman GA, Kopecky KK. Intravenous cholan giography with helical CT: comparison with endoscopic retrograde cholangiopancreatography. Radiology 1994; 192:675–680.
Page 435
52. Van Beers BE, Lacrosse M, Trigaux JP, de Cannière L, De Ronde T, Pringot J. Noninvasive imaging of the biliary tree before or after laparoscopic cholecystectomy: use of threedimensional spiral CT cholangiography. AJR 1994; 162:1331–1335. 53. Goldberg HJ. Helical cholangiography: complementary or substitute study for endoscopic retrograde cholangiography? Radiology 1994; 192:615–616. 54. Soto JA, Velez SM, Guzman J. Choledocholithiasis: diagnosis with oralcontrastenhanced CT cholangiography. AJR 1999; 172:943–948. 55. Macaulay SE, Schulte SJ, Sekijima JH, et al. Evaluation of a nonbreath hold MR cholangiography technique. Radiology 1995; 196:227–232. 56. Guibaud L, Bret PM, Reinhold C, Atri M, Barkun AN. Diagnosis of choledocholithiasis: value of MR cholangiography. AJR 1994; 163:847–850. 57. Guibaud L, Bret PM, Reinhold C, Atri M, Barkun AN. Bile duct obstruction and choledocholithiasis: diagnosis with MR cholangiography. Radiology 1995; 197:109–115. 58. Soto JA, Barish MA, Yucel EK, Siegenberg D, Ferrucci JT, Chuttani R. Magnetic resonance cholangiography: comparison to endoscopic retrograde cholangiopancreatography. Gastroenterology 1996; 110:589–597. 59. Reinhold C, Taourel P, Bret PM, Barkun AN, Atri M. MR cholangiography of choledocholithiasis by using a multicoil array and highresolution imaging parameters (abstr). Radiology 1995; 197(P):342. 60. Jeffrey RB, Federle MP, Laing FC, et al. Computed tomography of choledocholithiasis. AJR 1983; 140:1179–1183. 61. Baron RL, Stanley RJ, Lee JKT, et al. Computed tomographic features of biliary obstruction. AJR 1983; 140:1173–178. 62. Gallstones and laparoscopic cholecystectomy. NIH Consensus Statement 1992; 10(3):1–26. 63. Fink AS. Commentary: controversies in the management of common duct calculi. Surg Clin North Am 1994; 74(4):949–950. 64. Tompkins RK, Pitt HA. Surgical management of benign lesions of the common bile ducts. Curr Probl Surg 1982; 19:327–98. 65. Cranley B, Logan H. Exploration of the common bile duct: the relevance of the clinical picture and the importance of preoperative cholangiography. Br J Surg 1980; 67:869–872. 66. Philips EH. Controversies in the management of common duct calculi. Surg Clin North Am 1994; 74(4):931–948. 67. Bilbao MK, Dotter CT, Lee TG, Katon RM. Complications of endoscopic retrograde cholangiopancreatography (ERCP). A study of 10,000 cases. Gastroenterology 1976; 70:314–320. 68. Silvis ES, Ansel HJ. Endoscopic retrograde cholangiography: application in biliary tract disease. In: Berk JE, ed. Bockus' Gastroenterology. Philadelphia: Saunders, 1985, pp 3569–3579. 69. Rieger R, Wayand W. Yield of prospective, noninvasive evaluation of the common bile duct combined with selective ERCP/sphincterotomy in 1930 consecutive laparoscopic cholecystectomy patients. Gastrointest Endosc 1995; 2(1):6–12. 70. Dowsett JF, Polydorou AA, Vaira D, et al. Needle knife papillotomy: how safe and how effective? Gut 1990; 31:905–08. 71. Hamilton I, Lintott DJ, Rothwell J, Axon ATR. Acute pancreatitis following endoscopic retrograde cholangiopancreatography. Clin Radiol 1983; 34:543–546. 72. Thoeni RF, Fel SC, Goldberg HI. CI detection of asymptomatic pancreatitis following ERCP. Gastrointest Radiol 1990; 15:291–295. 73. Soto JA, Yucel EK, Barish MA, Chuttani R, Ferrucci JT. MR Cholangiopancreatography after unsuccessful or incomplete ERCP. Radiology 1996; 199:91–98. 74. Forbes A, Cotton PB. ERCP and sphincterotomy after Billroth II gastrectomy. Gut 1984; 25:971–974.
Page 436
75. Osnes M, Rosseland AR, Aabakken L. Endoscopic retrograde cholangiography and endoscopic papillotomy in patients with a previous BillrothII resection. Gut 1986; 27:1193–1198. 76. Cunningham JT. Endoscopic papillotomy and stent insertion: BII technique and limitations. In: Barkin JS, O'Phelan CA, eds. Advanced Therapeutic Endoscopy. New York: Raven Press, 1990, pp 193–200. 77. Pavone P, Laghi A, Catalano C, Broglia L, Messina A, Passariello R. Threedimensional MR cholangiography: a unique diagnostic procedure for evaluation of bile ducts in patients with biliaryenteric anastomosis. Radiology 1995; 197(P):312. 78. Outwater EK, Gordon SJ. Imaging the pancreatic and biliary ducts with MR. Radiology 1994; 192:19–21. 79. McDermott VG, Nelson RC. Re: MR cholangiopancreatography: efficacy of threedimensional turbo spinecho technique. AJR 1995; 165:301–302.
Page 437
19— Endoscopic Ultrasound of the Gallbladder and Bile Ducts Brian R. Stotland Boston University School of Medicine, Boston, Massachusetts I— Introduction Endoscopic ultrasound (EUS) is a relatively new imaging modality that provides detailed images of structures both within the wall of the luminal gastrointestinal tract and in adjacent areas. By incorporating an ultrasonic transducer into the tip of an endoscope or through the use of probes passed through the working channel of an endoscope, ultrasound can be performed from within the body cavity. The advantage of this approach over conventional, transcutaneous ultrasound is that a higher frequency of ultrasound may be employed. Higherfrequency ultrasound provides greater image resolution, but with less depth of penetration. In EUS, this limitation is circumvented by placing the transducer in close proximity to the structure of interest. Thus, by using an ultrasound frequency of between 7.5 to 20.0 MHz, structures as small as 2 mm can be distinguished. EUS can also be performed by passing an ultrasound probe through the working channel of a standard endoscope. While there has been a virtual explosion of publications on the use of EUS in the past decade, comparatively few publications have evaluated its role in biliary tract disease. This chapter reviews the use of this exciting modality and suggests its practical role in clinical practice. II— Technique Most echoendoscopes have oblique viewing optics; the esophagus is intubated and the scope is advanced in a fashion similar to the passage of a duodenoscope for endoscopic retrograde pancreatography (ERCP). Standard EUS imaging of the gallbladder and bile ducts is performed by placing the transducer into the duodenum or gastric antrum. From within the descending duodenum, the intrapancreatic common bile duct (CBD) is identified adjacent and parallel to the pancreatic duct. At this level, particularly if the lumen is filled with water, the ampulla itself can be identified. As the scope is withdrawn into the duodenal bulb, the bile duct can be traced to the CBD, but in general the hilum is less well visualized and the right and left biliary ducts are difficult to identify. The cystic can be identified coming and into and then out of view alongside the CBD and can sometimes be traced back into the gallbladder. The gallbladder can be readily identified from within the duodenal bulb or gastric antrum (Fig. 1). The wall normally appears threelayered with a hypoechoic center corresponding to the muscularis pro
Page 438
Figure 1 Endoscopic ultrasound view of the normal gallbladder, as imaged from within the duodenal bulb.
pria, bordered by hypoechoic layers on either side. The procedure is contraindicated in patients who cannot tolerate conscious sedation or who are at risk for perforation from an endoscopy. The presence of a duodenal diverticulum may obstruct some fields of EUS imaging. In patients who have a biliary or pancreatic stent in place, attention must be given on echoendoscope withdrawal to avoid inadvertently dislodging the stent. III— Evaluation for Gallstones It is well established that transabdominal ultrasound (TUS) should be the first imaging modality chosen when evaluating for the presence of stones within the gallbladder. However, TUS accuracy is less than 100% even in the best of hands (1). There is evidence that EUS may be able to detect cholelithiasis when TUS is normal, particularly in obese patients (2). In addition, EUS may be valuable in evaluating for the presence of microlithiasis. Biliary microlithiasis has been shown to be a cause of pancreatitis and perhaps biliary pain due to cholecystitis and is not detectable by TUS. In a study by Dill et al. (3), 66 patients with biliarytype pain and negative TUS underwent EUS. At operation, 61 of the patients had cholecystitis documented histologically, and 58 of these had preoperative EUS evidence of stones or sludge. When combined with stimulated biliary drainage for bile analysis, EUS had a 92% sensitivity and 100% predictive value; 90.5% of patients with preoperative EUS findings had resolution of biliary pain. In choledocholithiasis, common bile duct (CBD) stones can be visualized with TUS in 70 to 90% of cases, depending upon where in the CBD the stone is lodged (Fig. 2) (4–6). Overlying bowel gas or obesity may impair visualization of the distal CBD. EUS appears to be a more sensitive means of detecting choledocholithiasis and may be comparable to endoscopic retrograde cholangiography (ERC) without the associated risk of pancreatitis (7). A
Page 439
Figure 2 EUS view of the distal common bile duct containing two small stones (arrowheads). (From Ref. 8.)
prospective series of 155 patients found EUS sensitivity for CBD stones to be 96%, significantly better than TUS (63%) or computed tomography (CT) (71%) (8). In a study by Prat et al. (9) comparing EUS to ERC in the evaluation of choledocholithiasis in 119 patients, the results were EUS sensitivity 93%, specificity 97%, positive predictive value 98%, and negative predictive value 88%. Corresponding values for ERC were 89, 100, 100, and 83%. Canto et al. (10) prospectively compared EUS and ERC for the diagnosis of choledocholithiasis in 64 patients. EUS had a 94% accuracy, compared with 97% for ERC. In this study, TUS and CT had only a 77% accuracy. Overall, complications were significantly less frequently related to EUS (1.6%) compared with ERC (12.5%). In this study, a strategy of performing ERC only if the EUS exam detected stones would cost $754 per patient, compared with $1109 per patient if ERC were performed as the initial evaluation. In a large retrospective study by Palazzo et al. (11), EUS had a sensitivity of 94.9%, a specificity of 97.8%, and an accuracy of 95.9%. All stones detected by ERC had been detected by EUS. These studies suggest that in experienced centers, EUS may be the only precholecystectomy procedure necessary in patients with a low to medium risk of choledocholithiasis, thus avoiding the risk of ERC. However, in patients with CBD stones detected by TUS or CT, EUS is unnecessary. Such patients are usually best managed with ERC. Magnetic resonance cholangiography (MRC) is emerging in parallel to EUS as a noninvasive means for detecting CBD stones. Its accuracy ranges from 89 to 94% (12,13). A prospective study comparing EUS to MRC in 32 patients reported accuracies of 96.9% for EUS and 81.2% for MRC (not statistically different) (14). Larger prospective studies are needed. An alternative approach to EUS evaluation for choledocholithiasis was reported by Wiersema et al. (15). They reported successful EUSguided needle puncture of the distal CBD and contrast ductography in 8 of 11 patients. While EUSguided pancreatography has also been
Page 440
reported, these techniques are considered purely experimental. However, because the procedure would not be expected to induce pancreatitis as a complication, it offers a theoretical advantage over ERC. IV— Neoplastic Lesions of the Biliary Tree Gallbladder carcinoma and cholangiocarcinoma are rare lesions and are generally associated with a poor prognosis—though, as with other gastrointestinal malignancies, survival depends upon stage (Tables 1 to 3). They are frequently difficult to image with TUS or CT. On EUS, gallbladder carcinoma appears as a hypoechoic mass that obliterates the normal walllayer pattern. However, gallbladder stones may prevent proper tumor staging in many cases. In addition, in may be difficult to distinguish adenomyomatosis from carcinoma. Mitake et al. (16) reported an accuracy of 77% for T stage and 90% for N stage, but only when gallstones did not obstruct visualization of the lesion. Mizuguchi et al. (17) reported—in a study evaluating 25 patients with various gallbladder lesions (seven cancers)—that EUS was superior to magnetic resonance imaging (MRI), CT, and TUS for preoperative cancer diagnosis. Carcinoma of the bile duct appears as a hypoechoic mass that may extend along the duct, obstruct the lumen, or invade surrounding structures. These lesions are sometimes surrounded by fibrotic tissue, which appears hyperechoic on EUS. Tio et al. (18), in a study of 76 patients, reported an 83% Tstage accuracy for CBD tumors and an 85% accuracy for Klatskin and common hepatic duct tumors. However, most other authors report difficulty in visualizing the proximal biliary tree with EUS. The development of endosonographic probes capable of cannulating the CBD may improve staging ability. These transducers operate at high frequencies (20– 30 MHz), allowing for an axial resolution of 0.03 mm, but with a limited depth of penetration. Theoretically, these probes will improve staging accuracy of proximal biliary lesions by introducing the probe into the malignant stricture. Using an intraductal probe, Yasuda et al. (19) reported a Tstaging accuracy of 83%, compared with 75% for EUS in 18 lesions. Percutaneous intraductal biliary ultrasound staging of cholangiocarcinoma has also been described (20). However, experience with intraductal sonography is limited, and the clinical utility is not yet proven. In addition, it is not anticipated that it will allow the differentiation of an early Table 1 Gallbladder TNM Staging Tx
Primary tumor cannot be assessed
T0
No evidence of primary tumor
Tis
Carcinoma in situ
T1
Tumor invades lamina propria
T2
Tumor invades perimuscular connective tissue
T3
Tumor perforates the serosa or directly invades an adjacent organ
T4
Tumor extends more than 2 cm into liver or into 2 or more adjacent organs
NX
Regional lymph nodes cannot be assessed
N0
No regional lymph node metastasis
N1
Metastasis in cystic duct, pericholedochal or hilar nodes
N2
Metastasis in peripancreatic, periduodenal, periportal, celiac or superior mesenteric lymph nodes
Mx
Distant metastasis cannot be assessed
M0
No distant metastasis
M1
Distant metastasis
Source: Adapted from Ref. 32.
Page 441 Table 2 Extrahepatic Duct TNM Staging Tx
Primary tumor cannot be assessed
T0
No evidence of primary tumor
Tis
Carcinoma in situ
T1
Tumor invades subepithelial connective tissue or fibromuscular layer
T2
Tumor invades perifibromuscular connective tissue
T3
Tumor invades adjacent structures: liver, pancreas, duodenum, gallbladder, colon, stomach
NX
Regional lymph nodes cannot be assessed
N0
No regional lymph node metastasis
N1
Metastasis in cystic duct, pericholedochal or hilar nodes
N2
Metastasis in peripancreatic, periduodenal, periportal, celiac, superior mesenteric or posterior pancreaticoduodenal lymph nodes
Mx
Distant metastasis cannot be assessed
M0
No distant metastasis
M1
Distant metastasis
Source: Adapted from Ref. 32.
stage cholangiocarcinoma from a benign stricture. Publication of large series demonstrating efficacy on these uncommon conditions will require multicenter studies. Due to the development of obstructive jaundice, ampullary carcinoma often presents early in its course, resulting in a relatively good prognosis. With EUS, the mass appears as hyperechoic; it may invade into the duodenal wall, pancreatic parenchyma, or adjacent structures. T1 lesions cannot be differentiated from an ampullary adenoma (Fig. 3). EUS Tstage accuracy ranges from 80 to 90% (21,22). Staging accuracy is in general lower for T1 and T4 lesions. Nstaging accuracy tends to be lower (55 to 70%). However, it is anticipated that when clinically relevant, EUSguided fineneedleaspiration techniques will improve N staging. In a retrospective series, Quirk et al. (23) reported both a sensitivity and specificity of 83% for distinguishing local disease (T2N0, or less) from advanced disease. Because patients with local disease could be treated with a local surgical resection rather than a Whipple resection, there Table 3 Ampulla of Vater TNM Staging Tx
Primary tumor cannot be assessed
T0
No evidence of primary tumor
Tis
Carcinoma in situ
T1
Tumor limited to the ampulla of Vater or sphincter of Oddi
T2
Tumor invades duodenal wall
T3
Tumor invades 2 cm or less into the pancreas
T4
Tumor invades more than 2 cm into pancreas and/or adjacent organs
NX
Regional lymph nodes cannot be assessed
N0
No regional lymph node metastasis
N1
Regional lymph node metastasis
Mx
Distant metastasis cannot be assessed
M0
No distant metastasis
M1
Distant metastasis
Source: Adapted from Ref. 32.
Page 442
Figure 3 A 3cm ampullary adenoma imaged by EUS. The lesion does not appear to invade the muscularis propria (MP); this was confirmed by surgical resection.
was a significant cost savings. However, not all studies have demonstrated EUS reliability. Cahen et al. (24) reported only a 44% accuracy for depth of invasion in 23 patients. Menzel et al. (25) found that the accuracy (88.9%) of intraductal ultrasound staging was superior to conventional EUS for ampullary tumors and also reported a low EUS accuracy (56.3%). A recent study demonstrated that EUS was more accurate than either CT or MRI, and also indicated that the presence of a transpapillary endobiliary stent reduced EUS Tstage accuracy (from 84 to 72%) (26). Since many patients with ampullary tumors are treated initially with biliary stents if they are jaundiced, the results suggest that EUS staging should be performed first. Discrepancies reported between studies on the accuracy of ampullary staging might be due to differences in technique, operator experience, or sample size. It is worth noting that endoscopic pinch biopsies from ampullary lesions are known to be subject to significant sampling error, such that pathological evidence of adenoma does not rule out the presence of invasive carcinoma. Therefore, EUS is suggested prior to endoscopic or local surgical excision to rule out the presence of invasive disease that would require a more aggressive approach. In the past several years, multiple studies have demonstrated that the technique of EUSguided realtime fineneedle aspiration (FNA) is a reliable, accurate, and safe procedure. It allows tissue sampling of small lesions that are often undetectable or unreachable by other radiographic means. The sensitivity of this technique ranges from 89 to 100% in experienced hands (27). It is particularly useful for obtaining tissue from the retroperitoneum. Nstaging accuracy is generally lower using EUS, in part due to the relative inability to distinguish benign inflammatory nodes from malignant adenopathy. Although there is limited experience specifically pertaining to the biliary tract, experience in other organs suggests that EUSguided FNA should improve N staging, resulting in greater overall staging accuracy.
Page 443
V— Other Benign Conditions EUS may be helpful to evaluate other abnormalities of the biliary tree. Cholecystitis appears as nonspecific gallbladder wall thickening. Mitake et al. (28) demonstrated that EUS visualization of the CBD and pancreatic ducts can detect anomalous connections between the biliary and pancreatic ducts, which may predispose patients to cholangitis and cholangiocarcinoma. Tanno et al. (29) reported that a thickened inner hypoechoic layer of the gallbladder indicated the presence of an anomalous pancreaticobiliary ductal communication. However, this finding is not specific. Anecdotal reports have indicated that EUS may be able to diagnose gallbladder adhesions (30) and choledochal cysts (31). VI— Conclusion EUS is emerging as a useful imaging modality in the management of biliary tract disease. Given its reliability in diagnosing choledocholithiasis, EUS may the best test when there is an intermediate probability of CBD stones. Larger studies prospectively comparing MRC to EUS are needed to define the roles of these modalities in this setting. Patients with clinically obvious CBD stones are best managed by ERC. Given the ability of EUS to also diagnose neoplasms of the bile duct, ampulla, gallbladder, and pancreas, it is ideally suited to diagnose and stage obstructive jaundice. Refinement of intraductal ultrasound techniques and use of EUSguided FNA for suspicious adenopathy should enhance staging accuracy. References 1. Peikin S, Feld R, Kastenberg D, et al. Role of endosonography in the diagnosis of gallstone disease in obese patients (abstr). Gastroenterology 1992; 104:A328. 2. Dill J, Callis J, Hill S, Evans P. Combined endoscopic ultrasound and endoscopic MeltzerLyon testing in the diagnosis of cholelithiasis. Am J Gastroenterol 1994; 89(6):956–957. 3. Dill JE, Hill S, Callis J, Berkhouse L, Evans P, Martin D, Palmer ST. Combined endoscopic ultrasound and stimulated biliary drainage in cholecystitis and microlithiasis—diagnosis and outcomes. Endoscopy 1995; 27:424–427. 4. Laing FC, Jeffrey RB, Wing VW. Improved visualization of choledocholithiasis by sonography. Am J Radiol 1982; 143:949–952. 5. Laing FC. Ultrasound diagnosis of choledocholithiasis. Semin Ultrasound CT MR. 1987; 8:103–113. 6. Cronan JJ. US diagnosis of choledocholithiasis: a reappraisal. Radiology 1986; 161:133–134. 7. Amouyal PG, Amouyal P, Levy P, et al. Diagnosis of choledocholithiasis by ultrasonography. Gastroenterology 1994; 106:1062–1067. 8. Sugiyama M, Atomi Y. Endoscopic ultrasonography for diagnosing choledocholithiasis: a prospective comparative study with ultrasonography and computed tomography. Gastrointest Endosc 1997; 45:143–146. 9. Prat F, Amoural PG, Amouyal P, et al. Prospective controlled study of endoscopic ultrasonography and endoscopic retrograde cholangiography in patients with suspected commonbile duct lithiasis. Lancet 1996; 347:75–79. 10. Canto MI, Chak A, Stellato T, Sivak MV. Endoscopic ultrasonography versus cholangiography for the diagnosis of choledocholithiasis. Gastrointest Endosc 1998; 47:439–448. 11. Palazzo L, Girollet PP, Salmeron M, Silvain C, Roseau G, Canard JM, Chaussade S, Coutieer D, Paolaggi JA. Value of endoscopic ultrasonography in the diagnosis of com
Page 444
mon bile duct stones: comparison with surgical exploration and ERCP. Gastrointest Endosc 1995; 42:225–231. 12. Soto JA, Barish MA, Yucel EK, Siegenberg D, Ferrucci JT, Chuttani R. Magnetic resonance cholangiography: comparison with endoscopic retrograde cholangiography. Gastroenterology 1996; 110:589–597. 13. Chan YL, Chan ACW, Lam WWM, Lee DWH, Chung SSC, Sung JJY, et al. Choledocholithiasis: comparison or MR cholangiography and endoscopic retrograde cholangiography. Radiology 1996; 200:85–89. 14. de Ledinghen V, Lecesne R, Raymond JM, Gense V, Amouretti M, Drouillard J, Couzigou P, Silvain C. Diagnosis of choledocholithiasis: EUS or magnetic resonance cholangiography? A prospective controlled study. Gastrointest Endosc 1999; 49:26–31. 15. Wiersema MJ, Sandusky D, Carr R, Wiersema LM, Erdel WC, Frederick PK. Endosonographyguided cholangiopancreatography. Gastrointest Endosc 1996; 43:102–106. 16. Mitake M, Nakazawa Y, Naitoh et al. Endoscopic ultrasonography in diagnosis of the extent of gallbladder carcinoma. Gastrointest Endosc 1990; 36:562–566. 17. Mizuguchi M, Kudo S, Fukahori T, Matsuo Y, Miyazaki K, Tokunaga O, Koyama T, Fujimoto K. Endoscopic ultrasonography for demonstrating loss of multiplelayer pattern of the thickened gallbladder wall in the preoperative diagnosis of gallbladder cancer. Eur Radiol 1997; 7:1323–1327. 18. Tio TLJ, Cheng OB, Wijers et al. Endosonographic TNM staging of extrahepatic bile duct cancer: comparison with pathological staging. Gastroenterology 1991; 100:1351–1361. 19. Yasuda K, Mukai H, Nakajima M, Kawai K. Clinical application of ultrasonic probes in the biliary and pancreatic duct. Endoscopy 1992; 24(suppl 1):370–375. 20. Duda SH, Huppert PE, Schott U, Brambs HJ, Claussen CD. Percutaneous transhepatic intraductal biliary sonography for lymph node staging at 12.5 MHz in malignant bile duct obstruction: work in progress. Cardiovasc Intervent Radiol 1997; 20:133–138. 21. Dancygier H, Lightdale CJ, Stevens PD. Endoscopic ultrasonography of the upper gastrointestinal tract and colon. In: Dancygier H, Lightdale CJ. Endosonography in Gastroenterology. Stuttgart, Germany: Thieme, 199:13–174. 22. Tio TL, Sie LH, Kallimanis G, Luiken GJHM, Kimmings AN, Huibregtse K, Tytgat GNJ. Staging of ampullary and pancreatic carcinoma: comparison between endosonography and surgery. Gastrointest Endosc 1996; 44:706–713. 23. Quirk DM, Rattner DW, Fernandezdel Castillo C, Warshaw AL, Brugge WR. The use of endoscopic ultrasonography to reduce the cost of treating ampullary tumors. Gastrointest Endosc 1997; 46:334–337. 24. Cahen DL, Fockens P, DeWit LTH, Offerhaus GJA, Obertop H, Gouma DJ. Local resection or pancreaticoduodenectomy for villous adenoma of the ampulla of Vater diagnosed before operation. Br J Surg 1997; 84:948–951. 25. Menzel J, Hoepffner N, Sulkowski U, Reimer P, Heinecke A, Poremba C, Domschke W. Polypoid tumors of the major duodenal papilla: preoperative staging with intraductal US, EUS, and CT—a prospective, histopathologically controlled study. Gastrointest Endosc 1999; 49:349–357. 26. Cannon ME, Carpenter SL, Elta GH, Nostrant TT, Kochman ML, Ginsberg GG, Stotland BR, Rosato EF, Morris JB, Eckhauser F, Scheiman JM. EUS compared with CT, magnetic resonance imaging, and angiography and, the influence of biliary stenting on staging accuracy of ampullary neoplasms. Gastrointest Endosc 1999; 50:27–33. 27. Stotland BR, Kochman ML. Diagnostic and therapeutic endoscopic ultrasonography: endoscopic ultrasoundguided fineneedle aspiration in clinical practice. Gastrointest Endosc 1997; 45:329–331. 28. Mitake M, Nakazawa S, Naitoh Y et al. Endoscopic ultrasonography in the detection of anomalous connections of the pancreatobiliary duct. Endoscopy 1991; 23:117–120. 29. Tanno S, Obara T, Maguchi H, Mizukami Y, Shudo R, Fujii T, Takahashi K, Nishino N, Arisato S, Saitoh Y, Ura H, Kohgo Y. Thickened inner hypoechoic layer of the gallbladder
Page 445
wall in the diagnosis of anomalous pancreaticobiliary ductal union with endosonography. Gastrointest Endosc 1997; 46:520–526. 30. Dill JE, Henretta TR, Berkhouse L. Endoscopic ultrasound in the diagnosis of gallbladder adhesions. Am J Gastroenterol 1995; 90:855–856. 31. Avunduk C, Weiss R, Hampf F, Navab F. Obstructing choledochocele: diagnosis by endoscopic ultrasound. Abdom Imaging 1995; 20:72–74. 32. AJCC. AJCC Cancer Staging Manual. Philadelphia: Lippincott Raven, 1997.
Page 447
20— The Silent Gallstone William R. Brugge Massachusetts General Hospital, Boston, Massachusetts I— Introduction The vast majority of patients with gallstones have no symptoms. In the past, the management of these patients has been controversial because little was known about the natural history, risk factors for development of symptoms, and how disease states modify the risks. Recently, a large number of studies have focused on these questions. Only with a complete understanding of the natural history of gallstones can we determine whether a therapy can modify the development of symptoms and complications. These questions are increasingly important as a large number of lowrisk treatments become available. II— Diagnosis The diagnosis of gallstones is usually made with abdominal ultrasonography, and this approach is ideal for identifying patients with asymptomatic gallstones. Large populations can be screened with ultrasonography and the prevalence of stones in a population can be determined. In addition, the ultrasound exams can be repeated over time and the true incidence of stones, including asymptomatic stones, can also be determined (Table 1). Although it may be difficult to correlate the presence of symptoms and the finding of stones in a large population, these type of studies are critical for investigating the factors responsible for the development of gallstone symptoms. III— Prevalence The prevalence of asymptomatic gallstones can be examined with necropsies or with screening ultrasonography. Necropsy studies for asymptomatic gallstones are subject to bias and have largely been replaced with ultrasonography studies. Although the presence of gallstones is relatively easy to establish, determining the presence or absence of biliary symptoms in a large population is difficult. In large Italian studies of gallstone patients, biliary colic has been defined as pain located in the epigastrium or right upper quadrant lasting more than 30 min (1). Using this definition, approximately 85% of screened volunteers in Italy have no biliary symptoms and the prevalence of silent stones in a combined cohort of 29,739 people from several villages was 6.5% (men) and 10.5% (women) (2).
Page 448 Table 1 Prevalence and Incidence of Gallstones in Large Populations
Population studied
Number of subjects
Author/Ref.
British
1,896 Heaton (17)
2/11.5
Hispanics in the United States
2,299 Maurer (6)
7.2/23.2 (Mexican Americans)
Italians
1,962 Misciagria (8)
Obese Romanians Italian villages Italians Cirrhotics Indigent Mexicans Chronic renal failure
157
Acalovschi (9)
29,739 Attili (2) 4,751 Attili (7)
Annual incidence (%)
Prevalence (%) male/females
9.7
17
31
23/28
23.3 14.1
12.9
93
—
2.6 6.5/10.5
3,505 Gonzalez (3) Korzets (5)
—
Del Olmo (4)
313
Symptoms (%)
3.4
8
The prevalence of asymptomatic stones in other populations have been examined, but without the attention to the symptomatic state of the subjects with gallstones. In 3505 lowincome subjects screened in Mexico City, 5.8% of men and 19.7% of women had gallstones (3). The prevalence of stones in select highrisk groups has also been studied using ultrasound. In Spain, the prevalence of asymptomatic stones in patients with cirrhosis was found to be 23.3%, compared to a prevalence in the general population of 16.8% (4). Chronic renal failure patients have also been examined and were found to have a lower prevalence—12.9%—not significantly greater than that in the general population (5). In the United States, specific ethnic groups have a higher prevalence. The Pima Indians are the best studied and have an impressive prevalence of 70% by the age of 25. More recently, Hispanics have been studied with screening ultrasonography (6). Mexican Americans were found to have the highest prevalence, ranging from 7.2% in men to 23.2% in women to 44% in older women. The prevalence in Mexican Americans was 1.7 and 1.8 times greater than in Cuban Americans and Puerto Ricans. The risk factors for the development of silent stones are probably similar to the risk factors for stones in general. In men, the significant risk factors are age and diabetes. In women, age, obesity, and parity are significant risk factors. However, the use of these risk factors and others (family history, use of oral contraceptives) underestimates the risk of gallstones by twoto threefold (7). IV— Incidence The ability to repeat ultrasound studies over time in a cohort population made it possible to determine the yearly incidence of gallstone development. In a small town in southern Italy, nearly 2000 people have been followed annually by ultrasonography (8). The annual incidence of stone formation was found to be 9.7 per 1000, and several risk factors were identified, including age, body mass index, weight change, diabetes, use of laxatives, smoking, consumption of oil and fat, and parity. Recently, this type of study has been performed in obese subjects followed annually with ultrasonography (9). The annual incidence was found to be 26 per 1000 and very few of the subjects had symptoms (9). Disease states can also be studied; in cirrhotics, the annual incidence was found to be 34 per 1000 (4). No longterm incidence study has been performed to examine the change in incidence over time, but preliminary studies from Europe suggest that there may be a recent decline (10). Similar trends may be appearing in the United
Page 449
States. Although cholecystectomy rates in the United States are increasing for the elderly, the rates are decreasing for the young (11). V— Symptoms The symptoms that arise in association with gallstones are often nonspecific. Nausea, bloating, and irregular bowl movements are the least specific. Colic or transient upper abdominal pain after eating is the most predictive of gallstones. The presence of gallstones may be associated with no symptoms, unrelated symptoms, or biliary colic. Jorgensen has reported on the correlation between symptoms and the presence of stones (12). He found that subjects with incidental stones had symptoms that were indistinguishable from those of the population as a whole. Furthermore, there was no correlation between the size, number, and motility of stones and the presence of symptoms. However, Diehl et al. have noted that pancreatitis may occur more frequently in the presence of stones with diameters of <5 mm, when there are more than 20 stones present in gallbladder, and when the stones have a mulberry shape (13). Biliary colictype pain is more predictive for the presence of gallstones than nausea, flatulence, fat intolerance, and a large number of other symptoms (14). Diehl and coworkers have found that specific features of abdominal pain were more suggestive of gallstones, such as pain with radiation into the back lasting more than 3 h (15). However, the symptom response to cholecystectomy suggests that perhaps more than biliarytype pain improved with cholecystectomy. Gui et al. found that bloating, dyspepsia, heartburn, fat intolerance, nausea, and vomiting improved after cholecystectomy (16). Others, using a large metanalysis, have disputed that gallstones may be associated with vague symptoms (14). The factors that determine whether gallstones cause symptoms have not been well described. However, several epidemiological observations have been made, including the possibility that there are two discrete subpopulations, one with symptoms and one comprising those who will never develop symptoms (17). Biliarytype pain as a symptom of gallstones occurs more frequently in women than in men, whether the pain is mild or severe (7). This has been confirmed by Heaton and may explain the higher cholecystectomy rates in women (17). There is also evidence that obesity and lack of exercise are independent risk factors for the development of symptoms related to gallstones (9,18). It has been speculated that in 34% of cases, the development of symptoms related to gallstones could be prevented by a daily 30min period of endurancetype training. The mechanism of this effect is unknown, but exercise may decrease the lithogenicity of bile or alter the motility of the gallbladder. VI— Natural History An understanding of the natural history of asymptomatic gallstones is critical when treatment for gallstones is recommended. The prophylactic treatment of gallstones can be advocated only if the treatment can reduce the risk of symptoms, complications (cholecystitis or obstruction), and additional costs. Unfortunately, it is very difficult to perform the necessary natural history studies, since they involve large numbers of healthy subjects who must be observed over many years. Previous observational studies of gallstone patients were retrospective and subject to bias. Ralston and Smith reported an annual incidence rate of 29% for the development of biliary pain and complications in gallstone patients followed for 15 to 30 years (19). McSherry and coworkers also performed a retrospective chart review in HMO patients with gallstones and found that the complication rate was considerably lower—7% over 5 years (20). Gracie and Ransohoff performed the first prospective observation study of asymptomatic gallstones by studying faculty members at the University of Michigan (21) (Table 2). The rate
Page 450 Table 2 Rate of Symptom Development in Subjects with Gallstones
Author/Ref. Gracie (21)
123
McSherry (20)
135
Percent with symptoms at 2 years
Number of subjects
—
Percent with symptoms at 5 years
Percent with symptoms at 10 years
Percent with complications at 5 years
10
15
<2
10
—
7
1,911
Thistle (24)
193
31
—
—
17 at 2 years
Attili (23)
151
11.9
16.5 @ 4 years
25.8
3 at 10 years
32
25 @ 3.5 years
—
—
1/8
123
—
18
30
5
Friedman (25)
16
Barbara (22)
Zubler (27)
17
(cholecystectomy)
of symptom development was markedly lower than that in previous studies, only 1 to 2% each year, and the complication rate was less than 2%. Overall, only 20% of the patients developed symptoms over approximately 20 years. However, the population studied was unique and comprised Caucasian professional men with ready access to medical care. Larger studies of gallstone subjects in a diverse population have been performed in Italy. The Sirmione study was the first of many investigations in Italy that determined the prevalence rate of gallstones and, with followup, the rate of symptom development (22). In the village of Sirmione, 22% of 132 subjects found to have gallstones during a screening exam had biliary symptoms. When the remaining asymptomatic subjects were followed for 10 years, 16% developed biliary symptoms. The development of symptoms leveled off after 5 years, so that the percentage of the population with symptoms remained approximately the same over 10 years. Similar studies have also been performed by GREPCO (Rome Group for Epidemiology and Prevalence of Cholelithiasis) (23). They followed 118 asymptomatic subjects with gallstones and determined the rate of biliary colic, complications, cholecystectomy, and death at 2year intervals. They found the rate of development of biliary symptoms to be greater than previous studies. There was a nearly constant annual incidence rate of 2 to 3%, and this resulted in 30% of the population having symptoms at 30 years, with a 3% complication rate at 10 years. Although it is difficult to investigate and follow asymptomatic gallstone subjects in the United States, three studies have attempted to determine the rate of symptom development. The original study was based on an observation by Thistle in the National Cooperative Gallstone Study (NCGS) (24). By following gallstone patients (divided into two groups: asymptomatic and symptomatic patients) for 24 months, the frequency of biliary symptoms could be compared. The symptomatic patients had a high rate of symptoms—68% at 2 years—and the asymptomatic patients had a markedly lower rate—31% at 2 years. The rate of symptom development in the NCGS was higher than in most other investigations, probably because it was more than an observational study. Unfortunately, there was no longterm followup. However, Friedman et al., using a retrospective study of HMO gallstone patients in the United States found that the rate of symptom development was similar: 18% at 5 years and 30% at 10 years, with an annual complication rate of 1% (25). Women were more likely to become symptomatic than men and obese subjects were more likely to have a complication. Diabetics did not seem to have a significantly different natural history in terms of symptoms or complications (26). Last, Zubler and colleagues, using a retrospective study, examined the symptom and complication rate of gallstone patients in the rural United States (27). Thirtytwo patients were followed at 5year intervals and 25% developed symptoms at an average interval of 3.5 years. The progression of symptom development from asymptomatic to nonspecific symptoms to biliary colic usually occurs in a stepwise progression prior to a complication such as acute
Page 451
cholecystitis. It is rare for acute cholecystitis to develop without antecedent symptoms. The stepwise progression of symptoms allows physicians to prevent complications of gallstones by performing cholecystectomy in symptomatic patients. VII— Treatment The treatment approach to patients with asymptomatic gallstones has not been well studied. Clearly, there is little role for routine prophylactic cholecystectomy in all patients, since this would be prohibitively expensive and would not improve survival. However, there might be highrisk groups that could potentially benefit. The classic example is the sickle cell anemia patient with recurrent abdominal pain and gallstones. Since it is difficult to differentiate between sickle cell crisis and acute cholecystitis, some advocate prophylactic cholecystectomy for patients with sickle cell anemia (28). Another highrisk group are American Indians, who have not only a high incidence of gallstones but also an increased risk of death from gallstones and gastrointestinal malignancies (29). A prophylactic cholecystectomy at an early age might prevent the complications of gallstone disease and gallbladder cancer, but this has not been documented. Last, Graham has suggested that transplant patients— who are at greater risk of infections, malignancies, and druginduced hepatobiliary diseases—should be considered for early cholecystectomy (30). Obviously, the approach can be individualized for specific conditions. Higherrisk patients—for example, an obese patient with other medical problems who has poor access to health care facilities—might be considered for prophylactic treatment (Table 3). Since it is difficult to perform largescale studies of various treatments such as prophylactic cholecystectomy or stone dissolution in asymptomatic patients, the other approach is to perform decision analysis (31). Ransohoff has examined the life expectancy of gallstone patients with various interventions at various ages. For example, if a prophylactic cholecystectomy were performed on a 50yearold woman, her life expectancy would be reduced by an average of 12 days. In no scenario was there an improvement in the life expectancy when a prophylactic cholecystectomy was used. An incidental cholecystectomy is another approach to the treatment of asymptomatic patients with gallstones. Incidental cholecystectomy is performed during abdominal surgery for another indication. Although good trials are not available, a morbidly obese patient undergoing a laparotomy for gastric stapling should probably have a cholecystectomy. Similarly, an incidental cholecystectomy has been advocated in patients with porcelain gallbladders or large stones because of the increased risk of gallbladder cancer (32). Incidental cholecystectomies can also be considered for patients that are increased risk for the development of gallstones, as in patients with chronic hemolysis (33). Some have suggested that an incidental cholecystectomy should be performed for patients free of gallstones who undergo laparotomy for another indication, but the postoperative complication rate may be slightly increased (34). Table 3 Possible Indications for Prophylactic or Incidental Cholecystectomy Sickle cell anemia American Indians Transplant patients Morbid obesity
Page 452
References 1. Capocaccia L and the GREPCO. Clinical symptoms and gallstone disease: lessons from a population study. In: Capocaccia L, Ricci G, Angelico F, Angelico M, Attili AF, eds. Epidemiology and Prevention of Gallstone Disease. Lancaster, UK: MTP Press, 1984, pp 153–157. 2. Attili AF, Carulli N, Roda E, Barbara B, Capocaccia L, Menotti A, Okoliksanyi L, Ricci G, Capocaccia R, Festi D, et al. Epidemiology of gallstone disease in Italy: prevalence data of the Multicenter Italian study on cholelithiasis. Am J Epidemiol 141:158–165, 1995. 3. Gonzalez Villalpando C, Rivera Martinez D, Arredondo Perez B, Martinez Diaz S, Gonzalez Villalpando ME, Haffner SM, Stern MP. High prevalence of cholelithiasis in a low income Mexican population: an ultrasonographic survey. Arch Med Res 28:543–547, 1997. 4. Del Olmo JA, Garcia F, Serra M, Maldonado L, Rodrigo JM. Prevalence and incidence of gallstones in liver cirrhosis. Scand J Gastroenterol 32:1061–1065, 1997. 5. Korzets Z, Golan E, BenChitrit S, Schneider N, Chagnac A, Carel R, Bernheim J. Prevalence of cholelithiasis in nondiabetic hemodialysis and continuous ambulatory peritoneal dialysis patients. Nephrology 78:44–47, 1998. 6. Maurer KR, Everhart JE, Ezzati TM, Johannes RS, Knowler WC, Larson DL, Sanders R, Shawker TH, Roth HP. Prevalence of gallstone disease in Hispanic populations in the United States. Gastroenterology 96:487–492, 1989. 7. Attili AF, Pazzi P, Galeazzi R. Prevalence of previously undiagnosed gallstones in a population with multiple risk factors. Dig Dis Sci 40:1770–1774, 1995. 8. Misciagna G, Leoci C, Guerra V, Chiloiro M, Elba S, Petruzzi J, Mossa A, Noviello MR, Coviello A, Minutolo MC, Mangini V, Messa C, Cavallini A, DeMichele G, Giorgio I. Epidemiology of cholelithiasis in southern Italy: Part II. risk factors. Eur J Gastroenterol Hepatol 8:585–593, 1996. 9. Acalovschi MV, Blendea D, Pascu M, Georoceanu A, Badea RI, Prelipceanu M. Risk of asymptomatic and symptomatic gallstones in moderately obese women: a longtidunal followup study. Am J Gastroenterol 92:127–131, 1997. 10. Norrby S, Fagerberg G, Sjodahl R. Decreasing incidence of gallstone disease in a defined Swedish population. Scand J Gastroenterol 21:158–162, 1986. 11. Diehl AK. Trends in cholecystectomy rates in the United States. Lancet 2:683, 1987. 12. Jorgensen T. Abdominal symptoms and gallstone disease: an epidemiological investigation. Hepatology 9:856–860, 1989. 13. Diehl AK, Holleman DR Jr, Chapman JB, Schwesinger WH, Kurtin WE. Gallstone Size and risk of pancreatitis. Arch Intern Med 157:1674–1678, 1997. 14. Kraagg N, Thijs C, Knipschild P. Dyspepsia—how noisy are gallstones? A metaanalysis of epidemiologic studies of biliary pain, dyspeptic symptoms, and food intolerance. Scand J Gastroenterol 30:411–421, 1995. 15. Diehl AK, Sugarek NJ, Todd KH. Clinical evaluation for gallstone disease: usefulness of symptoms and signs in diagnosis. Am J Med 89:29–33, 1990. 16. Gui GP, Cheruvu CV, West N, Sivaniah K, Fiennes AG. Is cholecystectomy effective treatment for symptomatic gallstones? Clinical outcome after longterm followup. Ann R Coll Surg Engl 80:25–32, 1998. 17. Heaton KW, Braddon FE, Mountford RA, Hughes AO, Emmett PM. Symptomatic and silent gallstones in the community. Gut 32:316–320, 1991. 18. Lietzmann MF, Giovannucci EL, Rimm EB, Stampfer MJ, Spiegelman D, Wing AL, Willett WC. The relation of physical activity to risk for symptomatic gallstone disease in men. Ann Intern Med 128:417–425, 1998. 19. Ralston DE, Smith LA. The natural history of cholelithiasis: a 15 to 30year followup of 116 patients. Minn Med 48:327–332, 1965.
Page 453
20. McSherry CK, Ferstenberg H, Calhoun WF, Lahman E, Virshup M. The natural history of diagnosed gallstone disease in symptomatic and asymptomatic patients. Ann Surg 202: 59–63, 1987. 21. Gracie WA, Ransohoff DF. The natural history of gallstones: the innocent gallstone is not a myth. N Engl J Med 307:798–800, 1982. 22. Barbara L, Sama C, Morselli Labate AM, et al. A population study on the prevalence of gallstone disease: the Sirmione Study. Hepatol 7:913–917, 1987. 23. Attili AF, De Santis A, Repice AM, Maselli S, GREPCO Group. The natural history of gallstones: the GREPCO experience. Hepatology 21:656–660, 1995. 24. Thistle JL, Cleary PA, Lachin JM, Tyor MP, Hersh T, the steering committee of the National Cooperative Gallstone Study Group. The natural history of cholelithiasis: the national cooperative gallstone study. Ann Intern Med 101:171–175, 1984. 25. Friedman GD, Raviola CA, Fireman B. Prognosis of gallstones with mild or no symptoms: 25 years of followup in a health maintenance organization. J Clin Epidemiol 42: 127–136, 1989. 26. Del Favero G, Caroli A, Meggiato T, Volpi A, Scalone P, Puglisi A, DiMario F. Natural history of gallstones in noninsulindependent diabetes mellitus. A prospective 5year followup. Dig Dis Sci 39:1704–1707, 1994. 27. Zubler J, Markowski G, Yale S, Graham R, Rosenthal TC. Natural history of asymptomatic gallstones in family practice office practices. Arch Fam Med 7:230– 233, 1998. 28. Ware RE, Kinney TR, Casey JR, Pappas TN, Meyers WC. Laparoscopic cholecystectomy in young patients with sickle hemoglobinopathies. J Pediatr 120:58– 61, 1992. 29. Grimaldi CH, Nelson RG, Pettitt PH, Knowler WC. Increased mortality with gallstone disease: results of a 20year populationbased survey in Pima Indians. Ann Intern Med 118:185–190, 1993. 30. Graham SM, Flower JL, Schweitzer E, Bartlett ST, Imbembo Al. The utility of prophylactic cholecystectomy in transplant candidates. Am J Surg 169:44–48, 1995. 31. Ransohoff DF, Gracie WA, Wolfensen LB, Neuhauser D. Prophylactic cholecystectomy or expectant management for silent gallstones. Ann Intern Med 99:199– 204, 1983. 32. Diehl AK. Gallstone size and the risk of gallbladder cancer. JAMA 250:2323–2326, 1983. 33. Bragg LE, Thompson JS. Concomitant cholecystectomy for asymptomatic cholelithiasis. Arch Surg 124:480–482, 1989. 34. Green JD, Birkhead G, Herbert J, Li M, Vogt RL. Increased motility in surgical patients undergoing secondary (incidental) cholecystectomy. Ann Surg 211:50– 54, 1990.
Page 455
21— Biliary Crystals, Microlithiasis, and Sludge Dieter Jüngst Klinikum Grosshadern, LudwigMaximiliansUniversity, Munich, Germany Christoph von Ritter Vinzentimm Ruhpolding Hospital, Ruhpolding, Germany I— Introduction The term sludge is used for a variety of conditions that involve solid material, dispersed in a liquid medium, that slowly settles to the bottom. A ''sandy" precipitate is frequently found in the gallbladder upon cholecystectomy in patients with symptomatic gallstone disease, and the stratification of bile is a wellknown phenomenon. However, the importance of biliary sludge was increasingly recognized only after the advent of ultrasonography. There is sufficient evidence to indicate that biliary sludge is an intermediate step in the formation of gallstones of different origin, at least in some patients. Furthermore, biliary sludge has been shown to play a role in the pathogenesis of cholecystitis, cholangitis, and pancreatitis. II— Definition Biliary sludge is defined as a suspension of precipitated particulate matter in bile. As synonyms, biliary sediments, microcrystalline disease, and microlithiasis have been proposed. Generally, biliary sludge may consist of the whole range, from single crystals, aggregated crystals, to microlithiasis dispersed in a viscous, mucinrich liquid phase. The chemical composition of biliary sludge depends on the various causes of sludge formation. The more common precipitates are cholesterol monohydrate crystals (Fig. 1), and bile containing these crystals is characterized by a high viscosity and an increased mucin and protein content (1–4). Other precipitates in human bile include calcium bilirubinate granules, calcium phosphate and calcium carbonate crystals, and calcium salts of fatty acids and ceftriaxone. We have found that the ultracentrifugation of bile allows quantification of the main components of biliary sludge (5). Our data showed that assessment of the differences in the concentrations of cholesterol, protein, mucin, and bilirubin between native and ultracentrifuged gallbladder bile samples—collected during laparoscopic surgery from patients with cholesterol, mixed, and pigment stones—allows an estimation of the content of these components in biliary sludge (Fig. 2). The amount of biliary sludge was higher in the gallbladder bile of patients with cholesterol gallstones than in that of patients with mixed or pigment stones. Furthermore, the chemical composition of the precipitates correlated well to the composition of the associated stones (Fig. 2) (6).
Page 456
Figure 1 Phasecontrast microscopy of a cluster of cholesterol monohydrate crystals (Ø 5 to 10 µm) in gallbladder bile.
III— Diagnosis The major method for the diagnosis of biliary crystals is the microscopic examination of duodenal bile after cholecystokinin stimulation, allowing single crystals with a diameter of 5 to 10 µm to be detected. Transabdominal ultrasound examination of the gallbladder permits the visualization of particles in bile only above a diameter of approximately 2 mm. These represent the comparably larger particles of biliary sludge, consisting of aggregated crystals or
Figure 2 Mean concentration difference between native and ultracentrifuged (100,000 g for 1 h) gallbladder bile of cholesterol, protein, mucin, and bilirubin. Samples were collected during surgery from 35 patients with cholesterol (>50% cholesterol), 18 patients with mixed (10 to 50% holesterol), and 15 patients with pigment stones (<10% cholesterol). (From Ref. 6.)
Page 457
microliths embedded in the mucinrich liquid phase. Therefore, both methods determine differentsized particles of the spectrum of biliary sludge. A— Transabdominal Ultrasound and Computed Tomography With the advent of grayscale ultrasound, it has been recognized that the gallbladder may contain layered bile, with the most dependent layer demonstrating echogenicity of low amplitude (Fig. 3a). The fluid level in the gallbladder changes when the patient is moved from a supine to a decubitus or erect position. The rate at which this level is reestablished after a positional change is often very slow, suggesting that the medium is relatively viscous. Whenever layering is observed in bile, the echogenic layer is dependent; rarely, the gallbladder bile is diffusely echogenic. The precise nature of the origin of echoes within biliary sludge has been reported in different in vitro studies (7–9). These studies showed that filtration of bile converts echogenic bile into echofree bile. Examination of the filtered residue by light microscopy revealed that the source of echoes in biliary sludge was predominantly pigment precipitates mixed with cholesterol crystals. The pigment precipitates consisted mainly of calcium
Figure 3 a. Transabdominal sonography of the gallbladder of a patient with microlithiasis (Ø < 3 mm). b. Abdominal computed tomography of the gallbladder revealed major calcification of the microliths, excluding bile acid dissolution therapy in this patient.
Page 458
bilirubinate (7–9). The presence of calcium salts in biliary sludge may be detected by highresolution computed tomography (Fig. 3b). B— Biliary Microscopy Gallbladder bile can be collected noninvasively by duodenal drainage after stimulation with magnesium sulfate, which leads to a dilatation of the sphincter of Oddi followed by contraction of the gallbladder (MeltzerLyon test) (10–12). Nowadays the collection of duodenal bile is usually performed during duodenoscopy after stimulation of the gallbladder with cholecystokinin (13). The particulate matter visible in the microscope consists mainly of cholesterol monohydrate crystals and calcium bilirubinate granules and less often microspherolites consisting of calcium carbonate (12,14,15). It has been established that the presence of cholesterol crystals in gallbladder bile allows prediction of cholesterol gallstone disease with a sensitivity and specificity above 80% (15,16). Pigment stones can be predicted by the presence of only calcium bilirubinate granules with a sensitivity and specificity above 70% (14–16). When bile is collected in the duodenum after stimulation with cholecystokinin, gallstones can be predicted with a sensitivity of approximately 60 to 70% by microscopic detection of crystals (17). C— Endoscopic Ultrasound Ultrasonographically, the presence of sludge in the gallbladders of patients with gallstones is detectable in approximately 2% of patients (6). Compared to microscopy, the sensitivity of transabdominal ultrasound to detect smallersized particles of biliary sludge is rather low. These problems can be overcome by the use of endoscopic ultrasonography. This technique has been compared with microscopic examination of duodenal bile in the diagnosis of microlithiasis in patients with normal conventional ultrasonography (18,19). Compared to operative results, both methods were equally sensitive (>70%) and equally specific (>85%) in the diagnosis of biliary sludge or microlithiasis. In conclusion, conventional ultrasonography and microscopy of gallbladder bile collected by duodenal drainage or endoscopically by aspiration after gallbladder contraction represent valuable methods for the diagnosis of biliary sludge or microlithiasis. However, owing to the differences in sensitivity between the two methods, ultrasound may miss many patients with clinically relevant biliary sludge, while biliary microscopy may result in overestimation of such cases. Computed tomography appears to be a valuable aid for the detection or exclusion of major calcification of biliary sludge. IV— Prevalence, Epidemiology, and Risk Factors In the largest series—17,021 patients—admitted to a department of medicine with different diseases, including cholecystolithiasis, gallbladder sludge was found by transabdominal ultrasound in 1.7% (20). Further studies also using transabdominal ultrasound detected biliary sludge in 30 (1%) of 2949 consecutive patients admitted to a department of medicine for various diseases (6). In 452 patients with gallbladder stones, sludge was detected in 2% (6). Using light microscopy after duodenal drainage of gallbladder bile following cholecystokinin stimulation, cholesterol monohydrate crystals or pigment granules were found in up to 70% of patients with gallstones (12). Higher prevalences of sonographically detectable biliary sludge in the gallbladder have been reported during pregnancy (30%), parenteral nutrition (50%), and weight loss (25%) or after ingestion of ceftriaxone (40%) (21–29). Within a population, biliary sludge and microlithiasis occur sporadically, and specific risk factors have been identified that predict their formation.
Page 459
A— Age and Gender Because biliary sludge may dissolve spontaneously, the cumulative prevalence of sludge does not necessarily increase with age. However, since cholesterol secretion into bile increases with age, this might affect the formation of biliary sludge and microlithiasis. Female sex is a prominent risk factor for cholesterol gallstone formation. However, of 286 patients in whom biliary sludge was detected by ultrasound, 157 were women and 129 were men (20). Thus female sex seems not to be a specific risk factor at least for the formation of ultrasonographically detectable biliary sludge. B— Obesity, Weight Loss, and Total Parenteral Nutrition Obesity is a wellknown risk factor for the cholesterol gallstones (30). Due to the low prevalence of sonographically detectable biliary sludge, the relation between obesity and biliary sludge formation is unknown. Rapid weight loss is a recently recognized risk factor for cholesterol gallstone formation. Approximately 25% of obese patients develop gallstones during dietary weight loss. After gastric bypass surgery, as many as 50% of patients form biliary sludge or gallstones (26,31). The pathogenesis of biliary sludge and gallstone formation after rapid weight loss and gastric bypass surgery appears to be multifactorial. It has been shown that hepatic cholesterol secretion increases during caloric restriction (32,33). Additional factors may include increased mucin production (a potent stimulator of cholesterol crystal nucleation) and decreased gallbladder motility (33,34). Impaired gallbladder motility also appears to be of major importance for the development of biliary sludge during total parenteral nutrition (TPN). As many as 23% of patients under TPN for a minimum of 3 months were found to develop biliary sludge (22,25,35). Moreover, in a prospective study, a 50% incidence of biliary sludge was detected by ultrasound within 4 to 6 weeks of TPN. Ten days of fasting following gastrointestinal surgery caused sludge in 32% of patients (31,34). The high percentage of sludge formation during fasting is believed to be primarily caused by gallbladder hypomotility and bile stasis. In addition failure of the sphincter of Oddi to relax, causing preferential bile flow into the gallbladder, may play a role. C— Pregnancy and Parity Pregnancy and parity are wellknown risk factors for cholesterol gallstone disease. Pregnancy is a greater risk factor for the development of biliary sludge and subsequent gallstones (36). During pregnancy, bile becomes more lithogenic as a result of increased serum estrogen levels, which cause increased cholesterol secretion and supersaturation of bile (37). In addition, due to gallbladder hypomotility, the residual volume of the gallbladder increases and stasis develops, which also promotes sludge and stone formation (36). The altered gallbladder motility during pregnancy is predominantly due to the smooth muscle inhibitory effect of progesterone and perhaps to diminished contraction in response to cholecystokinin (38). Sludge formation can develop in the first trimester and increases steadily until delivery (21). D— Biliary Stasis In addition to impaired motility of the gallbladder, bile stasis is an important cause of sludge formation. Sludge was analyzed in six patients with common duct obstruction caused by chronic pancreatitis, pancreatic pseudocyst, or carcinoma (27). Microscopy of sludge showed calcium bilirubinate granules but no cholesterol crystals. During stasis of gallbladder bile, bilirubin glucuronide is hydrolyzed to bilirubin, which then forms the pigment precipitate. When unconjugated bilirubin precipitates from bile, it forms complexes with inorganic ions, mostly calcium. In an animal model, it has been shown that the ligation of gallbladders resulted in the formation of pigment sludge and stones (39). Three days after ligation, the gallbladders
Page 460
contained copious amounts of viscous black material that was microscopically identified as calcium bilirubinate granules. The major stimulus for this formation of pigment stones was hypersecretion of gallbladder mucin. E— Drugs The effect of estrogen on cholesterol gallstone formation has been extensively studied. Exogenous estrogen increases biliary cholesterol secretion by 40%, causing cholesterol supersaturation in the bile (40). No such relation has been documented between the intake of estrogen and the formation of biliary sludge. Octreotide, the somatostatin analogue, has been shown to cause a 28% incidence of gallstones in patients receiving this drug for the treatment of acromegaly. Decreased gallbladder motility is the most probable cause for this octreotide effect (41,42). Ceftriaxone, a thirdgeneration cephalosporin with a long duration of action, is excreted into urine. However, as much as 40% of the drug is secreted in unmetabolized form into bile, reaching biliary concentrations 100 to 200fold higher than the concentration in serum (43). Once the biliary saturation level is exceeded, ceftriaxone complexes with calcium and forms an insoluble salt (29). Formation of biliary sludge has been reported in 43% of children receiving highdose ceftriaxone; 19% of these patients were symptomatic (44). Microscopic examination of surgically collected bile specimens revealed fine precipitates 20 to 250 µm in diameter. There were a small number of cholesterol monohydrate crystals and calcium bilirubinate granules among an abundant amount of crystalline material, which predominantly consisted of calcium salts of ceftriaxone (29). F— Liver Transplantation A wellknown complication after liver transplantation is the development of biliary sludge. In the largest series, filling defects of the bile duct were detected by cholangiography in 94 (5.7%) of 1650 patients (45). On the basis of the cholangiographic appearance, the bile duct filling defects were categorized as sludge or cast in 53 (56%), stones in 32 (34%), and necrotic debris in nine (10%) patients. The formation of biliary sludge is considered to be a serious, lifethreatening complication. The major reasons for sludge formation were biliary strictures and prestenotic dilatations of the bile ducts. As in bile duct necrosis with debris formation due to bile duct ischemia caused by hepatic artery occlusion, sludge formation may also be caused by ischemia in some cases (45). V— Pathogenesis The formation of cholesterol monohydrate crystals is believed to be crucial in the pathogenesis of cholesterol gallstones (46,47). A variety of factors that promote or inhibit cholesterol crystal nucleation in model bile have been reported (48). Although virtually insoluble in water, cholesterol is made soluble in bile through carriers that include bile salts and phospholipids (49). In unsaturated bile, cholesterol is primarily transported in simple and mixed micelles (17). As cholesterol saturation increases in bile, more cholesterol is carried in larger phospholipid cholesterol vesicles (Fig. 4). Unilamellar vesicles can coalesce into multilamellar vesicles, which tend to be less stable and allow the growth of cholesterol crystals from the surface (50,51). These vesicles may interact with soluble mucin, which acts as an annealing agent, favoring the further nucleation and agglomeration of cholesterol monohydrate crystals (52,53). These crystals are entrapped in the soluble or gel form of mucin. Thus, one form of biliary sludge represents an early event in cholesterol gallstone formation with cholesterol monohydrate crystals embedded in biliary mucin. Calcium bilirubinate granules represent another major component of biliary sludge. These pigment granules are typically found in the gallbladder bile of patients with pigment stones
Page 461
Figure 4 Transmission electron microscopy of phospholipid cholesterol vesicles (30 to 100 nm) isolated from gallbladder bile by gelfiltration chromatography.
(14–16). In liver cirrhosis, the risk of developing biliary sludge consisting of calcium bilirubinate granules is increased (54). Hypersecretion of bilirubin conjugates (especially monoglucuronides) into the bile is the most important factor for the formation of biliary sludge consisting of calcium bilirubinate granules. In an animal model, the presence of hemolysis increased the biliary secretion of bilirubin monoglucuronides more than 10fold (55). Unconjugated monohydrogenated bilirubin is formed by the action of endogenous betaglucuronidase, which can coprecipitate with calcium as a result of supersaturation (56). While no defects in gallbladder motility have been found in patients with pigment stones (57), biliary stasis is an important factor, resulting in increased levels of unconjugated bilirubin. The cytotoxic effect of unconjugated bilirubin may favor mucin hypersecretion from the biliary epithelial cells (58). VI— Natural History In a prospective study, 96 patients with sonographically detected biliary sludge were followed for a mean of 3 years by serial ultrasound scans (59). In 17 patients (18%) biliary sludge disappeared and did not recur for at least 2 years. In 58 patients (60.4%), biliary sludge disappeared, but it recurred during the observation period, while 8 patients (8.3%) developed asymptomatic gallstones (59). Only 6 patients (6.3%) developed symptomatic gallstones that required treatment by cholecystectomy. Another 6 patients (6.3%) suffered from attacks of biliary pain in part associated with recurrent pancreatitis; they also had to be treated by elective cholecystectomy. In a further study, 56 patients with gallbladder sludge were followed by ultrasound (20). Within a mean of 2 months, 40 patients (71.4%) were free of sludge and showed normal sonographic gallbladder findings. Gallbladder stones without sludge developed in 5 patients (8.9%) within a mean of 2.5 months, and gallstones with persistence of sludge were observed in 2 further patients (3.6%) after 6.1 and 30.7 months, respectively. None of these patients became symptomatic in the course of the followup period. Acute cholecystitis developed in 4 further patients (7.1%). No cases of acute pancreatitis were observed in this study (20). The incidence of sludge and stone formation during pregnancy is approximately 30 and 2%, re
Page 462
spectively (60). Both sludge and stones are usually "silent," but when biliary pain develops, it is generally associated with the presence of stones and not with sludge. After delivery, gallbladder motility returns to normal and sludge disappears in 60 to 70% of cases (60). The incidence of gallbladder sludge amounts to 50% in patients with total parenteral nutrition of more than 6 weeks' duration (31). Normal nutrition causes disappearance of sludge in most cases within a few weeks; however, because of the development of acalculous cholecystitis or symptomatic gallstones, cholecystectomy is required in 15% of cases (31). Although biliary sludge in patients receiving ceftriaxone may cause symptoms in about 19%, in most patients sludge spontaneously disappears after the antibiotic treatment is withdrawn (44). Taken together, these studies show that biliary sludge is a reversible condition. Upon withdrawal of the noxious agent or condition, sludge dissolves or is discharged in the majority of cases. However, a minority of patients develop gallstones, which may become symptomatic. By far the most serious complication of biliary sludge is the development of biliary pancreatitis. VII— Clinical Manifestations of Biliary Sludge The hepatobiliary tract is a lowpressure/lowflow hydraulic excretory pathway for hydrophobic, waterinsoluble substances. Because of the lowflow nature of the system and the tenuous solubility of the bile constituents, bile is prone to precipitation and formation of solid material such as sludge (61). The clinical manifestations of microcrystalline disease (a proposed synonym for biliary sludge and microlithiasis) are only poorly understood. Biliary sludge or microlithiasis can cause symptoms mainly via two mechanisms: obstruction of either the cystic duct or the common bile duct. The presence of cholesterol monohydrate crystals is a constant feature of cholesterol gallstone disease (14–16,46). There is evidence to suggest that gallbladder bile rich in cholesterol and cholesterol crystals may cause epithelial hyperplasia of the gallbladder and result in hypersecretion of mucins (1,2,62–64). In longstanding cholesterol gallstone disease, the consequence of the persistent secretion of lithogenic bile leads to severe disturbance of gallbladder function, with impaired motility and loss of concentrative function (57,65,66), both believed to be caused by cholesterol crystals. Moreover, cholesterol crystals in bile may also lead to chronic inflammation of the cystic duct or the sphincter of Oddi, with subsequent fibrosis and sclerosis (H.U. Kloer, personal communication, 1998). A— Acalculous Biliary Pain Acalculous biliary pain, an intense episode of rightupper abdominal pain that begins suddenly, rises in intensity over a 15min period and continues at a steady plateau for several hours before slowly subsiding. The attacks of pain are frequently but not always precipitated by meals and may be accompanied by vomiting. The symptoms mimic those of a typical biliary colic. Between attacks, the physical examination is usually normal, with the possible exception of residual upper abdominal tenderness. The striking preponderance of patients with acalculous biliary pain who are usually young, parous females closely parallels the epidemiology of cholesterol gallstones. This suggests that there may be similar risk factors for the two conditions. Indeed, some studies of resected gallbladder specimens have shown that as many as onehalf of the patients with acalculous biliary pain actually have microscopic cholecystolithiasis, indicating that the original sonogram yielded a falsenegative result (67). Typically, the MeltzerLyon test reveals the presence of cholesterol monohydrate crystals in duodenal bile after the stimulation of gallbladder contraction. The resected gallbladders of a subset of patients with acalculous biliary pain show histological evidence of cholesterolosis. Although usually thought to be an incidental pathological
Page 463
finding, cholesterolosis of the gallbladder may in some patients impair normal gallbladder contraction and result in clinical features identical to those of acalculous biliary pain (68). B— Acute Acalculous Cholecystitis Acute acalculous cholecystitis may be caused by biliary sludge or microlithiasis. In most cases acute acalculous cholecystitis is associated with a triad of prolonged fasting, immobility, and hemodynamic instability. In this setting, the gallbladder epithelium is exposed to one of the most noxious environments endogenous to the body, which comprises a concentrated solution of detergent bile acids known to solubilize lipids. Normally, the gallbladder empties concentrated bile several times per day and is replenished with dilute hepatic bile. With prolonged fasting, the gallbladder never receives the adequate cholecystokinin (CCK)mediated stimulus required to void its contents; thus concentrated bile lingers in its lumen. Together with splanchnic vasoconstriction resulting from, for instance, septic shock, the stage is set for an ischemic/chemical injury to the gallbladder epithelium (69). The clinical features of acute acalculous cholecystitis differ from those of cholecystitis resulting from stone disease. Although rightupperquadrant pain, fever, localized tenderness over the gallbladder, and leukocytosis may be evident in classic presentations, some or all of these features are frequently lacking in elderly postoperative patients. Compared to ordinary calculous cholecystitis, the clinical course of acute acalculous cholecystitis is more fulminant. By the time the diagnosis is made, at least half of the patients have already experienced complications such as gangrene or localized perforation of the gallbladder (70). C— Cholesterolosis Cholesterolosis of the gallbladder is an acquired histological abnormality of the gallbladder mucosa involving excessive accumulation of cholesterol ester within epithelial macrophages (71). Although the cause of the accumulation of cholesterol esters and cholesterol in the gallbladder mucosa is unclear, it has been unequivocally shown that the gallbladder epithelium is capable of absorbing cholesterol from bile (72–74). However, it is unknown why, in some patients, resorbed biliary cholesterol is esterified and then stored in foamy macrophages, resulting in cholesterolosis (72). The lesion is frequently but not always found in gallbladders exposed to bile that is supersaturated with cholesterol, and the disorder probably shares a common pathogenetic mechanism with cholesterol gallstone disease (75). D— Cholangitis and Pancreatitis The most relevant clinical manifestation of microlithiasis or biliary sludge is obstruction of the common biliary duct with acute cholangitis or pancreatitis. Gallstone pancreatitis is often related to small stones, which may not be detected by conventional cholecystographic techniques (76–79). In a recent study, it was hypothesized that, in patients with acute pancreatitis of unknown cause, their condition could be caused by microlithiasis or cholesterolosis of the gallbladder (80). In a prospective study, evidence for this hypothesis was sought by microscopic examination of centrifuged duodenal bile in 51 patients with acute pancreatitis. Clusters of cholesterol monohydrate crystals, calcium bilirubinate granules, and calcium carbonate microspherolites were found in 67% of the patients. In contrast, biliary drainage showed no abnormal findings in 12 patients suffering from a bout of known alcoholic pancreatitis. Examination of gallbladder bile at cholecystectomy, or abdominal ultrasonography of the gallbladder for up to 12 months, showed that 73% of the patients with unexplained pancreatitis had biliary sludge or microlithiasis. Biliary crystals/solid markers both predicted biliary pancreatitis with a sensitivity and specificity of 86% and a predictive value of 95% (78). In a second prospective study, Lee et al. determined the frequency of biliary sludge in patients with
Page 464
acute idiopathic pancreatitis (77). The study comprised 86 patients with no known cause of pancreatitis and no ultrasonographic evidence of gallstones or dilatation of the biliary duct. The pancreatitis was considered idiopathic in 31 of the 86 patients (36%), of whom 23 had microscopic evidence of biliary sludge. Biliary sludge was detected by ultrasonography in only 11 of the 23 patients (48%). The sludge detected by ultrasonography was composed of calcium bilirubinate granules in 10 patients and cholesterol monohydrate crystals in 1. Calcium bilirubinate granules were found more frequently in men (9 men versus 4 women). The presence of biliary sludge was clearly associated with recurrent attacks of pancreatitis. Therefore, occult microlithiasis should be strongly suspected in cases of acute pancreatitis of unknown origin, especially when there are frequent relapses. E— Dysfunction of the Sphincter of Oddi Chronic exposure of the sphincter of Oddi to bile containing cholesterol crystals may lead to dysfunction of this sphincter, a wellknown clinical entity in patients with longstanding gallstone disease (H.U. Kloer, personal communication, 1998). VIII— Treatment of Biliary Sludge or Microlithiasis As with gallstone disease, treatment is necessary only in patients whose biliary sludge causes symptoms or complications. In the case of accidentally detected biliary sludge with no biliary symptoms, expectant management is warranted. In accordance with the natural history of the majority of such patients, biliary sludge will dissolve or be discharged spontaneously. However, in patients whose biliary sludge is causing biliary pain or complications such as acalculous cholecystitis, cholangitis, or recurrent pancreatitis, immediate treatment is warranted. A— Ursodeoxycholic Acid Patency of the cystic duct as documented by sonographically normal gallbladder contraction following a test meal or by opacification of the gallbladder after oral cholecystography is the precondition for oral bile acid dissolution therapy. Furthermore, the exclusion of calcified biliary sludge by a computed tomography scan of the gallbladder is valuable for the selection of appropriate patients. Treatment should be initiated with ursodeoxycholic acid (UDCA) at a dose of approximately 10 mg/kg/day. After successful dissolution of biliary sludge, a maintenance therapy with 500 mg/day is recommended (78). B— Laparoscopic Cholecystectomy In patients with persistent biliary sludge despite dissolution treatment with UDCA—and, in particular, in patients with symptomatic microlithiasis and impaired contractile function of the gallbladder or other complications—laparoscopic cholecystectomy is indicated. C— Endoscopic Papillotomy An alternative approach for patients with biliary sludge and recurrent biliary outflow obstruction associated with either recurrent cholangitis or pancreatitis is endoscopic papillotomy (81–83). This treatment may be combined with maintenance treatment including UDCA or laparoscopic cholecystectomy (84). In the more specific situation of symptomatic sludge formation during pregnancy or after
Page 465
ceftriaxone application, laparoscopic cholecystectomy may be indicated. After delivery a 3 to 6month course of fulldose UDCA may be a treatment option. Biliary sludge after treatment with ceftriaxone will, in most cases, dissolve rapidly. In patients with persistent sludge and biliary symptoms despite dissolution therapy, laparoscopic cholecystectomy is the treatment of choice. Biliary sludge after liver transplantation is a serious complication and should be treated primarily by oral chemolysis with UDCA. Unfortunately, medical treatment often fails and more aggressive approaches—such as percutaneous transhepatic biliary drainage followed by irrigation with heparinized saline solutions, intraluminal chemolysis with glycerylmonooctanoate and bile salt ethylenediaminetetraacetic acid (EDTA), or basket extraction—become necessary. Endoscopic intervention and surgery are alternative approaches (45,85). In patients with symptomatic sludge after weight loss or those receiving TPN, strategies such as bile acid dissolution therapy or laparoscopic cholecystectomy may be applied. However, since the development of sludge in patients undergoing weight loss and TPN is highly predictable, and in view of the potentially serious complications, prophylactic therapy is indicated (22,23). In patients on low calorie diets, 500 mg of UDCA daily will minimize the risk of sludge development (86). In patients receiving TPN, a daily stimulation of gallbladder contraction with intravenous cholecystokinin is highly effective in the prevention of biliary sludge (87). In patients with biliary strictures, the formation of sludge is secondary to the stricture and may itself further impair bile flow. A combined approach using endoscopic dilatation of the stricture, biliary drainage, and oral administration of UDCA to improve bile flow is a possible approach to this sometimes serious problem. There is evidence that biliary crystals, microlithiasis, and sludge represent intermediate events in gallstone development. Biliary cholesterol monohydrate crystals or calcium bilirubinate granules in gallbladder bile are associated with overt gallstone disease in approximately 80% of patients (12,14–16). Clinically, it is often impossible to distinguish whether symptoms in patients with gallstones are derived from visible stones or from sonographically occult microlithiasis. Epidemiological studies suggest that the most severe complications of gallstone disease, such as acute cholecystitis or acute biliary obstruction with either cholangitis or pancreatitis, are caused by small stones (79). In patients with cholesterol gallstone disease that may be diagnosed by appropriate clinical correlates (88), small stones or microliths are amenable to treatment with UDCA. As mentioned above, computed tomography scanning of the gallbladder may be helpful to identify suitable patients. Maintenance treatment with UDCA in patients with gallstones may improve the natural course of the disease by avoiding the complications caused by microlithiasis. Recently, the longterm effects of maintenance therapy with UDCA on the natural course of cholesterol gallstone disease was examined using a Cox proportional hazard model analysis (89). A total of 434 patients with cholesterol gallstones were prospectively followed for up to 12 years with either UDCA (600 mg/day, 168 patients) or no treatment (266 patients). The longterm cumulative occurrence rates of biliary colic was low in the primarily asymptomatic patients (8 and 16% at 5 and 10 years, respectively) but high in initially symptomatic patients (62 and 80% at 5 and 10 years, respectively). These rates were markedly reduced in patients taking UDCA, amounting to 0 and 5% at 5 and 10 years, respectively in the asymptomatic group of patients, and to 16 and 46% at 5 and 10 years, respectively in the symptomatic patients. Complete gallstone dissolution rates in patients treated with UDCA remained comparably low (22 and 24% at 5 and 10 years, respectively). These data show that maintenance therapy with UDCA could improve the natural course of gallstone disease in symptomatic patients and to a lesser degree also in asymptomatic patients. Since in the above study the effect of UDCA is presumably based predominantly on the dissolution of sludge, the results indirectly support an important role of biliary crystals, microlithiasis, and sludge in the pathogenesis of the symptoms and complications of cholesterol gallstone disease. Very recently, it was confirmed that bile abnormalities can be successfully corrected with UDCA therapy in patients with microlithiasis (90).
Page 466
Acknowledgments The authors thank Alan F. Hofmann, Nair Sreejayan, and GerdAchim KullakUblick for critically reviewing the manuscript and Robert Lugmaier for expert technical assistance. References 1. Gallinger S, Taylor RD, Harvey PTC, Petrunka CN, Strasberg SM. Effect of mucous glycoprotein on nucleation time of human bile. Gastroenterology 1985; 89:648–658. 2. Gallinger S, Harvey PRC, Petrunka CN, Ilson RG, Strasberg SM. Biliary proteins and the nucleation defect in cholesterol cholelithiasis. Gastroenterology 1987; 92:867–875. 3. Harvey PRC, Rupar CA, Gallinger S, Petrunka CN, Strasberg SM. Quantitative and qualitative comparison of gallbladder mucus glycoprotein from patients with and without gallstones. Gut 1986; 27:374–381. 4. Jüngst D, Lang T, von Ritter C, Paumgartner G. Role of high total protein in gallbladder bile in the formation of cholesterol gallstones. Gastroenterology 1991; 100:1724–1729. 5. Jüngst D, del Pozo R, Christoph S, Miquel JF, Eder MI, Lange V, Frimberger E, von Ritter C, Paumgartner G. Sedimentation of biliary ''sludge": effect on composition of gallbladder bile from patients with cholesterol, mixed or pigment stones. Scand J Gastroenterol 1996; 31:273–278. 6. Christoph S. Gallenblasensludge—Pathophysiologie und klinische Bedeutung. M.D. thesis, LudwigMaximiliansUniversity, Munich, Germany, 1994. 7. Conrad MR, Jones RO, Dietchy J. Significance of low level echoes within the gallbladder. Am J Roentgenol 1979; 132:967–972. 8. Filly RA, Allen B, Minton MJ, Bernhoft R, Lawrence WW. In vitro investigation of the origin of echoes within biliary sludge. J Clin Ultrasound 1980; 8:193–200. 9. Jennings WC, Drabek GA, Miller KA. Significance of sludge and thickened wall in ultrasound evaluation of the gallbladder. Surg Gynecol Obst 1992; 174:394– 398. 10. Meltzer SJ. The disturbance of the law of contrary innervation as a pathogenetic factor in the diseases of the bile ducts and the gallbladder. Am J Med Sci 1917; 153:469. 11. Lyon BBV. Diagnosis and treatment of diseases of the gallbladder and biliary ducts. JAMA 1919; 73:980–982. 12. Juniper K, Burson EN Jr. Biliary tract studies: II. The significance of biliary crystals. Gastroenterology 1957; 32:175–211. 13. Susann PW, Sheppard F, Baloga AJ. Detection of occult gallbladder disease by duodenal drainage collected endoscopically: a clinical and pathologic correlation. Am Surg 1985; 51:162–165. 14. Poupon R, Chrétien Y, Darnis F. Cristaux de cholestérol, saturation de bile en cholestérol et lithiasise biliaire. Gastroenterol Clin Biol 1984; 8:260–263. 15. Ros E. Navarro S, Fernández I, Reixach M, Ribó JM, Rodés J. Utility of biliary microscopy for the prediction of the chemical composition of gallstones and the outcome of dissolution therapy with ursodeoxycholic acid. Gastroenterology 1986; 91:703–712. 16. Ramond MJ, Dumont M, Belghiti J, Erlinger S. Sensitivity and specificity of microscopic examination of gallbladder bile for gallstones recognition and identification. Gastroenterology 1988; 95:1339–1343. 17. Carey MC, Small DM. The physical chemistry of cholesterol solubility in bile: relationship to gallstone formation and dissolution in man. J Clin Invest 1978; 61:998–1026. 18. Dill JE, Hill S, Callis J, Berkhouse L, Evans P, Martin D, Palmer ST. Combined endoscopic ultrasound and stimulated biliary drainage in cholecystitis and microlithiasis—diagnosis and outcomes. Endoscopy 1995; 27:424–427. 19. Dahan P, Andant C, Lévy P, Amouyal P, Amouyal G, Dumont M, Erlinger S, Sauvanet A, Belghiti J, Zins M, Vilgrain V, Bernades P. Prospective evaluation of endoscopic
Page 467
ultrasonography and microscopic examination of duodenal bile in the diagnosis of cholecystolithiasis in 45 patients with normal conventional ultrasonography. Gut 1996; 38: 277–281. 20. Janowitz P, Kratzer W, Zemmler T, Tudyka J, Wechsler JG. Gallbladder sludge: spontaneous course and incidence of complications in patients with stones. Hepatology 1994; 20:291–294. 21. Maringhini A, Ciambra M, Bacceliere P, Raimondo M, Pagliario I. Sludge, stones and pregnancy. Gastroenterology 1988; 95:1160–1161. 22. Messing B, Bories C, Kunstlinger F, Bernier J. Does total parenteral nutrition induce gallbladder sludge formation and lithiasis? Gastroenterology 1983; 84:1012– 1019. 23. Bolondi L, Gaiani S, Testa S, Labò G. Gallbladder sludge formation during prolonged fasting after gastrointestinal tract surgery. Gut 1985; 26:734–738. 24. Cano N, Cicero F, Ranieri F, Martin J, di Costanzo J. Ultrasonographic study of gallbladder motility during total parenteral nutrition. Gastroenterology 1986; 91:313–317. 25. Gafá M, Sarli L, Miselli A, Pietra N, Carreras F, Peracchia A. Sludge and microlithiasis of the biliary tract after total gastrectomy and postoperative total parenteral nutrition. Surg Gynecol Obstet 1987; 165:413–418. 26. Liddle RA, Goldstein RB, Saxton J. Gallstone formation during weightreduction dieting. Arch Intern Med 1989; 149:1750–1753. 27. Allen B, Bernhoft R, Blanckaert N, Svanvik J, Filly R, Gooding G, Way L. Sludge is calcium bilirubinate associated with bile stasis. Am J Surg 1981; 141:51–56. 28. Pitt HA, Doty JE, den Besten LW, Kuchenbecker SL. Stasis before gallstone formation: altered gallbladder compliance or cystic duct resistance. Am J Surg 1982; 143:144–149. 29. Park HZ, Lee SP, Schy AL. Ceftriaxoneassociated gallbladder sludge. Identification of calciumceftriaxone salt as a major component of gallbladder precipitate. Gastroenterology 1991; 100:1665–1670. 30. Stampfer MJ, Maclure KM, Colditz GA, Manson JE, Willett WC. Risk of symptomatic gallstones in women with severe obesity. Am J Clin 1992; 55:652–658. 31. Shiffman ML, Sugerman HJ, Kellum JM, Brewer WH, Moore EW. Gallstone formation after rapid weight loss: a prospective study in patients undergoing gastric bypass surgery for treatment of morbid obesity. Am J Gastroenterol 1991; 86:1000–1005. 32. Marks JW, Bonorris GG, Albers G, Schoenfield LJ. The sequence of biliary events preceding the formation of gallstones in humans. Gastroenterology 1992; 103:566–570. 33. Shiffman ML, Shambruck RD, Schwartz CC, Sugerman HJ, Kellum JM, Moore EW. Gallbladder mucin, arachidonic acid and bile lipids in patients who develop gallstones during weight reduction. Gastroenterology 1993; 105:1200–1208. 34. Inoue K, Fuchigami A, Higashide S, Sumi S, Kogire M, Suzuki T, Tobe T. Gallbladder sludge and stone formation in relation to contractile function after gastrectomy. Ann Surg 1992; 215:19–26. 35. Pitt HA, King W 3d, Mann LL, Roslyn JJ, Berquist WE, Ament ME, Den Besten L. Increased risk of cholelithiasis with prolonged total parenteral nutrition. Am J Surg 1983; 145:106–112. 36. Tsimoyiannis EC, Antoniou NC, Tsaboulas C, Papanikolaou N. Cholelithiasis during pregnancy and lactation: prospective study. Eur J Surg 1994; 160:627–631. 37. Lynn J, Williams L, O'Brien J, Wittenberg J, Egdahl RH. Effects of estrogen upon bile: implications with respect to gallstone formation. Ann Surg 1973; 178:514– 524. 38. Hould FS, Fried GM, Fazekas AG, Tremblay S, Mersereau WA. Progesterone receptors regulate gallbladder motility. J Surg Res 1988; 45:505–512. 39. Bernhoft RA, Pellegrini CA, Broderick WC, Way LW. Pigment sludge and stone formation in the acutely ligated dog gallbladder. Gastroenterology 1983; 85:1166–1171. 40. Henriksson P, Einarsson K, Eriksson A, Kelter U, Angelin B. Estrogeninduced gallstone formation in males: relation to changes in serum and biliary lipids during hormonal treatment of prostatic carcinoma. J Clin Invest 1989; 84:811–816.
Page 468
41. Van Liessum PA, Hopman WPM, Pieters GFFM, Jansen JBMJ, Smals AGH, Rosenbusch G, Kloppenborg PWC, Lamers CBHW. Postprandial gallbladder motility during long term treatment with the longacting somatostatin analog SMS 201–995 in acromegaly. J Clin Endocrinol Metab 1989; 69:557–562. 42. Montini M, Gianola D, Pagani MD, Pedroncelli A, Caldara R, Gherardi F, Bonelli M, Lancranjan I, Pagani G. Cholelithiasis and acromegaly: therapeutic strategies. Clin Endocrinol 1994; 40:401–406. 43. Arvidsson A, Alvan G, Angelin B, Borga O, Nord CE. Ceftriaxone: renal and biliary excretion and effect on the colon microflora. J Antimicrob Chemother 1982; 10:207–215. 44. Schaad UB, WedgwoodKrucko J, Tschaeppeler H. Reversible ceftriaxoneassociated biliary pseudolithiasis in children. Lancet 1988; 2:1411–1413. 45. Sheng R, Ramirez CB, Zajko AB, Campbell WL. Biliary stones and sludge in liver transplant patients: a 13year experience. Radiology 1996; 198:243–247. 46. Sedaghat A, Grundy SM. Cholesterol crystals and the formation of cholesterol gallstones. N Engl J Med 1980; 302:1274–1277. 47. Holan KR, Holzbach RT, Hermann RE, Cooperman AM, Claffey WY. Nucleation time: a key factor in the pathogenesis of cholesterol gallstone disease. Gastroenterology 1979; 77:611–617. 48. Holzbach RT. Recent progress in understanding cholesterol crystal nucleation as a precursor to human gallstone formation. Hepatology 1986; 6:1403–1406. 49. Admirand WH, Small DM. The physicochemical basis of cholesterol gallstone formation in man. J Clin Invest 1968; 47:1043–1052. 50. Sömjen GJ, Gilat T. Changing concepts of cholesterol solubility in bile. Gastroenterology 1986; 91:772–775. 51. Schriever CE, Jüngst D. Association between cholesterolphospholipid vesicles and cholesterol crystals in human gallbladder bile. Hepatology 1989; 9:541–546. 52. Smith BF. Human gallbladder mucin binds biliary lipids and promotes cholesterol crystal nucleation, in model bile. J Lipid Res 1987; 28:1088–1097. 53. Afdahl NH, Niu N, Gantz D, Small DM, Smith BF. Bovine gallbladder mucin accelerates cholesterol monohydrat crystal growth in model bile. Gastroenterology 1993; 104:1515–1523. 54. Acalovschi M, Badea R, Pascu M. Incidence of gallstones in liver cirrhosis. Am J Gastroenterol, 1991; 86:1179–1181. 55. Trotman BW, Bernstein SE, Bove KE, Wirt GD. Studies on the pathogenesis of pigment gallstones in hemolytic anemia: description and characteristics of a mouse model. J Clin Invest 1980; 65:1301–1308. 56. Cahalane MJ, Neubrand MW, Carey MC. Physicalchemical pathogenesis of pigment gallstones. Semin Liver Dis 1988; 8:317–328. 57. Behar J, Lee KY, Thompson WR, Biancani P. Gallbladder contraction in patients with pigment and cholesterol stones. Gastroenterology 1989; 97:1479–1484. 58. Trotman BW, Bernstein SE, Balistreri WF, Wirt GD, Martin RA. Hemolysisinduced gallstones in mice: increased unconjugated bilirubin in hepatic bile predisposes to gallstone formation. Gastroenterology 1981; 81:232–236. 59. Lee SP, Maher K, Nicholls JF. Origin and fate of biliary sludge. Gastroenterology 1988; 94:170–176. 60. Maringhini A, Ciambra M, Baccelliere P, Raimondo M, Orlando A, Tine F, Grasso R, Randazzo MA, Barresi L, Gullo D, Musico M, Pagliaro L. Biliary sludge and gallstones in pregnancy: incidence, risk factors, and natural history. Ann Intern Med 1993; 119:116–120. 61. Carey MC. Pathogenesis of gallstones. Am J Surg 1993; 165:410–419. 62. Doty JE, Pitt HA, Kuchenbecker SL, PorterFink V, Den Besten LW. Role of gallbladder mucus in the pathogenesis of cholesterol gallstones. Am J Surg 1983; 145:54–61.
Page 469
63. Lee SP, Nicholls JF. Nature and composition of biliary sludge. Gastroenterology 1986; 90:677–686. 64. Carey MC, Cahalane MJ. Whither biliary sludge. Gastroenterology 1988; 95:508–523. 65. Pomeranz IS, Shaffer EA. Abnormal gallbladder emptying in a subgroup of patients with gallstones. Gastroenterology 1985; 88:787–791. 66. Li YF, Moody FG, Weisbrodt NW, Zalewsky CA, Coelho JC, Senninger N, Gouma D. Gallbladder contractility and mucus secretion after cholesterol feeding in the prairie dog. Surgery 1986; 100:900–904. 67. HerreraBallester A, CanellesGamir P, MedinaChulia E, SolerRos JJ, OrtiOrtin E, OrtegaGonzalez F, QuilesTeodoro F. Cholecystectomy: a choice technique in biliary microlithiasis. Ann Med Int 1995; 12:111–114. 68. Kmiot WA, Perry EP, Donovan IA, Lee MJ, Wolverson RF, Harding LK, Neoptolemos JP. Cholesterolosis in patients with chronic acalculous biliary pain. Br J Surg 1994; 81:112–115. 69. Warren BL. Small vessel occlusion in acute acalculous cholecystitis. Surgery 1992; 111: 163–168. 70. Johnson LB. The importance of early diagnosis of acute acalculous cholecystitis. Surg Gynecol Obstet 1987; 164:197–203. 71. Weedon D. Cholesterolosis: Pathology of the Gallbladder. New York: Masson, 1984: 161. 72. Sahlin S, Stahlberg D, Einarsson K. Cholesterol metabolism in liver and gallbladder mucosa of patients with cholesterolosis. Hepatology 1995; 21:1269–1275. 73. Jacyna MR, Ross PE, Bakar MA, Hopwood D, Bouchier IA. Characteristics of cholesterol absorption by human gallbladder: relevance to cholesterolosis. J Clin Pathol 1987; 40: 524–529. 74. Tilvis RS, Aro J, Strandberg TE, Lempinen M, Miettinen TA. Lipid composition of bile and gallbladder mucosa in patients with acalculous cholesterolosis. Gastroenterology 1982; 82:607–615. 75. Braghetto I, Antezana C, Hurtado C, Csendes A. Triglyceride and cholesterol content in bile, blood, and gallbladder wall. Am J Surg 1988; 156:26–28. 76. Negro P, Flati G, Flati D, Porowska B, Tuscano D, Carboni M. Occult gallbladder microlithiasis causing acute recurrent pancreatitis. Acta Chir Scand 1984; 150:503–506. 77. Lee SP, Nicholls JF, Park HZ. Biliary sludge as a cause of acute pancreatitis. N Engl J Med 1992; 326:589–593. 78. Ros E, Navarro S, Bru C, GarciaPugés A, Valderrama R. Occult microlithiasis in "idiopathic" acute pancreatitis: prevention of relapses by cholecystectomy or ursodeoxycholic acid therapy. Gastroenterology 1991; 101:1701–1709. 79. Diehl AK, Holleman DR, Chapman JB, Schwesinger WH, Kurtin WE. Gallstone size and risk of pancreatitis. Arch Intern Med 1997; 157:1674–1678. 80. Paricio PP, Olmo DG, Franco EP, González AP, González LC, López JB. Gallbladder cholesterolosis: an aetiological factor in acute pancreatitis of uncertain origin. Br J Surg 1990; 77:735–736. 81. Davidson BR, Neoptolemos JP, CarrLocke DL. Endoscopic sphincterotomy for common bile duct calculi in patients with gallbladder in situ considered unfit for surgery. Gut 1988; 29:114–120. 82. CarrLocke DL. Role of endoscopy in gallstone pancreatitis. Am J Surg 1992; 165:519–521. 83. Welbourn CRB, Beckly DE, EyreBrook IA. Endoscopic sphincterotomy without cholecystectomy for gallstone pancreatitis. Gut 1995; 37:119–120. 84. Ricci F, Castaldini G, de Manzoni G, Borzellino G, Rodella L, Kind R. Minimally invasive treatment of acute biliary pancreatitis. Surg Endosc 1997; 11:1179– 1182. 85. Barton P, Steininger R, Maier A, Mühlbacher F, Lechner G. Biliary sludge after liver transplantation: 2. Treatment with interventional techniques versus surgery and/or oral chemolysis. Am J Roentgenol 1995; 164:865–869.
Page 470
86. Shiffman ML, Kaplan GD, BrinkmanKaplan V, Vickers FF. Prophylaxis against gallstone formation with ursodeoxycholic acid in patients participating in a very lowcalorie diet program. Ann Intern Med 1995; 122:899–905. 87. Sitzmann JV, Pitt HA, Steinborn PA, Pasha ZR, Sanders RC. Cholecystokinin prevents parenteral nutrition induced biliary sludge in humans. Surg Gynecol Obstet 1990; 170: 25–31. 88. Diehl AK, Schwesinger WH, Holleman DR Jr, Chapman JB, Kurtin WE. Clinical correlates of gallstone composition: distinguishing pigment from cholesterol stones. Am J Gastroenterol 1995; 90:967–972. 89. Tomida S, Abei M, Yamaguchi T, Shoda J, Matsuzaki Y, Tanaka N, Osuga T. Longterm effects of ursodeoxycholic acid in patients with cholecystolithiasis in comparison with the natural course: a Cox proportional hazard model analysis. Gastroenterology 1997; 112(4 suppl)A524. 90. Sharma BC, Agarwal DK, Dhiman RK, Baijal SS, Choudhuri G, Saraswat VA. Bile lithogenicity and gallbladder emptying in patients with microlithiasis: effect of bile acid therapy. Gastroenterology 1998; 115:124–128.
Page 471
22— Biliary Colic and Acute Cholecystitis Robert V. Rege University of Texas, Southwestern Medical Center, Dallas, Texas I— Introduction In the United States, 10% of the population have gallstones. Each year, more than 1 million people develop symptoms attributable to their gallstones, and 500 to 600 thousand patients per year require cholecystectomy to treat their disease. About 10 to 20% of these procedures are performed for acute cholecystitis. Gallstones and gallstonerelated diseases are therefore a significant medical and economic problem in our society. Similarly, gallstones are among the most common disorders affecting humanity throughout the world. Although dissolution therapy and extracorporeal shockwave lithotripsy can successfully treat selected patients with gallstones, cholecystectomy is the mainstay of gallstone treatment. It is without doubt the treatment of choice in individuals who develop acute cholecystitis. Laparoscopic cholecystectomy, introduced within the last 10 years, decreases postoperative pain and hastens postoperative recovery but does not decrease the morbidity and mortality of cholecystectomy. Its role in treating patients with complicated gallstone disease has not been fully established and its evolving. This chapter describes biliary colic, the most common symptom of gallstone disease, and compares it with the signs and symptoms of acute cholecystitis, the most common complication of gallstones. The diagnosis and treatment of acute cholecystitis are also discussed. Most patients with gallstones do not have symptoms. The majority of gallstones remain silent and undetected for many years, although some are incidentally detected by ultrasound, computed tomography (CT) of the abdomen, abdominal xrays ordered for another reason, or during another abdominal operation when the surgeon palpates them. In the past, it was believed that all gallstones eventually caused symptoms and that patients with untreated gallstones were at great risk for complications of gallstone disease. Cholecystectomy was, therefore, uniformly recommended for patients with known but asymptomatic gallstones. Several studies (1– 4) over the past two decades have addressed the natural history of asymptomatic gallstones and have found that "silent" gallstones remain asymptomatic for long periods of time. Only about 1 to 2% of patients per year with asymptomatic gallstones develop symptoms or complications requiring operative intervention, and the risk of developing symptoms appears to plateau after about 15 to 20 years. Careful risk analysis using data from such studies and outcomes from operative therapy indicate that, in general, the risk of simply observing patients with asymptomatic gallstones is equal to or less than that associated with prophylactic cholecystectomy, and currently cholecystectomy is not recommended for most patients with silent gallstones. On the other hand, surgery is recommended for patients who have symptoms caused by their gallstones.
Page 472
II— Biliary Colic: The Most Common Sign of Gallbladder Disease A— The Clinical Presentation of Biliary Colic Some patients present initially with a complication of their gallstones, such as acute cholecystitis, gallstone pancreatitis, or obstructive jaundice. However, three quarters of patients with gallstones present with attacks of pain, usually in the right upper quadrant or epigastrium, before a complication of gallstone disease occurs. Patients with intermittent attacks of rightupperquadrant abdominal pain and gallstones have symptoms due to gallstones and are said to have chronic cholecystitis. Patients with symptomatic cholelithiasis are clear candidates for cholecystectomy, since the majority of symptomatic patients develop recurrent symptoms, and symptoms will be severe enough in more than 5 to 10% of these patients per year to necessitate cholecystectomy. Moreover, patients with symptomatic gallstones are at higher risk for development of complications of gallstones, such as acute cholecystitis, obstructive jaundice, gallstonerelated pancreatitis, and cholangitis. Gallstones may also be responsible for mild, nonspecific gastrointestinal symptoms such as transient abdominal pain or discomfort, nausea, dyspepsia, and/or flatulence. The latter symptoms are not specific for gallstones, since they may be caused by other gastrointestinal disorders, including gastroesophageal reflux disease, peptic ulcer disease, and spastic colitis. Patients with mild, nonspecific symptoms are similar to patients with asymptomatic gallstones. They are unlikely to develop more significant symptoms or complications and usually do not require operative therapy (1,3). Operative therapy for "mildly" symptomatic patients with gallstones should be reserved for patients with frequent episodes without evidence of another cause after complete evaluation. Even with thorough evaluation, there is a high (10 to 50%) chance that cholecystectomy will not completely relieve the symptoms. Gallstones typically cause intermittent episodes of moderate to severe rightupperquadrant and/or epigastric abdominal pain beginning 15 to 60 min after a meal. These episodes of pain are commonly referred to as biliary colic, although the pain does not come in paroxysms, like those associated with renal calculi. Rather, the pain is constant and unremitting. The attack of pain is often associated with a meal and sometimes with specific foods, including fatty, fried, and spicy foods or dairy products. However, many patients are unable to identify a precipitating event. The pain is often quite severe and may bring the patient to an emergency room. It is often associated with nausea and vomiting, and patients may feel relief after vomiting. Vomiting of bilious (bittertasting) material is commonly described. Vomiting does not persist, as it does with intestinal obstruction; it usually subsides as the abdominal pain improves. In some patients, pain will not be localized to a specific quadrant of the abdomen and may be felt as generalized abdominal pain in the upper or even the lower abdomen. Pain can sometimes radiate to the back or right shoulder. Atypical symptoms from gallstones can mimic pain from acute hepatic disorders, peptic ulcers, reflux esophagitis, perforated viscus, bowel obstruction, and even myocardial ischemia. Likewise, these disorders can cause symptoms that mimic gallstone disease. An episode of biliary colic typically lasts for 15 min to 1 h and rarely lasts longer than 3 to 4 h. If the episode lasts for more than 6 h, one should suspect a complication of gallstone disease, such as acute cholecystitis, choledocholithiasis, or gallstone pancreatitis; otherwise another nonbiliary cause for the patient's abdominal pain should be considered. Unremitting epigastric pain radiating to the center back is highly suggestive of choledocholithiasis or gallstone pancreatitis. True colicky pain suggests renal calculi or early intestinal obstruction. Pain from a perforated peptic ulcer, perforated appendicitis, or diverticulitis usually becomes generalized rapidly as peritonitis spreads throughout the abdominal cavity, and these disorders are associated with significant abdominal tenderness. Pancreatitis may also cause diffuse abdominal pain and tenderness. Attacks of biliary colic occur randomly and are unpredictable. They may be separated by just a few days or by intervals of months or years. The frequency of attacks is often quite
Page 473
stable in a given patient for long periods of time. However, patients who experience frequent attacks or an increase in the frequency or severity of their attacks are likely to continue to experience symptoms, to develop more severe attacks, or to develop complications of gallstone disease. These patients are clear candidates for prompt though elective operative intervention. B— The Diagnosis of Chronic Cholecystitis in Patients with Biliary Colic Typical episodes of biliary colic are characteristic of and very specific for gallstone disease. The diagnosis can frequently be made on the basis of clinical history and physical examination alone, although the presence of gallstones or signs of gallstone disease must be confirmed by ultrasound, cholescintigraphy, or xray studies before treatment is undertaken. Episodes of biliary colic are rarely associated with fever or an elevated white blood cell count since the gallbladder is not acutely inflamed during an attack of biliary colic. Physical examination shows mild to moderate tenderness in the right upper quadrant of the abdomen or epigastrium during an attack, but the tenderness resolves as the pain subsides. Tenderness is never severe and peritoneal signs, such as percussion and rebound tenderness, are absent. If fever, an elevated white blood cell count, significant tenderness, or peritoneal signs are present, a complication of gallstone disease or another diagnosis should be considered (Table 1). Pathological changes in the gallbladder wall do not necessarily correlate with the type or degree of symptoms exhibited. Patients with symptoms of recent onset often have marked chronic inflammation and fibrosis on microscopic examination when their gallbladders are removed, while patients with a long history of symptoms or with severe symptoms may exhibit minimal pathological changes. Likewise, patients with increased white blood cell counts, fever, and edematous gallbladders at operation clearly have acute cholecystitis but may have chronic, not acute changes on pathological examination. Preoperative clinical findings reliably predict intraoperative and pathological findings, but the reverse is not necessarily true. The diagnoses of acute and chronic cholecystitis should be based on clinical presentation and intraoperative findings rather than pathological diagnosis (5). C— Causes of Biliary Colic The cause of biliary colic is not known, but it is hypothesized that the pain occurs when a gallstone becomes lodged in the cystic duct. Gallbladder distention and contractions of the gallbladder in an attempt to eliminate the stone are thought to result in pain. The attack subsides when the stone becomes dislodged. In some patients, small gallstones may actually pass through Table 1 Distinguishing Biliary Colic from Acute Cholecystitis
Biliary colic
Acute cholecystitis
Present
Present
Abdominal tenderness
Absent or mild
Moderate to severe (+) Murphy's sign
Fever
Absent or lowgrade
Usually present
White blood cell count (>11,000)
Absent
Usually present
Duration of symptoms
<4 h
>6 h
Ultrasound
Gallstones
Gallstones Wall thickening Ultrasound Murphy's sign
Radionucleotide cholescintigraphy
Gallbladder visualized within 4 h
No visualization of the gallbladder
Rightupperquadrant abdominal pain
Page 474
the cystic duct and then through the common bile duct into the duodenum, causing the symptoms. Passage of gallstones into and through the common bile duct may or may not be associated with elevations in liver function tests, amylase, or lipase. Pain in these patients more often radiates to the back and across the epigastrium into the left abdomen. Although these mechanisms are plausible and likely cause pain in the majority of patients with gallstones, it is interesting that patients with gallbladder polyps, cholesterolosis of the gallbladder, or acalculous chronic cholecystitis often have attacks of biliary colic. Biliary colic in these patients is indistinguishable from the pain experienced by individuals with cholelithiasis, even though there are no gallstones to obstruct the gallbladder outlet. The cause of biliary colic when gallstones are not present is poorly understood. Interestingly, biliary colic responds promptly to nonsteroidal antiinflammatory drugs (NSAIDs). A single dose of 75 mg of diclofenac provided satisfactory pain relief for attacks of biliary colic and decreased the progression to acute cholecystitis (6). These data suggest that inflammation and inflammatory mediators may play a key role in the development of symptomatic gallstones and complications associated with gallstone disease. Neither the inflammatory events leading to pain or the exact mechanism of action of NSAIDs is currently known. D— Treatment of Biliary Colic Most attacks of biliary colic are selflimiting and resolve spontaneously. Severe attacks and pain that persists lead patients to seek medical advice, often through an emergency room. These patients are in distress and require pain medication. Although narcotics theoretically increase biliary pain by causing spasm of the sphincter of Oddi, patients with biliary colic usually respond promptly to a single dose of narcotic analgesia. As mentioned above, NSAIDs provide effective treatment for biliary colic and may avert progression to acute cholecystitis (6). Patients with significant nausea and vomiting require nasogastric decompression. Further therapeutic interventions are rarely needed, but establishment of gallstones as the cause for the pain is required. Persistence or recurrence of symptoms after adequate pain medication suggests a complication of gallstone disease or another diagnosis. Further evaluation and treatment for patients with suspected acute cholecystitis is described below. III— The Pathogenesis and Diagnosis of Acute Cholecystitis Acute cholecystitis is an acute inflammatory disease of the gallbladder that results in edema of the gallbladder wall, infiltration of the gallbladder with acute or chronic inflammatory cells, infection, vascular compromise, gallbladder wall necrosis, and/or perforation of the gallbladder with abscess formation or peritonitis. In approximately 95% of patients, acute cholecystitis is due to gallstones, but in 5% gallstones are absent. Acalculous cholecystitis can be a particularly virulent form of the disease and may be more difficult to diagnose. It should be considered in any patient with suggestive symptoms who does not have gallstones on imaging studies. If untreated, both calculous and acalculous acute cholecystitis can progress to gangrene and perforation, two complications which are life threatening. The diagnosis and prompt treatment of acute cholecystitis is thus important if the morbidity and mortality of gallstone disease is to be minimized. A— Pathogenesis of Acute Cholecystitis Several factors have been identified that contribute to the development of acute cholecystitis (Fig. 1). Obstruction of the cystic duct is the most common abnormality associated with acute cholecystitis. However, such obstruction is not present in all patients with acute cholecystitis, and not all patients with cystic duct obstruction develop acute cholecystitis. Other factors—
Page 475
Figure 1 Several factors have been associated with the development of acute cholecystitis, including alterations in the composition of the bile when it is supersaturated, obstruction of the cystic duct with increased intraluminal pressure and gallbladder distention, bacteria in bile, and decreased blood flow to the gallbladder mucosa and wall. Although each of these factors may in itself cause an episode of acute cholecystitis, most patients who develop acute cholecystitis have two or more factors contributing to their disease.
such as supersaturated bile, biliary sludge, biliary crystals, and bacteria—may also cause or contribute to the pathogenesis of acute cholecystitis. In the majority of patients, the development of acute cholecystitis requires the presence of two or more factors. Most patients with acute cholecystitis have complete obstruction of the cystic duct. Usually, impaction of a gallstone in the cystic duct is the cause of the obstruction, but tumor (7), edema, and fibrosis of the duct may also obstruct the gallbladder outlet. Based on clinical and gross pathological observations, obstruction of the cystic duct with stasis of bile in the gallbladder lumen is believed to be the precipitating event for acute cholecystitis in most although not all patients. About 1 to 3% of patients with acute cholecystitis do not have cystic duct obstruction. Patients without cystic duct obstruction account for falsenegative radionucleotide scans (see below). In some individuals, cystic duct obstruction leads to hydrops of the gallbladder rather than acute cholecystitis. The role of cystic duct obstruction in acute cholecystitis has been explored in animal models of acute cholecystitis induced by ligating the cystic duct. Gallbladders containing normal bile develop hydrops, not acute cholecystitis. In contrast, ligation of the cystic duct in animals fed a lithogenic diet uniformly results in acute cholecystitis (8), indicating that the presence of supersaturated bile in the gallbladder lumen is required for acute cholecystitis to develop in animals. These findings are pertinent to humans, since most patients with gallstones have bile supersaturated with cholesterol, calcium bilirubinate, or another component. Clinical and experimental observations strongly suggest that stasis alone is insufficient for the development of acute cholecystitis and that the contents of the gallbladder are important in the development of acute inflammation in the gallbladder wall. The development of acute inflammation in the gallbladder wall requires a break in the normal defense mechanisms that protect the gallbladder epithelium from its luminal contents. As stated above, cystic duct obstruction is the most common precipitating factor and facilitates progression of the disease. Obstruction distends the gallbladder, increases intraluminal gallbladder pressure, breaks the integrity of the gallbladder epithelium, and increases exposure of the gallbladder wall to components of gallbladder bile or to bacteria or noxious agents in it.
Page 476
Proinflammatory factors in the lumen then induce an intense inflammatory response in the gallbladder wall, leading to pain, fever, and an increased white blood cell count. On the other hand, the pathogenesis of acute cholecystitis in unobstructed, undistended gallbladders is not well understood. Acalculous acute cholecystitis is more common in individuals on parenteral alimentation and in critically ill patients. The gallbladder becomes enlarged during parenteral alimentation because of a lack of the hormonal stimulation that accompanies oral feeding. Likewise, critically ill patients are often unable to take oral feedings, leading to gallbladder stasis. It is hypothesized that stasis of bile in the gallbladder leads to accumulation of toxic agents in the gallbladder lumen. Prolonged exposure to concentrated bile damages the gallbladder mucosa, leading to inflammation and acute cholecystitis. The factors responsible for inflammation have not been clearly defined, but several potential factors have been identified. Since most patients with acalculous cholecystitis do not have patent cystic ducts, it is likely that gallbladder obstruction plays a role in the pathogenesis of many of their disorders also. Obstruction of the cystic duct is likely due to swelling and edema caused by inflammation in the vicinity of the duct. The toxicity of bile seems to be greater if it is supersaturated with cholesterol or calcium bilirubinate. Patients with prolonged gallbladder stasis often develop ''gallbladder sludge," which consists of concentrated biliary lipids (bile salts and phospholipids), mucus, and biliary crystals composed of cholesterol, calcium bilirubinate, or calcium salts with the inorganic ions, carbonate, or phosphate and small stones (microlithiasis) (9–12). Gallbladder sludge may be eliminated from the gallbladder when the patient's illness and gallbladder stasis resolves, or it may progress to gallstone formation if it is not emptied from the gallbladder. Gallbladder sludge is also associated with an increased risk of the biliary complications usually attributed to gallstones, especially acute cholecystitis and gallstone pancreatitis. Sludge alone is probably capable of inducing an inflammatory response in the gallbladder wall. Vascular compromise has also been implicated in the development of acute cholecystitis, especially in critically ill patients, who are subject to episodes of hypotension with reduced blood flow to visceral organs, including the gallbladder. Reduced blood flow, ischemia, and reperfusion injury damages the gallbladder and decreases its natural resistance to injurious agents within its lumen. Although decreased blood flow to the gallbladder may be important in critically ill patients, vascular compromise does not initially play a role in most cases of acute cholecystitis. More than half of patients with acute cholecystitis develop hyperemia of the gallbladder wall due to the inflammatory response associated with the disease (7,13). Decreased blood flow to the gallbladder wall occurs later, when the patient's problem progresses from uncomplicated acute cholecystitis to gangrene. Colonization of bile with bacteria is common in otherwise asymptomatic patients; bacteria do not cause infection in the biliary ducts or gallbladders under normal circumstances. Usually a break in biliary epithelial integrity or a decrease in gallbladder defense mechanisms is required for invasive bacterial infection to occur. Although, when they are present, bacteria play an important role in the progression of acute cholecystitis. They are cultured from bile in only about 50% of patients with acute cholecystitis but less often early during the course of the disease. It is felt that they play a secondary rather than a primary role in acute cholecystitis. Later infectious complications of acute cholecystitis—such as cholangitis, perforation, abscess formation, peritonitis, and sepsis—are clearly caused by bacterial overgrowth and are largely responsible for the morbidity and mortality associated with this disease. While the majority of patients with bacteria in their bile do not develop clinical infections without other mitigating factors, particularly virulent strains of Escherichia coli, Clostridium species, or Salmonella species are sometimes primarily responsible for acute cholecystitis. In addition to causing acute cholecystitis in the presence of gallstones, these organisms are also responsible for some of the cases of acute acalculous cholecystitis. Acute cholecystitis differs from chronic cholecystitis by the intense inflammatory response that is present in the gallbladder wall. This inflammatory response is first manifest by edema of the gallbladder wall and later by the infiltration of leukocytes. The combination of
Page 477
increased intraluminal pressure and inflammation leads to damage of the mucosa. Inflammation initially causes hyperemia, but eventually perfusion to the gallbladder decreases as edema and increased intraluminal gallbladder pressure compromises venous outflow and then arterial inflow to the gallbladder wall. Bacteria in bile invade the gallbladder wall when defense mechanisms are overwhelmed, establishing an invasive infection. If unchecked, the process will progress to transmural necrosis of the gallbladder wall and to gallbladder perforation. 1— Mediators of Gallbladder Inflammation Several components of normal and saturated gallbladder bile have been implicated in the development of gallbladder inflammation. Clearly, bacteria induce inflammation when they invade the gallbladder wall, but a lack of disease in most patients with bacteribilia emphasizes that the presence of bacteria in bile alone usually does not lead to acute cholecystitis. As mentioned above, bacteria likely play a secondary role in this disease and are not thought to initiate the process in the great majority of patients. Clearly, infection is important later, as the disease progresses from uncomplicated to complicated acute cholecystitis. The observation that supersaturated bile is critically important for the development of acute cholecystitis in animals implies that some components of supersaturated bile may be injurious to gallbladder epithelia or capable of inducing an inflammatory response in the gallbladder wall. Bile salts, the primary lipid in bile, are responsible for the solubility of cholesterol and bilirubin in bile. They act as detergents, which, with phospholipids and cholesterol, form mixed micelles. Hydrophobic compounds intercalate into or bind onto micelles and are thus solubilized in what is otherwise an aqueous medium. On the other hand, the detergent properties of bile salts may be deleterious. Bile salts are capable of damaging epithelial layers by leaching phospholipids and cholesterol from cell membranes. Considering this, it is quite remarkable that the gallbladder, which is exposed to high concentrations of bile salts (50 to 250 mM), does not selfdigest. The normal gallbladder is extremely resistant to the effects of bile salts. Phospholipid and cholesterol in bile normally ameliorate the toxic effects of bile salts by fully saturating the micelles. Bile salts may contribute to damage when the gallbladder epithelium is compromised, and supersaturated bile containing altered concentrations of biliary lipids, shifts in bile salt profile from trihydroxy to more toxic dihydroxy bile salts, and alterations in the composition of biliary phospholipids may be even more toxic. Lysolecithin is a toxic phospholipid that is normally present in bile in small amounts. Its concentration increases in gallstone patients and patients with obstruction of the gallbladder or who undergo injury to the gallbladder mucosa. Lysolecithin, formed from lecithin by the enzyme phospholipase A, has been recognized as a potential mediator of gallbladder epithelial damage and acute cholecystitis for more than two decades (14–16). Alterations in lysolethicin concentrations, especially in combination with cystic duct obstruction or decreased blood flow/ischemia, likely contribute to gallbladder epithelial damage and inflammation of the gallbladder wall. Prostaglandins have also been suggested as inflammatory mediators of acute cholecystitis, but they probably do not initiate the process. They are known mediators of inflammation in other organs, and prostaglandin concentrations are increased in inflamed gallbladders. In addition, inhibitors of prostaglandin synthesis, including indomethicin and aspirin, ameliorate the inflammatory response accompanying an attack of acute cholecystitis (17–21). It is likely that prostaglandins play a secondary role in this disease. Elevated prostaglandin concentrations in the gallbladder wall are probably triggered by any injury to the gallbladder wall and by other inflammatory mediators. Recently, our laboratory examined the inflammatory response observed in the gallbladder wall during early gallstone formation in all animal models. Our studies clearly show that biliary crystals composed of cholesterol and bilirubin are "inflammatory" crystals. That is, they are capable of inducing an inflammatory response from polymorphonuclear leukocytes and monocytes in vitro and in the gallbladder wall in vivo (22–25). Since crystals are commonly found
Page 478
in individuals who have supersaturated bile and stasis, our data suggest that the presence of crystals in gallbladder bile contributes to inflammation of the gallbladder wall in acute and chronic cholecystitis, as they do during gallstone formation and growth in animals. Crystal induction of inflammation is not proven, but is is an attractive hypothesis to explain the roles played by supersaturated bile and gallbladder sludge in the development of clinical gallbladder disease. IV— Signs, Symptoms, and Laboratory Abnormalities Accompanying Acute Cholecystitis Acute cholecystitis commonly manifests itself as right upper quadrant abdominal pain with characteristics similar to biliary colic (Table 1). Pain is sometimes mild but more often severe. The pain is perceived in the right upper quadrant, sometimes in the epigastrium, or occasionally diffusely in the upper abdomen. Referred pain may also be experienced in the right shoulder, the back, or the chest. Nausea and vomiting often accompany the pain, but vomiting does not result in pain relief, as it does with biliary colic. An attack of acute cholecystitis is often preceded by a history of attacks of biliary colic, pointing to the gallbladder as the source of the acute problem. Atypical pain is sometimes mistaken for the pain caused by perforated ulcer, acute pancreatitis, acute gastritis, or even myocardial ischemia. Acute cholangitis can also mimic the findings of acute cholecystitis, but the patient usually has a high white blood cell count, significant fever, and jaundice. The triad of right upperquadrant pain, jaundice, and fever (Charcot's triad) is characteristic of cholangitis and distinguishes it from acute cholecystitis. The one factor that clearly distinguishes pain due to acute cholecystitis from biliary colic is the duration of the pain. Biliary colic rarely lasts for more than several hours, while pain from acute cholecystectomy persists. Acute cholecystitis should be considered if pain lasts longer than 6 h. In addition, the pain from acute cholecystitis is accompanied by localized rightupperquadrant tenderness. The tenderness is usually well localized directly over the gallbladder. Typically, tenderness increases under a physician's palpating hand placed just below the costal margin in the midclavicular line as the patient takes a deep breath. The increased tenderness results from the inflamed gallbladder descending below the costal margin and striking the examiner's hand. The increase in localized tenderness with this maneuver is referred to as Murphy's sign and is very specific for acute cholecystitis. The localized tenderness and Murphy's sign observed with an attack of acute cholecystitis must be distinguished from the pain caused by acute inflammation and swelling of the liver with acute hepatitis or with liver congestion due to right heart failure. Liver tenderness with the latter disorders is diffuse and elicited along the entire edge of the liver. It extends into the left upper quadrant of the abdomen, since the left lobe of the liver is also involved. Although tenderness also increases with a deep breath, the response is evoked diffusely and is not localized specifically over the gallbladder. Careful examination of the patient's abdomen is imperative to determine whether abdominal tenderness is diffuse or localized and whether the tenderness truly lies in the area where the gallbladder is expected. Lowerlobe pneumonia can also cause rightupperquadrant abdominal pain and shoulder pain if the process irritates the diaphragm. Tenderness in the right upper quadrant is absent. Pain due to pneumonia is distinguished from acute cholecystitis by symptoms of a respiratory problem, findings consistent with pneumonia on examination of the chest and consolidation of a segment of the right lower lung on chest xray. Tenderness in the right upper quadrant of the abdomen may be associated with localized percussion and rebound tenderness, but peritoneal signs are rarely generalized until a complication of acute cholecystitis has occurred. Diffuse peritoneal signs indicate generalized peritoneal irritation and suggest other acute abdominal disorders, such as perforated viscous, acute pancreatitis, small bowel obstruction, acutely ischemic bowel, diverticulitis, or acute appen
Page 479
dicitis. However, perforation of the gallbladder may also cause diffuse peritonitis. It can sometimes be distinguished from other abdominal catastrophes by the history of the illness. Peritonitis is a relatively early event (several hours to 3 days) with perforated viscous, acute pancreatitis, diverticulitis, and acute appendicitis, but gallbladder perforation is rare until 7 to 10 days after the symptoms of acute cholecystitis are apparent. However, a subgroup of patients with acute cholecystitis, especially those with acalculous cholecystitis, progress rapidly to gangrene and perforation. Pain and tenderness are usually but not always associated with fever and an increased white blood cell count. Temperature is rarely higher than 102°C and white blood cell count higher than 15,000 with uncomplicated acute cholecystitis. However, the absence of fever and leukocytosis does not exclude acute cholecystitis, especially in elderly and immunocompromised patients. Blood pressure is usually normal. Hypotension and confusion associated with rightupperquadrant abdominal pain, jaundice, and fever (Reynolds' pentad) is characteristic of acute cholangitis rather than acute cholecystitis. The gallbladder is usually not palpable, but it does become distended and surrounded by omentum and the right colon. The gallbladder or the gallbladder with surrounding phlegmon may be palpated late in the course of the disease. A mass of the right upper abdomen may also be present after gallbladder perforation if an abscess develops in the subhepatic space. A palpable gallbladder from acute cholecystitis must be distinguished from a palpable gallbladder due to chronic obstruction from bile duct tumors and benign strictures. In the latter case, the palpable gallbladder is not tender and jaundice is present. Bile duct tumors and strictures also cause dilatation of the intra and extrahepatic bile ducts. Symptoms and physical findings associated with acute cholecystitis may be greatly reduced in elderly patients (26–28). Of 168 patients 65 years of age or older who presented to an emergency room, 141 (84%) did not have rightupperquadrant or epigastric pain. Eight patients had no pain. Over half of the patients were afebrile and about 40% did not have increased white blood cell counts (26). Although Prousalidis et al. (28) noted that the clinical presentation of 47 patients with acute cholecystitis in 62 patients above age 70 was mild, 10 patients had gangrenous cholecystitis and 7 had choloperitoneum. The classic signs and symptoms of acute cholecystitis are frequently absent in elderly patients, and the presentation of elderly patients is often atypical. Thus, the clinical presentation does not correlate with the severity of the disease in elderly patients. A high index of suspicion for acute cholecystitis is important in any elderly patient who presents with abdominal complaints and/or sepsis. Acute cholecystitis should be part of the differential diagnosis in every elderly patient who presents with unexplained abdominal pain and tenderness. When diagnosed, acute cholecystitis should be treated aggressively in this age group. Although most patients with acute cholecystitis develop symptoms severe enough to warrant hospitalization, a few patients present with less impressive findings. They are sometimes said to have subacute cholecystitis. Typically, these patients present to an emergency room or physician with clear rightupperquadrant pain and tenderness. White blood cell counts are normal and the patient does not have fever, although nausea and anorexia are commonly present. The patient's pain responds to pain medication initially, a diagnosis of biliary colic is made, and the patient is discharged home. However, careful questioning later reveals that the patient's pain and tenderness never completely resolved or that it returned as soon as the effects of the pain medication subsided. The patient limits his or her diet to liquids, because a regular diet worsens symptoms; several days of discomfort then follow before the patient presents to another physician. At operation, these patients exhibit marked inflammation of the gallbladder and have acute cholecystitis indistinguishable from a more typical presentation of the disease. Recognition that these patients have acute cholecystitis is important, because they are at risk for complications of the disease and, with time, progression of inflammation increases the difficulty of both open and laparoscopic cholecystectomy. Patients discharged from the emergency room after "resolution" of an episode of presumed biliary colic should be instructed to return if their symptoms recur or persist. Persistence of symptoms may be the only evidence
Page 480
that the patient has developed a complication of gallstone disease such as acute cholecystitis, choledocholithiasis, or gallstone pancreatitis, requiring prompt and appropriate treatment. Acute cholecystitis is more common in critically ill and postoperative patients, but the diagnosis is difficult to establish. Critically ill patients frequently cannot respond appropriately to questioning about symptoms or to physical examination. If not mentally impaired by their illness, they are often heavily sedated in order to facilitate ventilatory assistance or to decrease agitation in the intensive care unit. Postoperative patients have pain due to their operation and are usually taking medications that mask the pain of acute cholecystitis. Fever and elevated white blood cell counts in both groups are attributed to the primary disease or to other complications, such as pneumonia, urinary tract infection, infected catheters, and wound infection. The only signs or symptoms of acute cholecystitis in these patients may be prolonged ileus, unexplained bacteremia, or ungoing sepsis. A— Confirmation of the Diagnosis of Acute Cholecystitis There are no specific laboratory tests for acute cholecystitis. An elevated white blood cell count confirms the presence of an acute inflammatory process but is not specific for gallbladder disease. Liver function tests are helpful in excluding other liver diseases, but liver enzymes and bilirubin may be mildly to moderately elevated with acute cholecystitis alone. Serum bilirubin levels below 3.0 mg/dL are common with severe acute cholecystitis, but values above 5 to 6 mg/dL are rarely due to acute cholecystitis and more reliably indicate that the patient has choledocholithiasis. Measurement of serum amylase or lipase is useful in excluding acute pancreatitis. Although mild to moderate elevations of transaminases, alkaline phosphatase, and bilirubin occur with acute cholecystitis, marked elevations are more consistent with hepatitis, cholangitis, and obstructive jaundice. Abdominal xrays are not useful in establishing the diagnosis of acute cholecystitis but may exclude free intraperitoneal air and small bowel obstruction. Occasionally, a calcified gallstone is seen in the gallbladder or air is noted in the gallbladder lumen or wall, indicating that the patient has emphysematous cholecystitis. Ultrasound is a noninvasive procedure that can detect gallstones in the gallbladder in more than 95% of patients. It is less able to detect stones of the common bile duct but reliably demonstrates dilated intra and extrahepatic bile ducts when present. The combination of typical signs and symptoms of acute cholecystitis and the presence of gallstones on ultrasound examination is diagnostic of acute cholecystitis in more than 90% of patients. When symptoms are less typical, the diagnosis is less reliable, since asymptomatic gallstones are common and may be incidentally demonstrated in individuals with upper abdominal symptoms from another problem unrelated to gallstones. Ultrasound can also demonstrate findings that are more specific for acute cholecystitis. Thickening of the gallbladder wall correlates well with inflammation and edema in the gallbladder wall but is often not present with acute cholecystitis. Sometimes, a thickened gallbladder wall is due to chronic fibrosis rather than acute inflammation. An ultrasonographic gallbladder Murphy's sign may also be helpful when present. The ultrasonographer visualizes the gallbladder with the ultrasound probe and then compresses the gallbladder under direct visualization. Production of tenderness specifically located over the gallbladder but not elsewhere with a deep breath suggests acute cholecystitis. However, the test is subject to examiner variability. Color Doppler imaging of the gallbladder wall in normal gallbladders and in gallbladders during acute cholecystitis has been studied (29,30). Most patients with acute cholecystitis have increased blood flow to the mid and fundal gallbladder wall. However, a patient with gangrene or necrosis of the gallbladder wall has no flow detected in the gallbladder wall, and Doppler imaging will not be useful in establishing the diagnosis. Jeffrey et al. demonstrated that the length of the cystic artery visualized with color Doppler imaging correlated with the diagnosis of acute cholecystitis, but only 26% of patients with acute cholecystitis had a cystic artery length longer than half of the gallbladder length, the criteria for a diagnosis of acute chole
Page 481
cystitis (29). Flow in the distal quartile of the gallbladder is particularly reliable in diagnosing acute cholecystitis, but occurs in only 19% of patients (29). Radionuclide cholescintigraphy with a derivative of iododiacetic acid or a related compound is the most specific test for acute cholecystitis. The radionuclide is injected intravenously and is rapidly extracted by the liver. The agent is then excreted into bile by the hepatocyte, thus making it possible to visualize the biliary tract. In a normal study, the cystic duct of the gallbladder is patent and the gallbladder fills with radionuclide. Visualization of the gallbladder excludes acute calculous cholecystitis in 98% of the cases. Filling of a normal gallbladder is rapid and usually occurs in the first hour. A chronically diseased gallbladder fills more slowly—thus, the need to wait 3 to 4 h before concluding that the test has failed to visualize the gallbladder. On the other hand, failure to visualize the gallbladder by 3 to 4 h indicates that the cystic duct is not patent, strongly suggesting acute cholecystitis. Radionucleotide cholescintigraphy is accurate in 97 to 98% of patients, but patients with acalculous cholecystitis do not always have cystic duct obstruction, and the gallbladder visualizes in about 1 to 3% of patients with acute cholecystitis. In addition, a radionuclide scan is not very accurate in critically ill patients or patients on parenteral nutrition. The gallbladder is distended in these patients and contains thick, concentrated bile that interferes with entry of radionuclide into the gallbladder. False positives (nonvisualization of the gallbladder) may be obtained from patients who do not have acute cholecystitis. Patients with hepatocellular disease or severe cholangitis may have poor uptake of radionuclide or poor secretion into the biliary tract from the liver. Poor visualization of the liver with delayed secretion of radionuclide into the biliary tract, allowing renal excretion of the radioactive tracer, suggests hepatocellular disease or ascending cholangitis. Such scans are best considered inconclusive, since they are do not exclude acute cholecystitis and are not specific for any hepatic or biliary tract disease. Finally, failure of radionuclide to enter the duodenum may be due to bile duct obstruction but is more often an artifact due to administration of pain medications that increase the tone of the sphincter of Oddi (31). In fact, narcotic medication is given by some to increase this tone and thus the likelihood of gallbladder filling in an attempt to decrease falsepositive tests for acute cholecystitis. Although not the primary test for acute cholecystitis, computed tomography (CT) is frequently performed in patients with unexplained abdominal pain or sepsis. CT is very useful in excluding other disorders that might mimic acute cholecystitis and may show specific signs of acute cholecystitis. Findings of acute cholecystitis include gallbladder wall thickening, pericholecystic stranding, distention of the gallbladder, pericholecystic fluid, highattenuation bile, and subserosal edema (32). Although established CT criteria are reliable when present, only 51% of patients met such criteria for acute cholecystitis (32). Therefore a normal CT scan does not exclude acute cholecystitis. Since the diagnosis of acute cholecystitis in critically ill patients is very difficult, we (33) and other (34–37) have used percutaneous aspiration of bile with Gram's stain and culture as a diagnostic test in these patients. Percutaneous drainage of the gallbladder following aspiration may also aid in the diagnosis as well as being therapeutic if the patient has acute cholecystitis (see below). Patients who have negative Gram's stains and cultures and who do not respond to drainage by 12 to 24 h often have another source of sepsis (33). V— Complicated Acute Cholecystitis Physicians should be aware of several variants of acute cholecystitis that may be more difficult to diagnose or have a more virulent course. A— Acute Acalculous Cholecystitis Acute acalculous cholecystitis is difficult to diagnose because it is rare, occurring in less than 5% of patients with acute cholecystitis. The diagnosis is often delayed because many physicians
Page 482
assume that the patient does not have acute cholecystitis if ultrasonography excludes gallbladder stones. Acalculous cholecystitis seems to be a more virulent disease and patients appear to progress to gangrene and perforation more rapidly than is usual with cases of acute cholecystitis in general. This observation seems to hold true even if delays in diagnosis are removed as a factor. Prompt diagnosis and timely treatment require that, unless another definitive cause for the symptoms can be found, the clinician caring for the patient consider this diagnosis in all patients with rightupperquadrant pain and tenderness who do not have gallstones by ultrasound. The diagnosis of acute acalculous cholecystitis is best established with radionuclide cholescintigraphy, but it should be understood that some patients will not have cystic duct obstruction and will have falsenegative examinations. In such patients, the surgeon may have to rely solely on clinical findings. Progression of rightupper quadrant tenderness, increasing white blood cell count, and persistent fever facilitates the decision to operate. In critically ill patients and those who develop acute cholecystitis after other operations, a decision to operate may be more difficult and more risky. Percutaneous transhepatic aspiration and drainage may be helpful (33,34–37) and therapeutic if the process has not progressed to gangrene. B— Emphysematous Cholecystitis Emphysematous cholecystitis occurs when the gallbladder is infected with gasforming bacteria. The gas may be seen in the gallbladder wall or in the lumen of the gallbladder on plain xray or CT scan. Emphysematous cholecystitis is more common in diabetics and immunocompromised patients. It is particularly virulent, progressing rapidly to gangrene and perforation. Appropriate antibiotic coverage should be instituted and fluid and electrolyte abnormalities corrected promptly. Emergent operative intervention is warranted to remove the source of infection and avoid perforation, with abscess formation or peritonitis. C— Gangrenous Cholecystitis Gangrenous cholecystitis occurs when vascular compromise of the gallbladder wall occurs. Although gangrene of the gallbladder may develop rapidly in some patients with virulent bacteria or with acute acalculous cholecystitis, it also occurs later in the course of untreated or partially treated acute cholecystitis. The transition from uncomplicated to gangrenous acute cholecystitis can be difficult to establish clinically. Once high fever, increasing pain and tenderness, and marked increases in white blood cell count occur, gangrenous cholecystitis is well established. In addition, elderly patients may present with minimal findings yet have gangrenous cholecystitis. Patients present with gangrenous acute cholecystitis in three ways: a virulent form of acute cholecystitis that progresses to gangrene early, gangrene resulting from a delay in diagnosis or treatment, and gangrene that develops in patients who appear to be adequately treated but who suddenly become worse. Cases of gangrene attributable to delay in diagnosis or inadequate treatment are avoidable. Early operative treatment of acute cholecystitis is the procedure of choice if the patient is a candidate for operative intervention because it avoids or promptly treats patients in all groups (see Sec. VI, "Treatment of Acute Cholecystitis"). D— Perforated Gallbladder Perforation of the gallbladder is a complication of acute cholecystitis that greatly increases morbidity and mortality. In the late stages of acute cholecystitis, vascular supply to the gallbladder wall becomes compromised, leading to devitalization of tissue. Eventually, increased intraluminal pressure leads to perforation and leakage of gallbladder contents. Purulent drainage and, if the cystic duct is patent, bile causes a severe inflammatory response. If this process is localized by surrounding organs (colon, liver, stomach, duodenum, and omentum), a pericholecystic or rightupperquadrant abscess results. Pericholecystic abscess in immunocompetent patients presents with marked rightupperquadrant tenderness, high white blood cell count (greater than 15,000), and persistent, high fever. Clinical and laboratory findings persist despite
Page 483
the institution of adequate medical treatment for acute cholecystitis. The palpable mass may be an enlarged gallbladder, but more often it consists of a phlegmon of the surrounding stomach, colon, and omentum—the organs that confine the process to the right upper quadrant of the abdomen. Immunocompromised and elderly patients may not exhibit expected symptoms and, as with other complications of acute cholecystitis, a high index of suspicion for gallbladder perforation is warranted in these patients. CT scan of the abdomen best diagnoses the abscess and may show evidence of gallbladder inflammation. Treatment consists of antibiotic therapy and prompt drainage of the abscess. Although percutaneous drainage of the abscess will stabilize septic patients, the necrotic gallbladder wall acts as a nidus of infection, prolonging recovery or resulting in failure of drainage. In individuals who are acceptable candidates for operation, the gallbladder should be removed promptly. In approximately onehalf of patients with gallbladder perforation, free perforation into the peritoneum occurs or a direct connection to another organ is established, causing formation of a fistula. Free perforation leads to diffuse abdominal tenderness, a rigid abdomen, and signs of generalized sepsis, since the contents of the gallbladder contain bacteria and—if the cystic duct is patent—bile. The exact cause of the peritonitis may not be apparent when the patient presents and other causes of peritonitis—including perforated viscus, acute appendicitis, and acute diverticulitis—must be considered. Emergent laparotomy is required as soon as fluid and electrolyte deficits are corrected. Mortality and morbidity are high, especially if sepsis and/or renal failure develop. Fistula formation most commonly develops between the gallbladder and the duodenum, stomach, or colon. A fistula between the gallbladder and the stomach or duodenum may actually relieve the patient's symptoms, and commonly causes no late clinical problems. A colonic fistula exposes the gallbladder and biliary tract to high concentrations of enteric organisms and may lead to persistent abscess, ascending cholangitis, or sepsis. In some patients, a large gallstone passes into the gastrointestinal tract through the fistula. The stone can migrate through the intestinal tract without sequelae, but if it becomes impacted at a narrow region of the intestine, such as the ileum or the sigmoid colon, intestinal obstruction ensues. The patient then presents with the typical signs and symptoms of a bowel obstruction. A history of biliary colic or recent acute cholecystitis may not be forthcoming. This entity, termed gallstone ileus, is diagnosed by abdominal xrays that demonstrate the classic findings of intestinal obstruction and the presence of air in the biliary tract. The air in the biliary tract outlines the ductal system and is located centrally. It must be distinguished from air in the portal system, which is found more peripherally in the liver parenchyma. In a minority of patients, a calcified gallstone may also be seen at the point of obstruction. Treatment of gallstone ileus is usually limited to laparotomy with removal of the gallstone to relieve the bowel obstruction, since recurrent gallstone ileus occurs in only 1 of 25 patients. Late complications attributable to gallbladder or the gallbladderintestinal fistula are rare. However, gallbladder removal at the initial operation may be considered if the patient is stable and the gallbladder contains another large gallstone on palpation, since many surgeons feel that the chance of recurrent intestinal obstruction or symptoms attributable to the gallbladder are increased in such patients. Expectant treatment of the gallbladder is especially recommended for elderly patients or those with significant comorbidities, since the risk of subsequent complications is low. Elective cholecystectomy is warranted for younger, healthy patients. In addition, patients who develop biliary tract symptoms while being observed are candidates for cholecystectomy. E— Salmonella Cholecystitis Salmonella infections have recently increased in the United States due to problems with food handling, increases in international travels, and the AIDS epidemic. Besides gastroenteritis, some patients develop localized infections involving the gastrointestinal tract, especially infections in the gallbladder. Salmonella cholecystitis is reported to occur in about 3% of patients with typhoid fever and is usually associated with preexisting cholelithiasis. Acalculous disease has also been reported to occur with both Salmonella typhi and nontyphoid serotypes. The
Page 484
typical patient develops symptoms of acute cholecystitis approximately 2 weeks after a case of typhoid fever, but acute cholecystitis may begin within days of the onset of gastroenteritis, especially with virulent nontyphoid strains. Medical therapy of Salmonellainduced acute cholecystitis is similar to treatment of acute cholecystitis in general, but antibiotic therapy should be tailored to cover Salmonella species. Ampicillin or chloramphenicol is the recommended initial antibiotic for enteric fever, bacteremia, and selected cases of gastroenteritis. Trimethoprimsulfamethoxole, a thirdgeneration cephalosporin (cefotaxime, ceftriazone, or cefoperazone), or, in adults, a quinolone (ciprofloxacin or norfloxacin) may be utilized, particularly if drug resistance is suspected or documented. Rapid progression of the disease, perforation of the gallbladder, gangrene, empyema, and cholangitis occur more frequently with Salmonellainduced acalculous cholecystitis. Therefore, in the absence of medical contraindications, early operative therapy is recommended in all patients with a clear diagnosis of Salmonellainduced acute cholecystitis. Laparoscopic cholecystectomy has been safely performed for Salmonellainduced acute cholecystitis (38–43). VI— Treatment of Acute Cholecystitis A— Medical Treatment of Acute Cholecystitis: Initial and Nonoperative Treatment Once the diagnosis of acute cholecystitis is established, the patient should be admitted to the hospital, given nothing by mouth, and begun on parenteral fluids. A nasogastric tube should be placed if the patients has persistent nausea and vomiting. Intravenous parenteral antibiotics should be started to avert serious infectious complications of acute cholecystitis. Antibiotic therapy should be directed to control the most common organisms that colonize bile. Enteric bacteria—including E. coli, Klebsiella aerogenes, Streptococcus faecalis, and Proteus species—are cultured from bile alone or in combination in approximately 90% of patients with biliary infections. Pseudomonas, Clostridium, bacteroides, and fungal species are also occasionally isolated from bile but are more commonly cultured from bile in diabetic patients, patients with cancer, and immunosuppressed patients. Classically, ampicillin and an aminoglycoside have been used to treat acute cholecystitis, but adequate coverage can be achieved in uncomplicated patients with a secondgeneration cephalosporin or ampicillin/sulbactam avoiding the nephrotoxicity of aminoglycosides. Broader coverage may be needed in immunosuppressed patients, patients with cancer, and diabetic patients to cover the unusual organisms that are more likely to be present in these patients. Broaderspectrum coverage is also indicated once a complication of acute cholecystitis has occurred. The patient's temperature, white blood cell count, and physical examination should be observed closely. A worsening clinical course necessitates urgent operation. Onefourth of patients develop a complication of acute cholecystitis or recurrent symptoms while under medical treatment. In the patients who respond to medical therapy, cholecystectomy is also indicated to avoid further complications of their gallbladder disease, although the recommended timing of cholecystectomy has varied over the past 30 years. B— Surgical Treatment 1— Delayed Treatment versus Early Cholecystectomy Twenty to thirty years ago, initial medical therapy of acute cholecystectomy followed by elective delayed cholecystectomy was uniformly recommended. It was felt that anesthetic and operative complications were more likely to occur when the patient had an acute illness and an inflamed gallbladder. However, approximately 20 to 30% of patients became worse or developed recurrent symptoms of biliary tract disease and required a more urgent operation. With improvements in medical, surgical, and anesthetic care, biliary surgeons noted that the outcomes after urgent operation when medical therapy had failed were not significantly different from outcomes after elective cholecystectomy and suggested that early cholecystectomy
Page 485 Table 2 Surgical Treatment of Acute Cholecystitis Rationale for delayed cholecystectomy 1. The operation is more difficult with acute inflammation. 2. The rate of bile duct injury is higher with acute cholecystitis. 3. Avoids urgent/emergent surgery. 4. The lowest morbidity and mortality in highrisk patients. Rationale for early cholecystectomy 1. The operation is often less difficult with edema versus fibrosis. 2. Avoids the 20 to 30% failure rate with medical therapy. 3. Studies do not demonstrate increased mortality or morbidity. 4. Lowest total days of hospitalization. 5. Avoids two hospital admissions.
might be advantageous (Table 2). Retrospective and randomized studies (44–48) established low, acceptable morbidity and mortality rates for cholecystectomy early during the course of acute cholecystitis. Moreover, bile cultures were less likely to be positive early during the course of the disease, urgent operations for worsening cholecystitis or for recurrent symptoms were avoided, and overall hospital stay and therefore costs were less with early as compared to delayed cholecystectomy. In fact, Archibald et al. (44) noted that the majority of complications and deaths occurred in the group of patients who underwent operation 7 days or more after hospital admission. Glenn (45) specifically commented that acute cholecystitis was a serious condition in elderly patients but that nonsurgical management resulted in even higher mortality than medical management. About 20 years ago, early cholecystectomy was established as the treatment of choice for acute cholecystitis if the patient was an acceptable operative candidate (Table 3). Table 3 Comparison of Open and Laparoscopic Cholecystectomy for Acute Cholecystitis
Delayed cholecystectomy
Open
Early cholecystectomy
Laparoscopic
Open
Laparoscopic
Positive bile cultures Pathological findings
Failure of medical therapy Conversion rate Medical Rx failure
Chronic inflammation fibrosis
Chronic inflammation fibrosis
20–30%
20–30%
—
4–9%
—
>30%
Acute/chronic inflammation edema —
Acute/chronic inflammation edema — 8–27%
—
Bile duct injury rate vs. elective surgery
— or
Failures medical Rx
—
—
—
—
Mortality/morbidity Failures medical Rx Highrisk patients 5–7 days
5–7 days
1–2 days
1–2 days
Postoperative hospital stay
3–5 days
1 day
3–5 days
1–2 days
Total hospital stay
8–12 days
6–8 days
4–7 days
2–4 days
Preoperative hospital stay
Page 486
It should, however, be understood that patients with significant comorbidities and critically ill patients might not be candidates for early operative intervention. Since acute cholecystitis resolves with medical therapy alone in 70 to 80% of patients, medical therapy with delayed cholecystectomy when the patient's acute medical problems are resolved is still an acceptable option for poor operative candidates. If medical therapy fails to treat the episode of acute cholecystitis in highrisk patients, the patient may be treated safely and effectively by percutaneous transhepatic drainage of the gallbladder (33,34). The introduction of laparoscopic cholecystectomy in the last decade has drastically altered the elective treatment of symptomatic gallstones. Patients have much less postoperative pain, markedly decreased length of hospitalization, and a more rapid return to normal activity compared to patients treated with open cholecystectomy. The technique was rapidly accepted as the procedure of choice by patients and surgeons alike for elective treatment of symptomatic cholelithiasis, but surgeons were more reluctant to use it to treat acute cholecystitis. It was not clear if laparoscopic cholecystectomy could be performed as safely when the gallbladder was acutely inflamed, if conversion rates would be higher than in elective operations, or if patients with acute cholecystitis would reap the benefits (shorter hospital stay and more rapid return to full activity) of laparoscopic cholecystectomy. 2— Open versus Laparoscopic Cholecystectomy a— Is laparoscopic cholecystitis safe for acute cholecystitis? Bender and coworkers (49) prospectively recorded data on 52 patients who underwent laparoscopic cholecystectomy for acute cholecystitis within in 24 h of admission. No patient died and only two complications were reported, a biloma in one patients and a wound infection in another. The conversion rate to open operation of 7.7% was only slightly higher than the reported conversion rates for elective laparoscopic cholecystectomy, and the mean postoperative length of hospital stay of 2.3 days was less than that with open operation. Lujan et al. (50) similarly noted minimal morbidity and mortality and a short postoperative hospital stay in 60 patients undergoing laparoscopic cholecystectomy for acute cholecystitis, but the conversion rate to open operation was somewhat higher, although acceptable, at 13%. Others (51–53) have directly compared patients with acute cholecystitis treated with early laparoscopic cholecystectomy (within 72–96 h of the onset of symptoms) with delayed cholecystectomy or open cholecystectomy. In general, morbidity and mortality after these operations were similar, but the length of hospitalizaiton was much less with laparoscopic cholecystectomy when compared to open operation. The rates of conversion to open operation ranged from 7 to 27%, but the course of patients converted to open operation does not differ from patients treated initially with open operation. On the other hand, Kum (54) compared laparoscopic cholecystectomy performed for acute cholecystitis with laparoscopic procedures performed for symptomatic gallstones. The operation for acute cholecystitis took an average of 22 min longer, the rate of common duct injury was higher (5.5 versus 0.2%) and the conversion rate was higher (13 versus 4%). Direct comparisons between early laparoscopic and open cholecystectomy for acute cholecystitis have not been made, although it is known the rate of bile duct injury is higher for patients treated for acute cholecystitis compared to those with uncomplicated disease. Moreover, the rate of bile duct injury correlates with the experience of the surgeon and the timing of the operation. b— Timing of Laparoscopic Cholecystectomy and Factors that Influence the Rate of Conversion Several investigators (53,55–58) have examined factors that predict conversion of laparoscopic cholecystectomy to open operation. The rate of conversion is higher in males (53,55); with a severely inflamed gallbladder (55); with gangrene, empyema, or hydrops (56); and with increasing duration of symptoms (55,56). Conversion rates may also be higher in elderly individuals (above 65 years of age), those with large gallstones, and when white blood cell counts are higher than 13,000 (56). Rates of conversion were highest in patients with gangrene (49%), hydrops (28.5%), and empyema (28.5%), but Lo et al. (58) did not observe increased conversion rates with severe inflammation, empyema, or gangrene. Male gender, large
Page 487
stones, white blood cell counts greater than 13,000, and fever were also associated with higher rates of postoperative complications. Garber et al. (57) noted that the rate of conversion was only 1.8% if patients were operated on within 4 days of the onset of symptoms, but that it rose to 31.7% after 4 days. Likewise, the complication rate rose from 2.7 to 13%, the mean operative time increased from 100 to 120 min, and postoperative days of hospitalization increased from 5.5 to 10.8 days when patients were operated on after 4 days. Eldar et al. (56) also noted a marked increase in the conversion rate from 4.7 to 23% if patients had symptoms for longer than 96 h. These data suggest that early laparoscopic cholecystectomy is optimally performed within 4 days of the onset of the symptoms of acute cholecystitis. The operation is easier to perform when edema is present and more difficult to perform later when fibrosis begins. The interpretation of current data in the literature is summarized in Table 3. Early cholecystectomy, performed with either open or laparoscopic techniques, decreases the rate of positive bile culture, avoids the higher morbidity and mortality in individuals who fail medical therapy, and decreases the total duration of hospitalization. The morbidity and mortality are similar with both operations, although the risk of injuring a common bile duct may be higher in patients treated laparoscopicaly, especially by inexperienced surgeons. Laparoscopic cholecystectomy for acute cholecystitis is more difficult than elective laparoscopic cholecystectomy, but it results in further decreases in the duration of hospitalization, especially if the patient is brought to the operating room as early as possible. Conversion rates to open operation range from 7 to 26%, but patients converted to open operation fair as well as those treated with open operation initially. Rates of conversion, operative complications, and operative times are minimized and approach the rates observed with elective cholecystectomy if the operation is performed less than 96 h after the onset of symptoms, the surgeon is experienced with both basic and advanced laparscopic techniques, and conversion is chosen promptly if the operation is not progressing. Excellent results also require the availability of stateoftheart laparoscopic equipment, angled laparoscopes, and intraoperative cholangiography (especially fluoroscopic cholangiography). Conversion to open operation should not be considered a failure or a complication, since prudent conversion with advanced disease decreases operative complications and operative time, and patients converted to open operation fare no worse than patients treated by open cholecystectomy initially. C— Alternatives to Medical and Surgical Treatment of Acute Cholecystitis Although cholecystectomy is the best option for patients who have acute cholecystitis that does not resolve with medical therapy alone, not all patients are acceptable candidates for anesthesia or surgery. Medical therapy fails because the obstructed gallbladder essentially becomes an abscess, and antibiotics cannot diffuse into the purulent material sequestered in its lumen. As in the case of abscesses elsewhere, adequate drainage of the gallbladder removes the purulent material that is acting as the focus of ongoing infection, and it decreases intraluminal pressure. Therefore, in most patients, gallbladder drainage treats an episode of acute cholecystitis successfully. Previously, drainage was accomplished operatively by placing a drainage tube, a socalled cholecystostomy tube, directly into the gallbladder. Surgical placement of a cholecystostomy tube requires a formal operation, and although it may be performed under local anesthesia, it can often be a challenging procedure in a critically ill patient. If a general anesthetic is needed, the patient's operative risk increases. The advantages of cholecystostomy is that it limits the extent of surgery and avoids blood loss compared to cholecystectomy. However, the morbidity and mortality following an open cholecystostomy tube is still significant. Percutaneous transhepatic placement of a drainage tube is now preferred. The procedure is performed under local anesthesia by invasive radiology. If necessary it can be performed at the bedside. The gallbladder is accessed transhepatically with ultrasound guidance using a fine
Page 488
needle. The needle enters the gallbladder through its hepatic surface and a wire is placed through it. Finally, a tube is placed over the wire and secured in place for continuous drainage. The procedure is invasive and may result in hemorrhage or leakage of bile from the liver. Perforation of adjacent organs may also occur. However, in experienced hands, gallbladder drainage is as effective as open cholecystectomy and complication rates are acceptably low. Aspiration of gallbladder contents, examination for leukocytes, and culture of bile can be diagnostic in atypical cases. Drainage will effectively treat 80 to 90% of patients but may not be effective is the gallbladder wall has already become necrotic. When they recover from their acute medical problems, patients who are treated percutaneously should have their gallbladders removed if they are acceptable operative candidates and their life expectancies warrant operative intervention to avoid future complications from their gallbladder disease. However, patients who do not meet these criteria may be treated by removal of the tube and observation or by longterm drainage of the gallbladder. Many patients will not have recurrence of symptoms or acute cholecystitis when the cholecystostomy tube is removed (33–35,37). References 1. Attili A, DeSantis A, Capri R, et al. The natural history of gallstones: the GREPCO experience. Hepatology 1995; 21:656–660. 2. Diehl A. Epidemiology and natural history of gallstone disease. Gastroenterol Clin North Am 1991; 20:1–19. 3. Friedman G. Natural history of asymptomatic and symptomatic gallstones. Am J Surg 1992; 165:399–404. 4. Gracie W, Ransohoff D. The natural history of silent gallstones: the innocent gallstone is not a myth. N Engl J Med 1982; 307:798–800. 5. Fitzgibbons R Jr, Tseng A, Wang H, Nguyen N, Sims K. Acute cholecystitis: does the clinical diagnosis correlate with the pathological diagnosis? Surg Endosc 1996; 10:1180–1184. 6. Akrivadis E, Hatzigavriel M, Kapnias D, Kirimlidis J, McAdam A, Garyfallos A. Treatment of biliary colic with diclofenac: a randomized, doubleblind, placebo controlled study. Gastroenterology 1997; 113:225–231. 7. Liu KJ, Richter H, Cho M, Jarad J, Nadimpalli V, Donahue P. Carcinoma involving the gallbladder in elderly patients presenting with acute cholecystitis. Surgery 1997; 122:748–754. 8. Rosyln JJ, DenBesten L, Thompson JE Jr, Silverman BF. Role of lithogenic bile and cystic duct obstruction in the pathogenesis of acute cholecystitis. Am J Surg 1980; 140:126–130. 9. Bolondi L, Gaini S, Testa S, Labo G. Gallbladder sludge formation during prolonged fasting after gastrointestinal tract surgery. Gut 1985; 26:734–738. 10. Carey MC, Cahalane MJ. Whither biliary sludge? Gastroenterology 1988; 95:508–523. 11. Doty JF, Pitt HA, PorterFink V, DenBesten L. The effect of intravenous fat and total parenteral nutrition on biliary physiology. J Parenter Enter Nutr 1984; 8:263–268. 12. Lee SP, Nicholls JF. Nature and composition of biliary sludge. Gastroenterology 1986; 90:677–686. 13. Uggowitzer M, Kugler C, Schramayer G, Kammerhuber F, Groll R, Hausegger K, Ratschek M, Quehenberger F. Sonography of acute, cholecystitis: comparison of color and power Doppler sonography in detecting a hypervascularized gallbladder wall. AJR 1997; 168:707–712. 14. Heuman R, Norrby S, Sjodahl R, Tiselius H, Tagesson C. Altered gallbladder bile composition in gallstone disease: relation to gallbladder wall permeability. Scand J Gastroenterol 1980; 15:581.
Page 489
15. Sjodahl R, Wetterfors J. Lysolecithin and lecithin in the gallbladder wall and bile: their possible roles in the pathogenesis of acute cholecystitis. Scand J Gastroenterol 1974; 9:525. 16. Duane WC, McHale AP, Sievert CE. Lysolecithinlipid interactions in disruption of the canine gastric mucosal barrier. Am J Physiol 1986; 250:G275–G279. 17. Jansson R, Thornell E, Svanvik J. Effects of indomethacin on gallbladder pressure in patients with acute cholecystitis. Scand J Urol Nephrol 1983; 755:51–53. 18. LaMorte WW, Lamont JT, Hale W, Booker ML, Scott TE, Turner B. Gallbladder prostaglandins and lysophospholipids as mediators of mucin secretion during cholelithiasis. Am J Physiol 1986; 251:G701–G709. 19. Myers SI, HaleyRussell D, Bartula LL, Nabzdyk W. Common bile ligation in the rabbit: an appropriate model for investigating the relationship of endogenous gallbladder prostanoid synthesis with evolving acute inflammation. Prostaglandins 1990; 40:165–185. 20. Svanvik J, Thornell E, Jivegård L. Treatment of biliary colic and acute cholecystitis with prostaglandin synthetase inhibitors. Dig Dis Sci 1990; 35:284–284. 21. Thornell E, Jivegard L, Bukhave K, Madsen JR, Svanvik J. Prostaglandin E2 formation by the gallbladder in experimental cholecystitis. Gut 1986; 27:370–373. 22. Prystowsky JB, Huprikar JS, Rademaker AF, Rege RV. Crystalline cholesterol, bilirubin, and calcium hydroxyapatite stimulate human peripheral blood mononuclear cells to release interleukin1 in vitro. Surg Forum 1993; 44:183–184. 23. Prystowsky JB, Rege RV. Crystalline cholesterol has inflammatory properties in the guinea pig gallbladder in vivo. Surg Forum 1994; 45:146–148. 24. Prystowsky JB, Huprikar JS, Rademaker AF, Rege RV. Human polymorphonuclear leukocyte phagocytosis of crystalline cholesterol, bilirubin, and calcium hydroxyapatite in vitro. Dig Dis Sci 1995; 40:412–418. 25. Prystowsky JB, Rege RV. The inflammatory effects of crystalline cholesterol in the guinea pig gallbladder in vivo. Surgery 1998; 123:258–263. 26. Parker JJ, Vukov LF, Wollan PC. Emergency department evaluation of geriatric patients with acute cholecystitis. Acad Emerg Med 1997; 4:51–55. 27. Adedeji O, McAdam W. Murphy's sign, acute cholecystitis, and elderly people. J R Coll Surg Edinb 1996; 41:88–89. 28. Prousalidis J, Fahadidis E, Apostalidis S, Katsohis C, Aletras H. Acute cholecystitis in aged patients. HPB Surg 1996; 9:129–131. 29. Jeffery RJr, NinoMurcia M, Ralls P, Davidson H. Color Doppler sonography of the cystic artery: comparison of normal controls and patients with acute cholecystitis. J Ultrasound Med 1995; 14:33–36. 30. Schiller V, Turner R, Sarti D. Color Doppler imaging of the gallbladder wall in acute cholecystitis: sonographicpathologic correlation. Abdom Imaging 1998; 21:233–237. 31. Joehl RJ, Koch KL, Nahrwold DL. Opioid drugs cause bile duct obstruction during hepatobiliary scans. Am J Surg 1984; 147:134–138. 32. Fidler J, Paulson E, Layfield L. CT evaluation of acute cholecystitis: findings and usefulness in diagnosis. AJR 1996; 166:1085–1088. 33. Werbel G, Nahrwold D, Joehl R, Vogelzang R, Rege R. Percutaneous cholecystostomy in the diagnosis and treatment of acute cholecystitis in the highrisk patient. Arch Surg 1989; 124:782–786. 34. Hamy A, Visset J, Likholatniikov D, Lerat F, Gibaud H, Savigny B, Paineau J. Percutaneous cholecystostomy for acute cholecystitis in critically ill patients. Surgery 1997; 121:398–401. 35. Melin M, Sarr M, Bender C, van Heerden J. Percutaneous cholecystostomy: a valuable technique in highrisk patients with presumed acute cholecystitis. Br J Surg 1995; 82:1274–1277. 36. Patterson E, McLoughlin R, Mathieson J, Cooperberg P, MacFarlane J. An alternative approach to acute cholecystitis: percutaneous cholecystostomy and interval laparoscopic cholecystectomy. Surg Endosc 1996; 10:1185–1188.
Page 490
37. van Overhagen H, Meyers H, Tilanus H, Jeekel J, Lameris J. Percutaneous cholecystostomy for patients with acute cholecystitis and an increased surgical risk. Cardiovasc Intervent Radiol 1996; 19:72–76. 38. Cohen JL, Bartlett JA, Corey GR. Extra intestinal manifestations of Salmonella infections. Medicine 1987; 66:349–388. 39. LopexTalavera JC, Buti M, Esteban R. Empiema vesicular por Salmonella enteritidis. Med Clin 1988; 90:352. 40. James JA, Morris G, Winter R, KestonJones M. Acalculous cholecystitis due to Salmonella Virchow. Br J Clin Pract 1990; 44:767–768. 41. Ocha J, Ricarte E, Carrasco M, Cabello J, Yanguela J. Complicacions de la gastroentritis aguda por Salmonella no typhi. Rev Esp Enf Ap Dig 1989; 75:262– 266. 42. Avalos M, Cerulli M, Lee R. Acalculous cholecystitis due to Salmonella typhi. Dig Dis Sci 1992; 37:1772–1775. 43. Lee J, McLoed M, Meyers W, Arthur J, Corey G. Successful laparoscopic management of perforated gallbladder associated with Salmonella javiana infection. N C Med J 1992; 53:594–595. 44. Archibald S, Colapinto N, Frost P. Early surgical management of acute cholecystitis. Can J Surg 1979; 22:464–466. 45. Glenn F. Acute cholecystitis. Surg Gynecol Obstet 1976; 143:56–60. 46. Lahtinen J, Alhava E, Aukee S. Acute cholecystitis treated by early and delayed surgery: a controlled clinical trial. Scand J Gastroenterol 1978; 13:673–678. 47. McArthur P, Cuschieri A, Sells R, Shields R. Controlled clinical trial comparing early with interval cholecystectomy for acute cholecystitis. Br J Surg 1975; 62:850–852. 48. Sianesi M, Ghirarduzzi A, Percudani M, Dell'Anna B. Cholecystectomy for acute cholecystitis timing of operation, bacteriologic aspects, and postoperative course. Am J Surg 1984; 148:609–612. 49. Bender J, Zenilman M. Immediate laparoscopic cholecystectomy as definitive therapy for acute cholecystitis. Surg Endosc 1995; 9:1081–1084. 50. Lujan J, Parrilla P, Robles R, Torralba J, GarciaAyllon J, Liron R, SanchexBueno F, Abrahamson J. Laparoscopic cholecystectomy in the treatment of acute cholecystitis. J Am Coll Surg 1995; 181:75–77. 51. Lujan J, Parrilla P, Robles R, Marin P, Torralba J, GarciaAyllon J. Laparoscopic cholecystectomy vs. open cholecystectomy in the treatment of acute cholecystitis: a prospective study. Arch Surg 1998; 133:173–175. 52. Lo C, Liu C, Lai E, Fan S, Wong J. Early versus delayed laparoscopic cholecystectomy for treatment of acute cholecystitis. Ann Surg 1996; 223:37–42. 53. Eldar S, Sabo E, Nash E, Abrahamson J, Matter I. Laparoscopic versus open cholecystectomy in acute cholecystitis. Surg Laparosc Endosc 1997; 7:407–414. 54. Kum C, Eypasch E, Lefering R, Paul A, Neugebauer E, Troidl H. Laparoscopic cholecystectomy for acute cholecystitis: is it really safe? World J Surg 1996; 20:43–48. 55. Bickel A, Rappaport A, Kanievski V, Vaksman I, Haj M, Geron N, Eitan A. Laparoscopic management of acute cholecystitis: prognostic factors for success. Surg Endosc 1996; 10:1045–1049. 56. Eldar S, Sabo E, Nash E, Abrahamson J, Matter I. Laparoscopic cholecystectomy for acute cholecystitis: prospective trial. World J Surg 1997; 21:540–545. 57. Garber S, Korman J, Cosgrove J, Cohen J. Early laparoscopic cholecystectomy for acute cholecystitis. Surg Endosc 1997; 11:347–350. 58. Lo C, Fan S, Liu C, Lai E, Wong J. Early decision for conversion of laparoscopic to open cholecystectomy for treatment of acute cholecystitis. Am J Surg 1997; 173:513–517.
Page 491
23— Laparoscopic Cholecystectomy A.C.T. Wan and A. Darzi Imperial College School of Medicine at St. Mary's, London, England I— Introduction In just over 10 years, the advent of keyhole surgery has initiated a worldwide diffusion of laparoscopic techniques to a multitude of surgical specialties. Endoscopic surgery continues to expand its horizon and experiences popularity in all areas of surgery, particularly surgery of the gastrointestinal tract. Since its inception, laparoscopic cholecystectomy has been well tested in the surgical arena among all minimalaccess surgical procedures. The use of laparoscopic techniques for other clinical entities remains controversial, but its successful application to gallstone diseases has set a high standard for others to follow. This chapter explores the development and the recent advances in this surgical field. And, because of the explosive growth of surgical and video technology, some of the indications for surgery are reconsidered. Since, internationally, an increasing number of procedures are being adopted and performed, an overview of the new technical breakthroughs and complications is provided. In addition, some of the potential problems arising from the introduction of this procedure to surgical training are assessed, along with the impact of this procedure on surgical trainees. II— History Conventional cholecystectomy, or ''open cholecystectomy," as it is now termed, has been the "gold standard" operation for gallstone diseases for over a century. Surgeons were once worshipped for their skills in relieving symptoms of biliary colic—the postoperative pain caused by the surgical procedure is a mere fraction of the crippling pain that can potentially be inflicted by gallstones. Although gynecologists have made good use of the laparoscope in recent time, it was not until the 1980s that the first laparoscopic cholecystectomy was carried out and expectedly greeted with much skepticism and hostility (1). During the early 1980s, fascinated by Semm's technique with laparoscopic appendectomy, Muhe came up with the idea of laparoscopic removal of gallstones. In 1984 he worked out the details of an operative laparoscope, the socalled Galloscope, and in 1985 he carried out the first laparoscopic cholecystectomy. Later, he modified his technique and operated through a trocar sleeve before designing an "open laparoscope" with a circular light. By March 1987, Muhe had conducted 97 endoscopic gallbladder removals (2). Although he published information about his technique and presented his work at various surgical meetings in Germany, his concept was ignored. The first laparoscopic cholecystectomy reported in the literature was performed by Mouret in Lyon, France, in March 1987 (3). Other French surgeons, like Dubois in Paris and
Page 492
Perrisat in Beaudeaux, polished the procedure in the following 2 years (4–6). During this period, the principle was adopted by American surgeon Reddick in Nashville, Tennessee, who performed and perfected the technique across the Atlantic (7). The advantages of the procedure became apparent over the years, with early discharge from hospital and early return to work as well as reduced wound pain and infection rates that are beneficial to both patients and hospitals. Subsequent years of experimentation and favorable clinical trials have promoted this procedure, and numerous surgeons are now practicing this technique as the procedure of choice for the majority of patients with symptomatic gallstone diseases. This progress in surgery to include laparoscopic procedures in our clinical practice has prompted the development of laparoscopic courses. Surgical colleges and recognized centers around the world have introduced skill courses, allowing trainees to learn the principles and techniques on simulators before putting their acquired skills to practice. III— Indications As laparoscopic cholecystectomy becomes the procedure of choice for patients with symptomatic cholelithiasis, many centers now offer an "all comers" policy (8). Patients with relative contraindications have undergone laparoscopic cholecystectomy without increasing morbidity; but as the conversion rate was higher, they should be warned of this potential change of management (9). In the absence of any absolute contraindications, few centers would offer open cholecystectomy as the firstline treatment and occasionally the only procedure for gallstone diseases. The latter is often limited to individual practices, particularly to surgeons who have not acquired the necessary skills to perform minimalaccess procedures. In a study conducted in United Kingdom, it was found that a "no conversion" policy could be adopted as the surgeon became more experienced (10). Laparoscopic cholecystectomy is now proposed to all patients requiring the removal of the gallbladder. A— Acute and Chronic, Early and Late Traditional criteria include patients with symptomatic gallstone diseases presenting with biliary colic, acute or chronic cholecystitis, and gallstone pancreatitis. Although elective surgery is still the most common scenario for the given procedure, it is also feasible in patients with acute cholecystitis (11). However, the conversion rate can reach as high as 40% in the case of gangrenous gallbladders (12). In a randomized controlled trial, Lo et al. demonstrated less technical modification and a shorter operative time for initial conservative management with interval procedures, but this does not reduce morbidity and conversion rate (13). In another study, early emergency surgery within 72 h results in a mortality of 0.3% but an increased conversion rate (14). Lai and colleagues found no significant difference in conversion and complication rates between early and delayed management, but the latter group had a shorter hospital stay (15). B— Acalculous Gallbladder Diseases The inclusion extends to acalculous cholecystitis and a group of gallbladder entities known as cholecystoses (i.e. polyps, diverticuli, and adenomas) (16). Following laparoscopic procedures, 171 patients with acalculous cholecystitis and biliary dyskinesia became symptomfree (17). C— Risk Factors 1— Age Some of the absolute contrindications proposed in the introductory phase have become relative contraindications as the techniques were modified. The older age groups with moderate to high
Page 493
anesthetic risks have benefited from laparoscopic cholecystectomy, with much lower postoperative complications (18,19). Magnuson's study found those elderly patients with uncomplicated gallstone disease to be excellent candidates for laparoscopic cholecystectomy, but early conversion or planned open cholecystectomy may be warranted in the elderly presenting with acute complications (20). 2— Previous Abdominal Surgery A history of previous abdominal surgery does not preclude laparoscopic cholecystectomy. Kumar demonstrated a subxiphoid approach using blunt finger dissection before primary port placement in 4 patients with more than 3 previous abdominal procedures (21). In another study, all 411 patients With previous infraumbilical intraperitoneal operations underwent successful laparoscopic removal of the gallbladder, whereas 10 patients with supraumbilical intraperitoneal procedures failed because of dense adhesions (22). Vezakis et al. demonstrated effective and safe management of gallstones by the laparoscopic method in patients who had undergone pancreatic debridement (23). 3— Pregnancy Until 1997, many believed that laparoscopic cholecystectomy should be reserved for complicated nonresolving biliary tract disease during pregnancy, as the disease of over 90% of patients with the acute process will resolve with conservative management (24). Pregnancy in the second and third trimesters no longer excludes the option of using a laparoscopic approach (25,26). Eight patients have delivered fullterm, healthy infants with no documented neonatal or maternal morbidity or mortality (27). In Glasgow's series, conservative treatment was attempted in 45 patients but failed in 36%; 14 proceeded to undergo laparoscopic cholecystectomy, with reducedpressure pneumoperitoneum used in half of these patients (28). Graham and colleagues described the safety of laparoscopic procedures throughout pregnancy but indicated that the second trimester is preferable, with a higher rate of fullterm delivery (29). 4— Pediatrics Children can present with symptomatic gallstones. Although gallstones are uncommon in the pediatric group, laparoscopic procedures for removal of the gallbladder are feasible and now preferable to open procedures as they are less invasive and more costeffective; but surgical technique is often modified to produce better cosmetic results (30). Johna et al. described 40 children aged 17 or less undergoing laparoscopic management of gallbladder diseases ranging from gallstone pancreatitis to sickle cell disease, reporting a 90% success rate (31). It is important to note that 3 patients continued to have postoperative abdominal pain, indicating the significance of accurate preoperative diagnosis. 5— Obese Patients The benefit of laparoscopic cholecystectomy is extending to the overweight and obese patients with symptomatic gallstone disease, and this procedure is becoming the procedure of choice among these patients (32). Longer laparoscopic ports and instruments with slight adjustments of port positions are recommended (33). In two studies, neither group found any significant difference in conversion and complication rates between normalweight and obese groups, but in Shirmer's study procedure time was longer in the morbidly obese group (34,35). 6— Other Comorbid Conditions Many other coexisting conditions may weigh against the treatment of symptomatic gallstones by the "keyhole" method. However, the advances in minimalaccess and anesthetics techniques
Page 494
have allowed many patients to undergo laparoscopic cholecystectomy with minimal morbidity. Patients with advanced liver and heart diseases have successfully undergone laparoscopic management of their gallstones. Sleeman reported 25 patients with Child's A and B cirrhosis having their gallstones managed laparoscopically with 30% morbidity (36). In another study, cirrhotics have more gallstonerelated complications than noncirrhotics but 75% underwent laparoscopic cholecystectomy, with 5 cases of intraoperative bleeding and 9 with postoperative complications (37). A cardiac transplant candidate with an ejection fraction of 15% endured the procedure after being medically optimized (38). Menegauz and colleagues, after performing successful procedures in all of their 15 patients, suggested offering elective laparoscopic management of gallstones to cardiac transplant candidates in their posttransplantation course (39). Alternation and careful monitoring of the anticoagulation regime is essential preoperatively; this combination, with meticulous techniques during surgery, can prevent the formation of hematomas in the operative and trocar sites (40). Reports of rarer clinical entities include the use of minimally invasive surgery to remove metastatic polyps in a gallbladder of a patient with malignant melanoma undergoing isolated limb perfusion at the same sitting (41). Other successful treatment of symptomatic stones includes patients with condition such as cystic fibrosis (42). Peritoneal dialysis is no longer a contraindication to this procedure, as demonstrated by Magnuson et al. (43). Cholecystoenteric fistula can be dealt with laparoscopically in skilled hands (44). Surgery in AIDS patients has been described in earlier literature, and laparoscopic treatment of gallstones among these patients is increasingly feasible, with acceptable morbidity and mortality (45,46). IV— Preparation for Surgery A— Investigations Patients who are jaundiced or have deranged liver functions will need further investigations before being considered for surgery. Liver serology should be determined in those with a history of travel, exposure to blood transfusion, intravenous drug abuse, and other factors predisposing to hepatitis. An abdominal ultrasound examination will confirm the status of the gallbladder, the adjacent biliary tree, and the liver. Recent advances in biliary imaging include stateoftheart computed tomography (CT) and magnetic resonance (MR) cholangiography (47,48). Details of gallbladder and biliary tree imaging are described in Chap. 18. Persistent jaundice and a dilated biliary tree with or without ductal stones are indications for preoperative endoscopic retrograde cholangiopancreatography (ERCP). Although preoperative cholangiography is mandatory for some surgeons to clarify anatomy, it is nonetheless usual to evaluate the biliary tree before surgery if the patient's clinical condition suggests the presence of ductal calculi. The advent of laparoscopic exploration of the common bile duct may alleviate the need for combined endoscopic and radiological evaluation prior to surgery, but successful application of this new technique is possible only in experienced hands. B— Patient Information Patients need to be informed on the pros and cons of this surgical technique. Surprisingly, there are some who may prefer the conventional type of procedure. Recent literature suggests that patients' postoperative recall of information did not correspond with the initial evaluation of their understanding of the surgical procedure and the process by which it was communicated to them (49). Nevertheless, it is our duty to obtain the appropriate informed consent from the patient. It is important to stress that conversion to open surgery is not a failure but it is the approach that gives the surgeon an optimal view and access to the gallbladder. The resultant postoperative state must also be addressed in order to avoid compromising any arrangements
Page 495
made by the patients during the recovery period. Temporary but minor side effects like shouldertip discomfort and bloating from residual gas in the peritoneal cavity should be mentioned in order to alleviate any postoperative anxiety. C— Prophylaxis—Anticoagulation and Antibiotic Patients undergoing laparoscopic cholecystectomy may be at increased risk of developing deep venous thrombosis due to the increased intraabdominal pressure accompanying the creation of pneumopertioneum and the relative venous stasis associated with the reverse Trendenlenburg position. Little is known about the rate of thromboembolism following laparoscopic cholecystectomy; however, a postal questionnaire to surgeons showed that thromboembolic deterrent (TED) stockings were used by 74% of surgeons, and a similar number prescribed heparin to their patients (50). The use of antibiotic prophylaxis for elective surgery remains controversial (51). Earlier studies were conflicting; one indicated a 22% infection rate without antibiotic usage and another suggested that prophylaxis is not necessary for lowrisk patients (52,53). A case of Clostridium gas gangrene of the abdominal wall was reported and the routine use of antibiotic in the perioperative period was recommended (54). More recently, in a randomized study involving 250 patients, Dobay et al. also found that antibiotic prophylaxis is not required in lowrisk patients (55). D— Anesthesia General anesthesia is the common mode of achieving muscle relaxation and assisted ventilation. Improved anesthetic techniques currently permit ASA grade III patients with ischemic heart disease to undergo laparoscopic procedures, but cardiorespiratory and blood gas parameters as well as acidbase balance must be carefully monitored in the perioperative period (56). Surgeons should be made aware of potential cardiovascular changes when using CO2 to create pneumoperitoneum (57). For highrisk patients, ASA grade III or IV, epidural anesthesia can be used effectively; for patients with severe chronic obstructive pulmonary disease (COPD), intraoperative shouldertip or abdominal pain is easily controllable with small doses of opioid analgesia (58). Patients are not routinely catheterized, although this is commonly done in some centers. A recent study showed that urinary catheterization is not necessary, especially if the bladder is not palpable (59). Patients are often asked to void and empty the bladder approximately 1 to 2 h before they are due to have the procedure. A nasogastric tube can also be placed to decompress the stomach; this maneuvre often improves the view of the biliary tree if it would otherwise be obstructed by a distended stomach and duodenum. V— Surgery A— Equipment 1— Video and Camera Equipment Advances in video and camera technology over the years have led to the rapid expansion of videoassisted laparoscopic surgery. The basic video equipment consists of the telescope, lighttransmission cable, video camera, processing unit, light source, and video monitors. The 10mm 0° wideangle laparoscope is the most commonly used telescope; and occasionally, a 30° telescope may improve the angle of view, especially if laparoscopic exploration of the common bile duct is considered. Telescopes with a diameter as small as 3 mm have recently been developed; this will no doubt lead to even smaller access wounds. The development of the chip video camera allows easy transfer of images from the telescope to the display monitor. Subsequent modification of the onechip camera to threechip technology gives improved resolution and color to images. Highresolution video monitors provide magnification of the im
Page 496
ages as well as greater clarity and definition, allowing the surgeon, assistants, and theater staff alike to view the procedure simultaneously. The newer threedimensional videoendoscopic system has the potential of overcoming the problems with depth perception during laparoscopic surgery. Birkett and coworkers found that structures were more easily defined and instrument manipulation was easier during clinical tasks when a threedimensional laparoscope was used. In addition, less time was required to complete complex inanimate tasks (60). However, Hanna et al. did not find any significant differences in execution time and errors made during the procedure in comparing a threedimensional with a twodimensional system (61). 2— Surgical Instruments Over the years, more and more laparoscopic tools have been developed to facilitate modification of the procedure. There has been a trend toward disposable instruments, although the associated cost is higher, in the region of $350 per case (62). Similarly, a German study found that surgical procedures with disposable instruments were faster and that the conversion rate was lower (63). A basic laparoscopic instrument set remains the same and should consist of a Veress needle, access ports, at least two grasping forceps, blunt dissectors, scissors, and a clip applicator. Suction and irrigation devices are used to maintain a clear operative field. 3— Room Setup and Patient Positioning The operative field is prepared and draped in the usual manner. However, the positioning of the patient depends solely on the surgeon. No new room setups have been introduced since the procedure became popularized. Two main room setups are recognized, and a variation or a combination of the two can be found in any operating theater that can accommodate minimalaccess procedures. The supine position for the patient is preferred in most of Europe and North America, with the surgeon operating from the left side of the patient, whereas the LloydDavies position is preferred in France and Germany, with the surgeon standing between the patient's legs (64). B— Cholecystectomy 1— Access Entry into the abdominal cavity commonly necessitates four small incisions (ranging from 5 to 12 mm) in the abdominal wall. Additional incisions can be made if extra instruments are required for the procedure (i.e., percutaneous cholangiography). Smaller access cannula ports ranging from 1.4 to 3 mm in diameter have been introduced (65). Micropuncture laparoscopic cholecystectomy was feasible in 25 patients using a 3mm access cannula (66). Ngoi and colleagues successfully performed 34 cases of needlescopic cholecystectomy, showing less wound pain and better cosmetic results (67). Although the usage of instruments smaller than those employed in the 3mm needlescopic method can be employed, this is presently restricted to uninflamed gallbladder. A threeport method was described by Tagaya and colleagues, with 119 out of 130 procedures proving successful (68). Recent development includes the usage of only two ports, further minimizing the number of incisions made (69). One example utilizes the umbilicus as the access route; the pain score and analgesia requirement were found to be much less (70). Two conventional techniques for port placement are practiced. The closed technique involves the introduction of the primary port after CO2 insufflation via a Veress needle and confirmation by the saline draw test. However, access problems may be encountered in patients with previous abdominal surgery (71). The second is an open technique that involves dissection through the small skin incision right down to the peritoneal cavity. A Hasson type cannula (blunt end) is then placed under direct vision before CO2 is introduced. Following complications with the closed technique when the procedure was first popularized, the latter technique
Page 497
achieves the principle of safe introduction of access ports under direct vision, thus minimizing unnecessary visceral injuries (72). Cogliandolo and coworkers in a randomized study involving 150 patients found that the open laparoscopic technique was safer and faster than the blind approach (73). Arguably, the open method will reduce potential complications in those patients with previous abdominal surgery. Although new preventive measures using biological or synthetic adhesiolytic agents may reduce the formation of adhesions, the risks of laceration and perforation of the viscera remains. 2— Pneumoperitoneum and Mechanical Lifters In order to create a working space to carry out the surgical procedure, gas is normally introduced into the peritoneal cavity. This is usually done via the Veress needle or the Hasson port before further port placement. CO2 is the gas of choice, as it causes the least physiological disturbance; it is nonflammable and readily absorbed. A randomized study shows that N2O is comparable with CO2 and may cause less cardiorespiratory disturbance in patients with premorbid cardiorespiratory diseases (74). A low CO2 insufflation rate of 2.5 L/min was found to significantly reduce postoperative shoulder pain in comparison with the highflow rate of 7.5 L/min and does not increase the operation time (75). Odeburg and associates found that pneumoperitoneum did not impair splanchnic circulation (76). Draper's study followed 57 patients who underwent laparoscopic cholecystectomy and found that bile spillage significantly reduced the duration of postoperative pneumoperitoneum; otherwise residual pneumoperitoneum resolves in 3 days in 81% and 7 days in 96% of cases (77). Various mechanical devices designed to lift the abdominal wall have surfaced over the years, but their use has been very much limited to the enthusiasts. The abdominal lift has been used successfully in patients with severely compromised cardiac function (78). Another study that reviewed the abdominal wall lift showed the advantages of limiting CO2 peritoneal insufflation, thus reducing unwanted side effects of a high intraabdominal pressure following insufflation (79). Finally, Nanashima and colleagues demonstrated similar physiological responses in the two groups, pneumoperitoneum and gasless abdominal lifting (80). The controversy remains. 3— Energy Source and Dissection Modes Conventional bipolar diathermy continues to be used for laparoscopic procedures. Other sources include the harmonic scalpel and ultrasonic dissecting instruments (81). Cavitational forces induced ultrasonically by the ultrasonic dissector were found to be valuable in isolating the hilar structures, particularly when they are edematous or embedded in fat (82). Naude and coworkers described the use of laparoscopic hydrodissection, using 50 mL of saline injected between the liver and the gallbladder with a cyst aspiration needle and resulting in less bleeding, fewer incidents of gallbladder damage and stone spillage, and a much shorter dissection time (83). Two hundred patients took part in a randomized trial of monopolar electrocautery versus ultrasonic activated coagulating shears, and the latter was found to be safer, easier to use, faster, and less prone to intraoperative complications and postoperative morbidity (84). Hershman and Rosin described 200 patients who were considered for laparoscopic laser cholecystectomy (85). A randomized trial of neodymium YAG laser versus monopolar electrosurgical dissection in 100 patients showed that the latter malfunctioned in 6 patients, requiring completion with electrosurgery, and estimated blood loss was more (86). Corbitt had similar findings in a randomized study involving 300 patients (87). 4— The Procedure Surgeons have adopted various surgical techniques over the years, but the fundamental principles of safe access to the peritoneal cavity and identification of appropriate anatomy, particularly Calot's triangle, has not changed (88–93). A diagnostic laparoscopy should be performed
Page 498
initially to look for synchronous pathology (94,95). In the absence of accurate tactile feedback, some pathology will be missed, although the presence of another pathology is uncommon (96). Following Reddick's method of exposing Calot's triangle, the Japanese surgeons devised various ways to maximize this exposure. The use of an atraumatic instrument to retract the caudal surface of the liver reduces tenting and angulation of the common bile duct and possibly reduces inadvertent injury to the biliary tree (97). In order to facilitate visualization, a tape or suture can be introduced and retract the gallbladder and round ligament from the extracoporeal side of the abdominal wall (98). Ligation of the cystic duct was also made easier by a movable tying invention known as the Ojigi spatula (99). Other methods of securing the cystic duct include the use of endoloops and endostaplers. Iatrogenic perforation of the gallbladder should be repaired early, and retrieval of stones followed by an intraperitoneal lavage has been recommended to minimize complications (100). In difficult cases, where extensive inflammation or fibrosis is encountered, a subtotal laparoscopic cholecystectomy can be performed (101,102). 5— Operative Cholangiography Cholangiograms have been obtained during laparoscopic procedures for reasons such as unclear anatomy or clinical and surgical features suggestive of common bile duct stone. This remains a controversial area, and the jury is still out in determining whether it should be done routinely (103). Two common methods are employed: the cholangiogram catheter is fed into the peritoneal cavity either via the lateral port or percutaneously. A policy of selective cholangiography appears sensible; this should be determined on the basis of abnormal liver function tests, a history of pancreatitis or jaundice, and dilatation of the common bile duct either at operation or by preoperative ultrasonsography (104). Young and colleagues have advocated the use of cholecystocholangiography as an alternative to cystic duct cholangiography because it is safe, successful, and quick (105). Longterm followup of 253 patients who underwent laparoscopic cholecystectomy with no preoperative stigmata of ductal stones revealed 2.3% of retained common bile duct stones (106). The advent of laparoscopic ultrasound may alter the management plan of common bile duct stones, especially if the surgeon is already skilled at laparoscopic exploration of the common bile duct (107). Many randomized studies have shown the superiority of this new procedure to intraoperative cholangiography (108–110). A detailed description of endoscopic ultrasound of the gallbladder and biliary tree has been reviewed in Chap. 19. 6— Delivery of Gallbladder The gallbladder is usually extracted via an epigastric or subumbilical incision. In the event of multiple calculi, it may be easier to suction some of the stones before retrieval or to place the detached gallbladder in a container such as an endoscopic bag to prevent the gallbladder from tearing as it exits through the abdominal wall (111). Large stones can be fragmented mechanically by a LaparoLith to facilitate delivery (112). Spilled stones can be similarly placed in a bag in a linear fashion to facilitate delivery (113). Stone leaking from the gallbladder can be placed in an intraperitoneal bag and "parked" on the liver while dissection continues (114). When abnormal signs or suspected gallbladder malignancy is present, gasless laparoscopy with abdominal wall lift and the use of an intraperitoneal bag for specimen extraction has been recommended (115). Retained stones in the peritoneal cavity are dealt with in Sec. V, below. 7— Postoperative Course Qureshi and associates have demonstrated a similar incidence of postcholecystectomy symptoms with both open and laparoscopic procedures (116). Ninetyfive percent of patients were considered to have had an overall symptomatic improvement by surgical treatment despite the persistence or de novo occurrence of symptoms (117). Rothwell et al. have shown that cholecystectomyinduced gastroesophageal reflux is not reduced by the laparoscopic approach
Page 499
(118). The laparoscopic technique did not alter the symptom of biliary reflux in patients with cholelithiasis (119). The introduction of intraperitoneal analgesia reduces postoperative pain (120). Pain scores were significantly reduced in patients with only intraperitoneal normal saline infusion subdiaphragmatically and, after its postdeflation suction, bupivacaine infusion in the same area (121,122). Improved pain scores were demonstrated in those groups using ketorolac and diclofenac as compared with the saline group (123). VI— Complications Over the last decade, following the inception of laparoscopic cholecystectomy, more than a dozen complications have been recognized. Some of these are shared with those of laparoscopy itself and others more specific to the procedure. There are some wellrecognized complications, whereas others remain as interesting case reports. They can be divided into two categories: at laparoscopy and at cholecystectomy. A— LaparoscopyRelated Complications The majority of problems occur as a result of the physiological and pressure effects of the pneumoperitoneum and the careless introduction of the cannula. Some of the cardiorespiratory effects from a CO2 pneumoperitoneum are described in Sec. V.B.2 above. The pressure effect can give rise to an alteration of liver function. Disturbances in alanine aminotransferase (ALT) occurred in 34% of patients and appear to be clinically insignificant in one study (124). Morino and colleagues found that the duration and level of intraabdominal pressure were responsible for changes of hepatic function during laparoscopic procedures and suggested that patients with severe hepatic failure should probably not be subjected to prolonged procedures (125). 1— Bowel Complications An interesting study demonstrated that normal a pneumoperitoneum pressure of 12 to 15 mmHg can cause severe splanchnic mucosal ischemia, expressing much concern for patients with coexisting peripheral vascular disease (126). Rare complications such as gastric outlet obstruction secondary to a postcholecystectomy biloma have been reported (127). Obstruction of the small bowel from a volvulus was found in two patients who had previous abdominal surgery (128). Jena et al. reported on a case of fatal injury secondary to duodenal perforation from inadvertent diathermy burns to the bowel wall (129). A case of colonic perforation with the newer and smaller trocar was managed conservatively and successfully (130). Mechanical bowel obstruction secondary to a spilled stone has also been reported (131). Small bowel obstruction secondary to herniation in a trocar puncture site was observed in an obese woman, but CT identified the cause of obstruction and exploratory laparotomy was avoided (132). 2— Vascular Complications Although death from vascular injury is rare, there is a report of a 46yearold man who sustained injury to the abdominal aorta and succumbed to hypovolemic shock despite resuscitation (133). In a study by Usal and associates, in 2589 patients over a 4year period, the incidence of vascular injury was 0.11% (134). A Swedish group reviewed 60 series of 153,832 patients among whom mortality from thromboembolism was 0.8%; from fatal pulmonary embolism, 0.02%; from pulmonary embolism, 0.06%; and from deep venous thrombosis, 0.03% (135). Increase in fibrinolytic activity in plasma as a result of the liberation of tissue plasminogen activator from the venous endothelium was found by MartinezRamos et al., indicating hypocoagulability during the immediate postoperative period and less thrombotic risk for patients undergoing this procedure (136). In a series comparing laparoscopic versus minilaparotomy
Page 500
cholecystectomy, the incidence of deep venous thrombosis remained low as long as adequate prophylaxis was provided (137). Cases of mesenteric artery and venous thrombosis were described in two studies, indicating the importance of exercising extra caution in treating highrisk patients (138,139). Renal vein thrombosis has also been described (140). Other complications include lacerations of vessels, causing hemobilia, and obstructive jaundice from pseudoaneurysms of the hepatic artery (141,142). 3— Wound Infection The infection rate associated with smaller incisions has decreased. Views on the use of antibiotic therapy for elective laparoscopic surgery are still controversial. In one study of 189 patients who underwent laparoscopic cholecystectomy, the overall rate of wound infection was 5.3%; however, all of these infections were minor (143). No major wound infection from the laparoscopic procedure was found in this study. Watkins and colleagues found, at 2 weeks, that there had been two wound infections; one resolved spontaneously and the other required removal of a gallstone from the subcutaneous tissue (144). In one study, 33% or 250 patients suffered perforation of the gallbladder but no late wound or intraabdominal infectious complications, and no patient required reoperation for intraabdominal sepsis (145). Diez and coworkers found that bile spillage increased the incidence of umbilical wound infection, particularly in the presence of remnants of stones, but there was no correlative increase in the incidence of intraabdominal collections or infections (146). 4— PortSite Metastasis Gallbladder cancer has been reported in 0.3 to 1.5% of cholecystectomies. Since the introduction of laparoscopic surgery, cholecystectomies have increased and the discovery of occult neoplasms may become more frequent. Calcified gallbladder, age 70 years and above, a long history of stones, and a thickened gallbladder all represent significant risk factors (147). Numerous cases of this phenomenon have been reported in the literature. Some metastases are from gallbladder primaries and others stem from a variety of sources, including unknown primaries (148–152). In a prospective study by the Swiss Association of Laparoscopic and Thoracoscopic Surgery, preoperatively undiagnosed adenocarcinoma of the gallbladder was found in 0.34% of patients; and recurrence of carcinoma at the port site was found in 5 of 37 patients (14%) (153). Delayed presentation can occur up to 47 months from the initial procedure (154). Portsite metastases seem to be secondary to multiple factors, including the type of gas used, local trauma, tumor manipulation, biological properties of the tumor, and the operator's surgical skills (155). 5— Hernia Early reports of portsite incisional hernia indicate that this complication has occurred mostly in the midline (156). Closure of fascia at the larger port sites is a mandatory procedure for many surgeons. Forced dilatation of the fasical layer can lead to postoperative herniation through the port site (157). The incidence of incisional hernia remains unchanged despite modification of the closure technique, including the use of some modern laparoscopic devices. One series had an incidence of 0.77% among 1300 cases (158). The occurrence of hernia may be attributed to poor technique or an iatrogenic injury to the intecostalis; the chimney effect from desufflation may encourage the herniation at large port sites (159,160). The use of smaller epigastric ports may reduce the occurrence of incisional hernia (161). In Nasser's series, all hernias except for one developed at the umbilicus; he also encouraged the avoidance of unnecessary wound extension and the use of nonabsorbable sutures for defects larger than 2 cm to reduce herniation through these defects (162).
Page 501
B— CholecystectomyRelated Complications These complications are also common to open procedures, but the introduction of videoassisted surgery has brought about an increased level of injury to the biliary tree. The learning curve of the procedure plays an important role, and it is proposed that complications tend to occur in the first 50 patients (163). 1— Bile Duct Injury This is the most feared complication arising from laparoscopic cholecystectomy. Morbidity continues to be high despite the fact that mortality has been somewhat reduced with improvements in new combined intervention and management. Singh and colleagues reported 37 patients with postcholecystectomy external biliary fistula, with 14 partial, 22 complete, and one subvesical duct of Luschka injury. Sixteen patients had a controlled external biliary fistula at presentation; 10 patients had intraabdominal collections; and 7 patients presented with peritonitis (164). In the worst scenario, Robertson described a patient undergoing liver transplantation for laparoscopic injury to the biliary tree (165). An audit of the Danish national database reviewed acute cholecystitis as the indication for laparoscopic cholecystectomy in 968 patients, with 1.3% sustaining laparoscopic bile duct injury; the incidence in patients with other indications was 0.62%. The frequency of bile duct injury in patients who had intraoperative cholangiography was not significantly different from that among those who did not (166). A recent publication from a group of Belgian surgeons showed a high rate of morbidity and mortality from biliary tract injury, comparable to that at open cholecystectomy. Multiple attempts at repair and other interventions were required in both the converted cases and those with recognized injury postoperatively (167). In the United States, a threecenter study of repair of a major biliary injury (ligation, avulsion, or resection) following laparoscopic cholecystectomy over a 6year period showed that 32 patients sustaining such injuries. The injury was recognized immediately in 10 patients. The remaining 22 patients had a combination of symptoms of pain, jaundice, and/or fever. Bismuth classification was as follows: 13% of patients in class I, 63% in class II, 7% in class III, 7% in class IV, and 10% in class V (168). Walsh et al. showed that onethird of injuries were diagnosed at surgery, and cystic duct leaks represented 25% of these. The level of injury has remained unchanged with bismuth types I and II in 37% and types III and IV in 38%. Excluding patients with cystic duct leaks, 58% were referred after a failed ductal repair (169). The role of intraoperative cholangiography remains debatable. A questionnaire survey of surgeons by Torkington et al. obtained 362 replies; among these, 58 bile duct resection injuries were reported by 48 surgeons. Of the bile duct resection injuries reported, 49 of 58 (85%) occurred when an intraoperative cholangiogram was not performed (170). In another postal survey in New Zealand, despite the increase in experience of surgeons, iatrogenic injury to the common bile duct persists at 2.8% for all injuries and around 1% for major duct injuries (171). Wright and Wellwood showed that four accessory ducts were sacrificed, and localized injury to the common hepatic or bile duct occurred in three patients. However, the investigators concluded that these injuries would not have been prevented by operative cholangiography (172). In the rural community setting, among 2654 patients over 5 years, 0.25% sustained bile duct injury (173). 2— Gallbladder Perforation, Bile Leak, Hematoma, and Abscess In the early days, gallbladder perforation occurred in 32% of patients, with increased operative time spent in irrigation and the retrieval of stones. However, no adverse complications resulted and conversion to open surgery was unnecessary (174). In more recent times, intraoperative gallbladder perforation occurred in 26.3% of the 110 patients during dissection of the gallbladder bed, retraction with grasping forceps, extraction from the abdomen, and slippage of cystic duct clips (175). If patients with perforated gallbladder are given appropriate perioper
Page 502
ative antibiotics and the spilt bile is properly aspirated, followed by peritoneal irrigation, the operative and postoperative courses are similar to those of patients with unperforated gallbladder (176). One patient underwent successful endoscopic nasobiliary drainage for bile leakage resulting from clip displacement of the cystic duct stump (177). Hepatic subcapsular hematoma formation is not common; Pietra and associates described their experience with the successful management of this complication (178). Intraperitoneal abscesses are mostly secondary to unretrieved stones. Freedman et al. reported a case of incarcerated hernia and an associated abscess cavity containing a large spilled gallstone, which, on CT scan suggested a possible tumor of the abdominal wall. Spilled stones should be removed whenever possible but should not be an indication for conversion to open operation (179). Abscesses are mostly secondary to persistence of infected bile, unresolved hematoma, or spilled stones. A paranephric abscess was found in a 63yearold woman 21 months after her procedure requiring laparotomy and drainage (180). Endoclips that remained in place after being misfired or displaced accidentally from the cystic duct can become the source of an intraperitoneal abscess (181). Surgicel is a bioabsorbable thrombogenic gauze that, given the proper clinical context, can be indistinguishable from an abscess on postoperative CT scans (182), Festekjian and peers described an abdominal wall biloma arising from the persistence of bile in the peritoneal cavity (183). 3— Stone Spillage Gallbladder perforation during laparoscopic cholecystectomy with spillage of bile and gallstones occurs in up to 40% of patients (184). Stones left in the abdominal cavity or trapped in trocar sites after laparoscopic cholecystectomy can cause serious late complications requiring repeated surgical interventions (185). The Swiss collected data on 10,174 patients, among whom 581 cases of gallstones spillage occurred; 34 of these were primarily converted to open procedures for stone retrieval. Of the remaining 547 patients, only 8 developed postoperative abscesses that required reoperation (186). Reported complications have included intraperitoneal abscesses and the formation of foreignbody granulomas (187–190). Horton and Florence reported four cases where surgical drainage, stone removal, and antibiotics were required and one case of trocarsite inflammatory masses requiring excision (191). Others have reported spilled stones presenting as masses. Steerman et al. has a patient presenting with pain and a mass in the inguinal region requiring exploration (192). Besides causing mechanical bowel obstruction, a retained intraabdominal stone has also led to obstructive cholangitis (193). Noda and colleagues described a case of infected gallstones that traversed the diaphragm, migrated into the lung parenchyma, and obstructed a segmental bronchus, causing pneumonia (194). Despite peritoneal lavage, another patient developed chronic rightupperquadrant discomfort and a pleural effusion over several months; her symptoms resolved only following the appearance of a pigmented bilirubin stone in her sputum (195). Empyema formation has also been described following migration of gallstone into the thoracic cavity (196). Graham reported the formation of an abdominal wall sinus tract secondary to gallbladder perforation and stone spillage (197). Pohl and associates have described a case of spillage of several gallstones through an abdominocutaneous sinus tract to the umbilicus, with spontaneous resolution (198). Lutken et al. presented a rare complication in a patient with a persistent urachus who, 9 months after intraperitoneal spillage, started to pass gallstones while urinating (199). Other unusual sequelae include gallstones deposited in the ovary, causing infertility, 20 months following the initial surgery (200). 4— Others A biliary origin of acute postcholecystectomy pancreatitis (APP) was established in 4 (12.5%) of 32 patients; this complication was associated with the surgeon's experience level and a high morbidity of 31.3%. Mortality of patients with APP decreased substantially compared with those undergoing open biliary surgery (201).
Page 503
VII— Discussion Literature related to this surgical procedure has been voluminous in the last decade. Many suggest that laparoscopic cholecystectomy should be the treatment of choice for the majority of patients with gallbladder diseases. The indication for surgery in cholelithiasis has not dramatically changed since the introduction of the laparoscopic technique: for those with symptomatic gallstones, early elective surgery is recommended (202). The European Association of Endoscopic Surgery (EAES) Consensus Development Conferences on laparoscopic cholecystectomy recommended that laparoscopic cholecystectomy be the procedure of choice for symptomatic cholelithiasis (203). Growth in cholecystectomy rates following the introduction of laparoscopic cholecystectomy was accompanied by evidence of lower clinical thresholds for performing surgery. Cholecystectomy rates increased by 22% from 1989 to 1993 (204). In the early '90s, numerous randomized prospective trials to compare laparoscopic with conventional cholecystectomy have failed because of the immense popularity of the laparoscopic procedure (205). Subsequently, early prospective audits of laparoscopic cholecystectomy showed that results had been satisfactory, with a median operative time of 90 min and a median postoperative stay of 2 days demonstrated by Wilson et al. (206). The conversion rate has remained under 5% in most series (207). The audit of the Royal College of Surgeons of England reported a low morbidity and mortality on 3319 cases, but bile duct injury was more common in laparoscopic than in open procedures (208). In this early period, two questionnaire surveys were sent out to members of the EAES; the returns of both confirmed that laparoscopic cholecystectomy was a better operation than the open procedure (209). Data collected from a Swiss audit of 10,174 patients from 82 surgical services showed that although laparoscopic cholecystectomy was a safe procedure, the rate of conversion to open cholecystectomy, depending both on the indication and on intraoperative complications, was still substantial. There was still a 10.4% morbidity associated with the procedure, but the incidence of common bile duct injuries, which decreases with growing laparoscopic experience, was relatively low (210). In the United States, 114,005 cases from 40 series were reviewed, indicating that the conversion rate was primarily related to inflammation. Bile duct stones were detected intraoperatively in 7.8% and a high rate of intraoperative cholangiography was associated with a lower incidence of bile leaks and duct injuries; the latter occurred in the first 50 patients of the surgeon's experience in about 91% of the cases (163). In the same series, between 1989 to 1995, 561 major bile duct injuries (0.5%) and 401 bile leaks from the cystic duct or liver bed (0.38%) were recorded. Intraoperative cholangiography was attempted in almost half of the laparoscopic cholecystectomies and was successful in 82.7%. In major bile duct injuries, the common bile duct and/or common hepatic duct were the most frequently injured (61.1%), and less than 2% of patients had complete transection. The morbidity for laparoscopic cholecystectomy, excluding bile duct injuries or leaks, was 5.4% and the overall mortality was 0.06%. It was also noted that the conversion rate to an open procedure was 21.6% (211). A— Alternatives There are reasonably large differences among hospitals, hospital groups, and regions of a state in the type of cholecystectomy used, even after adjustment for differences in a number of variables (212). There are techniques such as minicholecystectomy as an alternative procedure, and the literature cites many conflicting results of this technique against laparoscopic cholecystectomy. A minilaparotomy has some advantages: it enables quick conversion to an open cholecystectomy, if required, it provides the surgeon with a threedimensional picture, and it is suitable for the surgical treatment of stones of the common bile duct (213). A randomized study by Barkun and coworkers suggests that the qualityoflife scores—such as hospital stay, convalescence, and return to work from laparoscopic cholecystectomy—are better than those
Page 504
associated with minicholecystectomy (214). Another randomized study by Squirrell and colleagues suggests that there is less tissue destruction and pain with the laparoscopic technique than with a smallincision cholecystectomy but that the antiemetic requirements were greater and that the procedure did not confer any advantage in the degree of postoperative respiratory impairment, length of hospital stay, or perceived postoperative health (215). A Finnish study on acute gallbladders reported that there was an increased level of technical difficulty using the laparoscopic approach but that is was nevertheless safe and effective in experienced hands. The mortality and morbidity rate seemed to be even lower than that of the open group and a moderately high conversion rate must be accepted (216). In the Third World, results of 737 minilaparotomy cholecystectomies suggested this method to be a safe, effective, and cheaper alternative to the laparoscopic approach (217). B— DayCase Surgery and CostEffectiveness Increasing evidence proposes laparoscopic cholecystectomy as a daycase procedure in selected patients (218–220). With less depression of lung volume and an earlier return of lung function to normal, there are fewer potential complications and patients can be discharged within 24 h (221). With appropriate selection criteria, patients can benefit from daycase laparoscopic procedures for gallbladder diseases; Prasad and Foley reported 51 cases, 3 requiring an overnight stay because of pain and drain placement (222). In Europe, daycase surgery is beginning to catch on; with technical advancement, a high percentage of patients can return to normal function with 24 h (223). Voitk has suggested that outpatient cholecystectomy is safe for the higherrisk patients and that ''precautionary" hospitalization may be harmful (224). Other studies have compared day surgery with observation; Keulemans and colleagues found that there were no differences with respect to the other in outcomes and that day care is therefore preferable, as it is less expensive (225). In 1994, Fullerton and associates estimated a cost of £2102 for open surgery compared to £2026 for the laparoscopic counterpart, but the cost appeared to decrease with experience (226). In another study, daycase surgery resulted in a reduction from $7800 to $4600 over a 3year period (227). Zegarra and colleagues reported an average charge for outpatient surgery of $3669, a 25% reduction with no increase morbidity (228). In 1996, a study involving 18 U.S. states showed that the average total charge for an inhospital laparoscopic cholecystectomy was $13,940; it was $15,380 for an open cholecystectomy. Per day charges averaged $4130 and $2510 for laparoscopic and open procedures, respectively (229). After the initial hype and excitement over minimalaccess surgery of the gallbladder, evaluation of its health economics gathered momentum (230,231). A German study concluded that the most effective savings might be achieved by shortening the hospital stay and the period away from work. It broke down the costs of laparoscopic cholecystectomy into different components. The costs for the operation itself represented 19%, hotel and medical treatment (except for the operation) represented 47%, and the loss of income accounted for another 33% of the total costs (232). Early reports of costeffectiveness were not substantiated, as few procedures were performed and complications were high due to the steep learning curve. Nicoll et al. reported that conventional lithotripsy appeared to be at least as costeffective as cholecystectomy for patients with less bulky stones but less costeffective for those with large stones (233). Darzi and associates demonstrated the high cost, high failure rate, and considerable recurrence rate of lithotripsy compared to laparoscopic cholecystectomy (234). Despite a shorter duration of disability than with the laparoscopic procedure, lithotripsy was associated with disappearance of stones in less than 50% of cases (235). A further study by Go and colleagues comparing extracorporeal shockwave lithotripsy, conventional cholecystectomy, and laparoscopic cholecystectomy showed that the latter is the most costeffective means of managing patients with symptomatic cholelithiasis (236). A low complication rate became an important issue, besides considering the cost of arbitrary equipment and the role of routine cholangiography, in order for laparoscopic cholecystectomy to prove costeffective (237). Bass et al. used
Page 505
a computerized model and estimated the probabilities of potential outcomes of each procedure, associated qualityoflife effects, and related direct medical charges. The model projected that the laparoscopic method would be more effective than the open equivalent in terms of total mortality and qualityadjusted survival (238). Two retrospective studies showed conflicting and controversial results regarding costs and effective outcomes offered by the procedure. McKellar and peers found that the shorter recovery period from laparoscopic cholecystectomy, allowing the patients an earlier return to full preoperative activities, contributed to its cost effectiveness compared to open cholecystectomy, despite the overall increased total charge (239). The total hospital cost was considered 10% lower in the laparoscopy group by Wenner and colleagues, $1864 as compared to $2030 per patient in the open group and a median sickness allowance of $516 per patient compared to $1424, respectively (240). Despite some discrepancy in costs, effects overall are better for patients and direct costs are lower for laparoscopic cholecystectomy than for the conventional open technique. Hospital costs are initially higher, but as the learning curve is overcome, operative time would be shortened, reducing the number of procedures required to make it costeffective. Other costsaving measures could be employed. Hospitals have begun to realize that potential savings could be made especially by employing reusables (241). The incidence of blood transfusion was 0.46% in a series of 2589 patients undergoing the laparoscopic procedure. By avoiding type and screen, substantial cost savings were achieved (242). C— Future 1— Training The learning curve for laparoscopic cholecystectomy remains varied. One study suggested similarities between trainees and seniors in successes for cholangiograms, conversion rates, complications, and postoperative stays, but a shorter operative time for senior surgeons was found (243). The performance of difficult laparoscopic cholecystectomy by residents with the assistance of attending surgeons carried a higher conversion rate to laparotomy than those performed by the attending, although seniority did not influence the conversion rate (244). From inception, various centers have incorporated this basic laparoscopic procedure into their surgical programs. Although there was an initial decline in the number of gallbladder procedures performed by the residents at the introduction of laparoscopic cholecystectomy, the outcomes that followed were found to be similar between the supervised residents and the surgeons (245). Zucker et al. described residents who are completing their training, have performed an average 50 to 75 laparoscopic procedures as the primary surgeon and 25 to 30 as the first assistant (246). Hodgson's study reported on conventional apprenticeship for this procedure in a traditional residency program by adequately trained attending surgeons (247). According to Namias and colleagues, there is a rising trend of laparoscopic cholecystectomies and all cholecystectomies, but there is a reciprocal decline in the number of open procedures performed over a 5year period (248). Another study reported comparable complication rates for graduating residents after 5 years of exposure to laparoscopic cholecystectomy (249). The fear of decreasing exposure to the open procedures haunted trainees and trainers in the early days. ScottConner et al. expressed concerns from their residents about training in open procedures. Their current and graduated residents considered didactic sessions, including animal laboratories and simulators, an important part of training (250). The lack of previous experience in open cholecystectomy did not appear to alter the complication or conversion rates for residents, but more intraoperative cholangiograms were performed, probably due to the fear of making mistakes (251). Also noted in a study of a group of 96 residents by Friedman et al. was that learning via graded experience throughout training achieved results similar to those found with the addition of an intensive course (252). In countries where laparoscopic cholecystectomy has not been adopted, opportunities exist for surgeons to attend courses held in recognized regional training centers. Visiting sur
Page 506
geons can offer valuable handson tips to those who have just been initiated in the procedures. Handson animal models and a period of preceptorship with dedicated laparoscopic surgeons from the region or abroad can improve local understanding and techniques among surgeons and trainees (253). Asbun and colleagues successfully set up laparoscopic training courses in developing countries such as Nicaragua and Bolivia, teaching welltrained and motivated local surgeons under financial and other constraints (254). However, numerous limitations in developing countries still hinder the development of this surgical technique. In the early days of minimalaccess gallbladder surgery, models and simulators were developed to improve understanding of the new procedure. Simulators made of pigs' gallbladders have been used (255). Other simulators include the prototype simulator known as the "visible man" liver, with data sets conformed to the "standard" patients (256). Validation of surgical skills and objective measurement of dexterity may become an important part of surgical training. In general, a skillfully performed operation is 75% decision making and 25% dexterity. In some specialties, such as minimally invasive surgery, dexterity becomes more important (257). 2— Robotics and Telemedicine With the aid of digital and satellite technology, the advent of robotics and telemedicine have led to a breakthrough in the amalgamation of minimal access surgery and telecommunication (258). Surgical education in clinical anatomy and learning of operative techniques will take on another meaning, using simulators in virtual reality and telepresence systems (259). Kouvoussi et al. began experimentation with telerobotics in the lab and clinically as far back as 1994 (260). A mobile telemanipulator with a humanmachine interface, incorporating modified laparoscopic instruments with pivotal tips, has made remote laparoscopic cholecystectomy in a phantom model possible (261). Robotic assistance can be activated by voice or by foot pedal. Allaf and associates showed that foot control was faster and had less operatorinterface failures, but voice control was more accurate (262). Laparoscopic maneuvering and suturing can be faster and just as precise when performed manually as when performed with the prototype robotic system (263). The arrival of advanced telecommunications and videoconferencing equipment brought surgical proctoring and telementoring to new heights, especially in the area of laparoscopic procedures. Although much work is in the experimental stage, some procedures have been performed in various battlefields and war zones (264). By using suitable transmission and compression standards, telesurgery for laparoscopic procedures will be possible (265). Although satellite and dedicated lines are available, the Integrated Services Digital Network (ISDN) is the best transmission option at present (266). Reports of successful telementoring for laparoscopic procedures between sites close to each other and very much apart are encouraging (267,268). Telementoring will continue to improve, and early studies on laparoscopic procedures appeared to be encouraging (269). Telementoring has also been shown to work for advanced laparoscopic procedures (270). Remote advice can also be offered to less specialized centers performing laparoscopic surgery (271). High cost may limit the usage to institutions that can invest in the equipment, but solo laparoscopic surgery can be practiced with ease by enlisting these aids (272,273). However, much research and development will be required before these stateof theart technologies are fully implemented. VIII— Conclusion Extensive development has taken place since the inception of laparoscopic cholecystectomy. From modification of techniques, introduction of new instruments, education of a new generation of trainees, to the use of robotics and telemedicine, all these will no doubt bring the surgical management of gallstones into a new dimension. Changes are progressive; though we might be reluctant, some of these advances will inevitably find their way into our practices. Will we look back in a decade and ask each other "Which robot took out your gallbladder?"
Page 507
The "maiden voyage" discovered a new world, but it will be evolution that takes this technique to great heights. References 1. Rosin R. History. In: Rosen R, ed. Minimal Access Medicine and Surgery Principles and Techniques. Oxford: Radcliffe Medical Press, 1993, pp 1–9. 2. Litynski GS. Erich Muhe and the rejection of laparoscopic cholecystectomy (1985): a surgeon ahead of his time. J Soc Laparoendosc Surg 1998; 2:341–346. 3. Mouret P. From the first laparoscopic cholecystectomy to the frontiers of laparoscopic surgery: the future prospectives. Dig Surg 1991; 8:124–125. 4. Dubois F, Icard P, Bethelot G, Levard H. Coelioscopic cholecystectomy: preliminary report of 36 cases. Ann Surg 1990; 211:60–62. 5. Dubois F, Bethelot G, Levard H. Laparoscopic cholecystectomy: historic perspective and personal experience. Surg Laparosc Endosc 1991; 4:52–57. 6. Perrisat J, Collet D, Belliard B. Gallstones; laparoscopic treatment; cholecystectomy; cholecystoscopy and lithotripsy. Surg Endosc 1990; 4:1–5. 7. Reddick E, Olsen D, Daniell J, et al. Laparoscopic laser cholecystectomy. Laser Med Surg News Adv 1989; 3:38–40. 8. Martin IG, Holdsworth PJ, Asker J, Baltas B, Glinatsis MT, SueLing H, Gibson J, Johnston D, McMahon MJ. Laparoscopic cholecystectomy as a routine procedure for gallstones: results of an "allcomers" policy. Br J Surg 1992; 79:807–810. 9. Frazee RC, Roberts JW, Symmonds R, Snyder SK, Hendricks J, Smith R, Custer MD. What are the contraindications for laparoscopic cholecystectomy? Am J Surg 1992; 164:491–495. 10. Wallace DH, O'Dwyer PJ. Effect of a noconversion policy on patient outcome following laparoscopic cholecystectomy. Br J Surg 1997; 84:1680–1682. 11. Fontes PR, Nectoux M, Eilers RJ, Chem EM, Riedner CE. Is acute cholecystitis a contraindication for laparoscopic cholecystectomy? Int Surg 1998; 83:28–30. 12. Eldar S, Sabo E, Nash E, Abrahamson J, Matter I. Laparoscopic cholecystectomy for the various types of gallbladder inflammation: a prospective trial. Surg Laparosc Endosc 1998; 8:200–207. 13. Lo CM, Liu CL, Fan ST, Lai EC, Wong J. Prospective randomized study of early versus delayed laparoscopic cholecystectomy for acute cholecystitis. Ann Surg 1998; 227:461–467. 14. Schwesinger WH, Sirinek KR, Strodel WE III. Laparoscopic cholecystectomy for biliary tract emergencies: state of the Art. World J Surg 1999; 23:334–342. 15. Lai PB, Kwong KH, Leung KL, Kwok SP, Chan AC, Chung SC, Lau WY. Randomized trial of early versus delayed laparoscopic cholecystectomy for acute cholecystitis. Br J Surg 1998; 85:764–767. 16. Alberti D, Callea F, Camoni G, Falchetti D, Rigamonti W, Caccia G. Adenomyomatosis of the gallbladder in childhood. J Pediatr Surg 1998; 33:1411–1412. 17. Frassinelli P, Werner M, Reed JR III, Scaglotti C. Laparoscopic cholecystectomy alleviates pain in patients with acalculous biliary disease. Surg Laparosc Endosc 1998; 8:30–34. 18. Lujan JA, SanchezBueno F, Parrilla P, Robles R, Torralba JA, GonzalezCostea R. Laparoscopic vs. open cholecystectomy in patients aged 65 and older. Surg Laparosc Endosc 1998; 8:208–210. 19. Maxwell JG, Tyler BA, Rutledge R, Brinker CC, Maxwell BG, Covington DL. Cholecystectomy in patients aged 80 and older. Am J Surg 1998; 176:627–631. 20. Magnuson TH, Ratner LE, Zenilman ME, Bender JS. Laparoscopic cholecystectomy: applicability in the geriatric population. Am Surg 1997; 63:91–96.
Page 508
21. Kumar SS. Laparoscopic cholecystectomy in the densely scarred abdomen. Am Surg 1998; 64:1094–1096. 22. Diez J, Delbene R, Ferreres A. The feasibility of laparoscopic cholecystectomy in patients with previous abdominal surgery. HPB Surg 10:353–356. 23. Vezakis A, Dexter SP, Martin IG, Larvin M, McMahon MJ. Laparoscopic cholecystectomy after pancreatic debridement. Surg Endosc 1998; 12(6):865–866. 24. Ghumman E, Barry M, Grace PA. Management of gallstones in pregnancy. Br J Surg 1997; 84:1646–1650. 25. Geisler JP, Rose SL, Mernitz CS, Warner JL, Hiett AK. Nongynecologic laparoscopy in second and third trimester pregnancy: obstetric implications. J Soc Laparoendosc Surg 1998; 2:235–238. 26. Thomas SJ, Brisson P. Laparoscopic appendectomy and cholecystectomy during pregnancy: six case reports. J Soc Laparoendosc Surg 1998; 2:41–46. 27. Gouldman JW, Sticca RP, Rippon MB, McAlhany JC Jr. Laparoscopic cholecystectomy in pregnancy. Am Surg 1998; 64:93–87. 28. Glasgow RE, Visser BC, Harris HW, Patti MG, Kilpatrick SJ, Mulvihill SJ. Changing management of gallstone disease during pregnancy. Surg Endosc 1998; 12:241–246. 29. Graham G, Baxi L, Tharakan T. Laparoscopic cholecystectomy during pregnancy: a case series and review of the literature. Obstet Gynecol Surv 1998; 53:566– 574. 30. Kasirajan K, Obermeyer RJ, Kehris J, Lopez J, Lopez R. Microinvasive laparoscopic cholecystectomy in pediatric patients. J Laparoendosc Adv Surg Tech A 1998; 8:131–135. 31. Johna S, Shaul D, Taylor EW, Brown CA, Bloch JH. Laparoscopic management of gallbladder disease in children and adolescents. J Soc Laparoendosc Surg 1997; 1:241–245. 32. Angrisani L, Lorenzo M, De Palma G, Sivero L, Catanzano C, Tesauro B, Persico G. Laparoscopic cholecystectomy in obese patients compared with nonobese patients. Surg Laparosc Endosc 1995; 5:197–201. 33. Collet D, Edye M, Magne E, Perissat J. Laparoscopic cholecystectomy in the obese patient. Surg Endosc 1992; 6:186–188. 34. Phillips EH, Carroll BJ, Fallas MJ, Pearlstein AR. Comparison of laparoscopic cholecystectomy in obese and nonobese patients. Am Surg 1994; 60:316–321. 35. Schirmer BD, Dix J, Edge SB, Hyser MJ, Hanks JB, Aguilar M. Laparoscopic cholecystectomy in the obese patient. Ann Surg 1992; 216:146–152. 36. Sleeman D, Namias N, Levi D, Ward FC, Vozenilek J, Silva R, Levi JU, Reddy R, Ginzburg E, Livingstone A. Laparoscopic cholecystectomy in cirrhotic patients. J Am Coll Surg 1998; 187:400–403. 37. Angrisani L, Lorenzo M, Corcione F, Vincenti R. Gallstones in cirrhotics revisited by a laparoscopic view. J Laparoendosc Adv Surg Tech A 1997; 7:213–220. 38. Jones PE, Sayson SC, Koehler DC. Laparoscopic cholecystectomy in a cardiac transplant candidate with an ejection fraction of less than 15%. J Soc Laparoendosc Surg 1998; 2:89–92. 39. Menegaux F, Dorent R, Tabbi D, Pavie A, Chigot JP, Gandjbakhch I. Biliary surgery after heart transplantation. Am J Surg 1998; 175:320–321. 40. Yoshida T, Kitano S, Matsumoto T, Bandoh T, Baatar D, Ninomiya K, Hadama T. Laparoscopic cholecystectomy in patients undergoing anticoagulant therapy. Surg Today 1998; 28:308–312. 41. Gould SW, Banwell PE, Rosin RD. Recurrent melanoma of the leg and polypoid lesions of the gallbladder—a management dilemma. Eur J Surg Oncol 1997; 23:368–369. 42. McGrath DS, Short C, Bredin CP, Kirwan WO, Rooney E, Meeke R. Laparoscopic cholecystectomy in adult cystic fibrosis. Ir J Med Sci 1997; 166:70–71. 43. Magnuson TH, Bender JS, Campbell KA, Ratner LE. Cholecystectomy in the peritoneal dialysis patient: unique advantages to the laparoscopic approach. Surg Endosc 1995; 9:908–909.
Page 509
44. Carlei F, Lezoche E, Lomanto D, Schietroma M, Paganini A, Sottili M, Nardovino M. Cholecystoenteric fistula is not a contraindication for laparoscopic cholecystectomy: report of five cases treated by laparoscopic approach. Surg Laparosc Endosc 1997; 7:403–406. 45. Leiva JI, Etter EL, Gathe J Jr, Bonefas ET, Melartin R, Gathe JC. Surgical therapy for 101 patients with acquired immunodeficiency syndrome and symptomatic cholecystitis. Am J Surg 1997; 174:414–416. 46. Ricci M, Puente AO, Rothenberg RE, Shapiro K, de Luise C, LaRaja RD. Open and laparoscopic cholecystectomy in acquired immunodeficiency syndrome: indications and results in fiftythree patients. Surgery 1999; 125:172–177. 47. Kwon AH, Inui H, Imamura A, Uetsuji S, Kamiyama Y. Preoperative assessment for laparosopic cholecystectomy: feasibility of using spiral computed tomography. Ann Surg 1998; 227:351–356. 48. Dwerryhouse SJ, Brown E, Vipond MN. Prospective evaluation of magnetic resonance cholangiography to detect common bile duct stones before laparoscopic cholecystectomy. Br J Surg 1998; 85:1364–1366. 49. Kriwanek S, Armbruster C, Beckerhinn P, Blauensteier W, Gschwantler M. Patients' assessment and recall of surgical information after laparoscopic cholecystectomy. Dig Surg 1998; 15:669–673. 50. Bradbury AW, Chan YC, Darzi A, Stansby G. Thromboembolism prophylaxis during laparoscopic cholecystectomy. Br J Surg 1997; 84:962–964. 51. Lippert H, Gastinger J. Antimicrobial prophylaxis in laparoscopic and conventional cholecystectomy: conclusions of a large prospective multicenter quality assurance study in Germany. Chemotherapy 1998; 44:355–363. 52. Garcia N, Kapur S, McClane J, Davis JM. Surgical infections and prophylactic antibiotics: 341 consecutive cases of gallbladder surgery in the era of laparoscopic surgery. J Laparoendosc Adv Surg Tech A 1997; 7:157–162. 53. Illig KA, Schmidt E, Cavanaugh J, Krusch D, Sax HC. Are prophylactic antibiotics required for elective laparoscopic cholecystectomy? J Am Coll Surg 1997; 184:353–356. 54. Samel S, Post S, Martell J, Becker H. Clostridial gas gangrene of the abdominal wall after laparoscopic cholecystectomy. J Laparoendosc Adv Surg Tech A 1997; 7:245–247. 55. Dobay KJ, Freier DT, Albear P. The absent role of prophylactic antibiotics in lowrisk patients undergoing laparoscopic cholecystectomy. Am Surg 1999; 65:226–228. 56. Darvas K, Molnar Z, Irto I, Tarjanyi M, Flautner L. Anaesthesiological indications and contraindications of minimally invasive surgery. Acta Chir Hung 1997; 36:72–75. 57. Reed DN Jr, Nourse P. Untoward cardiac changes during CO2 insufflation in laparoscopic cholecystectomies in lowrisk patients. J Laparoendosc Adv Surg Tech A 1998; 8:109–114. 58. Pursnani KG, Bazza Y, Calleja M, Mughal MM. Laparoscopic cholecystectomy under epidural anesthesia in patients with chronic respiratory disease. Surg Endosc 1998; 12:1082–1084. 59. Majeed AW, Plura M, Priest S, Johnson AG. Is it necessary to catheterise the bladder before laparoscopy? Surg Laparosc Endosc 1998; 8:157–158. 60. Birkett DH, Josephs LG, EsteMcDonald J. A new 3D laparoscope in gastrointestinal surgery. Surg Endosc 1994; 8:1448–1451. 61. Hanna GB, Shimi SM, Cuschieri A. Randomised study of influence of twodimensional versus threedimensional imaging on performance of laparoscopic cholecystectomy. Lancet 1998; 351:248–251. 62. Apelgren KN, Blank ML, Slomski CA, Hadjis NS. Reusable instruments are more costeffective than disposable instruments for laparoscopic cholecystectomy. Surg Endosc 1994; 8:32–34. 63. Paolucci V, Schaeff B, Gutt CN, Encke A. Disposable versus reusable instruments in
Page 510
laparoscopic cholecystectomy: a prospective, randomised study. Endosc Surg Allied Technol 1995; 3:147–150. 64. Rosin D. Laparoscopic cholecystectomy. In: Zinner MJ, Schwartz SI, Ellis H eds. Maingot's Abdominal Surgery. Norwalk, CT: Appleton & Lange, 1997, pp 1855–1865. 65. Uranus S, Peng Z, Kronberger L, Pfeifer J, Salehi B. Laparoscopic cholecystectomy using 2mm instruments. J Laparoendosc Adv Surg Tech A 1998; 8:255– 259. 66. Davides D, Dexter SP, Vezakis A, Larvin M, Moran P, McMahon MJ. Micropuncture laparoscopic cholecystectomy. Surg Endosc 1999; 13:236–238. 67. Ngoi SS, Goh P, Kok K, Kum CK, Cheah WK. Needlescopic or ministie cholecystectomy. Surg Endosc 1999; 13:303–305. 68. Tagaya N, Kita J, Takagi K, Imada T, Ishikawa K, Kogure H, Ohyama O. Experience with threeport laparoscopic cholecystectomy. J Hepatobil Pancreat Surg 1998; 5:309–311. 69. Ramachandran CS, Arora V. Twoport laparoscopic cholecystectomy: an innovative new method for gallbladder removal. J Laparoendosc Adv Surg Tech A 1998; 8:303–308. 70. Bresadola F, Pasqualucci A, Donini A, Chiarandini P, Anania G, Terrosu G, Sistu MA, Pasetto A. Elective transumbilical compared with standard laparoscopic cholecystectomy. Eur J Surg 1999; 165:29–34. 71. Halpern NB. Access problems in laparoscopic cholecystectomy: postoperative adhesions, obesity, and liver disorders. Semin Laparosc Surg 1998; 5:92–106. 72. Hashizume M, Sugimachi K. Needle and trocar injury during laparoscopic surgery in Japan. Surg Endosc 1997; 11:1198–1201. 73. Cogliandolo A, Manganaro T, Saitta FP, Micali B. Blind versus open approach to laparoscopic cholecystectomy: a randomized study. Surg Laparosc Endosc 1998; 8:353–355. 74. Aitola P, Airo I, Kaukinen S, Ylitalo P. Comparison of N2O and CO2 pneumoperitoneums during laparoscopic cholecystectomy with special reference to postoperative pain. Surg Laparosc Endosc 1998; 8:140–144. 75. Berberoglu M, Dilek ON, Ercan F, Kati I, Ozmen M. The effect of CO2 insufflation rate on the postlaparoscopic shoulder pain. J Laparoendosc Adv Surg Tech A 1998; 8:273–277. 76. Odeberg S, Ljungqvist O, Sollevi A. Pneumoperitoneum for laparoscopic cholecystectomy is not associated with compromised splanchnic circulation. Eur J Surg 1998; 164:843–844. 77. Draper K, Jefson R, Jongeward R Jr, McLeod M. Duration of postlaparoscopic pneumoperitoneum. Surg Endosc 1997; 11:809–811. 78. Matsumoto T, Kitano S, Yoshida T, Sato K, Bandoh T, Ninomiya K, Baatar D, Hadama T. Laparoscopic cholecystectomy using the abdominal wall lift in a cardiac patient. Hepatogastroenterology 1998; 45:1476–1478. 79. Carry PY, Gallet D, Francois Y, Perdrix JP, Sayag A, Gilly F, Eberhard A, Banssillon V, Baconnier P. Respiratory mechanics during laparoscopic cholecystectomy: the effects of the abdominal wall lift. Anesth Analg 1998; 87:1393–1397. 80. Nanashima A, Yamaguchi H, Tsuji T, Yamaguchi E, Sawai T, Yasutake T, Nakagoe T, Ayabe H. Physiologic stress responses to laparoscopic cholecystectomy: a comparison of the gasless and pneumoperitoneal procedures. Surg Endosc 1998; 12:1381–1385. 81. Beneveto A, Carcano G, Cuffari A. New techniques in laparoscopic cholecystectomy: ultrasonography, CUSA and argon coagulator. Br J Surg 1993; 80 (suppl):543. 82. Wetter LA, Payne JH, Kirshenbaum G, Podoll EF, Bachinsky T, Way LW. The ultrasonic dissector facilitates laparoscopic cholecystectomy. Arch Surg 1992; 127:1195–1198. 83. Naude GP, Morris E, Bongard FS. Laparoscopic cholecystectomy facilitated by hydrodissection. J Laparoendosc Adv Surg Tech A 1998; 8:215–218. 84. Tsimoyiannis EC, Jabarin M, Glantzounis G, Lekkas ET, Siakas P, StefanakiNikou S. Laparoscopic cholecystectomy using ultrasonically activated coagulating shears. Surg Laparosc Endosc 1998; 8:421–424.
Page 511
85. Hershman MJ, Rosin RD. Laparoscopic laser cholecystectomy: our first 200 patients. Ann R Coll Surg Engl 1992; 74:242–247. 86. Bordelon BM, Hobday KA, Hunter JG. Laser vs electrosurgery in laparoscopic cholecystectomy: a prospective randomized trial. Arch Surg 1993; 128:233– 236. 87. Corbitt JD Jr. Laparoscopic cholecystectomy: laser versus electrosurgery. Surg Laparosc Endosc 1991; 1:85–88. 88. Cushieri A. Minimal invasive surgery for gallstone disease: laparoscopic cholecystectomy. In: Carter, Russell, Pitt, eds. Atlas of General Surgery. London: Chapman & Hall Medical, 1996, pp 422–455. 89. MacIntyre IM, Wilson RG. Laparoscopic cholecystectomy. Br J Surg 1993; 80:552–559. 90. CheslynCurtis S. General Surgery. In: Rosin R, ed. Minimal Access Medicine and Surgery Principles and Techniques. Oxford: Radcliffe Medical Press, 1993, pp 94–121. 91. Russell RCG. General surgery. Br Med J 1993; 307:1266–1269. 92. Zollinger RM Jr, Zollinger RM. Laparoscopic cholecystectomy. In: Zollinger RM Jr, Zollinger RM, eds. Atlas of Surgical Operations. Singapore: McGrawHill, 1993, pp 174–181. 93. Darzi A, Talamini MA, Dunn D. Cholecystectomy. In: Darzi A, Talamini MA, Dunn D, eds. Atlas of Laparoscopic Surgical Technique. London: Saunders, 1997, pp 31–46. 94. Salky BA, Edye MB. The difficult cholecystectomy: problems related to concomitant diseases. Semin Laparosc Surg 1998; 5(2): 107–114. 95. Gal I, Szivos J, Jaberansari MT, Szabo Z. Laparoscopic cholecystectomy: risk of missed pathology of other organs. Surg Endosc 1998; 12:825–827. 96. Junger W, Junger WG, Hutter J, Miller K, Moritz E. Delayed diagnosis of malignant tumors missed at laparoscopic cholecystectomy. Surg Endosc 1997; 11:1010–1012. 97. Sekimoto M, Tomita N, Tamura S, Ohsato H, Monden M. New retraction technique to allow better visualization of Calot's triangle during laparoscopic cholecystectomy. Surg Endosc 1998; 12:1439–1441. 98. Hasuike Y, Tomita N, Sakon M, Tamura S, Umeshita K, Kanai T, Gotoh M, Monden M. A new technique for exposing Calot's triangle during laparoscopic surgery: usefulness of extracorporeal lifting of the round ligament and gallbladder. Surg Laparosc Endosc 1996; 6:307–309. 99. Hashimoto D, Nayeem SA, Kajiwara S, Hoshino T, Fukuyo T, Hashimoto D, Nayeem SA, Kajiwara S, Hoshino T, Fukuyo T. Ojigi tying spatula: a newly developed movable spatula for laparoscopic ligation. Surg Laparosc Endosc 1993; 3:459–461. 100. Kimura T, Goto H, Takeuchi Y, Yoshida M, Kobayashi T, Sakuramachi S, Harada Y. Intraabdominal contamination after gallbladder perforation during laparoscopic cholecystectomy and its complications. Surg Endosc 1996; 10:888–891. 101. Ransom KJ. Laparoscopic management of acute cholecystitis with subtotal cholecystectomy. Am Surg 1998; 64:955–957. 102. Michalowski K, Bornman PC, Krige JE, Gallagher PJ, Terblanche J. Laparoscopic subtotal cholecystectomy in patients with complicated acute cholecystitis or fibrosis. Br J Surg 1998; 85:904–906. 103. Stuart SA, Simpson TI, Alvord LA, Williams MD. Routine intraoperative laparoscopic cholangiography. Am J Surg 1998; 176:632–637. 104. Corder AP, Scott SD, Johnson CD. Place of routine operative cholangiography at cholecystectomy. Br J Surg 1992; 79:945–947. 105. Young C, Moont M. Routine cholecystocholangiography: a viable alternative during laparoscopic cholecystectomy. Aust N Z J Surg 1998; 68:425–427. 106. Braghetto I, Debandi A, Korn O, Bastias J. Longterm followup after laparoscopic cholecystectomy without routine intraoperative cholangiography. Surg Laparosc Endosc 1998; 8:349–352. 107. Wu JS, Dunnegan DL, Soper NJ. The utility of intracorporeal ultrasonography for
Page 512
screening of the bile duct during laparoscopic cholecystectomy. J Gastrointest Surg 1998; 2:50–60. 108. Birth M, Ehlers KU, Delinikolas K, Weiser HF. Prospective randomized comparison of laparoscopic ultrasonography using a flexibletip ultrasound probe and intraoperative dynamic cholangiography during laparoscopic cholecystectomy. Surg Endosc 1998; 12:30–36. 109. Catheline JM, Turner R, Rizk N, Barrat C, Buenos P, Champault G. Evaluation of the biliary tree during laparoscopic cholecystectomy: laparoscopic ultrasound versus intraoperative cholangiography: a prospective study of 150 cases. Surg Laparosc Endosc 1998; 8:85–91. 110. Thompson DM, Arregui ME, Tetik C, Madden MT, Wegener M. A comparison of laparoscopic ultrasound with digital fluorocholangiography for detecting choledocholithiasis during laparoscopic cholecystectomy. Surg Endosc 1998; 12:929–932. 111. Leslie KA, Rankin RN, Duff JH. Lost gallstones during laparoscopic cholecystectomy: are they really benign? Can J Surg 1994; 37:240–242. 112. Stoblen F, Buess G, Manncke K, Mentges B, Lirici MM, Starlinger M, Becker HD. LaparoLith: a new instrument for stone fragmentation in laparoscopic cholecystectomy. Surg Endosc 1992; 6:309–312. 113. Kanehira E, Omura K, Kawai K, Ishikawa N, Watanabe Y. A new extraction bag to facilitate stone extraction in laparoscopic cholecystectomy: in vitro experiments. Endosc Surg Allied Technol 1994; 2:297–299. 114. Patterson EJ, Nagy AG. Don't cry over spilled stones? Complications of gallstones spilled during laparoscopic cholecystectomy: case report and literature review. Can J Surg 1997:40:300–304. 115. Ohmura Y, Yokoyama N, Tanada M, Takiyama W, Takashima S. Port site recurrence of unexpected gallbladder carcinoma after a laparoscopic cholecystectomy: report of a case. Surg Today 1999; 29:71–75. 116. Qureshi MA, Brindley NM, Leahy AL, Osborne DH, Broe PJ, BouchierHayes DJ, Grace PA. Postcholecystectomy symptoms after laparoscopic cholecystectomy. Ann R Col Surg Engl 1993; 75:349–353. 117. Van der Velpen GC, Smith SM, Cuschieri A. Outcome after cholecystectomy for symptomatic gallstone disease and effect of surgical access: laparoscopic v open approach. Gut 1993; 34:1448–1451. 118. Rothwell JF, Lawlor P, Byrne PJ, Walsh TN, Hennessy TP, Cholecystectomyinduced gastroesophageal reflux: is it reduced by the laparoscopic approach? Am J Gastroenterol 1997; 92:1351–1354. 119. Maddern GJ, Baxter PS. The effect of laparoscopic cholecystectomy on gastroduodenal reflux. Aust N Z J Surg 1997; 67:703–705. 120. Chundrigar T, Hedges AR, Morris R, Stamatakis JD. Intraperitoneal bupivacaine for effective pain relief after laparoscopic cholecystectomy. Ann R Coll Surg Engl 1993; 75:275–280. 121. Tsimoyiannis EC, Glantzounis G, Lekkas ET, Siakas P, Jabarin M, Tzourou H. Intraperitoneal normal saline and bupivacaine infusion for reduction of postoperative pain after laparoscopic cholecystectomy. Surg Laparosc Endosc 1998; 8:416–420. 122. Tsimoyiannis EC, Siakas P, Tassis A, Lekkas ET, Tzourou H, Kambili M. Intraperitoneal normal saline infusion for postoperative pain after laparoscopic cholecystectomy. World J Surg 1998; 22:824–828. 123. Fredman B, Olsfanger D, Jedeikin R. A comparative study of ketorolac and diclofenac on postlaparoscopic cholecystectomy pain. Eur J Anaesthesiol 1995; 12 (5):501–504. 124. Andrei VE, Schein M, Margolis M, Rucinski JC, Wise L. Liver enzymes are commonly elevated following laparoscopic cholecystectomy: is elevated intra abdominal pressure the cause? Dig Surg 1998; 15:256–259. 125. Morino M, Giraudo G, Festa V. Alterations in hepatic function during laparoscopic surgery: an experimental clinical study. Surg Endosc 1998; 12:968–972.
Page 513
126. Windsor MA, Bonham MJ, Rumball M. Splanchnic mucosal ischemia: an unrecognized consequence of routine pneumoperitoneum. Surg Laparosc Endosc 1997; 7:480–482. 127. Dev V, Shah D, Gaw F, Lefor AT. Gastric outlet obstruction secondary to post cholecystectomy biloma: case report and review of the literature. J Soc Laparoendosc Surg 1998; 2:185–188. 128. Lay PS, Tsang TK, Caprini J, Gardner A, Pollack J, Norman E. Volvulus of the small bowel: an uncommon complication after laparoscopic cholecystectomy. J Laparoendosc Adv Surg Tech A 1997; 7:59–62. 129. Jena GP, Monoharan GR. Minimally invasive, finally fatal—duodenal injury complicating laparoscopic cholecystectomy. S Afr Med J 1998; 88:732–733. 130. Berry MA, Rangraj M. Conservative treatment of recognized laparoscopic colonic injury. J Soc Laparoendosc Surg 1998; 2:195–196. 131. Tekin A. Mechanical small bowel obstruction secondary to spilled stones. J Laparoendosc Adv Surg Tech A 1998; 8:157–159. 132. Kopelman D, Schein M, Assalia A, Hashmonai M. Small bowel obstruction following laparoscopic cholecystectomy: diagnosis of incisional hernia by computed tomography. Surg Laparosc Endosc 1994; 4:325–326. 133. Tseng YH, Ho WM, Hung WT. Hypovolemic shock induced by laparoscopic cholecystectomy—a case report. Acta Anaesthesiol Sin 1997; 35:247–252. 134. Usal H, Sayad P, Hayek N, Hallak A, Huie F, Ferzli G. Major vascular injuries during laparoscopic cholecystectomy: an institutional review of experience with 2589 procedures and literature review. Surg Endosc 1998; 12:960–962. 135. Lindberg F, Bergqvist D, Rasmussen I. Incidence of thromboembolic complications after laparoscopic cholecystectomy: review of the literature. Surg Laparosc Endosc 1997; 7:324–331. 136. MartinezRamos C, LopezPastor A, NunezPena JR, RuizCaravaca ML, Ruiz de Gopegui M, SanzLopez R, Jorgensen TW, TamamesEscobar S. Fibrinolytic activity in laparoscopic cholecystectomy. J Laparoendosc Adv Surg Tech A 1998; 8:417–423. 137. Lord RV, Ling JJ, Hugh TB, Coleman MJ, Doust BD, NivisonSmith I. Incidence of deep vein thrombosis after laparoscopic vs minilaparotomy cholecystectomy. Arch Surg 1998; 133:967–973. 138. Sternberg A, Alfici R, Bronek S, Kimmel B. Laparoscopic surgery and splanchnic vessel thrombosis. J Laparoendosc Adv Surg Tech A 1998; 8:65–68. 139. Klugewitz K, Rehermann B, Seifert U, Boker KH, Stolte M, Meier PN, Wagner S, Manns MP. A rare case of bloody diarrhoea: thrombosis of the V. mesenterica inferior following laparoscopic cholecystectomy. Z Gastroenterol 1998; 36:35–39. 140. Lopez Cubillana P, Asensio Egea LJ, Rigabert Montiel M, Prieto Gonzalez A, Server Pastor G, Robles Campos R, Perez Albacete M, Parrilla Paricio D. Renal vein thrombosis caused by laparoscopic cholecystectomy. Scand J Urol Nephrol 1998; 32:56–57. 141. Balsara KP, Dubash C, Shah CR. Pseudoaneurysm of the hepatic artery along with common bile duct injury following laparoscopic cholecystectomy: a report of two cases. Surg Endosc 1998; 12:276–277. 142. Ribeiro A, Williams H, May G, Fulmer JT, Spivey JR. Hemobilia due to hepatic artery pseudoaneurysm thirteen months after laparoscopic cholecystectomy. J Clin Gastroenterol 1998; 26:50–53. 143. den Hoed PT, Boelhouwer RU, Veen HF, Hop WC, Bruining HA. Infections and bacteriological data after laparoscopic and open gallbladder surgery. J Hosp Infect 1998; 39:27–37. 144. Watkin DS, Wainwright AM, Thompson MH, Leaper DJ. Infection after laparoscopic cholecystectomy: are antibiotics really necessary? Eur J Surg 1995; 161:509–511. 145. Jones DB, Dunnegan DL, Soper NJ. The influence of intraoperative gallbladder perforation on longterm outcome after laparoscopic cholecystectomy. Surg Endosc 1995; 9:977–980.
Page 514
146. Diez J, Arozamena CJ, Ferraina P, Franci JM, Ferreres A, Lardies JM, Gutierrez VP. Relation between postoperative infections and gallbladder bile leakage during laparoscopic cholecystectomies. Surg Endosc 1996; 10:529–532. 147. Contini S, Dalla Valle R, Zinicola R. Unexpected gallbladder cancer after laparoscopic cholecystectomy: an emerging problem? Reflections on four cases. Surg Endosc 1999; 13:264–267. 148. Ohmura Y, Yokoyama N, Tanada M, Takiyama W, Takashima S. Port site recurrence of unexpected gallbladder carcinoma after a laparoscopic cholecystectomy: report of a case. Surg Today 1999; 29:71–75. 149. Reber PU, Baer HU, Patel AG, Schmied B, Buchler MW. Port site metastases following laparoscopic cholecystectomy for unsuspected carcinoma of the gallbladder. Z Gastroenterol 1998; 36:901–907. 150. Ohtani T, Takano Y, Shirai Y, Hatakeyama K. Early intrperitoneal dissemination after radical resection of unsuspected gallbladder carcinoma following laparoscopic cholecystectomy. Surg Laparosc Endosc 1998; 8:58–62. 151. Mintz Y, Lotan C, Goitein D, MuggiaSullam M. Laparoscopic port site metastasis of an undetected primary tumor. Surg Laparosc Endosc 1999; 9:68–69. 152. Rieger N, Mclntosh N. Port site metastasis from synchronous primaries of the colon and ovary following laparoscopic cholecystectomy. Eur J Surg Oncol 1998; 24:144–145. 153. Z'graggen K, Birrer S, Maurer CA, Wehrli H, Klaiber C, Baer HU. Incidence of port site recurrence after laparoscopic cholecystectomy for preoperatively unsuspected gallbladder carcinoma. Surgery 1998; 124:831–838. 154. Jeon HM, Kim JS, Lee CD, Kim EK, Kim SN. Late development of umbilical metastasis after laparoscopic cholecystectomy for a gallbladder carcinoma. Oncol Rep 1999; 6:283–287. 155. Schaeff B, Paolucci V, Thomopoulos J. Port site recurrences after laparoscopic surgery: a review. Dig Surg 1998; 15:124–134. 156. Plaus WJ. Laparoscopic trocar site hernias. J Laparoendosc Surg 1993; 3:567–570. 157. De Giuli M, Festa V, Denoye GC, Morino M. Large postoperative umbilical hernia following laparoscopic cholecystectomy: a case report. Surg Endosc 1994; 8:904–905. 158. Azurin DJ, Go LS, Arroyo LR, Kirkland ML. Trocar site herniation following laparoscopic cholecystectomy and the significance of an incidental preexisting umbilical hernia. Am Surg 1995; 61:718–720. 159. Korenkov M, Rixen D, Paul A, Kohler L, Eypasch E, Troidl H. Combined abdominal wall paresis and incisional hernia after laparoscopic cholecystectomy. Surg Endosc 1999; 13:268–269. 160. Gamal EM, Asztalos I, Sipos P, Miko I, Furka I, Metzger P. Late epigastric incisional hernias following laparoscopic cholecystectomy. Acta Chir Hung 1997; 36:95–6. 161. Schwartzman A, Cirocco WC, Alfonso AE. Minimizing trochar site herniation in laparoscopic cholecystectomy. J Laparoendosc Surg 1995; 5:157–161. 162. Nassar AH, Ashkar KA, Rashed AA, Abdulmoneum MG. Laparoscopic cholecystectomy and the umbilicus. Br J Surg 1997; 84:630–633. 163. Vecchio R, MacFadyen BV, Latteri S. Laparoscopic cholecystectomy: an analysis on 114,005 cases of United States series. Int Surg 1998; 83:215–219. 164. Singh V, Kacker LK, Sikora SS, Saxena R, Kapoor VK, Kaushik SP. Postcholecystectomy external biliary fistula. Aust N Z J Surg 1997; 67:168–172. 165. Robertson AJ, Rela M, Karani J, Steger AC, Benjamin IS, Heaton ND. Laparoscopic cholecystectomy injury: an unusual indication for liver transplantation. Transplant Int 1998; 11:449–451. 166. Adamsen S, Hansen OH, FunchJensen P, Schulze S, Stage JG, Wara P. Bile duct injury during laparoscopic cholecystectomy: a prospective nationwide series. J Am Coll Surg 1997; 184:571–578.
Page 515
167. Topal B, Aerts R, Penninckx F. The outcome of major biliary tract injury with leakage in laparoscopic cholecystectomy. Surg Endosc 1999; 13:53–56. 168. Bauer TW, Morris JB, Lowenstein A, Wolferth C, Rosato FE, Rosato EF. The consequences of a major bile duct injury during laparoscopic cholecystectomy. J Gastrointest Surg 1998; 2:61–66. 169. Walsh RM, Henderson JM, Vogt DP, Mayes JT, GrundfestBroniatowski S, Gagner M, Ponsky JL, Hermann RE. Trends in bile duct injuries from laparoscopic cholecystectomy. J Gastrointest Surg 1998; 2:458–462. 170. Torkington J, Pereira J, Chalmers RT, Horner J. Laparoscopic cholecystectomy, bile duct injury and the British and Irish surgeon. Ann R Coll Surg Engl 1998; 80:119–121. 171. Windsor JA, Pong J. Laparoscopic biliary injury: more than a learning curve problem. Aust N Z J Surg 1998; 68:186–89. 172. Wright KD, Wellwood JM. Bile duct injury during laparoscopic cholecystectomy without operative cholangiography. Br J Surg 1998; 85:191–194. 173. JonesMonahan K, Gruenberg JC. Bile duct injuries during laparoscopic cholecystectomy: a community's experience. Am Surg 1998; 64:638–642. 174. Soper NJ, Dunnegan DL. Does intraoperative gallbladder perforation influence the early outcome of laparoscopic cholecystectomy? Surg Laparosc Endosc 1991; 1:156–161. 175. Kimura T, Goto H, Takeuchi Y, Yoshida M, Kobayashi T, Sakuramachi S, Harada Y. Intraabdominal contamination after gallbladder perforation during laparoscopic cholecystectomy and its complications. Surg Endosc 1996; 10:888–891. 176. Assaff Y, Matter I, Sabo E, Mogilner JG, Nash E, Abrahamson J, Eldar S. Laparoscopic cholecystectomy for acute cholecystitis and the consequences of gallbladder perforation, bile spillage, and ''loss" of stones. Eur J Surg 1998; 164:425–431. 177. Hanazaki K, Igarashi J, Sodeyama H, Matsuda Y. Bile leakage resulting from clip displacement of the cystic duct stump: a potential pitfall of laparoscopic cholecystectomy. Surg Endosc 1999; 13:168–171. 178. Pietra N, Sarli L, Costi R, Violi V. Intrahepatic subcapsular hematoma: a rare postoperative complication of laparoscopic cholecystectomy. Surg Laparosc Endosc 1998; 8:304–307. 179. Freedman AN, Sigman HH. Incarcerated paraumbilical incisional hernia and abscess—complications of a spilled gallstone. J Laparoendosc Surg 1995; 5:189– 191. 180. Nishiyama T, Terunuma M, Sasaki K, Hanyu S. Right paranephric abscess as a rare late complication of laparoscopic cholecystectomy. Int J Urol 1998; 5:174– 175. 181. Labuski MR, Wise SW. Recurrent abdominal abscess secondary to a dropped laparoscopic clip: CT imaging. Abdom Imaging 1999; 24:191–192. 182. Turley BR, Taupmann RE, Johnson PL. Postoperative abscess mimicked by Surgicel. Abdom Imaging 1994; 19:345–346. 183. Festekjian JH, Hassantash SA, Taylor EW. Abdominal wall biloma: an unusual complication of laparoscopic cholecystectomy. J Soc Laparoendosc Surg 1997; 1:353–355. 184. Memon MA, Jenkins HJ Jr. Fitzgibbons RJ Jr. Spontaneous erosion of a lost intraabdominal gallstone through the back eight months following laparoscopic cholecystectomy. J Soc Laparoendosc Surg 1997; 1:153–157. 185. Zamir G, Lyass S, Pertsemlidis D, Katz B. The fate of the dropped gallstones during laparoscopic cholecystectomy. Surg Endosc 1999; 13:68–70. 186. Schafer M, Suter C, Klaiber C, Wehrli H, Frei E, Krahenbuhl L. Spilled gallstones after laparoscopic cholecystectomy: a relevant problem? A retrospective analysis of 10,174 laparoscopic cholecystectomies. Surg Endosc 1998; 12:305–309. 187. Sinha AN, Shivaprasad G, Rao AS, Sinha A. Subphrenic abscess following laparoscopic cholecystectomy and spilled gallstones. Indian J Gastroenterol 1998; 17:108–109. 188. Groebli Y, Hebert D, Tschantz P. The migrating spilled stone. Int Surg 1998; 83:31–32. 189. Lauffer JM, Krahenbuhl L, Baer HU, Mettler M, Buchler MW. Clinical manifestations of lost gallstones after laparoscopic cholecystectomy: a case report with review of the literature. Surg Laparosc Endosc 1997; 7:103–112.
Page 516
190. ParraDavila E, Munshi IA, Armstrong JH, Sleeman D, Levi JU. Retroperitoneal abscess as a complication of retained gallstones following laparoscopic cholecystectomy. J Laparoendosc Adv Surg Tech A 1998; 8:89–93. 191. Horton M, Florence MG. Unusual abscess patterns following dropped gallstones during laparoscopic cholecystectomy. Am J Surg 1998; 175:375–379. 192. Steerman PH, Sklar E, Troutman M. Unretrieved gallstone presenting as an inguinal mass. Surg Laparosc Endosc 1998; 8:239–240. 193. Petit F, Vons C, Tahrat M, CoulombL'Hermine A, Capron F, Franco D. Jaundice following laparoscopic cholecystectomy: an unusual complication of spilled stones. Surg Endosc 1998; 12:450–451. 194. Noda S, Soybel DI, Sampson BA, DeCamp MM Jr. Broncholithiasis and thoracoabdominal actinomycosis from dropped gallstones. Ann Thorac Surg 1998; 65:1465–1467. 195. Chan SY, Osborne AW, Purkiss SF. Cholelithoptysis: an unusual complication following laparoscopic cholecystectomy. Dig Surg 1998; 15:707–708. 196. Kelty CJ, Thorpe JA. Empyema due to spilled stones during laparoscopic cholecystectomy. Eur J Cardiothorac Surg 1998; 13:107–108. 197. Graham MD, Anderson PG, Toouli J. Abdominal wall sinus: a late complication of gallstone spillage during laparoscopic cholecystectomy. HPB Surg 1997; 10:163–164. 198. Pohl D, Milman PJ, Rothenberg H. Umbilical sinus tract with gallstone extrusion after laparoscopic cholecystectomy. Surg Endosc 1997; 11:846–847. 199. Lutken W, Berggren P, Maltbaek J. Passing of gallstones via the urethra: a complication of laparoscopic cholecystectomy. Surg Laparosc Endosc 1997; 7:495– 497. 200. Vadlamudi G, Graebe R, Khoo M, Schinella R, Gallstones implanting in the ovary: a complication of laparoscopic cholecystectomy. Arch Pathol Lab Med 1997; 121:155–158. 201. Z'graggen K, Aronsky D, Maurer CA, Klaiber C, Baer HU. Acute postoperative pancreatitis after laparoscopic cholecystectomy: results of the Prospective Swiss Association of Laparoscopic and Thoracoscopic Surgery Study. Arch Surg 1997:132:1026–1030. 202. Ho HS, Mathiesen KA, Wolfe BM. The impact of laparoscopic cholecystectomy on the treatment of symptomatic cholelithiasis. Surg Endosc 1996; 10:746– 750. 203. Neugebauer E, Troidl H, Kum CK, Eypasch E, Miserez M, Paul A. The EAES Consensus Development Conferences on laparoscopic cholecystectomy, appendectomy, and hernia repair. Consensus statements—September 1994. The Education Committee of the European Association for Endoscopic Surgery. Surg Endosc 1995; 9:550–563. 204. Escarce JJ, Chen W, Schwartz JS. Falling cholecystectomy thresholds since the introduction of laparoscopic cholecystectomy. JAMA 1995; 273:1581–1585. 205. Attwood SEA, Hill ADK, Mealy K, Stephens RB. A prospective comparison of laparoscopic versus open cholecystectomy. Ann R Coll Surg Engl 1992; 74:397–400. 206. Rees BI, Williams HR. Laparoscopic cholecystectomy: the first 155 patients. Ann R Coll Surg Engl 1992; 74:233–236. 207. Wilson RG, Macintyre IMC, Nison SJ, et al. Laparoscopic cholecystectomy as a safe and effective treatment for severe acute cholecystitis. BMJ 1992; 305:394–396. 208. Dunn D, Nair R, Fowler S, McCloy R. Laparoscopic cholecystectomy in England and Wales: results of an audit by the Royal College of Surgeons of England. Ann R Coll Surg Engl 1994; 76:269–275. 209. Jakimowicz JJ. Current state and trends in minimal access surgery in Europe. J R Coll Surg Edinb 1995; 40:397–406. 210. Z'graggen K, Wehrli H, Metzger A, Buehler M, Frei E, Klaiber C. Complications of laparoscopic cholecystectomy in Switzerland: a prospective 3year study of 10,174 patients. Swiss Association of Laparoscopic and Thoracoscopic Surgery. Surg Endosc 1998; 12:1303–1310. 211. MacFadyen BV Jr, Vecchio R, Ricardo AE, Mathis CR. Bile duct injury after laparoscopic cholecystectomy: the United States experience. Surg Endosc 1998; 12:315–321.
Page 517
212. Hannan EL, Imperato PJ, Nenner RP, Starr H. Laparoscopic and open cholecystectomy in New York State: mortality, complications, and choice of procedure. Surgery 1999; 125:223–231. 213. Rakos G, Markus B, Szell K. Cholecystectomy through a mini laparotomy alongside laparoscopic technique. Acta Chir Hung 1997; 36:286–288. 214. Barkun JS, Barkun AN, Sampalis JS, Fried G, Taylor B, Wexler MJ, Goresky CA, Meakins JL. Randomised controlled trial of laparoscopic versus mini cholecystectomy. Lancet 1992; 340:1116–1119. 215. Squirrell DM, Majeed AW, Troy G, Peacock JE, Nicholl JP, Johnson AG. A randomized, prospective, blinded comparison of postoperative pain, metabolic response, and perceived health after laparoscopic and small incision cholecystectomy. Surgery 1998; 123:485–495. 216. Kiviluoto T, Siren J, Luukkonen P, Kivilaakso E. Randomised trial of laparoscopic versus open cholecystectomy for acute and gangrenous cholecystitis. Lancet 1998; 351:321–325. 217. Sharma AK, Rangan HK, Choubey RP. Minilap cholecystectomy: a viable alternative to laparoscopic cholecystectomy for the Third World? Aust N Z J Surg 1998; 68:774–777. 218. Stephenson BM, Callander C, Sage M, Vellacott KD. Feasibility of "day case" laparoscopic cholecystectomy. Ann R Coll Surg Engl 1993; 75:249–251. 219. Cuschieri A. Daycase (ambulatory) laparoscopic surgery: let us sing from the same hymn sheet. Surg Endosc 1997; 11:1143–1144. 220. Voyles CR, Berch RB. Selection criteria for laparoscopic cholecystectomy in an ambulatory care setting. Surg Endosc 1997; 11:1145–1146. 221. Ali J, Gana TJ. Lung volumes 24 h after laparoscopic cholecystectomy—justification for early discharge. Can Respir J 1998; 5:109–113. 222. Prasad A, Foley RJ. Day case laparoscopic cholecystectomy: a safe and cost effective procedure. Eur J Surg 1996; 162:43–46. 223. Caimpanelli G, Cavagnoli R, Cioffi U, De Simone M, Fabbiani M, Pietri P. Can laparoscopic cholecystectomy be a day surgery procedure? Hepatogastroenterology 1998; 45:1422–1429. 224. Voitk AJ. Is outpatient cholecystectomy safe for the higherrisk elective patient? Surg Endosc 1997; 11:1147–1149. 225. Keulemans Y, Eshuis J, de Haes H, de Wit LT, Gouma DJ. Laparoscopic cholecystectomy: daycare versus clinical observation. Ann Surg 1998; 228:734– 740. 226. Fullerton GM, Darling K, Williams J. Evaluation of the cost of laparoscopic and open cholecystectomy. Br J Surg 1994; 81:124–126. 227. Narration PK, Demurs EJ. Initial results of a prospective trial of outpatient laparoscopic cholecystectomy. Surg Endosc 1997; 11:1091–1094. 228. Zegarra RF II, Saba AK, Peschiera JL. Outpatient laparoscopic cholecystectomy: safe and cost effective? Surg Laparosc Endosc 1997; 7:487–490. 229. Mushinski M. Average charges for cholecystectomies in the United States, 1996. Stat Bull Metrop Ins Co 1998; 79:16–25. 230. Guillou PJ, Windsor AJ, Nejim A. Clinical economics review: the healthcare economic implications of minimal access gastrointestinal surgery. Aliment Pharmacol Ther 1996; 10:707–713. 231. Orlando R 3rd, Russell JC. Managing gallbladder disease in a costeffective manner. Surg Clin North Am 1996; 76:117–128. 232. Ure BVM, Lefering R, Troidl H. Costs of laparoscopic cholecystectomy: analysis of potential savings. Surg Endosc 1995; 9:401–406. 233. Nicholl JP, Brazier JE, Milner PC, Westlake L, Kohler B, Williams BT, Ross B, Frost E, Johnson AG. Randomised controlled trial of costeffectiveness of lithotripsy and open cholecystectomy as treatments for gallbladder stones. Lancet 1992; 340:801–807.
Page 518
234. Darzi A, Geraghty JG, Williams NN. The pros and cons of laparoscopic cholecystectomy and extracorporeal shock wave lithotripsy in the management of gallstone disease. Ann R Coll Surg Engl 1994; 76:42–46. 235. Barkun AN, Barkun JS, Sampalis JS, Caro J, Fried GM, Meakins JL, Joseph L, Goresky CA. Costs and effectiveness of extracorporeal gallbladder stone shock wave lithotripsy versus laparoscopic cholecystectomy: a randomized clinical trial. McGill Gallstone Treatment Group. Int J Technol Assess Health Care 1997; 13:589–601. 236. Go PM, Stolk MF, Obertop H, Dirksen C, van der Elst DH, Ament A, van Erpecum KJ, van Berge Henegouwen GP, Gouma DJ. Symptomatic gallbladder stones: costeffectiveness of treatment with extracorporeal shockwave lithotripsy, conventional and laparoscopic cholecystectomy. Surg Endosc 1995; 9:37–41. 237. Mowschenson PM. Improving the costeffectiveness of laparoscopic cholecystectomy. J Laparoendosc Surg 1993; 3:113–19. 238. Bass EB, Pitt HA, Lillemoe KD. Costeffectiveness of laparoscopic cholecystectomy versus open cholecystectomy. Am J Surg 1993; 165:466–471. 239. McKellar DP, Johnson RM, Dutro JA, Mellinger J, Bernie WA, Peoples JB. Costeffectiveness of laparoscopic cholecystectomy. Surg Endosc 1995; 9:158– 162. 240. Wenner J, Graffner H, Lindell G. A financial analysis of laparoscopic and open cholecystectomy. Surg Endosc 1995; 9:702–705. 241. Kesteloot K, Penninckx F. The costs and effects of open versus laparoscopic cholecystectomies. Health Econ 1993; 2(4):303–312. 242. Usal H, Nabagiez; J, Sayad P, Ferzli GS. Cost effectiveness of routine type and screen testing before laparoscopic cholecystectomy. Surg Endosc 1999; 13:146–147. 243. Lekawa M, Shapiro SJ, Gordon LA, Rothbart J, Hiatt JR. The laparoscopic learning curve. Surg Laparosc Endosc 1995; 5:455–58. 244. Bickel A, Rappaport A, Hazani E, Eitan A. Laparoscopic cholecystectomy for acute cholecystitis performed by residents in surgery: a risk factor for conversion to open laparotomy? J Laparoendosc Adv Surg Tech A 1998; 8:137–141. 245. Schol FP, GO PM, Gouma DJ, Kootstra G. Laparoscopic cholecystectomy in a surgical training programme. Eur J Surg 1996; 162:193–197. 246. Zucker KA, Bailey RW, Graham SM, Scovil W, Imbembo AL. Training for laparoscopic surgery. World J Surg 1993; 17:3–7. 247. Hodgson WJ, Byrne DW, Savino JA, Liberis G. Laparoscopic cholecystectomy: the early experience of surgical attendings compared with that of residents trained by apprenticeship. Surg Endosc 1994; 8:1058–1062. 248. Namias N, McKenney MG, Sleeman D, Hutson DG. Trends in resident experience in open and laparoscopic cholecystectomy. Surg Laparosc Endosc 1997; 7:245–247. 249. Ferzli GS, Fiorillo MA, Hayek NE, Sabido F. Chief resident experience with laparoscopic cholecystectomy. J Laparoendosc Adv Surg Tech A 1997; 7:147– 150. 250. ScottConner CE, Hall TJ, Anglin BL, Muakkassa FF, Poole GV, Thompson AR, Wilton PB. The integration of laparoscopy into a surgical residency and implications for the training environment. Surg Endosc 1994; 8:1054–1057. 251. Zaraca F, Catarci M, Gossetti F, Carboni M. Senior versus proctored young and resident surgeons' experience in laparoscopic cholecystectomy: is there any need of previous exposure to open biliary surgery? J Laparoendosc Surg 1995; 5:303–307. 252. Friedman RL, Pace BW. Resident education in laparoscopic cholecystectomy. Surg Endosc 1996; 10:26–28. 253. Ooi LL. Training in laparoscopic surgery—have we got it right yet? Ann Acad Med Singapore 1996; 25:732–736. 254. Asbun HJ, Berguer R, Altamirano R, Castellanos H. Successfully establishing laparoscopic surgery programs in developing countries: clinical results and lessons learned. Surg Endosc 1996; 10:1000–1003. 255. Majeed AW, Reed MW, Johnson AG. Simulated laparoscopic cholecystectomy. Ann R Coll Surg Engl 1992; 74:70–71.
Page 519
256. Fasel JH, Gingins P, Kalra P, MagnenatThalmann N, Baur C, Cuttat JF, Muster M, Gailloud P. Liver of the "visible man." Clin Anat 1997; 10:389–393. 257. Darzi A, Smith S, Taffinder N. Assessing operative skills needs to become more objective. BMJ 1999; 318:887–888. 258. Go PM, Payne JH Jr. Endoscopic surgery teleconferencing. Int Surg 1996; 81:18–20. 259. Satava RM. Virtual reality, telesurgery, and the new world order of medicine. J Image Guid Surg 1995; 1:12–16. 260. Kavoussi LR, Moore RG, Partin AW, Bender JS, Zenilman ME, Satava RM. Telerobotic assisted laparoscopic surgery: initial laboratory and clinical experience. Urology 1994; 44:15–9. 261. Schurr MO, Breitwieser H, Melzer A, Kunert W, Schmitt M, Voges U, Buess G. Experimental telemanipulation in endoscopic surgery. Surg Laparosc Endosc 1996; 6:167–175. 262. Allaf ME, Jackman SV, Schulam PG, Cadeddu JA, Lee BR, Moore RG, Kavoussi LR. Laparoscopic visual field: voice vs foot pedal interfaces for control of the AESOP robot. Surg Endosc 1998; 12:1415–1418. 263. GarciaRuiz A, Gagner M, Miller JH, Steiner CP, Hahn JF. Manual vs robotically assisted laparoscopic surgery in the performance of basic manipulation and suturing tasks. Arch Surg 1998; 133:957–961. 264. Bowersox JC, Cordts PR, LaPorta AJ. Use of an intuitive telemanipulator system for remote trauma surgery: an experimental study. J Am Coll Surg 1998; 186:615–621. 265. Hiatt JR, Shabot MM, Phillips EH, Haines RF, Grant TL. Telesurgery: acceptability of compressed video for remote surgical proctoring. Arch Surg 1996; 131:396–401. 266. Smithwick M. Network options for widearea telesurgery. J Telemed Telecare 1995; 1:131–138. 267. Schulam PG, Docimo SG, Saleh W, Breitenbach C, Moore RG, Kavoussi L. Telesurgical mentoring: initial clinical experience. Surg Endosc 1997; 11:1001– 1005. 268. Lee BR, Bishoff JT, Janetschek G, Bunyaratevej P, Kamolpronwijit W, Cadeddu JA, Ratchanon S, O'Kelley S, Kavoussi LR. A novel method of surgical instruction: international telementoring. World J Urol 1998; 16:367–370. 269. Moore RG, Adams JB, Partin AW, Docimo SG, Kavoussi LR. Telementoring of laparoscopic procedures: initial clinical experience. Surg Endosc 1996; 10:107–110. 270. Rosser JC, Wood M, Payne JH, Fullum TM, Lisehora GB, Rosser LE, Barcia PJ, Savalgi RS. Telementoring: a practical option in surgical training. Surg Endosc 1997; 11:852–855. 271. Cheriff AD, Schulam PG, Docimo SG, Moore RG, Kavoussi LR. Telesurgical consultation. J Urol 1996; 156:1391–1393. 272. Johanet H. Voicecontrolled robot: a new surgical aide? Thoughts of a user. Ann Chir 1998; 52:918–921. 273. Baca I. Robot arm in laparoscopic surgery. Chirurgie 1997; 68:837–939.
Page 521
24— Overview of Nonsurgical Therapy of Gallstones Dominique E. Howard and Hans Fromm The George Washington University Medical Center, Washington, D.C. I— Introduction Since gallstones are very common and often treated surgically both at a considerable cost and with a risk of complications, the role of medical management requires careful scrutiny. Surgical intervention (in most cases, laparoscopic cholecystectomy) should be reserved for symptomatic gallstone patients. The term symptomatic refers to patients with biliary pain ("biliary colic"). If a careful history is obtained from the patient, abdominal symptoms related to irritable bowel syndrome, peptic ulcer disease, and other conditions can be distinguished from biliary pain and unnecessary surgery avoided. Unfortunately, cholecystectomy is often performed for symptoms that are either nonspecific or caused by conditions other than gallstones. Therefore serious attention to a thorough clinical evaluation of the patient must be the basis for a choice between surgery and medical management. The fact that the majority of gallstones are either silent or associated with symptoms that are not of biliary origin underscores the importance of a careful evaluation of the patient's medical history. The patient's choice between medical and operative treatment is another major factor in the the decision making. If patients choose conservative management over surgery in spite of the presence of biliary pain or complications, one must consider steps that could reduce the recurrence of symptoms or complications. Recent studies by Tomida et al. indicate that ursodeoxycholic acid (ursodiol, UDCA) significantly reduces both the incidence and recurrence of biliary pain in gallstone patients (1). In addition to the patient's choice and medical history, the selection of medical treatment is based on both gallstone and gallbladder criteria, which are discussed briefly below, in relation to the different therapeutic modalities. Details concerning patient selection and treatment results are the subject of the respective chapters by other authors. II— Management of Asymptomatic Stones Silent or asymptomatic stones account for twothirds of gallstones (2). Since asymptomatic stones rarely lead to serious complications when they become symptomatic, they are usually managed conservatively (3). The risk of asymptomatic stones becoming symptomatic is approximately 1 to 2% per year (4). In elderly patients, who are at a greater surgical risk, watchful waiting is the rule (5).
Page 522
III— Management of Symptomatic Stones Symptomatic stones produce a rightupperquadrant or epigastric pain that is often nocturnal at onset (6–8). The pain can last anywhere from 30 min to several hours. Food ingestion usually does not precipitate the pain (8). Patients with gallstones can also present with atypical or nonspecific symptoms, such as dyspepsia, bloating, nausea, indigestion, and intolerance of fatty foods (6,7). Another factor thought to influence pain and its recurrence is the patient's persona. Patients who tend to favor surgical intervention may actually exaggerate their symptoms, whereas those who fear surgery will minimize them (4). The management of symptomatic stones has traditionally been surgical. More recently, studies suggest that perhaps surgical intervention is not warranted and may actually pose a greater risk for a subset of patients. The rate of recurrence of biliary pain has been shown to be 41% per year throughout a patient's life. Symptomatic gallstone patients have been shown to develop complications at a fairly constant rate of 3% per year (9,10). One study revealed that 29% of patients with symptomatic stones will remain asymptomatic for up to 10 years following the initial attack (11). As far as the timing of surgical intervention is concerned, a patient who has had his or her firsttime attack of biliary pain can be managed expectantly (9–12). The cumulative death rate for 30yearold patients undergoing immediate cholecystectomy has been estimated to be 0.11%. The respective death rate for expectant management until the biliary pain recurs or a complication develops was estimated to be very similar: 0.14% (9). Elderly patients seem to be at increased risk from surgical management. The cumulative lifetime risk of mortality from gallstones is 2.3% and applies mainly to patients 65 years of age or older (9). Since the risk for expectant management is low, decisions regarding management need not be hasty (9). Surgical management has become the standard therapy for patients with persistent recurrent biliary pain. With the advent of laparoscopic cholecystectomy in 1987, the number of cholecystectomies performed has risen remarkably (13,14). Cholecystectomy is currently the most common abdominal procedure, performed at considerable cost (15,16). A laparoscopic cholecystectomy currently costs an average of $9792 (17). The significant increase in this operation is in part due to improper indications. Gallstone patients are frequently taken to the operating room for symptoms unrelated to gallstones, such as dyspepsia and indigestion. Surgical intervention is not without risk or expenses. As stated earlier, the mortality risk of surgery increases in patients who are 65 years of age or older as well as in those with complications or comorbidities (18,19). The experience of the surgeon, or lack thereof, also influences the risk. The incidence of bile duct injury, which is more common in laparoscopic than in open cholecystectomy, has been reported to be 0.95% (20). For those patients who have a firsttime episode of pain, those who are not surgical candidates, or those who refuse surgery, several nonsurgical options can be considered. IV— Nonsurgical Management A— Oral Dissolution Therapy with Bile Acids Ursodeoxycholic acid (UDCA), which can be given in combination with chenodeoxycholic acid (CDCA), constitutes the main method of nonsurgical treatment in selected patients with small and noncalcified gallstones. UDCA is usually given alone because it is very safe, in contrast to CDCD, although UDCA lessens the adverse effects of CDCA if given concomitantly (21–25). Bile acid dissolution therapy is most effective when used for noncalcified stones less than 5 mm. in diameter. Computed tomography is most sensitive in detecting calcifications of gallstones but may not be costeffective for the selection of patients for bile acid dissolution therapy (26). The average rate of stone dissolution in patients who respond to the treatment is 1 mm per month (27). The success of dissolution therapy varies with stone size. Very small stones may dissolve in up to 80 to 90% of cases (21–24). With larger stones, the success rate
Page 523
is much lower. Gallstones that develop after rapid weight loss can be prevented by UDCA therapy (28). After dissolution, gallstones may recur in 30 to 50% of patients at a yearly rate of 10% during the first 3 to 5 years (29). B— Extracorporeal ShockWave Lithotripsy Although used successfully in Europe and Asia, extracorporeal shockwave lithotripsy (ESWL) of gallstones is currently not FDAapproved in the United States. It offers an attractive and safe treatment option for selected patients with symptomatic gallstones, in particular for those with noncalcified single stones that are below 2 cm in diameter (30–36). ESWL is much more effective than oral dissolution therapy with bile acids for gallstones larger than 5 mm in diameter, and it can be performed on an outpatient basis (30,34). Gallstones can recur after successful treatment—that is, after disappearance of the gallstone fragments. Recurrence is more likely in patients with multiple stones (35,36). ESWL is usually used in conjunction with UDCA (30–37). C— Topical Dissolution Therapy Topical treatment with methyl tertbutyl ether (MTBE), a powerful, volatile C5 ether and an organic solvent of cholesterol, is experimental (38,39). MTBE is most successful in patients with cholesterol stones that are not calcified (40,41). This solvent is effective in dissolving both small and large stones and is infused directly into the gallbladder through a pigtail catheter placed endoscopically or transhepatically (38). Stones can dissolve within several hours of infusion. Given the nature of this solvent, it should be reserved for patients who are not eligible for the other nonsurgical treatment methods, who are surgical risks, and who are appropriate candidates. Ethyl propionate (EP), another topical solvent, is also a C5 ethyl ether organic solvent that is successful in dissolving gallstones and currently under investigation. This solvent appears to be safer then MTBE and is less volatile (42,43). EP has been shown to have fewer side effects, with less gastrointestinal irritation than MTBE; it is promising in its use for gallstone patients who are at increased surgical risk (42,44). V— Summary Gallstones can be either asymptomatic or symptomatic. Asymptomatic stones are managed conservatively. Symptomatic stones are usually treated surgically. With the advent of laparoscopic surgery, the rate of cholecystectomies has increased markedly. Often, patients are operated on unnecessarily for nonspecific symptoms. Patients' personalities may also influence their symptoms. Surgical intervention is not without risks, complications, or costs. Elderly patients and patients with comorbidities are at increased risk from surgery. In these patients, bile acid therapy and potentially ESWL and topical dissolution may provide the safest modes of treatment. References 1. Tomida S, Abei M, Yamaguchi T, Matsuzaki Y, Shoda J, Tanaka N, Osuga T. Longterm ursodeoxycholic acid therapy is associated with reduced risk of biliary pain and acute cholecystitis in patients with gallbladder stones: a cohort analysis. Hepatology 1999; 30: 6–13.
Page 524
2. Ricci G, the GREPCO Group. The GREPCO research programmes: aims and prevalence data. In: Cappocaccia L, Ricci G, Angelico F, Angelico M, Attili AF, eds. Epidemiology and Prevention of Gallstone Disease. Lancaster, England: MTP Press, 1984, p 9. 3. Yoon P, Fromm H. Medical dissolution therapy of gallstones. In: Braasch J, Tompkins R, eds. Surgical Disease of the Biliary Tract and Pancreas— Multidisciplinary Management. St Louis: MosbyYear Book, 1994, pp 207–219. 4. Friedman G. Natural history of asymptomatic and symptomatic gallstones. Am J Surg 1993; 165:399–404. 5. Watkins J, Blatt C, Layden T. Gallstones: choosing the right therapy despite vague clinical clues. Geriatrics 1993; 48(8):48–54. 6. Lund J. Surgical indications in cholelithiasis: prophylactic cholecystectomy elucidated on the basis of longterm follow up on 526 nonoperated cases. Ann Surg 1960; 151:153–161. 7. Price WH. Gallbladder dyspepsia. BMJ 1963; 2:138–141. 8. Riges B, Torosis J, McDongall CJ, et al. The circadian rhythm of biliary colic. J Clin Gastroenterol 1990; 12:409–414. 9. Ransohoff D. Management of patients with symptomatic gallstones: a quantitative analysis. Am J Med 1990; 88:154–160. 10. Ransohoff DF, Gracie WA. Treatment of gallstones. Ann Intern Med 1993; 119:606–619. 11. Newman HF, Northup JD, Rosenblum M, Abrams H. Complications of cholelithiasis. Am J Gastroenterol 1968; 50(6):476–496. 12. American College of Physicians. Guidelines for the treatment of gallstones. Ann Intern Med 1993; 119:620–622. 13. National Center for Health Statistics. National Hospital Discharge Survey. Advance Data from the Vital and Health Statistics of the National Center for Health Statistics. Rockville MD: US Department of Health, Education, and Welfare, Public Health Service and Health Resources Administration, 1998. 14. Reddick EJ, Olsen DO. Laparoscopic laser cholecystectomy: a comparison with minilap cholecystectomy. Surg Endosc 1989; 3:131–133. 15. Bruckstein AH. Nonsurgical management of cholelithiasis. Arch Intern Med 1990; 150: 960–964. 16. Weinstein MC, Coley CM, Richter JM. Medical management of gallstones: a costeffective analysis. J Gen Intern Med 1990; 5:277–284. 17. Klar RM, Kongstevdt PR. Comment. JAMA 1994; 271(7):500–501. 18. Glenn F. Surgical management of acute cholecystitis in patients 65 years of age and older. Ann Surg 1981; 193:56–59. 19. Hidalgo LA, Capella G, PiFigueras J, et al. The influence of age on early surgical treatment of acute cholecystitis. Surg Gynecol Obstet 1989; 169:393–395. 20. Targarona EM, Marco C, Balague C, et al. How, when, and why bile duct injury occurs; a comparison between open and laparoscopic cholecystectomy. Surg Endosc 1998; 12: 322–326. 21. Schoenfield LJ, Marks J. Oral and contact dissolution of gallstones. Am J Surg 1993; 165:427–430. 22. Fromm H. Gallstone dissolution and the cholesterolbile acidlipoprotein axis: propitious effects of ursodeoxycholic acid. Gastroenterology 1984; 87:229–233. 23. Fromm H. Gallstone dissolution therapy: current status and future prospects. Gastroenterology 1986; 91:1560–1567. 24. Fromm H, Roat JW, Gonzalez V, et al. Comparative efficacy and side effects of ursodeoxycholic and chenodeoxycholic acids in dissolving gallstones: a double blind controlled study. Gastroenterology 1983; 85:1257–1264. 25. Podda M, Zuin M, Battezzati PM, et al. Efficacy and safety of a combination of chenodeoxycholic and ursodeoxycholic for gallstone dissolution: a comparison with ursodeoxycholic acid alone. Gastroenterology 1989; 96:222–229.
Page 525
26. Sarva RP, Farivar S, Fromm H, Poller W. Study of the sensitivity and specificity of computerized tomography in the detection of calcified gallstones which appear radiolucent by conventional roentgenograph. Gastrointest Radiol 1981; 6:165–167. 27. Senior JR, Johnson MF, DeTurck DM, et al. In vivo kinetics of radiolucent gallstone dissolution by oral dihydroxy bile acids. Gastroenterology 1990; 99:243– 251. 28. Sugerman HJ, Brewer WH, Shiffman ML, et al. A multicenter, placebocontrolled, randomized, doubleblind, prospective trial of prophylactic ursodiol for the prevention of gallstone formation following gastricbypassinduced rapid weight loss. Am J Surg 1995; 169:91–97. 29. Fromm H, Malavoti M. Gallstone recurrence after medical therapy. Viewpoints Dig Dis 1992; 24(1):1–7. 30. Nahrwold DL. Gallstone lithotripsy. Am J Surg 1993; 165:431–434. 31. Sauerbruch T, Delius M, Paumgartner G, et al. Fragmentation of gallstones by extracorporeal shock waves. N Engl J Med 1986; 314:818–822. 32. Buttmann A, Adamek HE, Weber J, Riemann JF. ESWL and oral dissolution therapy: what factors influence results. Dig Dis Sci 1993; 38(9): 1702–1711. 33. Brand B, Groth J, Lerch L, Stange EF. Intensified ESWL of gallstones: dissociation of pulverisation, pain relief and stoneclearance. Hepatogastroenterology 1998; 45:70–76. 34. Albert MB, Fromm H, Borstelmann R, et al. Successful outpatient treatment of gallstones with piezoelectric lithotripsy. Ann Intern Med 1990; 113:164–166. 35. Dion YM, Morin J, Fraser W. Extracorporeal shockwave lithotripsy of gallstones: clinical experience with 170 patients. Can J Surg 1992; 35(2): 145–150. 36. Wehrmann T, Marek S, Hanisch E, et al. Causes and management of recurrent biliary pain after successful nonoperative gallstone treatment. Am J Gastroenterol 1997; 92:132–138. 37. Ertan A, Hernandez RE, Campeau RJ, et al. Extracorporeal shockwave lithotripsy and ursodiol versus ursodiol alone in the treatment of gallstones. Gastroenterology 1992; 103: 311–316. 38. Allen MJ, Borody TJ, Bugliosi TF, et al. Rapid dissolution of gallstones by methyl tertbutyl ether: preliminary observation. N Engl J Med 1985; 312:217–220. 39. Leuschner U, Hellstern A, Schmidt K, et al. Gallstone dissolution with methyl tertbutyl ether in 120 patientsefficacy and safety. Dig Dis Sci 1991; 36:193–199. 40. Lu DSK, Ho CS, and Allen LC. Gallstone dissolution in methyl tertbutyl ether after mechanical fragmentation: in vitro study. AJR 1990; 155:67–72. 41. Thistle JL, May GR, Bender CE, et al. Dissolution of cholesterol gallbladder stones by methyl tertbutyl ether administered by percutaneous transhepatic catheter. N Engl J Med 1989; 320:633–639. 42. Hofmann AF, Amelsberg A, Esch O, et al. Successful topical dissolution of cholesterol gallbladder stones using ethyl propionate. Dig Dis Sci 1997; 42:1274– 1282. 43. Hofmann AF, Schteingart CD, vanSonnenberg E, et al. Contact dissolution of cholesterol gallstones with organic solvents. Gastroenterol Clin North Am 1991; 20:183–199. 44. Clerici C, Gentili G, Zakko S, et al. Local and systemic effects of introduodenal exposure to topical gallstone solvents ethyl propionate and methyl tertbutyl ether in the rabbit. Dig Dis Sci 1997; 42(3):497–502.
Page 527
25— Biliary Lithotripsy M.W. Neubrand and Tilman Sauerbruch Medizinische Klinik and Poliklinik, Rheinische FriedrichWilhelmsUniversity, Bonn, Germany I— Introduction Extracorporeal shockwave lithotripsy (ESWL) has revolutionized the therapy of urolithiasis. After the convincing success of ESWL in urology, it was tempting to extend this technology to the treatment of gallstones. The first in vivo experiments in animals proved the feasibility of fragmenting stones located in the gallbladder, with few side effects (1). Further studies in humans showed that gallbladder stones as well as bile duct stones can be disintegrated by extracorporeally induced shock waves and that fragments may either pass spontaneously into the duodenum or may be successfully dissolved by adjuvant bile salt therapy (2,3). Since these first treatments had been performed in 1985, numerous phase II studies and some phase III studies have defined the role of ESWL of gallstones within the context of different treatment options available to date (4–7). It is the purpose of the present review to summarize this knowledge. Gallstone disease represents an important health care problem in the western world. The overall prevalence of gallstones lies between 10 and 15% (8–10). It increases with age and is higher in women. Most gallstones do not cause specific symptoms and therefore need no treatment (11–14). However, due to the high prevalence of gallstones, cholecystectomy is one of the most frequent abdominal operations. During the 1980s, more than 500,000 such procedures were performed each year in the United States, and the numbers have been rising slightly beyond that since the introduction of laparoscopic cholecystectomy (15). Although cholecystectomy is the ''gold standard" in the treatment of symptomatic gallbladder stones, it does, on the one hand, carry a small risk of fatal complications (0.2% for patients younger than age 70 and up to 5% for patients above that age) and of bile duct injury; on the other hand, there is a corresponding risk of 0.1 to 0.2% for conventional cholecystectomy and a risk of up to 0.5% for laparoscopic cholecystectomy (15–17). This is the main reason why nonsurgical alternatives are still under investigation. Numerous studies have shown that the ESWL of gallbladder stones produces virtually no mortalities. This has to be balanced against the fact that its efficacy is limited to wellselected ideal candidates. This chapter discusses the technical and physical aspects of shockwave treatment as well as the in vitro experiments that have provided a better understanding of the principles of ESWL. It summarizes the in vivo findings on the efficacy and costeffectiveness ESWL. The adverse effects of ESWL are discussed, as is the subject of stone recurrence after successful treatment. Last, but not least, the role of ESWL in the treatment of bile duct stones is considered.
Page 528
II— Physical Properties of Shock Waves Shock waves are sound waves with highpressure amplitudes ranging from 500 to 1000 bar. The period of time in which this pressure rises is less than a nanosecond, followed by an exponential pressure decline with a negative tensile component (18) (Fig. 1). Shock waves propagate faster than the sound in the medium in which they are generated (19). The effects of shock waves were first noticed on supersonic aircraft, whose metal surfaces were sprinkled with tiny pits and craters after they had traveled at speeds higher than that of sound. The positive component of such pressure waves exerts tear and shear forces on the gallstone, whereas the negative component probably leads to the formation and collapse of bubbles in the fluid surrounding of the stone surface—a process known as cavitation (20,21). Both phenomena cause a gradual fragmentation. For the purpose of stone disintegration, shock waves are generated in water. Figure 1 shows the pressure wave form of an electrohydraulic device that is used to generate shock waves. Three different principles are employed to generate shock waves: electrohydraulic sources, electromagnetic sources, and piezoelectric sources (Fig. 2). Electrohydraulic sources generate spherical shock waves, which are set off by an electrohydraulic spark gap discharge via a capacitor. Their site of generation is located at one focus of a metal ellipsoid. The expanding plasma between the electrode tips generates shock waves, which are reflected onto the second focus of the semiellipsoid, where the stone must be positioned for fragmentation. In the electromagnetic system, an impulse from a strong electric current flows through a flat coil, where it induces a magnetic field that causes a very rapid movement of a metal membrane overlying the coil. This generates a plane pressure wave in a waterfilled tube attached to the device. When the wave is focused by transmission through a biconcave acoustic lens, it can be used for lithotripsy (Fig. 2). The output of the shock wave is determined by the generator power setting. Piezoceramic sources use ceramic crystals, which are arrayed in a bowlshaped dish. Discharge of a capacitor leads to the deformation of each crystal. These crystals, together, then induce a converging selffocusing spherical pressure wave. The power in this system is determined by the aperture of the spherical array and, as noted above, is characterized by peak positive and negative amplitudes, pulse rise time, and pulse width. Apart from the focal dimensions of the lithotripter, these physical characteristics, in particular peak pressure, are of clinical importance. These parameters determine the efficacy
Figure 1 Pressure profile of a shock wave of the Dornier lithotripter MPL 9000 at 22 kV measured with a needle hydrophone. The diameter of the active area is <1 mm. The pressure rises to 600 bar within 80 ns and is followed by a slower decrease (700 ns) of the pressure and a tensile phase with a maximum strength of about 50 bar. (From Ref. 35.)
Page 529
Figure 2 Three different principles of shockwave generation. In the spark gap system, the shock wave is generated under water by spark discharge. The wavefront is reflected by an ellipsoidal reflector and focused into the second focus of the ellipsoid, where the gallstone is located. In the piezoelectric system, discharge of a capacitor leads to the deformation of pie zoceramic crystals arrayed in a bowlshaped spherical disk; together, these induce a converging selffocusing spherical pressure wave. In the electromagnetic system, an electric current flows through a flat coil, where it induces a magnetic field that causes a very rapid movement of a metal membrane overlying the coil. This generates a plane pressure wave, which is focused by transmission through an acoustic lens.
of stone fragmentation as well as the biological effects on human tissues. The three systems differ in peak positive pressure, pulse rise time, and pulse width (22). This results in different energy distributions within the focal areas of the three systems. Focal area is defined as the space in which at least 50% of the maximum pressure can be measured (Fig. 3). In general, the shockwave generators that produce low peak pressures have large focal areas. Provided that the peak pressure is above the threshold energy for stone fragmentation, these generators fragment the entire stone without the need to move the focal position, because the focal area is generally larger than the crosssectional area of the stone in these systems. However, there is a higher potential for tissue damage around the stone. These types of lithotripters may also produce more pain, because the pressure that reaches the skin is greater than that caused by generators with higher peak pressure but smaller focal area (piezoceramic systems). The total energy that passes through a stone can be calculated from the timepressure curve in the focal area of a lithotripter and the size of the stone located in the focal area (23) (Fig. 4). Due to the nonlinear pressure distribution in the focal area, the energy augments in a nonlinear relation to the size of stone that is covered by the shock waves. This results from a 50% decrease in the maximum energy at 2.5 mm. lateral from the focal point for an electrohydraulic lithotripter (Fig. 3). The pressure front of the shock waves expands relatively unhindered through the medium provided that the acoustic impedance remains unchanged. The acoustic impedance is the product of the medium's sound velocity and its density.
where Z is the acoustic impedance (106 kg/m2 × s), C is the sound velocity in the medium
Page 530
Figure 3 Lateral pressure distribution of a Dornier lithotripter MPL 9000 at 22 kV. The peak pressure of 600 bar reduces at lateral distance of around 2.5 mm to 50%. (From Ref. 35.)
(m/s), and ro is the density of the medium (103 kg/m3). The acoustic impedance of normal human tissue is similar to that of water (in the range of 1.5 × 106 kg/m2 × s). If the shock wave strikes a material not similar to water, such as human gallstones, part of the wave is transmitted through the stone and part is reflected. The energy fraction that is reflected is given by Eq. (2):
where rI corresponds to the fraction of energy that is reflected, z1 to the acoustic impedance in
Figure 4 Dependence of the acoustic energy passing through a given stone of radius r0 (lateral distance) at a certain energy setting. This curve shows the total amount of energy passing through a stone with a given radius (lateral distances) at four different voltage settings of the lithotripter. For example, a stone of 20mm diameter (lateral distance 10 mm) is subjected to a total energy of approximately 50 mJ per pulse at an energy setting of 22 kV, whereas at 18 kV this energy is reduced to 30 mJ per shock wave. With a stone of 15mm radius, the total acoustic energy per shock wave increases to 65 and 40 mJ in 22 and 18 kV settings, respectively. (From Ref. 23.)
Page 531
the surrounding medium, and z2 to the acoustic impedance within the stone. The fraction of energy that is transmitted to the stone is given by Eq. (3):
where tI is the fraction of energy transmitted and rI is the fraction reflected on the stone surface. The transmitted pressure is then partially reflected at the back surface of the stone. The fraction of pressure tI may exceed the compressive strength of the stone, and the fraction of energy of the negative reflected wave may exceed the stone's tensile strength. Generally it is believed that the initial fragmentation of the stone is due mainly to the tensile forces induced by the reflected wave, since the compressive strength of gallstones greatly exceeds the tensile strength. In urinary stones, the higher acoustic impedance as compared to that of the gallstones leads to a higher proportion of reflected shock waves. This explains why kidney stones generally disintegrate more readily than gallstones. To summarize, positive pressure forces and negative tensile forces as well as the formation of cavitation bubbles are responsible for the shockwave fragmentation of gallstones, but also, however, for tissue damage. III— In Vitro Studies The aim of shockwave fragmentation is to create the smallest fragments possible while causing the least tissue damage. The parameters involved in stone fragmentation include the chemical composition, structure, microhardness, elasticity, volume, and number of the stones as well as the shockwave characteristics of the surrounding medium (see above). A— Influence of the Surrounding Medium From Eqs. (2) and (3) it is evident that changes in the surrounding medium and thereby its acoustic impedance should influence disintegration, since the effects of tensile and compressive forces on the stone surface change with the difference in acoustic impedance between the stone and the surrounding medium. Unfortunately most in vitro experiments on shockwave fragmentation are performed in water, which does not exactly reflect the situation in bile. Delius and coworkers investigated the formation of cavitation bubbles in viscous fluids. They used polyvinylalcohol (PVA) for their experiments because its acoustic impedance is nearly identical to that of water, whereas its viscosity is much higher. The number of pits on aluminum foil caused by cavitation in PVA was significantly lower than that in water. Nitsche et al. studied the degree of stone fragmentation in relation to the density and viscosity of the surrounding medium (24). They found that the density of the fluid had no monotone correlation to the number of shock waves required. But, in contrast, there was a high direct correlation between the viscosity of the fluid and the number of shock waves needed. Bile viscosity differs considerably from patient to patient (25). To what degree medical interventions that might change the viscosity of bile [e.g., ursodeoxycholic acid (UDCA) or mucous secretion (e.g. acetyl salycilatic acid)] can affect the efficacy of lithotripsy has not been studied systematically to date. B— Influence of Chemical Composition, Radiological Stone Features, and Stone Structure Efforts have been made to find out which radiological features predict stone composition and stone disintegration. Computed tomography (CT) is useful in discriminating between cholesterol and calcified pigment stones, although the cutoff points for different CT systems as well as different media cannot be compared. Most authors describe a cutoff point between 100 and
Page 532
150 HU to discriminate between cholesterol stones and pigment stones (26). The overall accuracy is estimated to be around 95%. Most in vitro studies show that the CT density of stones is of minor importance for fragmentation (27–29). However, a stone density distribution index of more than 60 HU in one stone, reflecting the heterogenicity of the stone, could be a determinant of satisfactory fragmentation. Barkun et al. established this value in a carefully performed study (30). Xray is another possible radiological method for detecting stone calcification. However, its sensitivity is not greater than that of CT. Even mammographic xray technique that has a higher sensitivity for detecting calcifications than classic xray, which could not identify characteristics predicting parameters for good stone fragmentation in vitro (23). Ultrasonographic features have been carefully investigated by a Japanese group. Tsuchiya et al. showed that the echo patterns of gallstones can distinguish between those with high versus low cholesterol content (31,32). This has been verified by other investigators (33). A smooth, homogeneous hyperecho pattern distributed over the whole cross section of the stone with a faint acoustic shadow suggests a high cholesterol content, whereas a gallstone with a crescentshaped echo and a hard hypoecho shadow suggests a low cholesterol content. Stones with a very high cholesterol content (>90%) and a very typical hyperecho homogeneous pattern show excellent fragmentation. However, such pure cholesterol stones are rare. To summarize, CT and xray are of minor importance in predicting good fragmentation. Possibly the sonographic pattern would be better able to discriminate between good and bad fragmentation. C— Influence of Stone Volume and Number of Stones In vitro studies have consistently shown that the total mass of the gallstones as determined by volume or diameter is the most important parameter for predicting the result of fragmentation under standardized conditions (23,28,34). Using artificial calculi, the amount of shockwave pulses necessary to fragment a stone was found to increase linearly with stone volume and to be inversely related to the energy per pulse (Fig. 5) (35). This is also true for human gallstones. Thus, with respect to an individual stone, a constant amount of material is disintegrated per shockwave pulse. However, the degree of fragmentation differs between stones from different patients at least by a factor of 8, despite similar chemical composition, size, and shape (23). Other factors, such as microhardness or the elasticity of the stone, may be responsible for this variance. In addition, the threshold energy—i.e., the energy per pulse below which no frag
Figure 5 Dependence of stone volume on the number of shock waves necessary to disintegrate the stone into fragments smaller than 2 mm. The increase of the curves is linear, but it differs from different gallbladder (various symbols) stone families. The linear increase indicates that a constant amount of stone material is disintegrated per pulse. (From Ref. 23.)
Page 533
mentation occurs—varies from stone to stone (23). This threshold energy, although rather low, might be important in an in vivo situation, as when a layer of fragmented stones in front blocks shockwave access to the uncrushed stones behind it and reduces the energy per pulse that reaches the stone to below threshold. Trials with artificial stones with an acoustic impedance similar to that of human gallstones have documented the fact that layers of small fragments hinder further disintegration of remaining larger fragments. The attenuation depends to a large extent on the original stone size and the acoustic energy per pulse. The critical thickness of the layer of fragmented stones that blocks intact stones or larger fragments lies between 2.5 and 5.0 mm, depending on the energy level used (35). When the fragmentation of multiple stones is compared to the fragmentation of a single stone, both of similar volume, the results are better for the single stone. This is probably caused by the attenuating effect of the fragment layer, which is of particular relevance, since the threshold energy for intact stones appears to be higher than that for fragments of the same volume. IV— In Vivo Results of ShockWave Lithotripsy Within the last 25 years, different modalities for treating gallbladder stones have been added to standard open cholecystectomy: medical dissolution using bile salts, direct dissolution using methyltertbutyl ether (MTBE), ESWL with and without adjuvant dissolution, minicholecystotomy, and laparoscopic cholecystectomy (36– 43). The evaluation of their role in the treatment of gallbladder stones requires either controlled trials, which are rare, or the standardization of certain parameters such as success rates (i.e., complete stone clearance), morbidity, mortality, and, last but in our days not least, costs. Ideally, a careful balance of these different parameters with respect to each individual patient should allow the optimal treatment in a given situation. Some of these different therapeutic options, however, have been abandoned due to their limited success, leaving only a few centers that still offer the whole range of gallstone treatments. The role of ESWL is defined in the following paragraphs. A— Success Rate Lithotripsy has two major objectives: first, to facilitate dissolution (3,44), and, second, to allow spontaneous passage of fragments into the intestine. The success rate in shockwave lithotripsy is defined as the successful fragmentation of gallbladder stones into pieces smaller than 3 to 5 mm and the subsequent complete dissolution or expulsion of the stone fragments (Fig. 6a to c). As reported from in vitro studies, by far the most important parameters determining the rate and extent of complete disintegration are original stone number and stone volume and, to a lesser extent, their chemical composition and physical properties (see above) (2,5,7,45–48). Stones that are difficult to treat with bile acid dissolution alone (stones larger than 1 cm in diameter) and stones that are not too large (stones smaller than 3 cm) or too numerous (<4 stones) should be selected. Furthermore the stones should be radiolucent, reflecting a high cholesterol content and the possibility of adjuvant litholytic therapy. Gallbladder function must be guaranteed as documented by a normal gallbladder wall, a patent cystic duct, and adequate contraction. This allows the passage or active expulsion of fragments on the one hand and the entrance of cholesteroldesaturated bile after treatment with litholytic bile salts on the other. Therefore only patients with symptomatic radiolucent stones and an opacifying contracting gallbladder are eligible for a combined treatment of lithotripsy and bile acids. Clinical studies suggest that the best candidates are patients with solitary radiolucent stones up to 2 cm in an opacifying gallbladder whose fasting volume contracts more than 60% (5,49–51). When ESWL is limited to these patients, probably 5% or less of all gallstone patients become eligible. About 70% of these patients are rendered stonefree within 5 to 8 months (5,47,49,50) (Fig. 7). For larger stones up to 3 cm, complete stone clearance can be seen in only 50%, and this number
Page 534
Figure 6 Successful therapy of a solitary stone prior to ESWL (a), during shockwave lithotripsy (b), and 1 day after ESWL (c).
is reduced to 30% within the same time period when multiple stones (up to three) were treated. Besides stone size and number, the success rate is greatly influenced by initial gallbladder motility (51). Patients with good gallbladder contractility (i.e., ejection fraction >60%) are the best candidates for a high success rate. Stone clearance in patients with an ejection fraction higher than 60% prior to ESWL was twice as high after 3 months as in those patients with an ejection fraction below 60% (50). In summary, after 13 to 18 months, nearly 90% of all patients with solitary stones up to 2 cm and gallbladder contractility of more than 60% are completely free of stones and debris. Despite an even lower stone mass in patients with multiple stones, the results are generally less satisfactory than those for single stones. These data are readily explained (see Sec. III, "In Vitro Studies," above). Apparently a layer of stone fragments reduces the shockwave energy to below the threshold needed for fragmentation of the intact stones. As described above, the success of nonsurgical treatment is highly dependent on precise selection. Although ultrasonography (US) is the first diagnostic modality to detect gallbladder
Page 535
Figure 6 Continued.
stones, it is not able to document all parameters predictive of a high success rate precisely enough. In the case of more than one stone, US can only roughly estimate the total number of stones and will usually underestimate their size. By contrast, in the case of one stone in the gallbladder, a major part of that information can be obtained with ultrasound. It is able to distinguish between stones smaller or larger than 20 mm, and it is possible to determine the ejection volume as a marker of the patency of the cystic duct and the contractility of the
Figure 7 Clearance of fragments after extracorporeal shockwave lithotripsy and adjuvant bile acid therapy in patients with solitary radiolucent stones 20 mm or less in diameter. (From Ref. 46.)
Page 536
gallbladder. In addition, some authors claim that the ultrasonographic pattern of the gallstone may predict the outcome of fragmentation in vivo and in vitro (23,31– 33,52). Oral cholecystography, although no longer performed in many centers, is a very efficient diagnostic approach prior to ESWL. A single examination makes it possible to assess all four important parameters for the selection of optimal candidates: (a) stone volume, (b) stone number, (c) radiolucency, and (d) gallbladder function. The use of adjuvant bile salt treatment is controversial. From in vitro studies, the positive effect of ursodeoxycholic acid on the acceleration of stone dissolution is evident, underlining the initial strategy to follow up ESWL with bile acid therapy (44). In addition, some in vivo studies clearly indicate a beneficial effect of bile acid therapy on the speed of gallbladder clearance. The combination of chenodeoxycholic acid and ursodeoxycholic acid has been no more effective than ursodeoxycholic acid alone (53). The largest randomized study, by Schoenfield et al., showed that the group given adjuvant bile acid therapy had a success rate twice as high as that of the placebo group (54). However, this study was flawed by the fact that, because of the rather low energy used, the amount of stone disintegration was low. In this situation, stone dissolution is more important than fragmentation for stone clearance. In contrast, stone fragmentation into very small pieces favors stone expulsion from the gallbladder rather than dissolution (3). Overall, sufficient stone disintegration is probably more crucial than adjuvant bile acid therapy. Therefore, some groups have reported complete stone clearance in patients with no adjuvant bile acid treatment—that is, to the same degree as with litholytic therapy (55–58). Patients underwent, on average, four to six ESWL sessions, receiving a total of several thousand pulses. These preliminary reports were tested in a recently published randomized multicenter study from the Munich group (56). In this study, no difference in stone clearance was observed between patients who obtained litholytic treatment together with ESWL compared to those who received only ESWL (Fig. 8). However, with multiple lithotripsy sessions, the difference between small single stones (<20 mm), large single stones (>20 mm), and multiple stones in the success rate still remains (Fig. 9). B— Morbidity and Mortality ESWL is very safe. The most frequent adverse effect is biliary colic, which occurs in onethird of the patients treated (2,46). It is usually not accompanied by any more dangerous
Figure 8 Probability of complete stone disappearance after repeated shockwave lithotripsy with and without adjuvant bile acid dissolution therapy. (From Ref. 56.)
Page 537
Figure 9 Probability of complete disappearance of stones from the gallbladder after repeated shockwave lithotripsy in patients with small single stones, large single stones, and multiple stones (actuarial analysis). (From Ref. 56.)
complications, which are mainly caused by the passage of fragments and occur in 3 to 5% of patients. Pancreatitis is seen in around 2%, cholestasis in 1%, and cholecystitis also in 1%. Cholecystectomy or emergency endoscopic sphincterotomy is required in 2 to 5% and 1 to 2% of treated patients, respectively. Furthermore, obstruction of the cystic duct occurs in 5% of the patients and resolves spontaneously in more than half of them (47). Fatalities following lithotripsy are extremely rare, although anecdotal reports of deaths are published (59). When shock waves are directed at liver, gallbladder, kidney, or lung tissue, they induce hemorrhages and hematomas (20,60–63). However, the stone is normally located in the focal area, absorbing most of the shockwave energy, and only slight lesions may be observed in the surrounding tissue. The most serious side effect observed in dogs was lung bleeding. These adverse effects are dependent on shockwave energy and rate (60). In humans, liver hematomas are observed in 0.1% of the patients. Petechiae (8%) of the skin at the entry site of the shock wave and gross hematuria (4%) were transient and disappeared after 1 to 4 days. One month posttreatment, laboratory values of liver enzymes (ALT, AST, gamma GT) or parameters indicating cholestasis (alkaline phosphatase, bilirubin) did not differ from pretreatment values (2). C— Stone Recurrence From clinical studies on oral bile salt dissolution, it is known that patients who have undergone noninvasive treatment with preservation of the gallbladder are prone to stone recurrence. Around 50% of patients will redevelop gallbladder stones within 5 years after successful litholytic therapy (64,65). The risk of stone recurrence is lower in patients with solitary stones. The actuarial risk of gallstone recurrence after successful shockwave lithotripsy is 7% after 1, 15% after 2, and 31% after 5 years (66). In later years, the recurrence rate levels off, and approximately 60% are likely to remain stonefree without prophylaxis. In the remaining patients, pathogenetic mechanisms leading to stone recurrence—such as cholesterol supersaturated bile, nucleation defects, or gallbladder motor dysfunction—are likely to persist (67). Identification of those patients at high risk for stone recurrence is very important for successful ESWL. Sex, age, and body mass index do not indicate a greater risk of stone recurrence. A high relative content of deoxycholic acid in the bile—normally linked to an increased choles
Page 538
terol secretion—is under discussion as being one major parameter for the enhanced possibility of stone recurrence (67). Furthermore gallbladder contractility, besides stone number, is one of the most important predictors of stone recurrence (49). Only 13% of patients with solitary stones up to 20 mm and an ejection fraction above 60% developed recurrent stones, whereas 53% of patients with an ejection fraction below 60% had stone recurrence within the same time period (2 years). Therefore, the same parameters that predict the chance of complete stone clearance (i.e., stone size, stone number, and gallbladder motility) also partially predict the risk of stone recurrence, and the careful selection of patients for ESWL is of crucial importance not only for a high initial success rate but also for a low recurrence rate. C— CostEffectiveness The relative costs of shockwave lithotripsy, standard cholecystectomy, and laparoscopic cholecystectomy largely depend on the length of adjuvant bile salt therapy after ESWL, which is greatly dependent on the selection criteria for stone patients (68–70). ESWL is more costeffective than cholecystectomy for elderly patients and for patients with single stones. Younger patients and patients with multiple stones should be treated with surgery when economic factors are of great importance. This is even more the case when treatment with ESWL is compared with laparoscopic cholecystectomy (70). V— Extracorporeal ShockWave Lithotripsy of Bile Duct Stones Endoscopic papillotomy (EPT) followed by stone extraction is still the treatment of choice in cholecystectomized patients with bile duct stones (71). Many doctors also favour EPT in frail, elderly patients who still have their gallbladders in place, whether with subsequent operation of the gallbladder or without. However, more recent controlled trials have shown that therapeutic splitting may be less optimal than immediate choledochotomy together with cholecystectomy. Other very recent type II trials suggest that complication of EPT, which range between 3 and 8% (fatal complication rate around 1%) (72), may be reduced by balloon dilation of the papilla together with subsequent stone extraction (73). However, treatment of bile duct stones—whether after sphincterotomy of the papilla or after balloon dilation (74)—may fail in cases where there is a disproportion between the stone's size and the diameter of the bile ducts through which the stone must pass. Several techniques to crush or to dissolve the stones are available for that situation: mechanical lithotripsy, electrohydraulic lithotripsy, contact lithotripsy, laser lithotripsy, extracorporeal shockwave lithotripsy, and contact dissolution of stones in the biliary tree using substances that dissolve cholesterol (e.g., monooctanoin) or solutions that can dissolve stones or stone compounds composed of pigment. Direct dissolution techniques have mostly been abandoned because they take too long, are often unsuccessful, and have a rather high rate of side effects. By contrast, lithotripsy techniques are still widely used, since 10 to 20% of bile duct stones are not amenable to successful nonsurgical treatment without these adjuvant techniques. This chapter tries to define the role of ESWL for treatment of stones in the biliary tree. Analogous to the lithotripsy of gallbladder stones, shock waves are generated outside the body using electrohydraulic, electromagnetic, or piezoceramic sources. The shock waves are focused by reflection, acoustic lenses, or the array of the piezoceramic elements. Coupling to the body is achieved by a water cushion or bath (Fig. 10). Unlike the case with gallbladder stones, location of the calculi and positioning into the focal area should be done by fluoroscopy, although ultrasound monitoring has been reported
Page 539
Figure 10 Positioning of a bile duct stone for fragmentation with an electromagnetic lithotripter. The shock waves are focused by an acoustic lens. Coupling to the body is achieved by a water cushion from the ventral side. In electrohydraulic lithotripters, coupling is achieved by a water bath and shock waves enter from the rear.
in smaller series (75,76). This renders lithotripters, originally designed for kidney stones, also suitable for bile duct stones. In patients with intrahepatic bile duct stones, ultrasound monitoring and positioning may also be feasible using lithotripters with an integrated ultrasound system, originally designed for the treatment of gallbladder stones. In the beginning, we used shockwave entry from the rear to minimize attenuation of the wave energy by gasfilled bowel loops. Later on, clinical practice showed that a large percentage of patients may also be treated in the prone position, with the shock waves entering the body from the ventral side. Most centers observed the following selection criteria: Bile duct stones not amenable to endoscopic or percutaneous treatment according to the experience of the individual investigator Successful positioning of the calculi into the focal area of the lithotripter Focal axis of the shock waves that avoids lung tissue, gasfilled bowel loops, vascular aneurysms, calcified vessels, or large bone areas No severe deviation of coagulation parameters from the normal range If radiography is used for targeting and positioning of the stones, the direct injection of a contrast medium is required for visualization via nasobiliary tubes, T tubes, or transhepatically placed catheters (Fig. 11a and b). Treatment with older electrohydraulic machines with large focal dimensions often necessitates the use of anesthesia, whereas patients treated with other lithotripters have often received only intravenous sedative analgesia. A large number of uncontrolled but prospective clinical trials have been published to date. Stones may be disintegrated, at least to some extent, in 70 to 90% of patients (71,77,78). The overall clearance rate in patients with bile duct stones primarily not amenable to routine endoscopic or transhepatic procedures ranges between 60 and nearly 90% (75,77,79). The highest success rate has been observed in patients under general anaesthesia treated with a focal area of large volume (electrohydraulic kidney lithotripter, Dornier Medizintechnik) (71,77). In most patients, endoscopic extraction of fragments is necessary. A recently published small but carefully performed randomized trial (30 patients in each group) compared intracorporeal lithotripsy using a pulsed dye laser with ESWL using an electromagnetic generator (80). Intracorporeal lithotripsy was more efficient with respect to complete bile duct clearance (97 versus 73%). This favorable result was achieved with fewer
Page 540
Figure 11 An impacted bile duct stone that is not accessible with a basket via ERCP (a) is fragmented by shockwave lithotripsy into small pieces, which can be extracted or pass spontaneously through the papilla Vateri (b).
treatment sessions (1.3 versus 3.0 on average), leading to a shorter treatment time (0.9 versus 3.9 days). Side effects were minor and there was no 30day mortality. The study tells us that intracorporeal lithotripsy is very efficient in expert hands. The group that performed the trial is well known for its endoscopic expertise using approaches via the papilla or percutaneously via the intrahepatic biliary tree. However, it is still not clear to us whether these findings can be generalized. Unlike intracorporeal lithotripsy, ESWL is easy to learn, has a low level of side effects, can be repeated, and requires only limited technical skill. Severe adverse effects are rare in patients undergoing ESWL for bile duct stones. The most important complication following ESWL was septic disease, possibly caused by the introduction of microorganisms into the bloodstream during treatment. Therefore, perioperative antibiotic prophylaxis is mandatory. Hemobilia and macrohematuria, directly attributable to shock wave—induced tissue damage, are rare and of no clinical relevance in most patients. Intestinal hematoma or venous thrombosis have been reported anecdotally. Since cardiac arrhythmia may occur during ESWL when electrohydraulic shock waves are used, shockwave triggering should be performed under electrocardiographic control. General anesthesia is less and less warranted, since patients are treated with new lithotripters that have a smaller focal dimension. On the other hand, these lithotripters are presumably less effective than the firstgeneration electrohydraulic kidney lithotripters.
Page 541
Stone recurrence after successful extraction of fragmented bile duct stones occurs for two reasons (81,82): (a) the passage of stones from the gallbladder into the bile duct if the gallbladder is still in situ and (b) the reformation of stones resulting from bacterial overgrowth in the bile ducts (83), which induces deconjugation of bilirubin and phospholipids and subsequent crystallization of calcium bilirubinate and calcium fatty acid soaps. These stones are usually clayish and soapy and are described in 2 to 20% of patients despite successful sphincterotomy and adequate drainage. VI— Future Role of Extracorporeal ShockWave Lithotripsy The future role of shockwave lithotripsy will depend on several factors. First, the role of laparoscopic cholecystectomy has not yet been fully defined. Although it is less painful than standard cholecystectomy and reduces the hospital stay, it seems to be accompanied by a higher rate of bile duct injury. If this is true, ESWL will again become an acceptable alternative to surgery for selected patients. Second, costeffectiveness will be a very important issue in the future, and any therapeutic modality that is able to treat stones on an outpatient basis is worthy of consideration. From the financial perspective, ESWL is certainly effective when wellselected patients (greater age, single stone below 20 mm, in size, good functioning gallbladder) are treated with ESWL. In this patient group, the risk of complication is low and the duration of adjuvant bile salt therapy is brief if it is needed at all. The lithotripter can be shared with the urologist, thus ensuring its amortization. In addition, in contrast to surgery, ESWL can be carried out by one doctor without any assistance. Third, the treatment of bile duct stones by surgery is complicated by a rather high risk of mortality, so that ESWL faces more competition from intracorporeal lithotripsy. However, laser or electrohydraulic intracorporeal lithotripsy requires great expertise and lithotripter devices are expensive. In the case of bile duct ESWL, lithotripter models used for kidney stones can be shared with the urologist, and thus costs can be reduced. References 1. Brendel W, Enders G. Shock waves for gallstones: animal studies. Lancet 1983; 1:1054. 2. Sauerbruch T, Delius M, Paumgartner G, Holl J, Wess O, Weber W, Hepp W, Brendel W. Fragmentation of gallstones by extracorporeal shock waves. N Engl J Med 1986; 314: 818–822. 3. Greiner L, Münks C, Heil W, Jakobeit C. Gallbladder stone fragments in feces after biliary extracorporeal shock wave lithotripsy. Gastroenterology 1990; 98:1620–1624. 4. Sackmann M, Delius M, Sauerbruch T, Holl J, Weber W, Ippisch E, Hagelauer U, Wess O, Hepp W, Brendel W, Paumgartner G. Shockwave lithotripsy of gallbladder stones: the first 175 patients. N Engl J Med 1988; 318:393–397. 5. Darzi A, Leahy A, O'Morain C, Tanner W, Keane F. Gallstone clearance: a randomized study of extracorporeal shock wave lithotripsy and chemical dissolution. Br J Surg 1990; 77:1265–1267. 6. Ell C, Kerzel W, Langer H, Heyder N, Foerster E, Domschke W. Fragmentation of biliary calculi by means of extracorporeally generated piezoelectric shockwaves. Gut 1989; 34: 1006–1010. 7. Ponchon T, Barkun A, Pujol B, Mestas J, Lambert R. Gallstone disappearance following extracorporeal shock wave lithotripsy and oral bile acid dissolution. Gastroenterology 1989; 97:457–463.
Page 542
8. Barbara L, Sama C, Morselli Labate A, Taroni F, Rusticali A, Festi D, Sapio C, Roda E, Banterle C, Puci A, Formentini F, Colasanti S, Nardin F. A population study on the prevalence of gallstone disease: the Sirmione Study. Hepatology 1987; 7:913–917. 9. Jensen K, Jorgensen T. Incidence of gallstones in a Danish population. Gastroenterology 1991; 100:790–794. 10. Thijs C, Knipschild P, van Engelshoven J. The prevalence of gallstone disease in a Dutch population. Scand J Gastroenterol 1990; 25:155–160. 11. Friedman G. Natural history of asymptomatic and symptomatic gallstones. Am J Surg 1993; 165:399–404. 12. Ralston D, Smith L. The natural history of cholelithiasis: a 15to 30year followup of 116 patients. Minn Med 1965; 48:327–332. 13. Friedman G, Raviola C, Fireman B. Prognosis of gallstones with mild or no symptoms: 25 years of followup in a health maintenance organization. J Clin Epidemiol 1989; 42: 127–136. 14. Wenckert A, Robertson D. The natural course of gallstone disease: elevenyear review of 781 nonoperated cases. Gastroenterology 1966; 50:376–381. 15. Steiner C, Bass E, Talamini M. Surgical rates and operative mortality for open and laparoscopic cholecystectomy in Maryland. N Engl J Med 1994; 330:403– 408. 16. Kestenholz P, Herzog U, Kocher W. Die Cholecystektomie: vierjährige Erfahrung mit der laparoskopischen Methode. Schweiz Med Wochenschr 1994; 124 (suppl 64):438. 17. Williams L, Chapman W, Bonau, R, McGee E, Boyd R, Jacobs J. Comparison of laparoscopic cholecystectomy with open cholecystectomy in a single center. Am J Surg 1993; 165:459–465. 18. Lubock P. The physics and mechanics of lithotripters. Dig Dis Sci 1989; 34:999–1005. 19. Delius M, Müller M, Vogel A, Brendel W. Extracorporeal shock waves: properties and principles of generation. In: Ferruci J, Delius M, Burhenne HJ, eds. Biliary Lithotripsy. Chicago: Year Book, 1989, pp 9–15. 20. Sass W, Bräunlich M, Hayler M, Matura E, Folberth W, Kettermann S, Seifert J. Analysis of shock wave destruction of stones by highspeed films and microscopy. In: Paumgartner G, Sauerbruch T, Sackmann M, Burhenne HJ, eds. Lithotripsy and Related Techniques for Gallstone Treatment. St. Louis: Mosby— Year Book, 1991, pp 17–26. 21. Delius M, Brendel W. A mechanism of gallstone destruction by extracorporeal shock waves. Naturwissenschaften 1988; 75:200–201. 22. Vergunst H, Terpstra O, Schröder F, Matura E. In vivo assessment of shockwave pressures: implication for biliary lithotripsy. Gastroenterology 1990; 99:1467– 1474. 23. Neubrand M, Greinwald, I, Lobentanzer H, Paumgartner G, Hermeking H, Sauerbruch T. Physical laws of cholesterol gallstone fragmentation. Eur J Clin Invest 1997; 27:234–241. 24. Nitsche R, Hinrichsen H, Wilhelm R, Fölsch U. Influence of density and viscosity of fluids on extracorporeal shock wave lithotripsy of gallstones in vitro. Eur J Med Res 1996; 1:204–208. 25. Bouchier IAD, Cooperband SR, El Kodsi BM. Mucous substances and viscosity of normal and pathological human bile. Gastroenterology 1965; 49:343–353. 26. Brakel K, Terpstra O, Laméris J, Steen G, Nijs H, Blijenberg B. Predicting gallstone composition with CT: in vivo and in vitro analysis. Radiology 1990; 174:337–341. 27. Schulte S, Baron R. Piezoelectric biliary lithotripsy: an in vitro study of factors affecting gallstone fragmentation. AJR 1990; 155:1211–1216. 28. Schachler R, Bird N, Sauerbruch T, Frost E, Sackmann M, Paumgartner G. Extracorporeal shock wave lithotripsy (ESWL) of gallstones: an in vitro comparison between an electrohydraulic and a piezoceramic device. Gut 1991; 32:312–315. 29. Zeman R, Marchand T, Davros W, Garra B, GlassRoyal M, Soloway R. Gallstone fragmentation during biliary lithotripsy: effect of stone composition and structure. AJR 1991; 156:493–499.
Page 543
30. Barkun A, Valette P, Montet J, Dai K, Chauvin F, Cathignol D, Ponchon T. Physicochemical determinants of in vitro shock wave biliary lithotripsy. Gastroenterology 1991; 100:222–227. 31. Tsuchiya Y, Ohto M, Yazawa T. Ultrasonic properties of gallstonesdifferentiation between cholesterol stones and pigment stones: the biliary tract and pancreas 1986; 7:1483–1491. 32. Tsuchiya Y, Saito H, Saito N, Abe A, Ukaji M, Kuniyuki H, Mikami S, Natsuki Y, Nishiarai H, Haniya K, Takanashi H, Ohto M. Sonographic patterns of radiolucent gallbladder stones for predicting successful shockwave lithotripsy. J Gastroenterol Hepatol 1995; 10:426–431. 33. Nitsche R, Schweinsberg V, Klengel H, Niedmann P, Fölsch U. Different modes of fragmenting gallstones in extracorporeal shockwave lithotripsy. Scand J Gastroenterol 1993; 28:229–234. 34. Schachler R, Sauerbruch T, Wosiewitz U, Holl J, Hahn D, Denk R, Neubrand M, Paumgartner G. Fragmentation of gallstones using extracorporeal shock waves: an in vitro study. Hepatology 1988; 8:925–929. 35. Lobentanzer H, Neubrand M, Hermeking H, Sauerbruch T. In vitro study to elucidate the physical laws concerning the fragmentation of both solitary and multiple artificial stones. Clin Invest 1993; 71:882–887. 36. Neubrand M, Holl J, Sackmann M, Klüppelberg U, Pauletzki J, Paumgartner G, Sauerbruch T. Combination of extracorporeal shock wave lithotripsy (ESWL) and dissolution of gallbladder stones by methyl tertiary butyl ether: a randomized study. Hepatology 1994; 19:133–137. 37. Hellstern A, Leuschner U, Benjaminov A, Ackermann H, Heine T, Festi D, Orsini M, Roda E, Northfield T, Jazrawi R, Kurtz W, SchmeckLindenau H, Stumpf J, Eidsvoll B, Aadland E, Lux G, Boehnke E, Wurbs D, Delhaye M, Cremer M, Sinn I, Höring E, v. Gaisberg U, Neubrand M, Sauerbruch T, Salomon V, Swobodnik W, v.Sanden H, Schmitt W, Käser T, Schomerus H, Wechsler J, Janowitz P, Lohmann J, Porst H, Attili A, Bartels E, Arnold W, Strohm W, Paul F. Dissolution of gallbladder stones with methyl tertbutyl ether and stone recurrence: a European survey. Dig Dis Sci 1998; 43:911–920. 38. Thistle J. Direct contact dissolution of gallstones. Sem Liv Dis 1987; 7:311–316. 39. Danzinger R, Hofmann A, Schoenfield L, Thistle J. Dissolution of cholesterol gallstones by chenodeoxycholic acid. N Engl J Med 1972; 286:1–8. 40. Danzinger R, Hofmann A, Thistle J, Schoenfield J. Effect of oral chenodeoxycholic acid on bile acid kinetics and biliary lipid composition in women with cholelithiasis. J Clin Invest 1973; 52:2809–2821. 41. Perissat J, Collet D, Belliard R. Gallstones: laparoscopic treatment, intracorporeal lithotripsy followed by cholecystostomy—a personal technique. Endoscopy 1989; 21:373–374. 42. Perissat J. Laparoscopic cholecystectomy: The European Experience. Am J Surg 1993; 165:444–449. 43. The Southern Surgeons Club. A prospective analysis of 1518 laparoscopic cholecystectomies. N Engl J Med 1991; 324:1073–1078. 44. Neubrand M, Sauerbruch T, Stellaard F, Paumgartner G. In vitro cholesterol gallstone dissolution after fragmentation with shock waves. Digestion 1986; 34:51– 59. 45. Elewaut A, Crape A, Afschrift M, Pauwels W, De Vos M, Barbier F. Results of extracorporeal shock wave lithotripsy of gallbladder stones in 693 patients: a plea for restriction to solitary radiolucent stones. GUT 1993; 34:274–278. 46. Sauerbruch T, Neubrand M. Nonsurgical management of gallstones. Progress in liver disease 1992; 193–218. 47. Sackmann M, Pauletzki J, Sauerbruch T, Holl J, Schelling G, Paumgartner G. The Munich gallbladder lithotripsy study. Results of the first 711 patients. Ann Intern Med 1991; 114: 290–296. 48. Ell C, Kerzel W, Schneider H, Benninger J, Wirtz P, Domschke W, Hahn E. Piezoelectric
Page 544
lithotripsy: Stone disintegration and followup results in patients with symptomatic gallbladder stones. Gastroenterology 1990; 99:1439–1444. 49. Pauletzki J, Althaus R, Holl J, Sackmann M, Paumgartner G. Gallbladder emptying and gallstone formation: A prospective study on gallstone recurrence. Gastroenterology 1996; 111:765–771. 50. Pauletzki J, Sailer C, Klüppelberg U, von Ritter C, Neubrand M, Holl J, Sauerbruch T, Sackmann M, Paumgartner G. Gallbladder emptying determines early gallstone clearance after shockwave lithotripsy. Gastroenterology 1994; 107:1496–1502. 51. Sackmann M, Eder H, Spengler U, Pauletzki J, Holl J, Paumgartner G, Sauerbruch T. Gallbladder emptying is an important factor in fragment disappearance after shock wave lithotripsy. J Hepatol 1993; 17:62–66. 52. Dryszka H, Patel S, Sanghavi B, Patel G, Byk C, Salen G. Sonographic gallstone patterns are of value in predicting the outcome of biliary lithotripsy. Am J Gastroenterol 1991; 86:1626–1628. 53. Sackmann M, Pauletzki J, Aydemir U, Holl J, Sauerbruch T, Hasford J, Paumgartner G. Efficacy and safety of ursodeoxycholic acid for dissolution of gallstone fragments: Comparison with the combination of ursodeoxycholic acid and chenodeoxycholic acid. Hepatology 1991; 14:1136–1141. 54. Schoenfield L, Berci G, Carnovale RaTDNLSG. The effect of ursodiol on the efficacy and safety of extracorporeal shockwave lithotripsy of gallstones. The Dornier National Lithotripsy Study. N Engl J Med 1990; 323:1239–1245. 55. Soehendra N, Nam V, Binmoeller KF, Koch H, Bohnacker S, Schreiber HW. Pulverisation of calcified and noncalcified gallbladder stones: extracorporeal shock wave lithotripsy used alone. GUT 1994; 35:417–422. 56. Sauter G, KullakUblick G, Schumacher R, Janssen J, Greiner L, Brand B, Stange E, Wengler K. Lochs H, Freytag A, Wissing A, Holl J, Sackmann M, Paumgartner G. Safety and efficacy of repeated schockwave lithotripsy of gallstones with and without adjuvant bile acid therapy. Gastroenterology 1997; 112:1603– 1609. 57. Tsuchiya Y, Takanashi H, Haniya K, Nishiarai H, Mikami S, Natsuki Y, Kuniyuki H, Saito H, Saito N, Ohto M. An early gallstone clearance following repeat piezoelectric lithotripsy. J Gastroenterol Hepatol 1994; 9:597–603. 58. Tsuchiya Y, Ishihara F, Kajiyama G, Nakazawa S, Ohto M, Tanimura H, Akura Y, Harada M, Hihara M, Kawai Y, Kono Y, Koshiyama H, Morita M, Nakajima M, Nishina K, Sagawa H, Sakai T, Shoji M, Sone K, Sugimoto Y, Sugiyama K, Takahara O, Takamura T, Tazuma S, Wakamatsu H. Repeated piezoelectric lithotripsy for gallstones with and without usodeoxycholic acid dissolution: A multicenter study. J Gastroenterol 1995; 30: 768–774. 59. Wenzel H, Greiner L, Jakobeit C. Späkomplikationen der ESWL von Gallenblasensteinen mit letalem Ausgang. Dtsch Med Wochenschr 1990; 115:77. 60. Delius M, Enders G, Heine G, Stark E, Remberger K, Brendel W. Biological effects of shock waves: lung hemorrhage by shock waves in dogspressure dependence. Ultrasound Med Biol 1987; 13:61–67. 61. Delius M, Enders G, Xuan Z, Liebich H, Brendel W. Biological effects of shock waves: kidney damage by shock waves in dogsdose dependence. Ultrasound Med Biol 1988; 14:117–122. 62. Delius M, Jordan M, Eizenhoefer H, Marlinghaus H, Heine G, Liebich H, Brendel W. Biological effects of shock waves: kidney hemorrhage by shock waves in dogsadministration rate dependence. Ultrasound Med Biol 1988; 14:689–694. 63. Delius M, Denk R, Berding C, Liebich H, Jordan M, Brendel W. Biological effects of shock waves: cavitation by shock waves in piglet liver. Ultrasound Med Biol 1990; 16: 467–472. 64. Lanzini A, Jazrawi R, Kupfer R, Maudgal D, Joseph A, Northfield T. Gallstone recurrence after medical dissolution. An overestimated threat? J Hepatol 1986; 3:241–246. 65. Villanova N, Bazzoli F, Taroni F, Frabboni R, Mazzella G, Festi D, Barbara L, Roda E.
Page 545
Gallstone recurrence after successful oral bile acid treatment. Gastroenterology 1989; 97: 726–731. 66. Sackmann M, Niller H, Klueppelberg U, von Ritter C, Pauletzki J, Holl J, Berr F, Neubrand M, Sauerbruch T, Paumgartner G. Gallstone recurrence after shockwave therapy. Gastroenterol 1994; 106:225–230. 67. Berr F, Mayer M, Sackmann M, Sauerbruch T, Holl J, Paumgartner G. Pathogenetic factors in early recurrence of cholesterol gallstones. Gastroenterology 1994; 106:215–224. 68. Bass E, Steinberg E, Pitt H, Saba G, Lillemoe K, Kafonek D, Gadacz T, Gordon T, Anderson G. Costeffectiveness of extracorporeal shockwave lithotripsy versus cholecystectomy for symptomatic gallstones. Gastroenterol 1991; 101:189–199. 69. Nicholl J, Brazier J, Milner P, Westlake L, Kohler B, Williams B, Ross B, Frost E, Johnson A. Randomised controlled trial of costeffectiveness of lithotripsy and open cholecystectomy as treatments for gallbladder stones. Lancet 1992; 340:801–807. 70. Sonnenberg A, Benninger J, Ell C. Kostenvergleich zwischen der laparoskopischen Cholecystektomie und der extrakorporalen Stoßwellenlithotripsie in der Behandlung von Gallenblasensteinen. DMW 1994; 119:1532–1537. 71. Sauerbruch T, Feussner H, Frimberger E, Hasegawa H, Ihse I, Riemann J, Yasuda H. Treatment of common bile duct stones. A consensus report. Hepato Gastroenterol 1994; 41:513–515. 72. Freeman M, Nelson D, Sherman S. Complications of biliary sphincterotomy. N Engl J Med 1996; 335:909–918. 73. Bergmann J, Rauws E, Fockens P, et al. Randomised trial of endoscopic balloon dilation compared to sphincterotomy for removal of bile duct stones. Lancet 1997; 349:1124–1129. 74. Komatsu Y, Kawabe T, Toda N, Ohashi M, Isayama M, Tateishi K, Sato S, Koike Y, Yamagata M, Tada M, Shiratori Y, Yamada H, Ihori M, Kawase T, Omata M. Endoscopic papillary baloon dilation for the management of common bile duct stones: experience of 226 cases. Endoscopy 1998; 30:12–17. 75. Adamek H, Maier M, Jakobs R, Riemann J. Management of retained bile duct stones: a prospective open trial comparing extracorporeal and intracorporeal lithotripsy. Gastrointest Endosc 1996; 44:40–47. 76. Ponchon T, Martin X, Barkun A, Mestase J, Chavaillin A, Boustière C. Extracorporeal lithotripsy of bile duct stones using ultrasonography for stone localization. Gastroenterol 1991; 100:1730–1736. 77. Sauerbruch T, Holl J, Sackmann M, Paumgartner G. Fragmentation of bile duct stones by extracorporeal shock wave lithotripsy: A five year experience. Hepatology 1992; 15: 208–214. 78. Yasuda I, Tomita E, Moriwaki H, Kato T, Wakahara T, Sugihara J, Nagura K, Nishigaki Y, Sugiyama A, Enya M. Endoscopic papillary balloon dilatation for common bile duct stones: Efficacy of combination with extracorporeal shock wave lithotripsy for large stones. Eur J Gastroenterol Hepatol 1998; in press. 79. Gilchrist A, Ross B, Thomas W. Extracorporeal shock wave lithotripsy for common bile duct stones. Br J Surg 1997; 84:29–32. 80. Neuhaus H, Zillinger C, Born P, Ott R, Allescher H, Rösch T, Classen M. Randomized study of intracorporeal laser lithotripsy versus extracorporeal shockwave lithotripsy for difficult bile duct stones. Gastrointest Endosc 1998; 47:327–334. 81. Hammarstrom L, Stridbeck H, Ihse I. Longterm followup after endoscopic treatment of bile duct calculi in cholecystectomized patients. World J Surg 1996; 20:272–276. 82. Prat F. Malak N, Pelletier G. Biliary symptoms and complications more than 8 years after endoscopic sphincterotomy for choledocholithiasis. Gastroenterol 1996; 110:894–899. 83. Bergmann J, van Berkel A, Groen A. Biliary manometry, bacterial characteristics, bile composition, and histologic changes fifteen to seventeen years after endoscopic sphincterotomy. Gastrointest Endosc 1997; 45:400–405.
Page 547
26— Topical Contact Dissolution of Gallbladder Stones Salam F. Zakko University of Connecticut Health Center, Farmington, Connecticut I— Historical Overview Throughout history, there have been many remedies for the treatment of gallstones, including magic, herbal remedies, and naturopathic treatments. The earliest written accounts of contact chemical gallstone dissolution date back to the late 1500s, when Orgardney attempted in vitro dissolution of gallstones using turpentine but refrained from trying it in patients. The first account of chemical contact dissolution of gallstones in vivo was published in Lancet by Walker (1). He described the dissolution of a gallstone in situ using diethyl ether. Pribram standardized the procedure of ether dissolution for retained common duct stones (2), which surgeons used well into the 1950s (3). Subsequently, its use rapidly declined because of the unacceptable pain it caused upon infusion into the bile ducts as it boils below body temperature at 34.5°C, causing its volume to increase by 120fold. When Walker used it on his patient 100 years ago, he overcame this problem by venting the gallbladder around the glass tube that he used to infuse the solvent into the gallbladder through the cholecystotomy. Ether also can cause induction of anesthesia, and there is the potential for flammability. Since these early reports, advancements in the field have been sluggish, especially with the emergence and perfection of cholecystectomy. Nevertheless, the interest continued, especially for the management of retained common duct stones after cholecystectomy. Sporadic reports, describing various compounds for the dissolution of retained common duct gallstones via a T tube, continued to emerge. Chloroform was proposed, but it led to a number of unwanted complications, including anesthesia, cholangitis, pancreatitis, and hermorrhagic duodenal ulcerations (4). Solutions of heparin (5) and clofibrate (6) were once felt to be useful; however, subsequent in vitro studies showed that they were not different from normal saline (7,8). The general feeling was that their initial perceived success was attributable to the physical flushing effects of the irrigating solution. Success rates of up to 50 to 60% have been reported in eliminating retained common duct stones by saline irrigation alone (9,10). In the early 1970s, Way et al. reported success in eliminating retained common duct stones by infusing a 100mM solution of the bile acid sodium cholate at a rate of 30 mL/h (11). Up to 14 days of infusion were necessary, and a significant number of complications were observed. These were mainly due to the fact that the solution was used to dissolve retained common duct stones by infusing it one way through a T tube, producing a buildup of the bile acid in the intestinal tract, where it led to diarrhea in most patients. Higher concentrations were more effective but led to more diarrhea and were toxic, causing severe bile duct inflammation and even death in the rhesus monkey (12). Additional bile acid solutions were studied with
Page 548
and without the addition of lecithin, but all were relatively toxic to the biliary tree and none ever proved clinically useful (13). II— MonoOctanoin for Dissolution of Bile Duct Stones In 1974, it was discovered that monooctanoin, which was being commercially manufactured as a steroid solvent, was an excellent solvent of cholesterol gallstones (14). In the mid1980s, it was approved by the FDA for perfusion into the common bile duct to dissolve residual cholesterol gallstones after cholecystectomy (15). Monooctanoin is a viscous oil, with a freezing point of 20°C. It has a high solubility for cholesterol at 11.7 g/100 mL, but because of its high viscosity and poor kinetic solubility, it acts very slowly. A significant number of reports have been published describing its clinical usefulness, but the most comprehensive study is probably that by Palmer and Hofmann, who reported on 343 patients with stones of the common bile duct. These patients were treated at a large number of centers under an investigational new drug (IND) application sponsored by Dr. Alan Hofmann at the University of California in San Diego (16). Under this unique arrangement, physicians from around the United States faced with patients who had retained common bile duct stones obtained monooctanoin from Dr. Hofmann's laboratory and supplied outcomes data in return. Treatment times ranged from 3 to 21 days (average 7.2 days). In 26%, the stones disappeared; in another 8%, they became smaller, enabling extraction; and in 20%, there was only a minimal response. While this may be reasonable for the treatment of retained common duct stones after surgery, it is not acceptable as primary therapy for gallbladder stones, since 1 to 3 weeks of solvent infusion with only a 26% total success rate cannot compete with the surgical alternative. Monooctanoin is currently marketed under the name Moctanin (15); it is indicated as a solubilizing agent, administered either through a nasobiliary catheter or a T tube, for the dissolution of retained cholesterol common duct stones. In practice, it is used only in the rare event that a bile duct stone that is too large to be removed endoscopically or percutaneously is encountered postcholecystectomy, especially in a patient who is a poor operative risk. In such a case, a catheter resistant to the solvent is placed with its pigtail loop next to the stone and monooctanoin is infused one way at a flow rate of 1 to 5 mL/h while intrabiliary pressure is monitored with a central venous pressure setup connected to the infusion line. The infusion pressure must not be allowed to rise above the hepatic secretory pressure head, which is normally 20 to 30 mL of water, as this may lead to backdiffusion of possibly infected bile into the hepatic sinusoids. This procedure is irritating to the intestinal mucosa and therefore contraindicated in patients with active ulcers. The most common side effect from this process is diarrhea caused by the monooctanoin load into the intestine. In the series by Palmer et al., side effects led to discontinuation of treatment in 9% of patients (16). Attempts to further improve the efficacy of monooctanoin by combining it with substances such as Dlimonene (17), palmidrol with pluronicF68 (18), Nacetyl cysteine (19), or water have been made through several in vitro studies (20). The addition of Nacetyl cysteine or 10% water proved to slightly enhance the efficacy of monooctanoin, but none of these mixtures were able to dissolve stones fast enough to make monooctanoin usable for primary gallbladder stone dissolution. With the advances in and availability of endoscopic mechanical, electrohydraulic, and pulse dye laser lithotripsy, the interest in chemical dissolution of bile duct stones has faded. III— The Discovery of Methyl TertButyl Ether and Gallbladder Catheterization It was not until the discovery that the gasoline additive methyl tertbutyl ether (MTBE) was a safe and effective cholesterol gallstone solvent (21) that Walker's original procedure (1) was
Page 549
revisited. MTBE, like diethyl ether, is an aliphatic ether but has a boiling point that, at 55.2°C, is well above body temperature, allowing it to be instilled safely into the gallbladder. Its solubility for cholesterol is 18 g/100 mL, but, more importantly, its viscosity is much lower than that of monooctanoin, giving it excellent kinetic solubility and making it capable of dissolving cholesterol gallstones at least 50 times faster (21). In both animal and human testing, MTBE produced no gross injury to the gallbladder or its mucosa (21,22). Another major advance that helped resurrect contact dissolution of gallbladder stones was the ability of interventional radiologists to place a small catheter percutaneously into the gallbladder and remove it safely (23,24). Therefore, it was appealing to use this combination of an excellent cholesterol solvent and simple percutaneous access to the gallbladder to eliminate gallstones in a nonsurgical procedure. In 1985, Allen et al. described a patient whose single 13mm intrahepatic stone was dissolved after 7 h of MTBE lavage through a nasobiliary catheter and another patient in whom multiple small gallbladder stones were partially dissolved by MTBE infused via a percutaneous transhepatic catheter (25). Since then, successful use of MTBE for percutaneous gallbladder stone dissolution has been described by several groups (26–28), but the results have been poor due to a number of problems that are perhaps the main reason behind the nondissemination of this technique. These problems are discussed below, along with some proposed solutions and recent developments. IV— The Procedure of Percutaneous Topical Gallstone Dissolution This procedure is currently considered investigational in the United States because no FDAapproved solvent is available. A smalldiameter catheter is placed into the gallbladder and a lipid solvent such as MTBE is then used to lavage and dissolve the stones in situ, taking care not to exceed either the gallbladder volume (27,28) or leakage pressure (29) to prevent the escape of solvent from the gallbladder. Hence, the application of the solvent is topical whereby the solvent is introduced only into the lumen of the gallbladder and not systemically. When the stones have dissolved, the percutaneous catheter is removed. No recuperation time is required and the gallbladder is spared. The average time until all stones are eliminated is dependent on the solvent, the method of solvent delivery, and the type of stones. Candidates for percutaneous topical dissolution must have mostly cholesterol gallstones. At a minimum, the stones must be radiolucent on plain abdominal xray. The size and number of stones and whether the gallbladder is functioning are irrelevant. This is because the solvents are instilled directly into the gallbladder—in contrast to medical therapy with oral bile acids, where gallbladder function is necessary for the bile acids to enter and become concentrated in the gallbladder and for residual debris to be expelled into the duodenum. In fact, when gallbladder function was assessed by oral cholecystography with sincalide stimulation, it was found to recover shortly after stones were eliminated by topical dissolution with MTBE (30). Normal coagulation and bleeding parameters must be verified before percutaneous catheterization is attempted. Also, adequate attachment of the gallbladder to the liver must be confirmed and the presence of hemangiomas, cysts, or tumors in the planned path of the catheter must be ruled out. This can easily be done using either ultrasonography or computed tomography. The night before the procedure, it is best if the patient is given six pills of oral contrast, such as Telepaque (iopanoic acid), as for an oral cholecystogram. This is in an attempt to visualize the gallbladder on fluoroscopy, which simplifies the catheterization procedure. If the gallbladder is not functioning and is not visualized fluoroscopically in response to the administration of contrast, then gallbladder catheterization may be achieved using ultrasonographic guidance. Catheter placement is achieved by using a coaxial guidewire catheter introduction system (24). After sterile prepping and draping, local anesthetic (1% lidocaine) is instilled to the level
Page 550
of the liver capsule. A 2mm skin nick is made and a 22gauge Accustick needle (Meditech, Watertown, MA) is introduced into the gallbladder. A 0.018in extra stiff stainless guidewire with a platinum tip is then exchanged for the needle. A coaxial dilator consisting of a 6Fr sheath over a 4Fr dilator containing a stainless steel stiffener (Meditech Accustick Introducer System) is then introduced over the wire. The 4Fr dilator and stiffener cannula are removed, leaving the 0.018in. wire coiled in the gallbladder lumen. All bile is aspirated through the 6Fr sheath by covering the protruding end of the 0.018in. wire with a K52 connector; the gallbladder is rinsed clear then filled to half its volume with 20% watersoluble contrast. The 0.018in. wire is then exchanged for a standard 0.038in. Tefloncoated guidewire and the sheath is exchanged for a 6 to 7Fr pigtail catheter, which is used for the dissolution. The catheter is preferably placed through the gallbladder/liver interface (the extraperitoneal area of attachment of the liver to the gallbladder). This ensures a good seal with hepatic tissue around the catheter, which helps stabilize it. It also helps prevent intraperitoneal bile leakage after catheter removal, as the liver tissue recoils to seal the puncture track when the catheter is taken out. It is possible to place the catheter directly into the gallbladder without traversing hepatic tissue, but, in this case, the catheter has to be left in place for more than 2 weeks for a mature track to develop. Otherwise, an intraperitoneal bile leak is very likely. In most cases, it is desirable to ascertain at least some degree of livertogallbladder attachment on ultrasonography or computed tomography before catheterization. Cholesterol gallstone solvents such as MTBE are organic solvents that dissolve plastics. Hence, all wettable surfaces such as the catheter and all instruments utilized for the procedure (connective tubing and syringes) must be made of glass, stainless steel, polyethylene, polyamide, or polytetrafluoroethylene (PTFE) (31). When the catheter is in place, dissolution begins with aliquots of solvent being infused into and out of the gallbladder. Several means of solvent delivery have been employed, ranging from a glass syringe (25–27), a fixedvolume automatic syringe pump (32), or a pressurecontrolled peristaltic pump system (29). The dissolution response is assessed at regular intervals using a combination of techniques that include evaluating the cholesterol output in the solvent effluent, cholecystography by contrast fluoroscopy, and ultimately ultrasonography. At the end, irrigation of the gallbladder with plain saline is usually beneficial in clearing any fine insoluble residue which frequently remains even after predominantly cholesterol stones are dissolved. V— Problems Associated with Topical Dissolution Therapy On the surface, the procedure of topical dissolution appears to be very simple and straightforward. However, in its apparent simplicity, it can be tricky, and it must not be underestimated. Several problems exist which are either general, relating to the procedure, or specific, relating to the use of MTBE as solvent. These problems are discussed in the three main categories outlined below. A— Problems Relating to Solvent Delivery Classically, solvent lavage is carried out manually using syringes or a fixedvolume syringe pump (27,28). In an attempt to prevent overflow into the biliary tract and intestine, MTBE was infused and aspirated, with the infusion volume being fixed and limited to less than that which initially filled the gallbladder (overflow volume). However, changes in this overflow volume due to spontaneous gallbladder contraction frequently occur, leading to inadvertent emptying of solvent into the common duct and then the duodenum. In a series of 26 patients treated with percutaneous solvent litholysis, we showed that the gallbladder capacity decreased by
Page 551
30% of its initial value after 2 h of MTBE therapy in 57% of patients. In fact, in 8% of these patients, the gallbladder volume decreased by 80% (33). Hence, in more than half the patients, reliance on the initial overflow volume will most probably lead to the escape of solvent into the duodenum during the course of the procedure. MTBE has no ill effects on the gallbladder mucosa (21,22) and is not appreciably absorbed from it (29); however, if it escaped through the cystic and common bile ducts into the duodenum, it leads to a number of toxic effects due to mucosal irritation (nausea, vomiting, and duodenal inflammation) and/or systemic absorption (somnolence, anesthesia, hemolysis, and the intense odor of MTBE on the breath) (26–28,34). This means that when a fixedvolume syringedelivery system is used, the operator will have to rely on the patient's symptoms in controlling the volume delivered at any given time. This is laborintensive and, in principle, is potentially hazardous. In the study by Thistle et al., more than 50% of the patients treated required intravenous analgesia during solvent infusion and 31% experienced nausea and emesis (27). Manual infusion of solvent and aspiration by syringe is also slow and timeconsuming. The Mayo group reported an average dissolution time of 12.5 h (range = 4 to 31) (27). Similar figures were reported in the German series by Leuschner and colleagues (28). Another problem with solvent delivery by syringe is that it often leads to incomplete stone elimination, since particles of insoluble residue are left behind. Some 70% of patients treated at the Mayo Clinic were left with residue, as were 34% of patients treated in the German series. Such residue may not only continue to cause symptoms (27) but can act as a nidus for early gallstone reformation (35). Finally, fixedvolume infusion and aspiration is inefficient, because increases in gallbladder capacity, resulting from stone dissolution, are not utilized to increase solvent delivery, which would enhance dissolution. This may be partially responsible for the prolonged times of dissolution reported (27,28). B— Patient and Gallstone Selection The problem here is that the currently available solvents for topical dissolution are organic cholesterol solvents, but not all gallstones are cholesterol. Even ''cholesterol gallstones" have varying degrees of noncholesterol material, which may leave residual debris that cannot be aspirated through the percutaneous catheter; the result is incomplete gallstone elimination. Noncholesterol gallstones and noncholesterol materials in cholesterol gallstones are composed of varying concentrations of calcium salts of bilirubinate, palmitate, phosphate, and carbonate, which fortunately are radiopaque (36). Because of this, the plain abdominal xray has been used to identify noncholesterol stones that will not dissolve. However, the plain xray lacks sensitivity because, frequently, the noncholesterol material in a gallstone may not be sufficiently dense to produce a radiopaque image on a plain abdominal xray. Computed tomography is more sensitive than plain xrays in detecting such calcific material, but in many instances it is too sensitive, and many stones that appear dense on a CT image may still be totally eliminated as a result of dissolution. Other imaging modalities may hint at the presence of cholesterol stones. The density of cholesterol stones is less than that of concentrated bile or contrast. Therefore, if the patient is examined in the upright position during an oral cholecystogram (OCG), the finding of floating stones in the form of a layer is highly indicative of stones that will respond very favorably to dissolution (Fig. 1). Floating stones can occasionally also be seen on ultrasonography (Fig. 2). Floating stones are found only in a small proportion of patients, and if this were the sole characteristic used for patient selection, many patients with otherwise soluble stones would be excluded. All of the above suggest that more accurate means for the selection of stones amenable to dissolution are needed.
Page 552
Figure 1 Floating stones on an upright oral cholecystogram.
C— Postdissolution Imaging Since any nonsurgical procedure leaves an intact gallbladder, it must be determined that all gallstones and debris have been totally dissolved or removed, because any remaining debris may continue to cause symptoms (27) and accelerate early gallstone reformation (35). It must also be determined that no other incidental pathology, such as ulcers or tumors, is present on the gallbladder mucosa. Although the incidence of carcinoma of the gallbladder is low, there seems to be a relationship between its presence and longstanding gallstone disease (37). Ultrasonography has been the standard imaging modality used to evaluate the gallbladder following dissolution therapy. The problem with ultrasonography is that it lacks sensitivity for minute residual fragments, it is particularly difficult in the presence of the percutaneous catheter, and it cannot visualize subtle mucosal changes such as ulcers or the early stages of carcinoma.
Figure 2 Floating cholesterol stones as observed on Cultrasonography.
Page 553
VI— Refinements and Advances in Topical Dissolution Several solutions have been and are being devised to overcome the previously stated problems in an effort to make the procedure of topical chemical dissolution clinically viable and able to compete favorably with the surgical alternatives. But before any workable solutions are developed, an understanding of several basic biomechanical principles is needed. The following is a discussion of a number of theoretical issues and a review of the most recent developments in the field. A— Biomechanical Considerations The flow of a confined fluid between compartments (e.g., from the gallbladder through the cystic duct, common bile duct, and the duodenum) is dependent on a force resulting from pressure gradients (38). When applied to gallbladder fluid dynamics, the gallbladder emptying pressure can be defined as follows: Gallbladder emptying pressure = cystic duct resistance + common bile duct pressure This emptying pressure is different from one patient to another, but when studied in a number of patients undergoing gallstone dissolution, it was found to be fairly constant in the same patient over time (33). This is not surprising, since the cystic duct has no significant musculature but only redundant mucosa that arranges itself in a series of folds called the valves of Heister. These valves are passive in their impedance to the flow of bile (39,40). Therefore, as long as intragallbladder pressure is maintained below the emptying pressure for a given patient, no fluid will pass from the gallbladder into the common bile duct regardless of the volume of the gallbladder. This principle is especially important to consider when dealing with compartments that undergo constant changes in volume, such as the gallbladder (33,41). The rate of dissolution of solids in liquids is determined by the following relationship (42,43):
Thus, to enhance the dissolution rate of gallstones, a lowviscosity solvent with high solubility for the targeted solute is optimal. Also, the solvent must be stirred or perfused at high rates to provide high solvent velocity. Continuous contact time between the solvent and solute is important in enhancing the dissolution. In the case of dissolving gallbladder stones, continuous contact between the solvent and the stones must be ensured by the constant removal or exclusion of bile. During solvent lavage, bile in the gallbladder may form a separation layer between the solvent and stones, reducing the contact time. This can be managed, during solvent delivery, by not allowing intragallbladder pressure to fall below that of the common bile duct, as otherwise bile would be drawn into the gallbladder. Cholesterol gallstones contain, in addition to cholesterol monohydrate, varying proportions of noncholesterol residue, which is insoluble in cholesterol solvents such as MTBE. Such residue, in the form of fine, sandlike macroscopic particles, is frequently left behind after manual MTBE therapy (27,28), mainly because of the residues' high density and the inefficiency of syringes or syringe pumps in evacuating such residue. This problem can be solved by achieving continuous flow of sufficient turbulence to keep such particles in suspension, promoting their aspiration. B— Solvent Delivery Systems After the initial report of the successful use of MTBE for topical gallstone dissolution using a manual syringe, the Mayo Clinic group cooperated with BaxterTravenol, Inc., in automating
Page 554
the procedure of handsyringing the solvent into the gallbladder. As a result, a volumecontrolled oscillating syringe pump was developed. However, in a controlled study versus the hand syringe, this device was not significantly better (32), probably because it lacked feedback control and suffered from the problems discussed earlier relative to fixedvolumesyringe solvent delivery. Independent work at the University of California, San Diego, by Zakko and Hofmann led to the description of an automated solvent delivery system that utilizes peristaltic pumps and a pressurecontrolled feedback loop. The system, which is referred to as the microprocessorassisted solvent transfer (MST) system, was developed to overcome the problems that relate to solvent delivery (44). It consists of a microprocessor that controls two peristaltic pumps, which circulate the solvent from a fixedvolume reservoir into the gallbladder and back (Fig. 3). A number of feedback loops continuously update the microprocessor controller with information on intragallbladder pressure, elapsed time, the revolutions per minute, and pumping direction of each pump. The microprocessor operates with a specific algorithm that is outlined elsewhere (44). The end result is automatic highflow solvent lavage into the gallbladder while intragallbladder pressure is monitored and adjusted to prevent the solvent from escaping beyond the confines of the gallbladder, where it could lead to toxic effects (44). The high flow rate enhances dissolution and produces the necessary turbulence, which helps to produce fragmentation and agitation, keeping the insoluble residue in suspension and promoting its aspiration. While a high solvent perfusion rate has no major impact when pure cholesterol gallstones are being dissolved, it is highly important for the efficient dissolution of mixed stones (Fig. 4) (29), which are found much more commonly. Prior to system operation, the gallbladder leakage pressure is determined for each particular patient by injecting dilute contrast into the gallbladder through the infusion lumen of
Figure 3 Schematic diagram of the microprocessorassisted solvent transfer system. (From Ref. 44.)
Page 555
Figure 4 The effects of solvent (methyl tertbutyl ether, or MTBE) perfusion rates on the time of total dissolution for gallstones with varying cholesterol contents. (From Ref. 31.)
the percutaneous catheter while observing the gallbladder fluoroscopically. The gallbladder leakage pressure is the pressure at which the contrast is observed to escape through the cystic duct or to leak around the catheter entry port. The gallbladder contents are then slowly aspirated until total evacuation is observed on fluoroscopy and bile is found in the aspirate. The pressure at this point is called the bile aspiration pressure. During its operation, the MST system maintains intragallbladder pressure between the gallbladder leakage pressure and the bile aspiration pressure. Hence, the system is able to achieve a high effective solvent flow rate of up to 200 mL/min while assuring that no solvent escapes beyond the gallbladder (29) and that no bile enters the gallbladder. In contrast to solvent delivery by fixedvolume manual syringe or syringe pump, the MST system constantly and automatically modifies the volume of solvent being infused to maintain a constant pressure and to correct for variations in gallbladder capacity. The feedback control loops also allow for a number of safety alarms that permit uninterrupted automatic operation. All this greatly simplifies the otherwise laborintensive procedure, and it is hoped that this will help in bringing it to the realm of everyday clinical practice. The first prototype of the system was constructed and used in the treatment of patients in San Diego (29). C— Computed Tomography (CT) for Gallstone Selection and a Predictive CT Index To address the issues relative to selection of patients with stones amenable to dissolution, several studies have looked at more objective parameters on CT to see whether they correlate with gallstone dissolvability and/or the chemical composition of the gallstones. While the average global density of the CT image has been shown to correlate with gallstone composition (36), it does not appear to correlate well with stone dissolvability (45,46). This is probably because a 90% cholesterol stone may have a small focus of densely packed noncholesterol material, such as calcium bilirubinate, that cannot be dissolved or fragmented to particles small enough to be aspirated through the percutaneous catheter.
Page 556
These findings have resulted in a reevaluation of the methodology used in the in vitro studies that look at the use of CT scanning for gallstone selection. While correlations were initially made with the stone's chemical composition (36), now stone dissolvability is used (47). The gallstones studied were exposed to the solvents in agitated test tubes (45,48–50), a setup that is totally different from and inferior to the methods used for solvent delivery when patients are actually treated. More recent studies use model gallbladders and the same solvent delivery systems used in treating patients. More importantly, earlier studies used the rate of cholesterol dissolution in terms of decrease in stone weight over time as an outcome measure (45,48–50). This introduced significant errors, since the cholesterol portion of a mostly noncholesterol stone will still dissolve at a fairly fast rate, yielding a decrease in weight. This may erroneously be interpreted as favorable dissolution, while the majority of the stone would remain untouched even after many hours of subsequent solvent infusion. Recognizing that the aim of therapy is to completely eliminate all stones and residue from the gallbladder, a more clinically applicable outcome of complete stone dissolution with elimination of all residue is now used as an endpoint measure (47,51,52). CT scanning of small objects such as gallstones is subject to a number of idiosyncracies that can limit its usefulness. Several fine details need to be observed before it can yield clinically useful diagnostic information with respect to gallstone dissolvability. First, localization of the gallstones in the gallbladder may be difficult. It is conceivable that a moderatesized gallstone may be missed on sectioning during the CT scanning process. This may be the case even when contiguous CT scan cuts of 3 to 5mm collimation are done. The problem here lies in the method employed to scan the gallstones in vivo, especially using oldergeneration machines. Before each CT cut, the patient is asked to stop breathing at a particular point in the breathing cycle in the hope that the gallbladder will assume the same position it had during the previous cut. In reality, no patient can hold his or her breath at the same spot twice. As a result, the gallbladder is never back to the same position it was at during the first cut. In the experience of the author, when the procedure is performed in this fashion, 5mm cuts will have a margin of error of up to ±30 mm. This is obviously associated with an increased incidence of missed gallstones. To correct for this, sequential 2.5mm contiguous cuts through the gallbladder using dynamic scanning with offline reconstruction can be carried out. This allows one CT scan cut every 2 s. Hence, during a 30s breathhold, 15 sequential cuts may be obtained with the gallbladder in the same position. More recently, the spiral CT scanner can perform all CT scan cuts in one breathhold. Another source of inaccuracy is the observed variability in the CT attenuation values from one machine to another and even on the same machine at different times. Obviously this would have a major impact, since the decisions made are based on CT attenuations. This can be overcome by calibrating the machine using a standard phantom prior to scanning each patient. The numbers from the phantom can then be used to correct for any variability. To improve on the specificity of the CT scan in selecting stones amenable to dissolution, a predictive CT index can be calculated for each stone from multiple correlative CTimage variables (51). This index was found to be highly predictive in selecting gallstones amenable to total dissolution with MTBE. It is calculated from a number of variables derived from the gallstone's CT image information. Such variables include the standard deviation of the gallstone's average CT density, the density of the most dense pixel, the density of the least dense pixel, the average CT density of dense pixels and its standard deviation. Although each of these variables on its own is only moderately predictive, all of them, when used collectively in a specifically derived mathematical equation, yield a highly specific index with a predictive value of up to 98%, a specificity of 93%, and a sensitivity of 85% when studied in vitro (51). This was confirmed in an in vivo study, which yielded even higher figures (52). When this index was used in vivo to select patients for topical dissolution with MTBE, successful complete gallstone dissolution and evacuation of residue was achieved in 30 of 31 patients treated (53). Fine residual debris, which was missed on ultrasonography in the presence of the percutaneous catheter, was detected after its removal in one patient.
Page 557
Figure 5 The minigallbladder endoscope. (From Ref. 54.)
D— Percutaneous Gallbladder Endoscopy To solve the problem of postdissolution imaging discussed above and to develop a more accurate imaging modality with which to examine the interior of the gallbladder after topical dissolution, a percutaneous gallbladder miniendoscope was developed and a procedure of percutaneous gallbladder endoscopy described (54). The main objective is to evaluate the gallbladder by direct visualization after topical dissolution of gallstones but prior to loss of the percutaneous access, so that, if residue is found, it may be dealt with. The gallbladder mucosa can also be carefully examined to look for possible mucosal lesions. The miniendoscope is only 2.2 mm in diameter (Fig. 5), essentially allowing it to be introduced into the gallbladder percutaneously without having to dilate the percutaneous puncture, which would prolong patient recovery time. In a prospective controlled study, the sensitivity of gallbladder endoscopy was compared with ultrasonography and aircontrast cholecystography in 18 patients who underwent percutanteous topical dissolution. All examinations were performed before catheter removal and after the gallbladder was deemed stonefree by the traditionally employed technique of singlecontrast cholecystography. Residual debris was detected in only 1 patient by ultrasonography and in none of the 18 patients by doublecontrast cholecystography, yet endoscopy showed stone fragments in 13 patients (Fig. 6). In all these patients, catheter repositioning and additional solvent perfusion resulted in elimination of the debris as assessed by further endoscopy. Two patients had endoscopically detected erosions. Doublecontrast fluoroscopy found only one of
Figure 6 Residual debris as observed endoscopically. (From Ref. 55.)
Page 558
Figure 7 Normal gallbladder mucosa as seen within minutes after infusion of MTBE. (From Ref. 55.)
these, whereas ultrasonography detected neither (55). Endoscopy has also helped to verify the normality of the gallbladder mucosa after exposure to gallstone solvents (Fig. 7) (55). The finding of an early gallbladder carcinoma by percutaneous endoscopy after topical dissolution has also been reported (56). VII— A Comprehensive Procedure for Topical Dissolution Studies are currently under way to evaluate the utility of a comprehensive procedure for topical dissolution in which patients are selected with the CT index, treated using the microprocessorassisted solvent transfer system, and examined with percutanteous gallbladder endoscopy prior to catheter removal. A preliminary report on the treatment of 52 patients showed encouraging results. The solvent MTBE was circulated from a 150mL reservoir at 180 mL/min for 2h periods until total dissolution and elimination of debris was achieved as assessed by percutaneous gallbladder endoscopy. Complete dissolution with no residue occurred in 51 patients after an average of 5.8 ± 0.6 h of treatment spread over 2 ± 0.2 days. Of the 52 patients treated, 17 were at high surgical risk and 9 had a nonfunctioning gallbladder on oral cholecystography. Most patients were treated as outpatients. No analgesia was required during the dissolution procedure. After catheter removal, three patients experienced a bile leak that responded to conservative therapy. One patient had a mild case of pancreatitis related to the postprocedure cholangiogram, three patients developed local infection, and one patient with an obstructing common duct stone had cholangitis and a transient episode of hepatitis (57). VIII— New Solvents The encouraging results with automated topical dissolution have led to the investigation of other solvents that may be less toxic for the patient and safer to handle than the ether homologue MTBE, which is highly volatile and explosive. Hofmann and colleagues found that three C5 esters—ethyl propionate, isopropyl acetate, and propyl acetate—were superior to MTBE with respect to their boiling points, flammability, taste, and odor (58,59). When matched cholesterol gallstones were subjected to topical dissolution in vitro with the microprocessorassisted solvent transfer system, ethyl propionate was found to exhibit a faster dissolution rate than MTBE and it showed a trend for being efficacious against a larger number of unselected human gallstones (60).
Page 559 Table 1 Comparison of Methyl TertButyl Ether (MTBE) and Ethyl Propionate (EP)
MTBE
Cholesterol solubility (g/100 mL)
EP 18
10
Density (g/mL)
0.76
0.89
Viscosity (cP)
0.29
0.50
Flash point (°C)
27.8
12
Vapor pressure at 25°C (mmHg)
245
40
LD50 (g/kg, rat)—oral (65,66)
4
8.73
>0.152
LD50 (g/kg, rat)—intraperitoneal (67,68)
1.2
Smell (59)
Intense stench
Fruity
Metabolism (59,69)
Methanol, tertbutanol, Ethanol and and formic acid propionic acid
Ethyl propionate (EP) is a naturally occurring compound found in grapes (var. Sauvignon), several types of wine, and cocoa. It has been in public use since the 1930s, was given GRAS (generally regarded as safe) status by the Federal Emergency Management Agency (FEMA) (1965), and approved by the FDA for food use (21 CFR 172.515). Its use in fragrances in the United States amounts to approximately 16,000 1b/year (61). In the Phillipines, among other countries, EP is marketed as a major component of a number of liquors such as Lambanog. EP has a substantially higher flash point and lower vapor pressure at room temperature than MTBE, making it safer to use in the hospital environment. Table 1 compares its properties to those of MTBE. Its LD50 is somewhat higher than that of MTBE, and it is less toxic when given intraperitoneally, intravenously, or intraduodenally (62–64). EP appears to have a less severe toxicity profile mainly because it is rapidly metabolized. It is hydrolyzed by esterases into ethanol and propionic acid (69) and rapidly metabolized on first pass through the liver, hence it was not detected in blood when introduced into the gallbladder of piglets (63). In contrast, MTBE is excreted mostly unchanged through the lungs and its metabolites, formic acid and methanol, are relatively more toxic (62). When infused into the duodenum of rats, it caused significantly less inflammation than MTBE (64), and functional studies on the rats' intestines showed less cytotoxicity with EP than MTBE (60). These favorable properties indicate that EP is a more promising solvent for the topical dissolution of gallstones. A recent preliminary study on gallstone patients in San Diego indicated that EP is more tolerated than MTBE. None of the patients treated had any detectable blood levels of EP or its metabolite, ethanol, and none experienced any foreign smell or taste during EP irrigation of the gallbladder. There were no changes in the patients' liver function tests or hematological parameters (70). As yet unpublished data from the authors' center seem to support this. IX— Endoscopic Retrograde Gallbladder Cannulation Although simple and safe, the procedure of percutaneous cholecystostomy is invasive by definition. This has led several investigators to evaluate the procedure of percutaneous endoscopic retrograde cannulation of the gallbladder for delivering gallstone solvents. Foerster et al. described a catheter with a specially formulated tip that may be used through the standard sideviewing duodenoscope during the procedure of endoscopic retrograde cholangiography to anchor the cystic duct's opening into the common bile duct (71). A slippery guidewire over which the dissolution catheter may be introduced into the gallbladder, is then used to negotiate the cystic duct. The initial reports have been encouraging, but several drawbacks exist. The long
Page 560
catheter has a large dead space, which can impede effective solvent exchange, especially in very small gallbladders. Once the catheter is in place, manipulation of the catheter pigtail loop in the gallbladder is difficult. Such manipulation is frequently necessary to place the catheter tip near the stones so as to achieve efficient and speedy dissolution. Most importantly, however, the procedure of endoscopic retrograde cholangiography may not be safer than percutaneous cholecystostomy. It requires conscious sedation, is very uncomfortable to the patient, and may be associated with a modest number of complications, some of which include pancreatitis, intestinal or biliary tract perforation, throat trauma, aspiration, and pneumonia. In contrast, none of these complications are known to occur with percutaneous cholecystostomy, which, in addition, does not require conscious sedation, since it is not uncomfortable to the patient. Controlled studies will be needed to evaluate these techniques. X— Gallstone Recurrence after Topical Dissolution Since gallstone dissolution preserves the gallbladder and does nothing to alter the defect that led to the gallstones in the first place, gallstone recurrence is both a theoretical and real possibility. Longterm followup studies after gallstone dissolution with oral bile acids have shown that recurrence is, on average, 10% per year for the first 5 years to a cumulative recurrence rate of 50%. If the gallstones have not recurred within the first 5 years, they are unlikely to do so (72). Recurrence is probably the most persuasive argument in favor of surgical therapy. However, recurrent stones seem to follow the same natural history of gallstone disease, and most patients with recurrent stones remain asymptomatic (73). Oral bile acid therapy will prevent recurrence (74), but this may not be very practical due to the expense and the need for lifelong administration. Also, it may not be reasonable to commit all patients to it when only 50% will have recurrence. Nonsteroidal antiinflammatory agents have been shown in sporadic studies to help (75), but their actual effectiveness remains to be proven, and it appears that the dose needed is relatively high. Recurrent stones are generally of the same type of the original stones (76). Hence, they will respond again to dissolution therapy. A good strategy is to wait and see which patients will develop recurrent stones and place only those who are at high surgical risk on longterm oral bile acid therapy. With recent advancements in topical dissolution, whereby endoscopy is used to leave the gallbladder clean of all residue, it is believed that recurrence will be significantly less. Preliminary data have shown that the rate of recurrence in patients who had no remaining residue as assessed endoscopically appears to be significantly less than that in those who were treated in the same fashion but without endoscopic verification; actuarial cumulative recurrence at 4 years was 15 versus 40%, respectively (77). It remains to be seen if these percentages will hold as larger number of patients are treated. Percutaneous gallbladder ablation by chemical sclerosis was advocated as a means of functionally eliminating the gallbladder and hence preventing recurrence. The procedure, as described by Becker et al. (78), involves the instillation of a mucosal sclerosant such as absolute alcohol, tetracycline, or hot saline through a percutaneous gallbladder catheter to sclerose and ablate the gallbladder in situ. First, however, obstruction of the cystic duct is induced using a percutaneous coagulation probe to prevent inadvertent spillage of sclerosants into the common bile duct and to minimize the possibility of subsequent recanalization of the gallbladder. This technique is most suitable after percutaneous gallstone dissolution because of the already present percutaneous catheter, through which the sclerosis may be performed. The initial patient experience as reported by Becker et al. has shown that the technique is safe and initially successful (79), but further studies will be needed to prove its longterm efficacy. A philosophical argument against gallbladder ablation is the fact that one of the main aims of topical dissolution therapy is to preserve gallbladder function. A significant improvement in gallbladder function has been observed after elimination of gallstones by topical dissolution (80). Although the loss of the gallbladder is generally considered to be benign, a
Page 561
number of studies are emerging to prove otherwise. About onethird of patients with no gallbladder will suffer from diarrhea, which in 12% will be severe enough to require medical attention (81). This is confirmed in a more recent study of patients after cholecystectomy, 40% of whom reported a greater than 100% increase in bowel movements and 18% of whom reported diarrhea (82). Although still controversial, there are data to indicate that there may be about a twofold increase in the rate of the development of rightsided colon cancer in patients with no gallbladder (83). These effects are probably a consequence of loss of the reservoir for the bile acid pool in the body and shift of this reservoir to the intestine. XI— The Future Topical dissolution has clear advantages in terms of being a true outpatient nonsurgical procedure, not requiring general anesthesia, suitable for highrisk patients, and preserving the organ. With the continued advances in its apparatus and techniques, it is conceivable that it may replace cholecystectomy for the treatment of gallstone disease in many patients. The automated solvent delivery system will mean that several patients may be treated at the same time, leading to the possible establishment of outpatient centers for this purpose. In theory, automated topical dissolution should be much less expensive than cholecystectomy since there are no costs for an operating room, general anesthesia, an anesthesia team, or hospitalization. In the highrisk patients who in general run a complicated course with surgery, the cost savings would be enormous. The development of safe and effective solvents for noncholesterol stones (84,85), will make it more applicable and further reduce its cost because preprocedural screening would be unnecessary. The near future will undoubtedly show a great deal of activity in this area. Acknowledgment The author wishes to thank Annette Weiss, RN, for her editorial review. References 1. Walker JW. The removal of gallstones by ether solution. Lancet 1891; 1:874–875. 2. Pribram BDC. The method for dissolution of common duct stones remaining after operation. Surgery 1947; 22:806–818. 3. Althan O, Kohler R. Ether treatment of retained postoperative biliary tree stones. Acta Chir Scand 1959; 116:437–449. 4. Best RR, Rasmussen JA, Wilson CE. Management of remaining common duct stones by various solvents and biliary flushing regimen. Arch Surg 1953; 67:839– 853. 5. Romero R, Butterfield WC. Heparin and gallstones. Am J Surg 1974; 127:687–688. 6. GarciaRomero E, LopezCantarero M, Quesada A, Arcelus IM. The nonoperative removal of retained common duct stones after biliary surgery with clofibrate. J Surg Res 1979; 26:129–133. 7. Cheung LY, Englert E, Moody FG, Wales EE. Dissolution of gallstones with bile salts, lecithin, and heparin. Surgery 1974; 76:500–503. 8. Toouli J, Jablonskin P, Watts JMcK. Dissolution of human gallstones: the efficacy of bile salt, bile salt plus lecithin and heparin solutions. J Surg Res 1975; 19:47– 53. 9. Catt PB, Hogg DF, Chenic GJA, Hardie IR. Retained biliary calculi: removal by a simple nonoperative technique. Ann Surg 1974; 180:247–251. 10. Castleden WM. Retained common bile duct calculi. Br J Surg 1976; 63:47–50.
Page 562
11. Way LW, Admirand WH, Dunphy JE. Management of choledocholithiasis. Ann Surg 1972; 176:347–359. 12. Mack E, Saito C, Goldfarb S, et al. Local toxicity of Ttube infused cholate in the rhesus monkey. Surg Forum 1977; 28:408–409. 13. Motson RW. Dissolution of common bile duct stones. Br J Surg 1981; 68:203–208. 14. Flynn GL, Shah Y, Prakongpan S, Kwan KH, Higuchi WI, Hofmann AF. Cholesterol solubility in organic solvents. J Pharm Sci 1979; 68:1090–1097. 15. Abranowicz M, ed. Monooctanoin for gallstones. Med Lett Drugs Ther 1987; 29:52. 16. Palmer KR, Hofmann AF. Intraductal monooctanoin for the direct dissolution of bile duct stones: experience in 343 patients. Gut 1986; 27:196–202. 17. Asakawa S, Igimi H, Shimura H. Development of a new solution for use through an endoscopically placed nasobiliary tube. Hepatology 1983; 3:810. 18. Leuschner U, Baumgartel H, Wurbs D. Auflosung von CholesterinGallenganssteinien mit einer modifizierten Capmul 8210Emulsion und einer EDTA Gallensalzlosung. Leber Magen Darm. 1980; 10:284–287. 19. Smith BF, LaMont JT. Identification of gallbladder mucinbilirubin complex in human cholesterol gallstone matrix: effects of reducing agents on in vitro dissolution of matrix and intact gallstones. J Clin Invest 1985; 76:439–445. 20. Bogardus JB. Importance of viscosity in the dissolution rate of cholesterol in monooctanoin solutions. J Pharm. Sci 1984; 73:906–908. 21. Allen MJ, Borody TJ, Bugliosi TF, May GR, LaRusso NF, and Thistle JL. Cholelitholysis using methyl tertiary butyl ether. Gastroenterology 1985; 88:122–125. 22. vanSonnenberg E, Zakko SF, Hofmann AF, D'Agostino HB, Jinich H, Hoyt DB, Moossa AR, Ramsby GR. Human gallbladder morphology after gallstone dissolution with methyl tertbutyl ether. Gastroenterology 1991; 100:1718–1723. 23. Lameris JS, Jeekel J, Havelar IJ, Von Seyen AJ. Percutaneous transhepatic cholecystostomy. Fortschr Roentgenstr 1985; 142:80–82. 24. vanSonnenberg E, Casola G, Varney RR, Zakko S, Wittich GR, Cox J, Hofmann AF. Interventional radiology in the gallbladder. Radiographics 1989; 9:39–49. 25. Allen MJ, Borody TJ, Bugliosi TF, May GR, LaRusso NF, and Thistle JL. Rapid dissolution of gallstones in humans using methyl tertbutyl ether. N Engl J Med 1985; 312:217–220. 26. vanSonnenberg E, Hofmann AF, Neoptolemus J, Wittich GR, Princenthal RA, Wilson SW. Gallstone dissolution with methyl tertbutyl ether via percutaneous cholecystostomy: success and caveats. AJR 1986; 146:865–867. 27. Thistle JL, May GR, Bender CE, Williams HJ, KeRoy AJ, Nelson PE, Peine CJ, Petersen BT, McCullough JE. Dissolution of cholesterol gallbladder stones by methyl tertbutyl ether administered by percutaneous transhepatic catheter. N Engl J Med 1989; 320:633–639. 28. Leuschner U, Hellstern A, Schmidt K, Fischer H, Leuschner M. Gallstone dissolution with methyl tertbutyl ether in 120 patients—efficacy and safety. Dig Dis Sci 1991; 36:193–199. 29. Zakko SF, Hofmann AF. Microprocessorassisted solvent transfer system for gallstone dissolution: in vitro and in vivo validation. Gastroenterology 1990; 99:1807–1813. 30. Cappa JA, Zakko SZ, Nitzberg MC, Ramsby GR, Chen HH, Guttermuth CF, Srb SM. Significant improvement in gallbladder (GB) motor function one week after gallstone elimination by percutaneous dissolution. Gastroenterology 1993; 104:A351. 31. Long CA, Teplick SK, Brandon JC, Harb GH, Yan K, Baker ML. Effects of gallstone solvents on commonly used catheters. J Vasc Intervent Radiol 1994; 5:479–484. 32. Leuschner U, Hellstern A, Ansell A, Gatzen M, et al. Manual and automatic gallstone dissolution with methyl tertbutyl ether. Dig Dis Sci 1994; 39:1302–1308. 33. Zakko S, Ramsby G. Pressure and flow dynamics in the human gallbladder: observations during automatic computerized solvent litholysis (ACSL) with MTBE. Proceedings of
Page 563
the Second International Meeting on Pathochemistry, Pathophysiology and Pathomechanics of the Biliary System, Bologna, Italy, March, 1990. 34. Ponchon T, Baroud J, Pugol B, Valette PJ, Parrot D. Renal failure during dissolution of gallstones by methyl tertbutyl ether (letter). Lancet 1988; 2:176–177. 35. Small DM: Cholesterol nucleation and growth in gallstone formation (editorial). N Engl J Med 1980; 302:1305. 36. Brakel K, Lameris JS, Nijs HG, Terpstra OT, Steen G, Blijenberg BC. Predicting gallstone composition with CT: In vivo and in vitro analysis. Radiology 1990; 174:337–341. 37. Lowenfels AB, Lindstrom CG, Conway MJ, Hastings PR. Gallstones and risk of gallbladder cancer. J Natl Cancer Inst 1985; 75:77–80. 38. Nave CR, Nave BC. Physics for the Health Sciences. 2nd ed. Philadelphia: Saunders, 1980. 39. Lichtenstein ME, Ivy AC. The function of the ''valves" of Heister. Surgery 1937; 1:38. 40. Otto WJ, Scott GW, Rodkiewicz CM. A comparison of resistances to flow through the cystic duct and the sphincter of Oddi. J Surg Res 1979; 27:68–72. 41. Lanzini A, Jazrawi RP, Northfield TC. Simultaneous quantitative measurements of absolute gallbladder storage and emptying during fasting and eating in humans. Gastroenterology 1987; 92:852–861. 42. Higuchi WI, Sjuib F, Mufson D, Simonelli AP, Hofmann AF. Dissolution kinetics of gallstones: physical model approach. J Pharm Sci 1973; 62:942–945. 43. Steinberger RL, Treyball RE. Mass transfer from a solid soluble sphere to a flowing liquid stream. J Am Inst Chem Eng 1960; 6:227–232. 44. Zakko SF, Hofmann AF. Microprocessorassisted solvent transfer system for effective contact dissolution of gallbladder stones. IEEE Trans Biomed Eng 1990; 37:410–416. 45. Baron RL, Kuyper SJ, Lee SP, Rohrmann CA, Jr, Shuman WP, Nelson JA. In vitro dissolution of gallstones with MTBE: correlation with characteristics at CT and MR imaging. Radiology 1989; 173:117–121. 46. Brink JA, Boston MA, Mueller PR, Simeone JF, Prien EL, Saini S, Tung G, Ferrucci JT. Limitations of CT in the prediction of gallstone composition: in vitro study and review. Radiology 1989; 173:246. 47. Scirica JC, Guttermuth CF, Zakko SF. MTBE dissolution of cholesterol gallstones via catheter is poorly predicted by the stone's cholesterol content. Gastroenterology 1991; 100:A339. 48. Sarva RP, Farivar S, Fromm H, Poller W. Study of the sensitivity and specificity of computerized tomography in the detection of calcified gallstones which appear radiolucent by conventional roentgenography. Gastrointest Radiol 1981; 6:165–167. 49. Hickman MS, Schwesinger WH, Bova JD, Kurtin WE. Computed tomographic analysis of gallstones: an in vitro study. Arch Surg 1986; 121:289–291. 50. Baron RL, Rohrmann CA, Jr, Lee SP, Shuman WP, Teefey SA. CT evaluation of gallstones in vitro: correlation with chemical analysis. AJR 1988; 151:1123– 1128. 51. Zakko SF, Moore D, Guttermuth C, Romano D, Ramsby G. A CT index for selection of gallstones amenable to percutaneous automatic computerized solvent litholysis (ACSL) with methyl tertbutyl ether (MTBE) using a microprocessorassisted solvent transfer (MST) system. Radiology 1989; 173:421. 52. Malkin M, Zakko SF, Guttermuth CF, Srb S, Romano D, Rashid S, Ramsby GR. A CTscan index highly predictive of gallstones (GS) amenable to contact dissolution with MTBE administered via catheter: in vivo validation. Gastroenterology 1991; 100:A770. 53. Zakko SF, Ramsby GR, Srb SM, Guttermuth CF. Automatic computerized solvent litholysis (ACSL) for gallbladder stones: experience with methyl tertbutyl ether (MTBE) and a microprocessorassisted solvent transfer (MST) system. Gastroenterology 1990; 98:A647. 54. Zakko SF, Rashid S, Ramsby GR. Diagnostic percutaneous cholecystoscopy after nonsurgical treatment of gallstones. Gastrointest Endosc Clin North Am 1991; 1:127–136.
Page 564
55. Zakko SF, Srb S, Ramsby GR. Sensitivity of percutaneous endoscopy compared with ultrasonography in the detection of residue or mucosal lesions after topical gallbladder stone dissolution. Gastroint Endosc 1995; 42:434–438. 56. MacGillivray DC, Zakko SF, Siegenthaler MP, Ramsby GR. Early carcinoma of the gallbladder diagnosed by percutaneous cholecystoscopy. Gastroint Endoscopy 1997; 45:207–210. 57. Zakko SZ, Ramsby GR, Chen H, Srb S, Guttermuth C. A comprehensive procedure for contact dissolution of gallbladder stones. Proceedings of the World Congresses of Gastroenterology, Los Angeles, CA, October 7, 1994. 58. Hofmann AF, Schteingart CD, Zakko SF, Hajjar JJ, Cohan J, Esch O, Breslin K, Singh E, Lillienau J. C5 esters: New solvents for contact dissolution of cholesterol gallstones (GS). Proceedings of the World Congresses of Gastroenterology, Sydney, Australia, August 30, 1990. 59. Hofmann AF, Schteingart CD, vanSonnenberg E, Esch O, Zakko SF. Contact dissolution of cholesterol gallstones with organic solvents. Gastroenterol Clin North Am 1991; 20:183–199. 60. Zakko SF, Scirica JC, Guttermuth MC, Dodge J, Hajjar JJ. Ethyl propionate is more effective and less cytotoxic than methyl tertbutyl ether for topical gallstone dissolution. Gastroenterology 1997; 113:232–237. 61. Opdyke, DLJ. Monographs on fragrance raw materials. Food Cosmet Toxicol 1978; 16:749–750. 62. Esch O; Spinosa JC; Hamilton RL; Crombie DL, et al. Acute effects of topical methyl tertbutyl ether or ethyl propionate on gallbladder histology in animals: a comparison of two solvents for contact dissolution of cholesterol gallstones. Hepatology 1992; 16:984–991. 63. Esch O, Schteingard CD, Pappert D, Kirby D, et al. Increased blood levels of methyl tertbutyl ether but not of ethyl propionate during instillation with contact gallstone dissolution agents in the pig. Hepatology 1993; 18:373–379. 64. Clerici C, Gentili G, Zakko SF, Baló S, Miglietti M, Giansanti M, Modesto R, Guttermuth CF, Morelli A. Local and systemic effects of intraduodenal exposure to the topical gallstone solvents ethyl propionate and methyl tertbutyl ether in the rabbit. Dig Dis Sci 1997; 42:497–502. 65. National Technical Information Service, U.S. Department of Commerce [PB87174603]. 66. Acute Toxic Data 1992; 1:174. 67. Akimoto R, Rieger E, Moossa AR, Hofmann AF, Wahlstrom HE. Systemic and local toxicity in the rat of methyl tertbutyl ether: a gallstone dissolution agent. J Surg Res 1992; 53:572–577. 68. Food Cosmet Toxicol 1978; 16:749. 69. Browning E. Esters. In: Toxicity and Metabolism of Industrial Solvents. Amsterdam: Elsevier, 1965, pp 522–591. 70. Hofmann AF, Amelsber A, Esch O, Schteingart CD, Lyche K, Jinich H, VanSonnenberg E, D'Agostino HB. Successful topical dissolution of cholesterol gallbladder stones using ethyl propionate. Dig Dis Sci 1997; 42:1274–1282. 71. Foerster EC, Matek W, Domschke W. Endoscopic retrograde cannulation of the gallbladder: direct dissolution of gallstones. Gastroint Endosc 1990; 36:444– 450. 72. Ruppin DC, Dowling RH. Is recurrence inevitable after gallstone dissolution by bileacid treatment? Lancet 1928; 181–185. 73. Lanzini A, Jazrawi, Kupfer RM, Maudgal DP, Joseph AEA, Northfield TC,. Gallstone recurrence after medical dissolution: an overestimated threat? J Hepatol 1986; 3:241–246. 74. Marks J, Lan SP et al. Lowdose chenodiol to prevent gallstone recurrence after dissolution therapy. Ann Intern Med 1984; 100:376–381. 76. Pereira SP; Hussaini SH; Kennedy C; Dowling RH. Gallbladder stone recurrence after medical treatment: do gallstones recur true to type? Dig Dis Sci 1995; 40:2568–2575.
Page 565
77. Zakko SF, Ramsby GR, Guttermuth CM, Weiss AM. The impact of percutaneous cholecystoscopy and residual debris on recurrence after topical gallbladder stone dissolution. Gastroenterology 1996; 110:A481. 78. Becker CD, Quenville NF, Burhenne HJ. Gallbladder ablation through radiologic intervention: an experimental alternative to cholecystectomy. Radiology 1989; 171:235–240. 79. Becker CD, Fache JS, Malone DE, Stoller JL, Burhenne HJ. Ablation of the cystic duct and gallbladder: clinical observations. Radiology 1990; 176:687–690. 80. Cappa JA, Zakko SF, Nitzberg MC, Ramsby GR, Chen HH, Guttermuth CF, Srb SM. Significant improvement in gallbladder motor function one week after gallstone elimination by percutaneous dissolution. Gastroenterology 1993; 104:A351. 81. Fort JM, Azpiroz F, Casellas F, Andreu J, Malagelada JR. Bowel habit after cholecystectomy: physiological changes and clinical implications. Gastroenterology 1996; 111:617–622. 82. Zakko SF, Guttermuth MC, Jamali SH, Estill M, Nasir L, Do SC, Weiss AM. A population study of gallstone composition, symptoms, and outcomes after cholecystectomy. Gastroenterology 1991; 116:A43. 83. Giovannucci E, Colditz GA, Stampfer MJ. A metaanalysis of cholecystectomy and risk of colorectal cancer. Gastroenterology 1993; 105:130–141. 84. Wosiewitz U, Guldutuna S, Fischer H, Leuschner U. Pigment gallstone dissolution in vitro: solubilization of brown bilirubinate and black polybilirubinate stone material by buffered solvents containing ethylenediaminetetraacetic acid, bile salts, and reducing thiols. Scand J Gastroenterol 1989; 24:373–380. 85. Guttermuth CF, Zakko SF, Malkin M, Petruff C. Successful contact dissolution of noncholesterol gallstones (GS) using chelosol in alternation with MTBE: an in vitro controlled study. Gastroenterology 1992; 102:A314.
Page 567
27— Common Bile Duct Stones Tony C.K. Tham Ulster Hospital, Belfast, Northern Ireland David R. Lichtenstein Boston University School of Medicine, Boston, Massachusetts I— Introduction Common bile duct (CBD) stones are found in 10 to 20% of patients undergoing cholecystectomy. Optimal management of these individuals with calculous biliary tract disease continues to require an integrated approach between endoscopists, surgeons, and interventional radiologists. Treatment selection is dependent on factors that include the clinical scenario, success and complication rates for the different techniques, as well as institutional expertise and availability. Currently, endoscopic management is preferred for extracting common duct stones in most clinical situations. Endoscopic management of choledocholithiasis was initially considered justifiable only in elderly postcholecystectomy patients with recurrent or retained CBD stones who were at high risk for serious complications from conventional surgical CBD exploration. With improved efficacy, demand, and training in therapeutic biliary endoscopic techniques, however, there has been an expanding role for the endoscopic management of calculous biliary tract disorders. Based on current evidence, there seems to be little doubt that endoscopic therapy is the treatment of choice for elderly patients with bile duct stones regardless of their clinical presentation or gallbladder status and in patients of all ages presenting with cholangitis or severe gallstone pancreatitis. Furthermore, early postoperative patients with retained stones can safely be treated by endoscopic sphincterotomy without the need to await maturation of a Ttube track. The role of ERCP continues to evolve in the new era of minimally invasive surgery, where a number of acceptable treatment algorithms have been proposed to manage patients undergoing laparoscopic cholecystectomy suspected of having CBD stones. II— Classification and Pathogenesis CBD stones are classified according to their site of formation. Primary bile duct stones arise de novo in the bile ducts and secondary bile duct stones form in the gallbladder and subsequently migrate into the bile duct (1). In the western world, secondary bile duct stones predominate, as 80 to 95% of patients with common duct stones have concomitant gallbladder stones (2). The composition of secondary bile duct stones reflects the composition of gallstones, with about 80% being of cholesterol origin. Black pigment stones also form in the gallbladder but infrequently migrate into the CBD.
Page 568
Primary bile duct stones are composed primarily of calcium bilirubinate and have a cholesterol content less than that of gallbladder stones. They are light brown, greenish brown, or black and soft, frequently laminated, and easily crushed to form "biliary mud" (3). They are often referred to as brown pigment stones or bile pigment calcium stones, in contrast to the blackpigmented bilirubinate or the yellow cholesterol stones that originate in the gallbladder (4). The precise pathogenesis of primary bile duct stones is unknown, but bacterial or parasitic infection (5), bile stasis (6), diet (7), foreign material in the duct (8), and juxtapapillary diverticula (9) all contribute to their formation. The most common organisms cultured from primary bile duct stones include the enteric organisms Escherichia coli, Klebsiella, Bacteroides, and Clostridium species (5). III— Clinical Presentation The clinical presentation of bile duct stones is broad and may range from asymptomatic to complications of biliary colic, jaundice, cholangitis, or pancreatitis. Approximately 1 to 5% of patients with symptomatic cholelithiasis without predictors of CBD stones will have asymptomatic choledocholithiasis. The natural history of asymptomatic CBD stones, although incompletely understood, appears to be less innocuous than that of asymptomatic gallstones. Thus the discovery of CBD stones, even when incidental, usually warrants active intervention to remove the stones—in contrast to the incidental finding of gallbladder stones, which can be followed expectantly (10). Johnson and Hosking reported greater than 50% of patients with retained bile duct stones developing symptoms and 25% developing resultant serious complications (11). However, a recent study suggests a more indolent course for asymptomatic CBD stones. Patients undergoing cholecystectomy without risk factors for choledocholithiasis were randomized to cholecystectomy or cholecystectomy combined with intraoperative cholangiography. CBD stones were discovered in 12% of patients in the cholangiography group. It was assumed that a similar percentage of patients in the group not undergoing cholangiography had stones, and none developed symptoms during a 3year followup period (12). CBD stone passage is felt to be dependent on size, with smaller stones having a greater propensity to migrate spontaneously (13). The pain of symptomatic bile duct stones is indistinguishable from that of symptomatic cholelithiasis. The pain is caused by sudden obstruction and distention of the bile duct, producing increased intrabiliary pressure. The pain is typically located in the epigastric region or right upper quadrant of the abdomen, often radiating to the flank or back. The term colic is a misnomer, because the pain is constant in nature. It is commonly accompanied by nausea and vomiting, unassociated with posture or gas passage, and typically lasts for 30 rain to several hours. Food ingestion may precipitate an attack of pain, but the composition of a meal is not a proven factor. Stone impaction in the distal CBD may lead to jaundice, cholangitis, or gallstone pancreatitis. The serum bilirubin level is usually elevated to less than 15 mg/dL and the alkaline phosphatase level may be normal or elevated to several times normal. Longstanding bile duct obstruction can rarely lead to secondary biliary cirrhosis, although an incidence of biliary cirrhosis in patients operated on for bile duct stones as high as 8% has been reported (14). If a stone obstructs the bile duct, intraductal infection may ensue, leading to acute cholangitis. Charcot's triad of fever, pain, and jaundice occurs in about 75% of such individuals (15). These classic features are not uniformly present, and a high index of suspicion must be maintained, especially in elderly patients, where more subtle presentations are common (15,16). IV— Diagnosis The diagnosis of CBD stones can be challenging and has become of increased importance with the establishment of laparoscopic cholecystectomy as the preferred treatment for symp
Page 569
tomatic cholelithiasis. A preoperative diagnosis is desirable. Transabdominal ultrasonography, computed tomographic (CT) scanning, cholescintigraphy, endoscopic retrograde cholangiopancreatography (ERCP), endoscopic ultrasonography (EUS), magnetic resonance imaging (MRI) cholangiography, intravenous cholangiography (17), intraoperative cholangiography, and intraoperative bile duct palpation have all been assessed for confirming the diagnosis of choledocholithiasis. In expert hands, the sensitivity of ERCP for detecting bile duct stones approaches 100%. Ultrasonography has a sensitivity of 50 to 75% for detecting choledocholithiasis, since gas in the duodenum can create an acoustic interference that obscures visualization of the distal CBD (18). The sensitivity of CT scanning for detecting choledocholithiasis is similar to that of ultrasonography (19); hence, direct cholangiography, transhepatic and endoscopic, is often used in patients when there is a strong suspicion of choledocholithiasis. An advantage of ERCP is the immediate therapeutic capability for those with confirmed CBD stones. Recently, MRI cholangiography (Fig. 1) (20,21) and endoscopic ultrasonography (Fig. 2) (22–25) have emerged as less invasive diagnostic alternatives to ERCP. With MRI cholangiography, no contrast material is administered, but native high signal intensity of fluid on T2weighted images permits imaging of the biliary tree. EUS combines endoscopy with realtime, highresolution ultrasonography. Unlike transabdominal ultrasonography, it provides excellent sonographic visualization of the extrahepatic biliary tree without interference from bowel gas or abdominal fat. Preliminary studies have found the sensitivity of MRI cholangiography and EUS for detecting choledocholithiasis to be higher than that of transabdominal ultrasonography or abdominal CT scanning, with detection rates of about 90 to 95% (20–25)—an accuracy comparable to that of ERCP for diagnosing choledocholithiasis. The clinical utility of these newer technologies remains to be determined, as both EUS and MRI cholangiography have no therapeutic potential; therefore the indication for these imaging techniques will likely be used to exclude
Figure 1 Magnetic resonance cholangiogram shows choledocholithiasis. CBD, common bile duct; PD, pancreatic duct; S, stones.
Page 570
Figure 2 Endoscopic ultrasonography shows two stones 4 to 5mm in diameter (arrowheads) without an acoustic shadow in the common bile duct.
stones so as to reduce the risks and costs of unnecessary diagnostic ERCP. Proposed strategies suggest utility for EUS and MRI cholangiography in individuals with a high acuity of illness who are at risk for an invasive ERCP procedure, following failed ERCP, in the pregnant patient, or for individuals with a low to intermediate likelihood for CBD stones where the likelihood for therapeautic necessity is low, thereby averting an invasive diagnostic evaluation (24,26,27). There is an extensive endoscopic and surgical literature to help us stratify patients with gallstones into those at low (less than 10%), intermediate (10 to 30%) or high (greater than 30%) risk for having CBD stones (28,29). The proven risk factors include the presence of obstructive jaundice, cholangitis, a dilated bile duct on imaging, elevated liver function tests (LFTs), and gallstone pancreatitis (28–32). Interestingly, recent uncomplicated gallstone pancreatitis is only of intermediate risk (10 to 20%), presumably because the offending bile duct stone has usually passed into the duodenum by the time the ERCP is performed (28,31,33). The prevalence of CBD stones also increases with age. In patients younger than 60 years of age, the prevalence of concomitant CBD stones removed at cholecystectomy is 4 to 7%, but this increases to 13 to 18% for those 60 to 79 years of age and to 33% for those above 80 years of age (1). V— Management Optimal management of individuals with calculous biliary tract disease continues to require an integrated approach among endoscopists, surgeons, and interventional radiologists. Selection is dependent on factors that include the clinical scenario, success and complication rates for the different techniques, as well as institutional expertise and availability. These techniques are discussed below, emphasizing management of specific clinical situations.
Page 571
A— Endoscopic Therapy There is extensive experience with endoscopic management of CBD stones. At present ERCP is the preferred technique for extracting common duct stones in most clinical situations. Prior to ERCP, coagulopathy should be corrected whenever possible. Prophylactic antibiotics are administered 1 h prior to biliary procedures when there is evidence of biliary obstruction (34), and highlevel disinfection of endoscopic equipment is essential for minimizing procedurerelated cholangitis. Conscious sedation of the patient during the procedure is established using a combination of intravenous medications, such as midazolam, demerol, fentanyl, and pethidine. In order to facilitate bile duct stone extraction, the biliary sphincter is incised with a sphincterotome utilizing a technique referred to as biliary papillotomy or sphincterotomy (Fig. 3). Selective cannulation of the CBD that is confirmed fluoroscopically is a fundamental requirement for sphincterotomy. In experienced hands, this can be achieved in about 95% of patients. Using a sphincterotome, a vertical incision is made beginning at the papillary orifice and extending in a cephalad direction along the intramural course of the common bile duct for a variable length depending on local anatomy, common bile duct diameter, and the size of the stone to be removed. Following biliary sphincterotomy, common bile duct stones are extracted using a Dormiatype basket (Fig. 3) or a Fogartytype balloon catheter. These methods are successful in clearing the bile duct of CBD stones in over 90% of patients (33,35). 1— Difficult Bile Duct Stones The most difficult circumstances encountered during endoscopic bile duct stone removal result from technical difficulties with achieving deep biliary cannulation or from the inaccessibility
Figure 3 Endoscopic sequence for choledocholithiasis management. Top left: Endoscopic view of the intact papilla. Top right: A sphincterotome with a partially bowed wire in the bile duct. Intermittent controlled pulses of current are applied to direct the incision in the 11 or 12 o'clock position. Bottom left: Completed sphincterotomy with the biliary orifice visible at its apex. Bottom right: Stone extraction with a biliary basket.
Page 572
of the papilla as related to variant anatomy from periampullary diverticula, ampullary tumors, ampullary stenosis, or surgically created Billroth II or RouxenY reconstruction (Table 1). In these situations, the success of CBD cannulation can be enhanced by using various endoscopic accessories, including precut papillotomy (36). The technique of precut papillotomy, also termed access papillotomy, involves cutting the roof of the papilla with a needle knife or a specifically designed precut papillotome, thereby exposing the opening to the distal CBD. Precut papillotomy is indicated only as a method to achieve selective bile duct cannulation when access to the CBD is imperative for purposes of completing a therapeutic procedure, as the rate and severity of complications are modestly increased when compared with standard sphincterotomy (37,38). Following precut sphincterotomy, access to the CBD will be obtained in approximately 90% of cases (38). When cannulation fails, patient management alternatives include obtaining a cholangiogram by the percutaneous route; surgical exploration with intraoperative cholangiography; noninvasive bile duct imaging with MRI cholangiography or endoscopic ultrasonography; and a second attempt ERCP by the same endoscopist or at another institution (38,39). Following successful endoscopic sphincterotomy, a variety of factors may hinder stone extraction, including large stone size (greater than 15 mm), number (multiple), consistency (hard), shape (piston), and location. Stones located proximal to a narrowed distal CBD, above a stricture, in a sigmoidshaped bile duct, in the cystic or intrahepatic ducts, or adjacent to a T tube may prove difficult to remove. To facilitate extraction of large bile duct stones, several adjuvant techniques have been developed to reduce stone size prior to endoscopic removal. Mechanical lithotripsy remains the best initial option for large stones that cannot be removed by conventional techniques (Fig. 4). Mechanical lithotripters, which are modifications of standard Dormia baskets, are of greater tensile strength, allowing removal of 80 to 95% of difficult bile duct stones, which are otherwise refractory to standard extraction techniques (40,41). When mechanical lithotripsy fails, treatment with extracorporeal shockwave lithotripsy (ESWL) and intracorporeal techniques (laser or electrohydraulic) appears to be acceptable to achieve fragmentation and removal of common duct stones (42–46). The choice between these various lithotripsy methods largely depends on local expertise, as high success rates of 80 to 90% and low morbidity are reported in welltrained hands. Chemical dissolution of biliary stones has Table 1 Difficult Bile Duct Stones Unfavorable stone characteristics Stones greater than 15 mm in diameter Pistonshaped stones Multiple stones Unfavorable stone location Impacted stones Stones proximal to a biliary stricture Intrahepatic stones Tortuous bile duct Disproportionate size of the bile duct and stone Difficult access to the papilla Billroth II reconstruction Duodenal diverticulum RouxenY reconstruction Upper gastrointestinal stricture Ampullary neoplasm Papillary stenosis
Page 573
Figure 4 Left: ERCP showing a large retained common bile duct stone after cholecystectomy. Right: The bile duct stone is captured in a biliary basket and mechanical lithotripsy from closure of the device against the metallic sheath results in stone fragmentation.
been attempted by perfusing the common bile duct with solvents administered via an indwelling nasobiliary tube, percutaneous transhepatic catheter, cholecystostomy tube, or through existing T tube (47,48). The results with the semisynthetic vegetable oil monooctanoin (47) and the newer organic solvent methyltertbutyl ether (MTBE) (48) have been disappointing owing to incomplete stone dissolution and the potential for complications. In the 5% or less of situations where stone extraction is incomplete, a nasobiliary tube or stent should be inserted to maintain biliary drainage and prevent stone impaction in the distal common bile duct (49,50). This serves as a temporizing therapy, allowing for improvement in the patient's clinical condition until complete stone clearance is achieved either via additional endoscopic maneuvers or subsequent surgery. Maxton et al. (51) repeated ERCP examinations at 2 to 3month intervals to perform biliary stent exchanges and repeat attempts at stone extraction. Fifty of seventynine (63%) patients had successful stone clearance after two to five additional ERCPs. Endobiliary stent placement should not be considered a longterm solution to choledocholithiasis owing to the resulting high rate of complications (49,50), the exception being the nonsurgical candidate who has failed vigorous endoscopic attempts at stone extraction. Bergman et al. (50) placed biliary stents as permanent therapy in 58 patients following unsuccessful stone extraction. During a median 36month follow up, complications developed in 40%, with most related to cholangitis; there was 16% mortality from biliaryrelated causes. 2— Complications Early complications of endoscopic sphincterotomy and CBD stone extraction occur in approximately 5% of patients and procedurerelated mortality ranges from 0.2 to 0.4% (35,37,52). The complications include pancreatitis (2 to 4%), hemorrhage (1 to 2%), cholangitis (1%),
Page 574
cholecystitis (0.5%), and retroduodenal perforation (0.3%) (35,37,52). Significant risk factors for complications from endoscopic sphincterotomy include suspected sphincter of Oddi dysfunction as an indication for the procedure, the presence of cirrhosis, difficulty in cannulating the bile duct, access to the bile duct by ''precut" sphincterotomy, and the use of a combined percutaneousendoscopic procedure (37). The overall risk of complications is not related to the patient's age, number of coexisting illnesses, or diameter of the bile duct. Endoscopists who perform many sphincterotomies experience lower complication rates (35,37). Although there are many reports assessing shortterm morbidity following endoscopic sphincterotomy, longterm results are quite variable and difficult to interpret. Periods of observation have been relatively short, ranging from a mean of 1 to 18 years. In these studies, recurrent biliary problems have developed in 10 to 25% of individuals, including recurrent CBD stones (10%), biliary sphincter stenosis (1 to 2%), and, infrequently, nonobstructive cholangitis (53–61). These complications are often managed endoscopically. The factors predictive of symptom relapse include the presence of gallstones, large bile duct size, and periampullary diverticula (60,61). Gallbladder nonfilling during cholangiography was initially thought to contribute to symptom recurrence (26); however, this has not been confirmed in recent series (60). Only two studies have addressed longterm postsphincterotomy followup in young patients under 60 years of age (58,62). Bergman et al. (58) found that during 3 to 18 years follow up, 22 patients (24%) developed biliary complications, with recurrent bile duct stones noted in 13 (14%) patients and stenosis of the sphincterotomy in 9 (10%). One patient underwent surgery after failed endoscopic treatment, one patient died of cholangitis prior to planned ERCP, and the other patients were successfully managed endoscopically. Tham et al. (62) included patients with CBD stones or dysfunction of the sphincter of Oddi and followed them for 7 months to 12 years. Three (10%) patients developed further problems, which included biliary sphincter stenosis and cholangitis. Longterm risk from bacterial colonization of the biliary tree, leading to malignancy, has not been reported in these studies. 3— Alternative Endoscopic Techniques Alternatives to endoscopic sphincterotomy include balloon dilatation of the sphincter (63,64) and, less commonly, pharmacological relaxation of the sphincter of Oddi (65,66). The aim of these techniques is to preserve the biliary sphincter and thereby avoid longterm postsphincterotomy complications. Staritz et al. (65) were able to remove small bile duct stones 6 to 12 mm in diameter following pharmacological dilatation of the sphincter by glyceryl trinitrate administered sublingually. Followup manometric examinations showed the papillary function to be well preserved. A Japanese group utilized intravenous infusion of isosorbide dinitrate and were able to extract small CBD stones (most <10 mm) in 15 of 18 (83%) patients. Pancreatitis occurred in 1 (6%) patient who had unsuccessful stone extraction (66). There have been two prospective randomized trials comparing endoscopic sphincterotomy to endoscopic balloon dilation of the papilla (67,68). The Amsterdam group reported successful extraction of CBD stones in 90 of 101 (89%) patients following balloon dilation and similarly in 92 of 101 (91%) patients following endoscopic sphincterotomy. The median diameter of the stones was 10 mm (range, 3 to 36 mm). Biliary sphincterotomy was necessary in 9% of the balloon sphincteroplasty group, and mechanical lithotripsy was required more often in this group (31 versus 13%) (67). Early complications occurred in 17% of the balloon sphincteroplasty and 24% of the sphincterotomy patients. A preliminary randomized U.S. multicenter study (68) reported successful stone extraction in 80 of 85 (94%) patients following balloon dilatation versus 100% of 92 patients undergoing sphincterotomy. More complications occurred in the balloon dilatation group than in the sphincterotomy group (12 versus 1%), with 2 deaths (3%) occurring following balloon sphincterotomy as a result of necrotizing pancreatitis. Furthermore, the procedure time required for balloon dilatation was longer. Thus, although balloon sphincter dilation appears to be a promising alternative to sphincterotomy, the concerns raised by the U.S. study would suggest that it should not be used in routine clinical practice until further data become available.
Page 575
B— Surgery 1— Open Exploration of the Common Bile Duct Open exploration of the CBD can be performed through either the cystic duct, a supraduodenal choledochotomy, or a transduodenal approach. Transcystic bile duct exploration is possible only if the cystic duct is large enough to admit a choledochoscope. It may not be possible to remove large CBD stones by the transcystic approach even though access with the choledochoscope is achieved. In these situations, a supraduodenal choledochotomy is recommended. To perform a supraduodenal choledochotomy, the gallbladder is first removed and a vertical incision is made in the lower portion of the bile duct above the duodenum. The stones may be expressed immediately and additional stones retrieved by a combination of flushing, forceps, or ballooncatheter extraction techniques. A postexploratory assessment of the bile duct should be made either by cholangiography or preferably choledochoscopy to assess the completeness of stone removal. Omission of this step will lead to a higher incidence of retained CBD stones (69). Retained bile duct stones following surgery occurred in 4 to 16% of patients in earlier series, but this frequency is now substantially lowered by peroperative choledochoscopy, with most recent series reporting an incidence of 1 to 5% (70). The standard closure of the CBD is over a T tube. A Ttube cholangiogram is obtained on about day 10, and if the biliary tree is free of stones, the T tube can be removed. At the time of CBD exploration, a drainage procedure should be considered if there are multiple bile duct stones or retained CBD stones following cholecystectomy. This can be achieved by a formal choledochoduodenostomy (71–76). A potential complication of choledochoenterostomy is the occurrence of "sump syndrome," in which the portion of the CBD distal to the anastomosis acts as a sump to collect bile, stones, food, and other debris. This may result in intermittent partial or complete obstruction of the stoma, leading to bacterial proliferation and most commonly giving rise to pain and repeated episodes of cholangitis and, less frequently, pancreatitis (77). The incidence of the sump syndrome ranges from 0.4 to 2.8%; the main factors contributing to its development include a long sump, the presence of ampullary dysfunction or stenosis, and a small choledochoenterostomy. Transduodenal exploration of the CBD is best performed for stones impacted at its lower end that are not readily accessible to the supraduodenal route. The duodenum is mobilized by a Kocher's maneuver and the head of the pancreas is exposed. Once the papilla is identified, a grooved director is passed through the ampulla into the CBD and the ampulla is incised along its superior border. The duct can be explored from below with forceps, a balloon, or flushing catheter. The opened ampulla can be left unsutured as a sphincterotomy or a formal sphincteroplasty performed. Ttube drainage is probably unnecessary, as any retained stones should readily pass spontaneously into the duodenum. In the early 1980s, surgical series reported an operative mortality for open bile duct exploration of 4 to 10% for elective procedures, increasing to 20% in the emergency setting for elderly or highrisk patients (78). However, in 12 series published from 1988 to 1992, mortality in patients over age 70 ranged from 0 to 9%; in eight series, it was less than 4% (79). In comparison, complications from endoscopic sphincterotomy occur in 5 to 10%, with a mortality of 0.2 to 0.4% and no definable agerelated increase in risk (37). Two recent prospective randomized studies comparing bile duct clearance rates for endoscopic sphincterotomy versus open surgery showed success rates of 88% versus 94% (80) and 90% versus 90% (81), respectively. Only one of these studies specifically considered the elderly and highrisk patient presenting with jaundice, cholangitis, and acute pancreatitis (80). The procedures were presumably done in a nonurgent setting with comparable morbidity of 16% versus 23% and mortality of 6% versus 4% for endoscopic and surgical approaches, respectively. However the endoscopic morbidity and mortality in this study (80) are inexplicably much higher than previously reported (37,70). The limited data currently available suggest that the longterm results of endoscopic sphincterotomy are similar to those of open surgery; however, these series are not strictly
Page 576
comparable, as the spectrum of patients, definition of complications, and completeness of followup vary considerably. In addition, most surgical series report complications only for those patients who require additional surgery, which is most unusual after endoscopic therapy. Longterm series following surgical bile duct exploration report a complication rate of 14 to 36% (71–76). However, if a drainage procedure is also performed, such as a choledochoduodenostomy or sphincteroplasty, the longterm complication rates are reduced. Following a choledochoduodenostomy, the incidence of longterm complications is reported to be 3 to 7% (71,74,82,83); following sphincteroplasty, it is said to be 2 to 9% (73,74,83). 2— Laparoscopic Common Bile Duct Exploration (LCBDE) LCBDE can be performed at the time of cholecystectomy through the cystic duct or through a supraduodenal route analogous to open CBDE approaches. Laparoscopic extraction of stones through the cystic duct is indicated for small stones in the distal CBD up to 10 mm in size, which accounts for the vast majority. LCBDE can be performed under fluoroscopic control or by direct visual guidance. The visually guided technique requires dilatation of the cystic duct followed by the introduction of a flexible 10Fr choledochoscope threaded over a guidewire into the CBD. The stones can be removed under direct vision with a basket placed through the biopsy channel of the choledochoscope, by entrapping and extracting them through the cystic duct, or by passing them into the duodenum. The transcystic technique cannot be used in patients with stones greater than 5 to 8 mm in diameter or with small cystic ducts or low insertion of the cystic duct into the CBD because of the attendant risks of duct laceration. Laparoscopic choledochotomy is the preferred technique for those in whom the cystic duct cannot be used to gain access to the CBD or for those patients with multiple, large, or proximal stones. A verticaloblique incision is made on the anterior wall of the CBD and a flexible choledochoscope is inserted into it through the choledochotomy. The entire biliary tract is inspected and any stones found within it are trapped and removed under vision through the choledochotomy by means of a basket. Drainage of the bile duct is essential following this procedure, as transient impairment of transpapillary drainage due to edema is expected for several days (84,85). In a series of 274 patients undergoing attempted LCBDE, a transcystic approach was indicated in 68%, a supraduodenal approach in 26%, conversion to open surgery in 6%, and postoperative ERCP in 5% (86). The reported experience from laparoscopic bile duct stone extraction is limited. In nonrandomized studies, the overall success of laparoscopic bile duct stone extraction ranges from 87 to 98%, morbidity 4 to 17%, mortality 0 to 1%, and retained stones in 1 to 5% (87–99). The results of laparoscopic bile duct clearance have been compared to endoscopic sphincterotomy. In two prospective randomized trials, the singlestage laparoscopic technique of laparoscopic cholecystectomy and LCBDE was equally effective as the endoscopic approach of ERCP and laparoscopic cholecystectomy in clearing the duct of stones, had similar morbidity, but was associated with a significantly shorter hospital stay (100,101). Whether the surgeons performing both procedures had comparable expertise must be questioned due to lower than expected endoscopic success rates, potentially biasing the results in favor of the surgical approach. In the situation where the patient has had previous cholecystectomy, there have been no randomized comparisons of endoscopic sphincterotomy versus open or laparoscopic bile duct exploration, although, bile duct explorations postcholecystectomy are potentially more difficult to perform. C— Percutaneous Management Percutaneous transhepatic techniques for biliary stone extraction are preferred when endoscopic therapy has failed and for intrahepatic biliary calculi such as seen in oriental cholangiohepatitis (102,103). When endoscopic failure is the result of inability to gain deep cannulation of the bile duct, a combined percutaneousendoscopic "rendezvous" technique is often performed
Page 577
(104). A variety of methods have been described for this technique, but the most commonly utilized involves obtaining percutaneous access followed by cholangiography and placement of a 400cm guidewire through a catheter into the bile duct and ultimately into the duodenum. Within the duodenum, the endoscopist places a snare over the tip of the guidewire and withdraws it up the endoscope channel, whereby it is utilized in a standard fashion to complete the procedure. The principal advantage of the transhepatic approach to biliary intervention is the control that can be achieved for manipulations within the ductal system. Transhepatic catheterization is not limited by previous surgical bypass procedures. A diagnostic cholangiogram is initially obtained and a guidewire advanced into the biliary tree. Graded dilators are passed over the wire to enlarge the tract. This process requires successive procedures with progressive enlargement of the tract to approximately 10 Fr. Once the tract is mature, manipulation and extraction of calculi within the ductal system may be completed. Smallcaliber choledochoscopes with therapeutic channels are available for passage through the tract into the ductal system. Multiple sessions are often required to clear the system of calculi. When the cholangiogram reveals the absence of stones, percutaneous catheter access can be removed, allowing the tract to close spontaneously. The major disadvantage of the transhepatic approach is the morbidity associated with the capsular puncture and transparenchymal tract. D— Clinical Indications 1— Laparoscopic Cholecystectomy Laparoscopic cholecystectomy has now replaced open cholecystectomy as the procedure of choice in the vast majority of individuals with symptomatic cholelithiasis. Prior to the introduction of laparoscopic cholecystectomy, preoperative ERCP was not considered advantageous in the otherwise healthy patient with suspected common duct stones because ERCP followed by cholecystectomy was not superior to combined cholecystectomy, cholangiography, and open common bile duct exploration (105, 106). Open CBDE increased the duration of hospitalization and time to return to work by an acceptably small amount compared with open cholecystectomy alone. However, compared with laparoscopic cholecystectomy alone, the addition of open CBDE substantially lengthens hospitalization and back towork times. The development of laparoscopic cholecystectomy, with its minimally invasive strategy, has renewed interest in less invasive approaches to CBD stone detection and treatment, leading to an expanding array of available treatment options that includes ERCP and laparoscopic CBD exploration. Optimal management of individuals with calculous biliary tract disease continues to require an integrated approach between endoscopists, surgeons, and interventional radiologists. There is now a consensus in the medical community that endoscopic removal of CBD stones is preferable to surgery in (Table 2) (a) patients who have undergone previous cholecystectomy, (b) highrisk surgical patients when the gallbladder is still present, (c) patients with acute cholangitis, (d) selected patients with acute biliary pancreatitis, and (e) as an integrated approach for selected patients with suspected choledocholithiasis undergoing planned laparoscopic cholecystectomy. Some 5 to 15% of patients undergoing cholecystectomy will be found to have choledocholithiasis. Patients more likely (greater than 10 to 20%) to have CBD stones can be selected based on established predictors that include biliary ductal dilation, elevated liver function tests (LFTs), presence of CBD stones on imaging, cholangitis, and gallstone pancreatitis (Table 3) (29–32). Patients without predictors for CBD stones have less than a 5% prevalence of CBD stones at the time of cholecystectomy (30–32). These patients do not require routine cholangiography to detect bile duct stones or to define ductal anatomy, as aberrant ductal anatomy is infrequently identified and rarely contributes to operative bile duct injury. Selective cholangiography is currently favored for patients with predictors of choledocholithiasis. Three primary strategies have emerged for the detection and management of bile duct stone (Fig. 5).
Page 578 Table 2 Risk Factors for Choledocholithiasis High likelihood Bile duct stone on imaging Cholangitis Persistent jaundice Severe gallstone pancreatitis Intermediate likelihood Recent jaundice Mild gallstone pancreatitis Mild or transient LFT elevations Dilated CBD Low likelihood Normal LFTs Nondilated CBD No history of jaundice or pancreatitis Key: LFT, liver function test; CBD, common bile duct.
These include ERCP and stone clearance prior to performance of laparoscopic cholecystectomy, laparoscopic cholecystectomy with intraoperative cholangiography followed by LCBDE or postoperative ERCP, or open cholecystectomy with intraoperative cholangiography and common duct exploration. The choice is dependent on the likelihood of CBD stones and local institutional expertise. ERCP performed prior to the laparoscopic cholecystectomy is justified in order to avoid prolonging the operative procedure with intraoperative cholangiography while maintaining the minimally invasive approach (107). If the ERCP is unsuccessful, the surgeon has the option of referring the patient to a more experienced endoscopist, performing laparoscopic cholecystectomy with intraoperative cholangiography and LCBDE, or performing an open surgical procedure. The disadvantage of this strategy is that less than 40% of patients will have bile Table 3 Indications for Endoscopic Management of Choledocholithiasis Postcholecystectomy (no Ttube) Postcholecystectomy (Ttube) Failed Ttube tract extraction Symptomatic patient with immature Ttube tract Gallbladder in situ Elderly patient Patient at high operative risk Patients choosing nonoperative management of gallstones Selected patients undergoing laparoscopic cholecystectomy Prior to laparoscopic cholecystectomy in those at high likelihood for CBD stones Following laparoscopic cholecystectomy for those with a positive IOC Gallstone pancreatitis When concomitant cholangitis suspected Severe disease Acute cholangitis Key: CBD, common bile duct; IOC, intraoperative cholangiogram.
Page 579
Figure 5 Algorithm for management of choledocholithiasis in patients undergoing laparoscopic cholecystectomy. 1. High likelihood = jaundice or CBD stone on imaging. 2. Intermediate likelihood = transient increase in LFTs, dilated CBD, history of jaundice or pancreatitis. 3. Low likelihood = no indicators present. ERCP, endoscopic retrograde cholangiopancreatography; IOC, intraoperative cholangiogram; LCBDE, laparoscopic CBD exploration.
Page 580
duct stones on preoperative ERCP; therefore, the majority will undergo an "unnecessary" ERCP and will be subjected to the added expense and risk of ERCPrelated pancreatitis. Laparoscopic cholecystectomy is recommended within 2 days of ERCP, as longer delays may allow further stone passage from the gallbladder into the CBD. Based on decision analysis studies, this strategy of preoperative ERCP should be restricted to those individuals who are most likely to have CBD stones— namely, those with persistent obstructive jaundice, cholangitis, stones on imaging, and severe gallstone pancreatitis (Table 3) (33,107,108). We undertook a retrospective study to define the role of ERCP based on the likelihood of bile duct stones when laparoscopic bile duct exploration was not routinely utilized (33). In 1847 patients, the likelihood of bile duct stones was considered highest for risk factors including serum bilirubin >2 mg/dL, alkaline phosphatase >150 IU/L, pancreatitis, or a dilated CBD or stone on ultrasonography or CT scanning. This was considered an indication for preoperative ERCP. A lower likelihood of bile duct stones was defined by mildly elevated LFTs (serum bilirubin level of 1.5 to 2 mg/dL, alkaline phosphatase of 110 to 150 IU/L, or alanine aminotransferase greater than twice the upper limit of normal), or a remote history of jaundice or pancreatitis. Intraoperative cholangiography was performed for these patients. We found that preoperative ERCP was performed in 135 (7.3%) patients and demonstrated CBD stones in 43 (32%). Selective intraoperative cholangiography was performed in 87 (5%) patients and stones were found in 2 (2%). We then defined "stricter criteria" for predicting the presence of CBD stones as either persistent jaundice or a demonstrated CBD stone on ultrasonography or CT scanning. Applying stricter criteria to select patients for preoperative ERCP was predictive of ductal stones in 56%, resulting in a 50% reduction in preoperative ERCPs, while only 3% of patients with stones would have been missed. The selection of patients for preoperative ERCP may be further refined in the future with the application of MRI cholangiography and EUS. Patients who have had transient LFT abnormalities or fluctuating jaundice fall into an "intermediaterisk" category for CBD stones (31). Interestingly, recent pancreatitis has repeatedly been shown to be an intermediate predictor for CBD stones (prevalence 10 to 20%), particularly if LFTs have normalized, presumably because the offending gallstone has passed spontaneously into the duodenum (28,33). In most tertiary referral centers, patients in the intermediaterisk group undergo laparoscopic cholecystectomy with intraoperative cholangiography rather than preoperative ERCP. The advantage of the strategy with intraoperative cholangiography is that most patients can be managed with one procedure because the incidence of finding bile duct stones in patients with an intermediate likelihood of bile duct stones is only approximately 10 to 30%. If a CBD stone is identified by intraoperative cholangiography, three treatment options can be considered: (a) LCBDE, (b) completion of the laparoscopic cholecystectomy followed by postoperative ERCP and stone extraction, or (c) conversion to an open CBDE. Conversion to an open procedure violates the minimally invasive strategy and exposes the patient to the added morbidity of the open operation. Laparoscopic transcystic cholangiography and CBD exploration with stone extraction can be performed at the time of laparoscopic cholecystectomy, but at present few surgeons have the experience or inclination to perform LCBDE for stone extraction. However, this is likely to evolve as skills and instrumentation improve. If LCBDE can be performed successfully, then this avoids a second endoscopic procedure or an open operation. The alternative strategy of postoperative ERCP (107,109) is typically favored and is dependent on endoscopic expertise, as failed stone removal—which occurs in less than 5% of patients in the hands of a skilled endoscopist—necessitates repeated attempts at ERCP or a second operative procedure. If the patient has a known anatomic anomaly that may render ERCP technically challenging (e.g., Billroth II gastrectomy or RouxenY biliary reconstruction), then prelaparoscopic cholecystectomy ERCP is justified. In order to reduce the risk of unsuccessful bile duct cannulation following laparoscopic cholecystectomy, a percutaneous transcysticcholedochopapillary drain could be inserted at the time of surgery (110). To summarize, we suggest the following algorithm for the management of patients with suspected bile duct stones (Fig. 5).
Page 581
2— Gallstone Pancreatitis The diagnosis of gallstone pancreatitis is supported in the setting of pancreatitis by the presence of gallstones on ultrasonography combined with a greater than threefold elevation in alanine aminotransferase (111). Indications for ERCP in gallstone pancreatitis include suspected cholangitis or the development of severe pancreatitis within 24 to 72 h of symptom onset. Gallstone pancreatitis is proposed to result from stone impaction in the common channel of the pancreatic and bile ducts resulting in obstruction of the two systems and reflux of bile into the pancreatic duct. In support of this theory, gallstones can be recovered from the feces of 85 to 95% of patients (112), and the incidence of CBD stones is as high as 80% in those undergoing urgent biliary intervention compared to a 5 to 30% incidence when the procedure is delayed (113)—a finding that suggests frequent spontaneous passage of stones. Patients with more severe pancreatitis are more likely to retain a CBD stone (114,115), and animal studies have shown that the severity of pancreatitis is proportional to the duration of obstruction of the acute pancreatic duct (116). This suggests that early intervention to reduce the duration of pancreatic duct obstruction or stone removal to prevent recurrent obstruction may improve the severity and outcome of acute gallstone pancreatitis. Patients with gallstone pancreatitis should be stratified into severity of illness based on wellestablished clinical criteria (e.g., Ranson, Imrie, Glasgow, and APACHE II scores) and dynamic contrastenhanced CT scanning performed to assess for the presence and extent of pancreatic necrosis in those patients deemed to have clinically severe disease. The majority of patients will have mild pancreatitis and do well with conservative therapy alone. Elective laparoscopic cholecystectomy with intraoperative cholangiography is performed during the index hospital stay. For those patients with predicted severe disease, early biliary surgical therapy is not recommended owing to the high operative morbidity and mortality. In a series by Kelly and Wagner (117), 165 patients with gallstone pancreatitis were prospectively randomized to early or delayed surgery. In the group with severe pancreatitis, they found a 48% mortality following urgent operative intervention, compared with an 11% mortality in patients in whom surgery was delayed for more than 48 h. An endoscopic approach to gallstone pancreatitis offers the theoretical advantage of immediate relief of ampullary obstruction and ductal clearance without the risks of general anesthesia or the surgical procedure. Urgent ERCP with endoscopic sphincterotomy and stone extraction can be performed safely and with good rationale in those individuals with suspected acute cholangitis, an increasingly recognized comorbidity that is suspected in the presence of fever and jaundice (118). More controversial is the utility of urgent ERCP for the "severely" ill patient with gallstone pancreatitis. Three of four prospective controlled series (114,115,119,120) support this recommendation, demonstrating the safety and efficacy of ERCP in reducing local pancreatic and systemic complications in the subgroup of patients with severe biliary pancreatitis when the procedure is performed during the initial 24 to 72 h of hospitalization. The first randomized controlled trial came from the United Kingdom (114). Patients with suspected gallstone pancreatitis were randomized to urgent ERCP within 72 h or conservative treatment. In the group with severe pancreatitis, ERCP reduced the morbidity (61 versus 24%), length of hospital stay (17 versus 9.5 days), and mortality (18 versus 4%), although the difference in mortality did not reach statistical significance. The outcome of patients with mild pancreatitis was the same in both groups. A second randomized controlled trial from Hong Kong involving 195 patients (115) confirmed the results of the U.K. study. In the patients with severe pancreatitis who underwent urgent ERCP, there was a significant reduction in morbidity from 54 to 13% and in mortality from 18 to 3%. The incidence of biliary sepsis was significantly lower in those patients with severe pancreatitis who underwent an ERPC (0%) compared to those who did not (20%). There were no clinical differences in those who had mild pancreatitis. The third study was from Poland and was published in abstract form only (121). All 280 patients with acute gallstone pancreatitis underwent ERCP within 24 h. Seventyfive patients were found to have impacted stones in the papilla and were treated by immediate sphincterotomy. The remaining patients were randomized to immediate sphincter
Page 582
otomy or conservative treatment. They found significantly fewer complications (17 versus 36%) and a lower mortality rate (2 versus 13%) in the group that underwent ERCP. They also found, unlike the prior studies, that the benefits of ERCP and sphincterotomy extended to patients predicted to have mild pancreatitis. The most recent and only negative study excluded those gallstone pancreatitis patients with evidence of obstructive jaundice (119). Stratification of patients according to the severity of pancreatitis did not affect the findings. This latter study might be interpreted to indicate that the subgroup of gallstone pancreatitis patients with no evidence of biliary obstruction can be treated conservatively without urgent ERCP; however, a confirmatory study is required before these findings are accepted into routine practice. Endoscopic sphincterotomy in lieu of cholecystectomy should be considered in highrisk elderly patients with intact gallbladders who have recovered from an attack of gallstone pancreatitis, even in the absence of documented CBD stones. The rationale for endoscopic sphincterotomy in the absence of ductal stones is to eliminate the common channel so that gallstones that migrate from the gallbladder pass unimpeded through the bile duct into the duodenum without an opportunity to obstruct the pancreatic duct and cause pancreatitis. Evidence from two endoscopic studies substantiates the efficacy of the endoscopic approach in reducing the incidence of recurrent pancreatitis to less than 5% during a mean followup of 4 years (122,123). Other scenarios of gallstone pancreatitis where sphincterotomy should be considered definitive therapy include circumstances where biliary lithiasis is thought to be a temporary condition, as in pregnancy, prolonged fasting, and rapid weight loss. 3— Postcholecystectomy with T Tube In Situ Bile duct stones less than 10 mm in diameter detected postoperatively on Ttube cholangiography may pass spontaneously or with hydrostatic pressure from flushing or perfusing the T tube. However, the majority of these stones will require additional mechanical manipulation. Secondary bile duct exploration is associated with increased morbidity and mortality. This has stimulated the development of alternative techniques to extract these stones, including hydraulic Ttube irrigation with or without pharmacological relaxation of the sphincter of Oddi (124), Ttube infusion of cholesterol solvents (124), Ttube tract choledochoscopy and lithotripsy (125), or percutaneous extraction of stones through a mature Ttube track (126). Success with the latter technique has ranged from 77 to 96%. However, multiple sessions are often required and complications such as sepsis, biliary trauma, and biliary leakage occur in 4 to 8% of individuals (126–129). Delays of 4 to 6 weeks are required prior to manipulation to allow maturation of the Ttube track. Alternatively, early ERCP and sphincterotomy can be safely and effectively performed after stone detection on Ttube cholangiography without the need for Ttube maturation, allowing earlier discharge from hospital. The results from eight endoscopic series totaling 337 patients indicate an overall endoscopic success rate of 90% with a morbidity of 7% and mortality of 0.6% (129–136). The choice between these two techniques, which appear to have similar efficacy and safety, depends on local expertise, as direct comparisons in controlled trials have not been made. 4— Postcholecystectomy without T Tube Retained bile duct stones following bile duct exploration occurred in 4 to 16% of patients in earlier series, but this frequency has been substantially lowered by the introduction of preoperative choledochoscopy to 1 to 5% (70). The incidence of retained stones is similar following LCBDE and stone extraction, with a rate of 1 to 5% (87,88,94–99). Endoscopic sphincterotomy remains the treatment of choice for the elderly patient presenting days to years after cholecystectomy, as these patients are at higher risk from further abdominal surgery (135,137,138). Although the treatment selection in the young, healthy, postcholecystectomy patient includes further surgery or endoscopic sphincterotomy, most clinicians would choose the latter.
Page 583
5— Selected Patients with Gallbladder In Situ (GBIS) Peroral endoscopic techniques provide an effective treatment alternative for bile duct stone management and may be safer than surgery in the elderly and highrisk groups. ERCPrelated morbidity and mortality are unchanged regardless of age (37). In deciding whether sphincterotomy is adequate treatment for choledocholithiasis, the efficacy of the procedure in removing the stones, the shortterm risks of the endoscopic sphincterotomy, and the longterm risks of leaving the GBIS should be compared to the conventional surgical alternative of cholecystectomy combined with CBDE (139). Ideally, a prospective, randomized, comparative study between surgery and ERCP is needed to confirm the superiority of the endoscopic approach. The shortterm risks of endoscopic sphincterotomy for CBD stones, as previously stated, include acute pancreatitis, bleeding, cholangitis, and perforation in less than 5% of cases, with a procedurerelated mortality of 0.2 to 0.4% (35,37,52). Longterm outcome with 5 to 15 years of followup, leaving the gallbladder in situ following endoscopic sphincterotomy and CBD stone removal, demonstrates variable results. On average, approximately 20 to 25% of patients will develop recurrent gallbladder or biliary tree problems (60,61,140). The main risks of leaving the gallbladder in situ are the development of acute cholecystitis in 3 to 8% and, over the long term, approximately 10 to 20% of patients will develop symptoms referable to their gallbladder, requiring cholecystectomy (70,80). An additional 10% will develop recurrent CBD stones; 1 to 2% will develop biliary sphincter stenosis and, infrequently, nonobstructive cholangitis (60,61). This compares favorably to the surgical alternative of open cholecystectomy and CBD exploration, which has a mortality rate of approximately 3% and a 10 to 20% recurrence rate for biliary problems (79). Compared to earlier series, more recent series have suggested that the morbidity and mortality after surgical bile duct exploration have decreased (79), and two recent prospective randomized studies have questioned the rationale for leaving the gallbladder in situ following endoscopic sphincterotomy (60,61). 6— Cholangitis Acute cholangitis is a lifethreatening condition demanding immediate rescuscitative measures and administration of intravenous antibiotics. The majority of patients with cholangitis of mild severity will defervesce rapidly, allowing for a semiurgent approach to endoscopic biliary decompression followed by elective laparoscopic cholecystectomy. Reynolds and Dargan in 1959 described a particularly lethal form of cholangitis, also referred to as suppurative cholangitis and manifest by a clinical pentad (Reynolds' pentad) of shock and mental confusion plus Charcot's triad of pain, fever, and jaundice. Emergency ERCP is the treatment of choice for patients with this severe form of cholangitis as well as for those individuals with mild cholangitis not responding within 24 h to antibiotic treatment. Endoscopic biliary decompression can be performed successfully in 85 to 95% of patients (141–145), with a lower morbidity and mortality compared with percutaneous transhepatic (146–149) or surgical drainage (150). In a critically ill patient, only brief attempts should be made to remove bile duct stones at the time of initial ERCP. Bile should be aspirated to decompress the biliary system prior to injection of contrast, and once the level of obstruction has been established, drainage should be performed with a nasobiliary tube or stent (151). When necessary, these drainage procedures can be completed without sphincterotomy, thus avoiding the risk of hemorrhage from an underlying bleeding diathesis, which often accompanies severe cholangitis (151). Elective sphincterotomy and stone extraction are performed once clinical improvement has been noted. Emergency surgery and percutaneous approaches should be reserved for endoscopic failures, as conventional surgical treatment in the context of severe cholangitis is associated with a morbidity and mortality of 20 to 40% (16). In a prospective, comparative trial, 82 consecutive patients with severe acute cholangitis due to choledocholithiasis were randomly assigned to undergo emergency ERCP or surgical biliary decompression (150). The hospital mortality rate was significantly lower in the endoscopically treated group (10 versus 32%). Furthermore,
Page 584
patients in the surgery group had more complications (66 versus 34%), greater need for mechanical ventilatory support (63 versus 29%), and a higher rate of retained CBD stones (29 versus 7%). 7— Pregnancy Symptomatic bile duct stones during pregnancy pose a diagnostic and therapeutic challenge. The frequency of choledocholithiasis in pregnancy requiring intervention has been reported to be as low as one in 1200 deliveries (152). There are emerging data on the safety and efficacy of endoscopic management (152–155). The largest singlecenter experience of 15 patients suggests safety provided that appropriate measures are taken to protect the fetus and mother (155). Fetal monitoring is performed and the pelvis of every patient is shielded. Fluoroscopy is minimized and hardcopy radiographs are taken only when essential to minimize fetal exposure to radiation. Dosimetry to calculate fetal radiation exposure is performed, with a mean dose of about 325 mrad reported. A subsequent multicenter study reported the experience of 20 pregnant patients undergoing therapeutic ERCP. The only ERCP complication was pancreatitis in one patient. There was one spontaneous abortion (3 months after ERCP) and one neonatal death, but a causal relationship to ERCP was not considered likely (152). References 1. DenBesten L, Doty JE. Pathogenesis and management of choledocholithiasis. Surg Clin North Am 1981; 61:893–907. 2. Madden JL. Common duct stones; their origin and surgical management. Surg Clin North Am 1973; 53:1095–1113. 3. Saharia PC, Zuidema GD, Cameron JL. Primary common duct stones. Ann Surg 1977; 185:598–604. 4. Glenn F. Post cholecystectomy choledocholithiasis. Surg Gynecol Obstet 1972; 134: 249–252. 5. Tabata M, Nakayama F. Bacteria and gallstones: etiological significance. Dig Dis Sci 1981; 26:218–224. 6. Imamoglu K, Perry JF, Wangesteen OH. Experimental production of gallstones by incomplete stricture of the terminal common bile duct. Surgery 1957; 42:623. 7. Nagase M, Hikasa Y, Soloway RD, Tanimura H, Setoyama M, Kato H. Gallstones in western Japan: factors affecting the prevalence of intrahepatic gallstones. Gastroenterology 1980; 78:684–690. 8. Whiting MJ, Watts JM. Chemical composition of common bile duct stones. Br J Surg 1986; 73:229–232. 9. Kennedy RH, Thompson MH. Are duodenal diverticula associated with choledocholithiasis? Gut 1988; 29:1003–1006. 10. NIH Consensus Conference on Gallstones and Laparoscopic Cholecystectomy. Am J Surg 1993; 165:390–547. 11. Johnson A, Hosking S. Appraisal of the management of bile duct stones. Br J Surg 1987; 74:555–560. 12. Murison MSC, Gartell PC, McGinn FP. Does selective peroperative cholangiography result in missed common bile duct stones? J R Coll Surg Edinb 1993; 38:220–224. 13. Chung RS, Chad V, Eisenstat M. Choledocholithiasis treated with laparoscopic stenting of the papilla followed by stent guided sphincterotomy. Gastrointest Endosc 1997; 45: A405. 14. Lindenauer SM, Child CG III. Disturbances of liver function in biliary tract disease. Surg Gynecol Obstet 1966; 123:1205–1211.
Page 585
15. Anciaux ML, Pelletier G, Attali P, Meduri B, Liguory C, Etienne JP. Prospective study of clinical and biochemical features of symptomatic choledocholithiasis. Dig Dis Sci 1986; 31:449–453. 16. Boey JH, Way LW. Acute cholangitis. Ann Surg 1980; 191:264–270. 17. Tham TCK, Collins JSA, Watson RGP, Ellis PK, McIlrath EM. Diagnosis of common bile duct stones by intravenous cholangiography: prediction by ultrasound and liver function tests compared with endoscopic retrograde cholangiography. Gastrointest Endosc 1996; 44:158–163. 18. Laing FC, Jeffrey RB, Wing VW. Improved visualization of choledocholithiasis by sonography. Am J Roentgenol 1984; 143:949–952. 19. Baron RL. Common bile duct stones: reassessment of criteria for CT diagnosis. Radiology 1987; 162:419–424. 20. Reinhold C, Bret PM. Current status of MR cholangiopancreatography. Am J Roentgenol 1996; 166:1285–1295. 21. Chan YL, Chan AC, Lam WW, Lee DW, Chung SS, Sung JJ, Cheung HS, Li AK, Metreweli C. Choledocholithiasis: comparison of MR cholangiography and endoscopic retrograde cholangiography. Radiology 1996; 200:85–89. 22. Sugiyama M, Atomi Y. Endoscopic ultrasonography for diagnosing choledocholithiasis: a prospective comparative study with ultrasonography and computed tomography. Gastrointest Endosc 1997; 45:143–146. 23. Palazzo L, Girollet PP, Salmeron M, Silvain C, Roseau G, Canard JM, Chaussade S, Couturier D, Paolaggi JA. Value of endoscopic ultrasonography in the diagnosis of common bile duct stones: comparison with surgical exploration and ERCP. Gastrointest Endosc 1995; 42:225–231. 24. Canto MIF, Chak A, Stellato T, Sivak MV. Endoscopic ultasonography versus cholangiography for the diagnosis of choledocholithiasis. Gastrointest Endosc 1998; 47:439–448. 25. Amouyal P, Amouyal G, Levy P, Tuzet S, Palazzo L, Vilgrain V, Gayet B, Belghiti J, Fekete F, Bernades P. Diagnosis of choledocholithiasis by endoscopic ultrasonography. Gastroenterology 1994; 106:1062–1067. 26. De Ledinghen V, Lecesne R, Raymond JM, Gense V, Amouretti M, Drouillard J, Couzigou P, Silvain C. Diagnosis of choledocholithiasis: EUS or magnetic resonance cholangiography? A prospective controlled study. Gastrointest Endosc 1999; 49:26–31. 27. Sahai AV, Mauldin PD, Marsi V, Hawes RH, Hoffman BJ. Bile duct stones and laparoscopic cholecystectomy: a decision analysis to assess the roles of intraoperative cholangiography, EUS, and ERCP. Gastrointest Endosc 1999; 49:334–343. 28. Santucci A, Natalini G, Sarpi L, Fiorucci S, Solines A, Morelli A. Selective ERCP and preoperative bile duct stone removal in patients scheduled for laparoscopic cholecystectomy: a prospective study. Am J Gastroenterol 1996; 91:1326–1330. 29. Barkun AN, Barkun JS, Fried GM, Ghitulescu G, Steinmetz O, Pham C, Meakins JL, Goresky CA. Useful predictors of bile duct stones in patients undergoing laparoscopic cholecystectomy. Ann Surg 1994; 220:32–39. 30. Onken JE, Brazer SR, Eisen GM, Haber GB, Herman ME, Dorsher PJ, Moore JP, Fennerty MB, Ryan ME, Shaw MJ, Lande JD, Pheley AM. Predicting the presence of choledocholithiasis in patients with symptomatic cholelithiasis. Am J Gastroenterol 1996; 91:762–767. 31. Roston AD, Jacobson IM. Evaluation of the pattern of liver tests and yield of cholangiography in symptomatic choledocholithiasis: a prospective study. Gastrointest Endosc 1997; 45:394–399. 32. Abboud PAC, Malet PF, Berlin JA, Staroscik R, Cabana MD, Clarke JR, Shea JA, Schwartz JS, Williams SV. Predictors of common bile duct stones prior to cholecystectomy: a metaanalysis. Gastrointest Endosc 1996; 44:450–459. 33. Tham TCK, Lichtenstein DR, Vandervoort J, Wong RC, Brooks D, Van Dam J, Ruymann F, Farraye F, CarrLocke DL. Role of endoscopic retrograde cholangiopancrea
Page 586
tography for suspected choledocholithiasis in patients undergoing laparoscopic cholecystectomy. Gastrointest Endosc 1998; 47:50–56. 34. Kullman E, Borch K, Lindstrom E, Ansehn S, Ihse I, Anderberg B. Bacteremia following diagnostic and therapeutic ERCP. Gastrointest Endosc 1992; 38:444– 449. 35. Loperfido S, Angelini G, Benedetti G, Chilovi F, Costan F, De Berardinis F, De Bernardin M, Ederle A, Fina P, Fratton A. Major early complications from diagnostic and therapeutic ERCP: a prospective multicenter study. Gastrointest Endosc 1998; 48:1–10. 36. Dowsett JF, Vaira D, Hatfield AR, Cairns SR, Polydorou A, Frost R, Croker J, Cotton PB, Russell RC, Mason RR. Endoscopic biliary therapy using the combined percutaneous and endoscopic technique. Gastroenterology 1989; 96:1180–1186. 37. Freeman ML, Nelson DB, Sherman S, Haber GB, Herman ME, Dorsher PJ, Moore JP, Fennerty MB, Ryan ME, Shaw MJ, Lande JD, Pheley AM. Complications of endoscopic sphincterotomy. N Engl J Med 1996; 335:909–918. 38. Shakoor T, Geenen JE. Precut papillotomy. Gastrointest Endosc 1992; 38:623–627. 39. Kumar S, Sherman S, Hawes RH, Lehman GA. Success and yield of second attempt ERCP. Gastrointest Endosc 1995; 41:445–447. 40. Chung SC, Leung JW, Leong HT, Li AK. Mechanical lithotripsy of large common bile duct stones using a basket. Br J Surg 1991; 78:1448–1450. 41. Shaw MJ, Mackie RD, Moore JP, Dorsher PJ, Freeman ML, Meier PB, Potter T, Hutton SW, Vennes JA. Results of a multicenter trial using a mechanical lithotripter for the treatment of large bile duct stones. Am J Gastroenterol 1993; 88:730–733. 42. Adamek HE, Maier M, Jakobs R, Wessbecker FR, Neuhauser T, Riemann JF. Management of retained bile duct stones: a prospective open trial comparing extracorporeal and intracorporeal lithotripsy. Gastrointest Endosc 1996; 44:40–47. 43. Adamek HE, Maier M, Jakobs R, Wessbecher FR, Neuhauser T, Riemann JF. Management of bile duct stones: a prospective open trial comparing extracorporeal and intracorporeal lithotripsy. Gastrointest Endosc 1996; 44:40–47. 44. Sauerbruch T, Stern M. Fragmentation of bile duct stones by extracorporeal shock waves: a new approach to biliary calculi after failure of routine endoscopic measures. Gastroenterology 1989; 96:146–152. 45. Neuhaus H, Zillinger C, Born P, Ott R, Allescher H, Rosch T, Classen M. Randomized study of intracorporeal laser lithotripsy versus extracorporeal shockwave lithotripsy for difficult bile duct stones. Gastrointest Endosc 1998; 47:327–334. 46. Cotton P, Kozarek R, Schapiro R. Endoscopic laser lithotripsy of large bile duct stones. Gastroenterology 1990; 99:1128–1133. 47. Palmer KR, Hofmann AF. Intraductal monooctanoin for the direct dissolution of bile duct stones: experience in 343 patients. Gut 1986; 27:196–202. 48. Neoptolemos JP, Hall C, O'Connor, Murray WR, CarrLocke DL. Methyltertbutylether for treating bile duct stones: the British experience. Br J Surg 1990; 77:32–35. 49. Chopra KB, Peters RA, O'Toole PA, Williams SG, Gimson AE, Lombard MG, Westaby D. Randomised study of endoscopic biliary endoprosthesis versus duct clearance for bile duct stones in highrisk patients. Lancet 1996; 348:791–793. 50. Bergman JJ, Rauws EAJ, Tijssen JGP, Tytgat GN, Huibregtse K. Biliary endoprostheses in elderly patients with endoscopically irretrievable common bile duct stones: report on 117 patients. Gastrointest Endosc 1995; 42:195–201. 51. Maxton DG, Tweedle DE, Martin DF. Stenting for choledocholithiasis: temporizing or therapeutic? Gut 1995; 36:446–449. 52. Cotton PB, Lehman G, Vennes JA, Geenen JE, Russell RCG, Meyers WC, Liguory C, Nickl N. Endoscopic sphincterotomy complications and their management: an attempt at consensus. Gastrointest Endosc 1991; 37:383–391. 53. Hawes RH, Cotton PB, Vallon AG. Followup 6 to 11 years after duodenoscopic sphincterotomy for stones in patients with prior cholecystectomy. Gastroenterology 1990; 98: 1008–1012.
Page 587
54. Riemann JF, Lux G, Forster P, Altendorf A. Longterm results after endoscopic papillotomy. Endoscopy 1983; 15(suppl. 1):165–168. 55. Ikeda S, Tanaka M, Matsumoto S, Yoshimoto H, Itoh H. Endoscopic sphincterotomy: longterm results in 408 patients with complete followup. Endoscopy 1988; 20:13–17. 56. Clark DW, Geenen JE, Hogan WJ, Johnson GK, Schmalz MJ, Catalano MF. Ten year followup of patients undergoing endoscopic sphincterotomy for common bile duct stones. Gastrointest Endosc 1994; 40:A104. 57. Cetta FM. The role of bacteria in pigment gallstone disease. Ann Surg 1991; 213:315–326. 58. Bergman JJ, van der Mey S, Rauws EAJ, Tytgat GNJ, Huibregste K. Late complications after endoscopic sphincterotomy for bile duct stones in patients younger than 60 years. Gastrointest Endosc 1996; 44:643–649. 59. Prat F, Abdel Malak N, Pelletier G, Buffet C, Fritsch J, Choury AD, Altman C, Liguory C, Etienne JP. Biliary symptoms and complications more than 8 years after endoscopic sphincterotomy for choledocholithiasis. Gastroenterology 1996; 110: 894–899. 60. PereiraLima JC, Jakobs R, Winter UH, Benz C, Martin WR, Adamek HE, Riemann JR. Longterm (7 to 10 years) of endoscopic papillotomy for choledocholithiasis: multivariate analysis of prognostic factors for the recurrence of biliary symptoms. Gastrointest Endosc 1998; 48:457–464. 61. Tanaka M, Takahata S, Konomi H, Matsunaga H, Yokohata K, Takeda T, Utsunomiya N, Ikeda S. Longterm consequence of endoscopic sphincterotomy for bile duct stones. Gastrointest Endosc 1998; 48:465–469. 62. Tham TCK, Kennedy R, O'Connor FA. Early complications and mean 8 year followup after endoscopic sphincterotomy in young fit patients. Eur J Gastroenterol Hepatol 1994; 6:621–624. 63. MacMathuna P, White P, Clarke E, Merriman R, Lennon JR, Crowe J. Endoscopic balloon sphincteroplasty (papillary dilation) for bile duct stones: efficacy, safety and followup in 100 patients. Gastrointest Endosc 1995; 42:468–474. 64. May GR, Cotton PB, Edmunds SE, Chong W. Removal of stones from the bile duct at ERCP without sphincterotomy. Gastrointest Endosc 1993; 39:749–754. 65. Staritz M, Ewe K, Meyer zum Buschenfelde. Endoscopic papillary dilatation (EPD) for the treatment of common bile duct stones and papillary stenosis. Endoscopy 1983; 15: 197–198. 66. Ibuki Y, Kudo M, Todo A. Endoscopic retrograde extraction of common bile duct stones with drip infusion of isosorbide dinitrate. Gastrointest Endosc 1992; 38:178–180. 67. Bergman JJ, Rauws EA, Fockens P, van Berkel AM, Bossuyt PM, Tijssen JG, Tytgat GN, Huibregtse K. Randomised trial of endoscopic balloon dilation versus endoscopic sphincterotomy for removal of bile duct stones. Lancet 1997; 349:1124–1129. 68. DiSario JA, Freeman ML, Bjorkman DJ, MacMathuna P, Petersen B, Sherman S, Lehman G, Hixson L, Jaffe P, AlKawas F, Moore J, Khandelwal and the EDES Study Group. Endoscopic balloon dilation compared to sphincterotomy (EDES) for extraction of bile duct stones: preliminary results. Gastrointest Endosc 1997; 45:A129. 69. Way LW, Admirand WH, Dunphy JE. Management of choledocholithiasis. Ann Surg 1972; 176:347–349. 70. Winslet MC, Neoptolemos JP. The place of endoscopy in the management of gallstones. Baillieres Clin Gastroenterol 1991; 5:99–129. 71. Lygidakis NJ. Surgical approaches to recurrent choledocholithiasis: choledochoduodenostomy versus Ttube drainage after choledochotomy. Am J Surg 1983; 145:636–639. 72. Larson RE, Hodgson JR, Priestley JT. The early and long term results of 500 consecutive explorations of the common duct. Surg Gynecol Obstet 1966; 122:744–750. 73. Peel ALG, Bourke JB, HermonTaylor J, MacLean ADW, Mann CV, Ritchie HD. How should the common bile duct be explored? Ann R Coll Surg 1975; 56:124–134. 74. Allen B, Shapiro H, Way L. Management of recurrent and residual common duct stones. Am J Surg 1981; 142:41–47.
Page 588
75. Saharia PC, Zuidema G, Cameron JL. Primary common duct stones. Ann Surg 1977; 185:598–604. 76. Braasch JW, Fender HR, Bonneval MM. Refractory primary common bile and duct stone disease. Am J Surg 1980; 139:526–530. 77. Polydorou A, Dowsett JF, Vaira D, Salmon PR, Cotton PB, Russell RC. Endoscopic therapy of the sump syndrome. Endoscopy 1989; 21:126–130. 78. Tierney S, Lillemoe KD, Pitt HA. The current management of common duct stones. Adv Surg 1995; 28:271–299. 79. Moreaux J. Traditional surgical management of common bile duct stones: a prospective study during a 20 year experience. Am J Surg 1995; 169:220–226. 80. Targarona EM, Ayuso RMP, Bordas JM, Ros E, Pros I, Martinez J, Teres J, Trias M. Randomised trial of endoscopic sphincterotomy with gallbladder left in situ versus open surgery for common bile duct calculi in highrisk patients. Lancet 1996; 347:926–929. 81. Hammarstrom LE, Holmin T, Stridbeck H, Ihse I. Longterm followup of a prospective randomized study of endoscopic versus surgical treatment of bile duct calculi in patients with gallbladder in situ. Br J Surg 1995; 82:1516–1521. 82. Baker AR, Neoptolemos JP, Leese T, James DC, Fossard DP. Long term followup of patients with side to side choledochoduodenostomy and transduodenal sphincterotomy. Ann R Coll Surg Engl 1987; 69:253–258. 83. Thomas CG, Nicholson CP, Owen J. The effectiveness of choledochoduodenostomy in the treatment of benign obstruction of the common duct. Ann Surg 1971; 173:845–846. 84. Holdsworth RJ, Sadek SA, Ambikar S, Baker PR, Cuschieri A. Dynamics of bile flow through the human choledochal sphincter following exploration of the common bile duct. World J Surg 1989; 13:300–306. 85. DePaula AL, Hashiba K, Bafutto M, Machado C, Ferrari A, Machado MM. Results of the routine use of a modified endoprosthesis to drain the common bile duct after laparoscopic choledochotomy. Surg Endosc 1998; 12:933–935. 86. Cuschieri A, Berci G. Laparoscopic treatment of common duct stones. In: Laparoscopic Biliary Surgery, 2nd ed. Oxford, England: Blackwell Scientific Publications, 1992, pp 155–169. 87. Paganini AM, Lezoche E. Followup of 161 unselected consecutive patients treated laparoscopically for common bile duct stones. Surg Endosc 1998; 12:23–29. 88. Berthou JC, Drouard F, Charbonneau P, Moussalier K. Evaluation of laparoscopic management of common bile duct stones in 220 patients. Surg Endosc 1998; 12:16–22. 89. Millat B, Atger J, Deleuze A, Briandet H, Fingerhut A, Guillon F, Marrel E, De Seguin C, Soulier P. Laparoscopic treatment for choledocholithiasis: a prospective evaluation in 247 consecutive unselected patients. Hepatogastroenterology 1997; 44:28–34. 90. Drouard F, PassoneSzerzyna N, Berthou JC. Laparoscopic treatment of common bile duct stones. Hepatogastroenterology 1997; 44:16–21. 91. Lezoche E, Paganini AM, Carlei F, Feliciotti F, Lomanto D, Guerrieri M. Laparoscopic treatment of gallbladder and common bile duct stones: a prospective study. World J Surg 1996; 20:535–541. 92. Lezoche E, Paganini AM. Singlestage laparoscopic treatment of gallstones and common bile duct stones in 120 unselected, consecutive patients. Surg Endosc 1995; 9:1070–1075. 93. Millat B, Fingerhut A, Deleuze A, Briandet H, Marrel E, de Seguin C, Soulier P. Prospective evaluation in 121 consecutive unselected patients undergoing laparoscopic treatment of choledocholithiasis. Br J Surg 1995; 82:1266–1269. 94. Phillips EH, Liberman M, Carroll BJ, Fallas MJ, Rosenthal RJ, Hiatt JR. Bile duct stones in the laparoscopic era. Is preoperative sphincterotomy necessary? Arch Surg 1995; 30:880–885. 95. Rhodes M, Nathanson L, O'Rourke N, Fielding G. Laparoscopic exploration of the common bile duct: lessons learned from 129 consecutive cases. Br J Surg 1995; 82: 666–668.
Page 589
96. Stoker ME. Common bile duct exploration in the era of laparoscopic surgery. Arch Surg 1995; 130:265–268. 97. DePaula AL, Hashiba K, Bafutto M. Laparoscopic management of choledocholithiasis. Surg Endosc 1994; 8:1399–1403. 98. Phillips EH, Rosenthal RJ, Carroll BJ, Fallas MJ. Laparoscopic transcystic duct common bile duct exploration. Surg Endosc 1994; 8:1389–1393. 99. Berci G, Morgenstern L. Laparoscopic management of common bile duct stones. A multiinstitutional SAGES study: Society of American Gastrointestinal Endoscopic Surgeons. Surg Endosc 1994; 8:1168–1174. 100. Cuschiere A and the EAES Ductal Stone Group. EAES ductal stone study—preliminary findings of multicentre prospective randomised trial comparing two stage versus single stage management. Gut 1996; 39(suppl 1):A43. 101. Rhodes M, Sussman L. Prospective randomised trial of laparoscopic common bile duct exploration versus postoperative ERCP. Gut 1997; 40(suppl 1):A68. 102. Geisinger MA. Percutaneous biliary stone extraction: radiologic and combined radiologic endoscopic techniques. Gastrointest Endosc Clin North Am 1991; 1:105–124. 103. Ponsky JL. Alternative methods in the management of bile duct stones. Surg Clin North Am 1992; 72:1099–1107. 104. Dowsett JF, Vaira D, Hatfield ARW, Cairns SR, Polydorou A, Frost R, Croker J, Cotton PB, Russell RCG, Mason RR. Endoscopic biliary therapy using the combined percutaneous and endoscopic technique. Gastroenterology 1989; 96:1180–1186. 105. Stiegman GV, Goff J, Mansour A, Pearlman N, Reveille RM, Norton L, Precholecystectomy endoscopic cholangiography and stone removal is not superior to cholecystectomy, cholangiography, and common duct exploration. Am J Surg 1992; 163:227–230. 106. Neoptolemos JP, CarrLocke DL, Fossard DP. Prospective randomised study of preoperative endoscopic sphincterotomy versus surgery alone for common bile duct stones. BMJ 1987; 294:470–474. 107. Erickson RA, Carlson B. The role of endoscopic retrograde cholangiopancreatography in patients with laparoscopic cholecystectomies. Gastroenterology 1995; 109:252–263. 108. Chan AC, Chung SC, Wyman A, Kwong KH, Ng EK, Lau JY, Lau WY, Lai CW, Sung JJ, Li AK. Selective use of preoperative endoscopic retrograde cholangiopancreatography in laparoscopic cholecystectomy. Gastrointest Endosc 1996; 43:212–215. 109. Traverso LW, Kozarek RA, Ball TJ, Brandabur JJ, Hunter JA, Jolly PC, Patterson DJ, Ryan JA, Thirlby RC, Wechter DG. Endoscopic retrograde cholangiography after laparoscopic cholecystectomy. Am J Surg 1993; 165:581–586. 110. Perissat J, Huibregtse K, Keane FB, Russell RC, Neoptolemos JP. Management of bile duct stones in the era of laparoscopic cholecystectomy. Br J Surg 1996; 83:755–757. 111. Tenner S, Dubner H, Steinberg W. Predicting gallstone pancreatitis with laboratory parameters: a metaanalysis. Am J Gastroenterol 1994; 89:1863–1866. 112. Acosta JM, Ledesma CL. Gallstone migration as a cause of acute pancreatitis. N Engl J Med 1974; 290:484–487. 113. Stone HH, Fabian TC, Dunlop WE. Gallstone pancreatitis: biliary tract pathology in relation to time of operation. Ann Surg 1981; 194:305–312. 114. Neoptolemos JP, CarrLocke DL, London NJ, Bailey IA, James D, Fossard DP. Controlled trial of urgent endoscopic retrograde cholangiopancreatography and endoscopic sphincterotomy versus conservative treatment for acute pancreatitis due to gallstones. Lancet 1988; 2:979–983. 115. Fan ST, Lai ECS, Mok FP, Lo CM, Zheng SS, Wong J. Early treatment of acute biliary pancreatitis by endoscopic papillotomy. N Engl J Med 1993; 328:228–232. 116. Runzi M, Saluja A, Lerch MM, Dawra R, Nishino H, Steer ML. Early ductal decompression prevents the progression of biliary pancreatitis: an experimental study in the opossum. Gastroenterology 1993; 105:157–164. 117. Kelly TR, Wagner DS. Gallstone pancreatitis: a prospective randomized trial of the timing of surgery. Surgery 1988; 104:600–605.
Page 590
118. Chang L, Lo SK, Stabile BE, Lewis RL, deVirgilio C. Gallstone pancreatitis: a prospective study on the incidence of cholangitis and clinical predictors of retained common bile duct stones. Am J Gastroenterol 1998; 93:527–531. 119. Folsch UR, Nitsche R, Ludtke R, Hilgers RA, Creutzfeldt W. Early ERCP and papillotomy compared with conservative treatment for acute biliary pancreatitis. N Engl J Med 1997; 336:237–242. 120. Nowak A, Blaszczynska M, Marek TA, NowakowskaDulawa E, Kaczor R. Prospective longterm followup after acute biliary pancreatitis in patients with gallbladder stones left in situ. Gastrointest Endosc 1998; 47:A125. 121. Nowak A, NowakowskaDulawa E, Marek TA, Rybicka J. Final results of the prospective, randomized, controlled study on endosopic sphincterotomy versus conventional management in acute biliary pancreatitis. Gastroenterology 1995; 108:A380. 122. Siegel JH, Veerappan A, Cohen SA, Kasmin FE. Endoscopic sphincterotomy for biliary pancreatitis: an alternative to cholecystectomy in highrisk patients. Gastrointest Endosc 1994; 40:573–575. 123. Welbourn CRB, Beckly DE, EyreBrook IA. Endoscopic sphincterotomy without cholecystectomy for gallstone pancreatitis. Gut 1995; 37:119–120. 124. Tritapepe R, di Padova C, Di Padova F. Noninvasive treatment for retained common bile duct stones in patients with T tube in situ: saline washout after intravenous ceruletide. Br J Surg 1988; 75:144–146. 125. Josephs LF, Birkett DH. Laser lithotripsy for the management of retained stones. Arch Surg 1992; 127:603–605. 126. Burhenne JH. Percutaneous extraction of retained biliary tract stones. Am J Radiol 1980; 134:888–898. 127. Cotton PB. Retained bile duct stones: Ttube in place, percutaneous or endoscopic management? Am J Gastroenterol 1990; 85:1075–1078. 128. Caprini JA. Biliary stone extraction. Am Surg 1988; 54:343–346. 129. Nussinson E, Cairns SR, Vaira D, Dowsett JF, Mason RR. A 10 year single centre experience of percutaneous and endoscopic extraction of bile duct stones with T tube in situ. Gut 1991; 32:1040–1043. 130. Tandon RK, Nijhawan S, Arora A. Management of retained common bile duct stones in patients with Ttube in situ: role of endoscopic sphincterotomy. Am J Gastroenterol 1990; 85:1126–1131. 131. Bickerstaff KI, Berry AR, Chapman RW, Britton J. Early postoperative endoscopic sphincterotomy for retained biliary stones. Ann R Coll Surg Engl 1988; 70:350–351. 132. Soehendra. N, Kempeneers I, Eichfuss HP, ReyndersFrederix V. Early postoperative endoscopy after biliary tract surgery. Endoscopy 1981; 13:113–117. 133. Simpson CJ, Gray GR, Gillespie G. Early endoscopic sphincterotomy for retained common bile duct stones. J R Coll Surg Edinb 1985; 30:288–289. 134. O'Doherty DP, Neoptolemos JP, CarrLocke DL. Endoscopic sphincterotomy for retained common bile duct stones in patients with Ttube in situ in the early postoperative period. Br J Surg 1986; 73:454–456. 135. Danilewitz MD. Early postoperative endoscopic sphincterotomy for retained common bile duct stones. Gastrointest Endosc 1989; 35:298–299. 136. Lambert ME, Martin DF, Tweedle DEF. Endoscopic removal of retained stones after biliary surgery. Br J Surg 1988; 75:896–898. 137. Brolin RE, Siemons GO, Fynan TM. Critical analysis of retained and residual common duct stones. Am Surg 1986; 52:588–593. 138. Cranley B, Logan H. Exploration of the common bile duct—the relevance of the clinical picture and the importance of preoperative cholangiography. Br J Surg 1980; 67:869–872. 139. Keulemans YCA, Rauws EAJ, Huibregtse K, Gouma DJ. Current management of the gallbladder after endoscopic sphincterotomy for common bile duct stones. Gastrointest Endosc 1997; 46:514–519.
Page 591
140. Hill J, Martin DF, Tweedle DEF. Risks of leaving the gallbladder in situ after endoscopic sphincterotomy for bile duct stones. Br J Surg 1992; 78:554–557. 141. Ditzel H, Schaffalitzky DE, Muckadell OB. Endoscopic sphincterotomy in acute cholangitis due to choledocholithiasis. Hepatogastroenterology 1990; 37:204– 207. 142. Leese T, Neoptolemos JP, Baker AR, CarrLocke DL. Management of acute cholangitis and the impact of endoscopic sphincterotomy. Br J Surg 1986; 73:988–992. 143. Leung JW, Chung SCS, Sung JJY, Banez VP, Li AKC. Urgent endoscopic drainage for acute suppurative cholangitis. Lancet 1989; 1:1307–1309. 144. Gogel HK, Runyon BA, Volpicelli NA, Palmer RC. Acute suppurative obstructive cholangitis due to stones: treatment by urgent endoscopic sphincterotomy. Gastrointest Endosc 1987; 33:210–213. 145. Ikeda S, Tanaka M, Itoh H, Kishikawa H, Nakayama F. Emergency decompression of bile duct in acute obstructive suppurative cholangitis by duodenoscopic cannulation: a lifesaving procedure. World J Surg 1981; 5:587–593. 146. Gould RJ, Vogelzang, Neimen HL, Pearl JG, Politcha SM. Percutaneous biliary drainage as an initial therapy of the biliary tract. Surg Gynecol Obstet 1985; 160:523–527. 147. Kadir S, Baassiri A, Barth KH, Kaufman SL, Cameron JL, White RT. Percutaneous transhepatic biliary drainage in the management of biliary sepsis. Am J Roentgenol 1982; 138:25–29. 148. Pessa ME, Hawkins IF, Vogel SB. The treatment of acute cholangitis: percutaneous transhepatic biliary drainage before definitive therapy. Ann Surg 1987; 205:389–392. 149. Nunez D, Guerra JJ, Alsheikh WA, Russell E, Mendez G Jr. Percutaneous biliary drainage in acute suppurative cholangitis. Gastrointest Radiol 1986; 11:85– 89. 150. Lai EC, Mok FP, Tan ES, Lo CM, Fan ST, You KT, Wong J. Endoscopic biliary drainage for severe acute cholangitis. N Engl J Med 1992; 326:1582–1586. 151. Sugiyama M, Atomi Y. The benefits of endoscopic nasobiliary drainage without sphincterotomy for acute cholangitis. Am J Gastroenterol 1998; 93:2065–2068. 152. Jamidar PA, Beck GJ, Hoffman BJ, Lehman GA, Hawes RH, Agrawal RM, Ashok PS, Ravi TJ, Cunningham JT, Troiano F. Endoscopic retrograde cholangiopancreatography in pregnancy. Am J Gastroenterol 1995; 90:1263–1267. 153. Baillie J, Cairns S, Putnam W, Cotton P. Endoscopic management of choledocholithiasis during pregnancy. Surg Gynecol Obstet 1990; 171:1–4. 154. Axelrad AM, Fleischer DE, Strack LL, Benjamin SB, AlKawas FH. Performance of ERCP for symptomatic choledocholithiasis during pregnancy: technique to increase safety and improve patient management. Am J Gastroenterol 1994; 89:109–112. 155. Vandervoort J, Tham TCK, Wong RCK, Roston AD, Slivka A, Ferrari Jr AP, Musa A, Lichtenstein DR, Van Dam J, Nawfel RD, CarrLocke DL. Is ERCP during pregnancy safe? Gastrointest Endosc 1996; 43:A400.
Page 593
28— Acalculous Cholecystitis David Nunes Boston University School of Medicine, Boston, Massachusetts I— Acute Acalculous Cholecystitis A— Introduction The term acalculous cholecystitis has been used to define both inflammatory conditions of the gallbladder as well as gallbladderrelated symptoms in the absence of gallstones. The term encompasses a wide variety of both acute and chronic diseases of the gallbladder of many causes, but in some cases—for instance, chronic acalculous cholecystitis—pathological examination of the gallbladder may be normal. Acute acalculous cholecystitis accounts for 2 to 17% of all cholecystectomies for cholecystitis (1–7). The importance of this condition is reflected by the high incidence of acalculous cholecystitis in patients who are acutely stressed or severely ill and by its high morbidity and mortality. The proportion of patients undergoing cholecystectomy for acalculous cholecystitis rises to 50% in those who develop postoperative acute cholecystitis and to 92% following major trauma (4,8,9). Data from the 1970s and 1980s show that the incidence of acute acalculous cholecystitis is increasing, with little improvement in overall mortality (10,11). Much of this high mortality is related to failure of diagnosis, delay in treatment, the condition of the patient, and the development of complications, including gallbladder gangrene and perforation. This is in marked contrast to the relatively benign outcome observed in patients with calculous disease. For these reasons it has been suggested that acalculous cholecystitis be referred to as necrotizing cholecystitis, differentiating it from acute calculous cholecystitis. B— Pathogenesis The term acute acalculous cholecystitis refers to an acute necroinflammatory condition of the gallbladder. From an etiological standpoint, four broad pathogenic mechanisms can be identified: (a) ischaemia, (b) infection, (c) chemical injury, and (d) gallbladder obstruction. In the majority of cases, one or several of these etiological factors is likely to be important. 1— Ischemia There is considerable evidence to support ischemia as an important cause of acalculous cholecystitis, particularly when there is progression to gangrene and perforation. Experimental data have shown that the gallbladder may be particularly susceptible to ischemic injury. An ischemic etiology is the primary cause in patients with primary vasculitides (12–14), embolic disease (15), and abdominal visceral ischemia (16). But it is also thought to play a causative role in a
Page 594
number of clinical scenarios associated with decreased splanchnic blood flow. For instance, it is well documented that shock (8), dehydration (17), hemorrhage (18), and hypoxemia are all risk factors (see Table 1). An increased incidence of acute acalculous cholecystitis is also reported in patients undergoing cardiac surgery, particularly when surgery results in diminished splanchnic circulation as occurs in the repair of a ruptured aortic aneurysm or with cardiac valve replacement. Furthermore, in these clinical settings, the incidence of cholecystitis is unaffected by the presence of gallstones (19–22). Mucosal ischemia can result from gallbladder distention with high intraluminal pressures. Distention may be due to (a) a switch from the normal absorptive function of the gallbladder to secretion, (b) accumulation of thick tenacious biliary sludge, (c) impaired gallbladder contraction, and (d) increased resistance to gallbladder emptying due to biliary sludge or mechanical resistance of the cystic duct or sphincter of Oddi (23). An unusual cause of gallbladder ischemia is torsion of gallbladder, which occurs predominantly in elderly patients with a ''floating gallbladder" or visceroptosis (24). Studies of the vasculature of gallbladders removed for both calculous and acalculous cholecystitis have shown that the blood supply in patients with acalculous disease is clearly abnormal (25). In calculous disease, dilatation of both the larger and smaller blood vessels with vascular congestion is seen. In contrast, in acute acalculous cholecystitis, arterial occlusion is almost always observed and probably explains the higher incidence of gangrene and perforation in these patients. Current evidence would suggest that the gallbladder vasculature is particularly susceptible to the effects of inflammatory mediators and activation of the coagulation system, resulting in vasospasm and vascular occlusion (26,27). The increased susceptibility of the gallbladder vasculature to injury is supported by several case reports of acalculous cholecystitis in patients undergoing intraarterial infusion of lipiodol and other chemotherapeutic agents, with relative sparing of other intraabdominal organs (28–30). Similar effects have been seen in patients receiving systemic interleukin2 therapy (40,41). 2— Infection Gallbladder infection may be either primary or secondary. Positive cultures of bile and the gallbladder wall, obtained at the time of cholecystectomy, are found in 28 to 60% of cases (31,32). In the majority of these, the histological findings are not compatible with primary infection; infection is thought to be a secondary event (33). It should be noted, however, that superinfection may intensify the inflammatory response and that the presence of bacteria is strongly associated with infective complications, including wound infection, septicemia, and subhepatic and phrenic abscesses. Escherichia coli is consistently the commonest pathogen isolated (3,7,34,35) and, together with Streptococcus Enterococcus and Klebsiella, accounts for over 75% of biliary bacteria identified. Mixed infections are common, and the frequency of anaerobic infections is probably underestimated. Using careful sample handling and culture techniques, up to 40% of patients will have anaerobic organisms identified. Whether conditions associated with increased biliary colonization, such as duodenal diverticula, predispose to acalculous cholecystitis or infective complication is unknown (36). Cholecystitis due to primary infection can occur as a direct result of bacterial, fungal, viral, protozoal, and parasitic infections (see Table 1). Immunosuppression appears to increase the risk of infective acalculous cholecystitis; in particular, there is a clear association of acalculous cholecystitis with the acquired immnnodeficiency syndrome (AIDS) and organ transplantation. Most cases occurring in those with AIDS are due to cytomegalovirus (CMV) and Cryptosporidium, though microsporidial and bacterial infections are also found. In the immunocompetent patient, acalculous cholecystitis is caused by organisms with a tropism for the gallbladder, such as the Salmonella species (37). Distant or systemic infection may also induce acalculous cholecystitis. For instance, lipopolysaccharide has been shown in animal models to be capable of inducing an inflammatory
Page 595 Table 1 Organisms Implicated as the Primary Cause of Acalculous Cholecystitis Organism
References
Salmonella typhi
37,124–126
Salmonella (nontyphoidal spp.)
128–131
Vibrio cholerae
259–261
Staphylococcus
137
Leptospira
133–135
Listeria
262
Pneumocystis cariniia
148
Mycobacterium avium intracellulare Mycobacterium tuberculosis a
a
a
201 201
Microspora (Enterocytozoon bieneusi and Septata intestinalis)
148,160,161
Cryptosporidiuma
148,155
a
148,263
Isospora
Cytomegalovirus
a
151–159
Candida
90,138–142
Ascaris
144
Echinococcus
143
a
Primarily seen in patients with the acquired immunodeficiency syndrome (AIDS).
response in the gallbladder (38,39), which can be inhibited by the administration of indomethacin. Similarly, the systemic release of proinflammatory mediators, as in shocked patients, would be expected to result in a similar inflammatory response. These observations are in keeping with the apparently increased susceptibility of the gallbladder vasculature to inflammatory mediators, as described above. It is therefore likely that increased systemic levels of proinflammatory and vasoconstrictive substances released in response to infection, burns, surgery, and trauma all play an important role in the pathogenesis of acute acalculous cholecystitis. 3— Chemical Injury Several of the constituents of bile have been shown to induce a gallbladder mucosal inflammatory response. These include bile salts, cholesterol, and lysolecithin. In animal models, cholesterolsupersaturated bile induces a gallbladder mucosal inflammatory response prior to the appearance of gallstones or cholesterol crystals (42). The mechanism by which supersaturated bile induces gallbladder inflammation has not been fully elucidated, but increased absorption of cholesterol by the gallbladder mucosa has been suggested. Several observations support a proinflammatory effect of cholesterol. First, cholecystitis at least in animal models, does not develop following cystic duct occlusion except in the presence of supersaturated bile (43). Second, tissue injection of cholesterolrich solutions results in an intense inflammatory response, triggering an arteritis (44). Third, an inflammatory response to tendon xanthomas is not an uncommon finding in patients with hypercholesterolemia (45). Lysolecithin is also an important mediator of inflammation and is likely to play a substantial role in the pathogenesis of acalculous cholecystitis. In animal models, lysolecithin has been shown to induce both cholecystitis and a gallbladder secretory response (38,46–48). In humans, relatively high levels of lysolecithin are found in acalculous as compared to calculous cholecystitis (49,50). In the healthy gallbladder, lysolecithin is rapidly cleared, protecting it from the harmful effects of high concentrations (51). However, in an injured or dysfunctional gallbladder, there is an accumulation of lysolecithin, resulting in an unfavorable and toxic ratio of lysolecithin to lecithin (31,52). The secretory response induced by lysolecithin and other
Page 596
inflammatory mediators also contributes to gallbladder distention, increased gallbladder wall tension and decreased vascular perfusion, as already discussed. The metabolism of native lecithin to lysolecithin probably occurs within the gallbladder as a result of the release of phospholipase A from lysosomes and bacteria. The trigger for the lysosomal release of phospholipase A is unclear but may be related to ischemia, obstruction, or as a response to systemic inflammatory mediators. Whether this is an early event or whether gallbladder injury secondary to some other process is required for the release of tissue phospholipases is unclear. Nevertheless, lysolecithin is likely to contribute to the severity of the gallbladder injury and may have particular importance for the pathogenesis of acalculous cholecystitis. Lysolecithin is also a mucin secretagogue (53). Hypersecretion of mucin is a prerequisite for the formation of biliary sludge, a tenacious material, which inhibits gallbladder emptying and promotes cholesterol crystal nucleation. Defects in both gallbladder emptying and biliary sludge are common findings in patients with acalculous cholecystitis and are early events in the pathogenesis of cholesterol gallstone disease (54). The third major group of chemical mediators of gallbladder inflammation is bile salts. Bile salts are directly injurious to membranes (55–57) and have been shown to induce inflammation in experimental systems (58). It has also been observed that the concentrations of the more toxic deoxycholates are higher in patients with gallstones than in those without stones. It is therefore proposed that high concentrations of toxic bile acids, increased concentrations of lysolecithin, and a reduction in lecithin concentration act in concert to induce a profound inflammatory response (59). A unifying theory would propose that many of the clinical factors that predispose to acalculous cholecystitis lead to cholesterol supersaturation of bile, increased production of lysolecithin, and alterations in the bile salt pool, which act together to produce gallbladder inflammation. 4— Obstruction Obstruction to gallbladder emptying is associated with cholecystitis and is felt to be one of the primary events in patients with calculous cholecystitis. Impaired gallbladder emptying is also thought to contribute to cholecystitis in patients with acalculous disease (60). Gallbladder obstruction may be caused by gallbladder tumors (61), biliary sludge (62), and compression of the cystic duct (63). Increased resistance to gallbladder emptying occurs as a result of the administration of opiates (60,64), papillary edema (23), and continuous positivepressure ventilation (60,65,66). Biliary stasis, which is associated with diminished gallbladder clearance and formation of biliary sludge, can result from fasting and parenteral feeding (67,68). Obstruction or failure of adequate gallbladder emptying alone is not sufficient to induce cholecystitis. In animal models, cystic duct ligation results in cholecystitis only when it performed in the presence of cholesterolsupersaturated lithogenic bile (43). Biliary sludge is commonly observed in critically ill patients (69) and is a key factor in the pathogenesis of acalculous cholecystitis. Sludge is a tenacious mixture of mucin, biliary proteins, cell debris, and cholesterol crystals that precedes the formation of mature cholesterol gallstones (62). Increased mucin secretion is thought to result from mucosal inflammation and is an early event in animals fed a cholesterolenriched diet (70). Many of the factors associated with the formation of gallbladder sludge are also risk factors for acalculous cholecystitis (69). Passage of sludge into the common bile duct as well as abnormalities of sphincter of Oddi function probably account for a significant proportion of the obstructive features observed in patients with acalculous cholecystitis (71). Accumulation of biliary sludge is due to both the high viscosity of sludge as well as impaired gallbladder emptying in response to physiological stimuli (72). Sludge usually resolves rapidly after resolution of the acute illness and reinstitution of a normal diet, but this is not universal (69,73). It should not, however, be considered a benign finding, as the incidence of subsequent gallstones and cholecystitis is significant, occurring in 19.6% of patients in one series (73).
Page 597
B— Pathology Pathological studies of acute acalculous cholecystitis show an acute inflammatory infiltrate of both the gallbladder wall and vessels (74). As already mentioned, vascular studies have shown that the vascular supply is frequently compromised (25), which may result in focal necrosis and gangrene of the gallbladder wall, with perforation in fulminant cases. While a polymorphoneutrophilic infiltrate predominates, a series evaluating the frequency of eosinophilic infiltration in cases of acute and chronic acalculous cholecystitis has shown that there is also a higher prevalence of eosinophils in acalculous cholecystitis as compared to calculous disease (75). As with eosinophilic enteritis, eosinophils may be found in the serosa, muscular layer, or mucosa. The reason for the presence of a dense eosinophilic infiltration remains unclear, but this may be an important contributor to the severity of injury. C— Clinical Presentation 1— The Clinical Setting From the above discussion, it is clear that the pathogenesis of acute acalculous cholecystitis is multifactorial in the majority of cases. In a small number, acalculous cholecystitis occurs as the direct result of a biliary infection (see Table 1) or involvement of the gallbladder by a vasculitis (12,14). Risk factors that predispose to the development of acalculous cholecystitis have been well documented and are shown in Table 2. However, these risk factors are identified in only 50% of cases; the remainder have no clearly identified cause (76). Of all cholecystectomies performed for acute cholecystitis, between 4 and 17% are for noncalculous disease (1–3,5,6,76). The percentage rises to almost 50% in postoperative cases and 92% in posttraumatic cases (9), implicating surgery and trauma as important causes of acute acalculous cholecystitis independent of the presence of gallstones. Studies from Vietnam Table 2 Clinical Risk Factors for the Pathogenesis of Acute Acalculous Cholecystitis Risk factors
References
Total parenteral nutrition
34,67,78,80,82
Septicemia, biliary infections
261,264
Major trauma
8,32,103–114
Burns
17,98–102
Nonbiliary surgery, cardiac aneurysm repair
19,21,88,185,265,266; 22,84
Childbirth
Multiple blood transfusions
74,92,267,268
Broadspectrum antibiotics
269,270
Mechanical ventilation
5,66,105,267,268,271
Opiates
64,101
Immunosuppression— chemotherapy, AIDS, transplantation
90–92,94,95,146,147,150,151,153,154, 156 162,201,263,272276
Diabetes mellitus
10,97
Ischemic heart disease
Peripheral vascular disease
277
Malignancy
61
Chemotherapy
30,40
Fasting
Page 598
have also identified repeated blood transfusion as a risk factor (18). Clinical factors that have been shown to induce gallbladder stasis—such as assisted ventilation, parenteral nutrition (34,64,67,77–82), and the administration of opiates that increase sphincter of Oddi pressures—have all been shown to be associated with an increased risk of acalculous cholecystitis. Impaired gallbladder emptying and a switch of mucosal function from absorption to secretion results in increased gallbladder wall tension and ischemia (8,23). In this setting, the hypotensive and hypoxic patients are particularly at risk of ischemic injury. 2— Critical Illness and Surgery Conditions associated with acute acalculous cholecystitis are detailed in Table 2. The earliest reports of acalculous cholecystitis were in severely ill postoperative patients (4,83,84). Since that time the incidence of acalculous cholecystitis has been rising. This is due in part to the increasing age of the population, a clear risk factor. Furthermore, surgical and medical procedures have become progressively more complex with prolonged intensive care and a high incidence of immunosuppression due to chemotherapy, illness, and HIV infection (74,85). In several studies, there is a clear predilection for acalculous cholecystitis to occur in men (84,86), while the incidence of gallstonerelated cholecystitis in critically ill patients has an equal gender incidence. The incidence of acute cholecystitis complicating surgical procedures is not insignificant, and the risk is particularly high in those undergoing emergency surgery (21). In a series of 703 patients who had undergone abdominal aortic aneurysm repair, acute cholecystitis occurred in 1.1%; approximately half of these had acalculous disease (19). Therefore the presence of gallstones is not a strong predictor of this complication. The reported incidence of 0.12% in patients undergoing elective cardiac surgery is significantly less, but again, approximately half of these occur in the absence gallstones (86,87). Overall the incidence is highest when surgery is performed on patients with diminished cardiac output or in cases where there has been associated shock or hypoxemia (22). In a report of 22 patients with acalculous cholecystitis, all were receiving both inotropic support and morphine and a large proportion required artificial ventilation (82%) or the use of an aortic balloon pump (88). Artificial ventilation is a clear risk factor, with the highest incidence in those requiring positive endexpiratory pressure (PEEP) (5,23,66,89). PEEP is not only related to the severity of the respiratory disease but also associated with decreased portal venous blood flow and increased hepatic vein pressures, both of which are likely to be contributing factors to acalculous cholecystitis. 3— Transplantation Many cases of acute acalculous cholecystitis have been reported following a variety of transplant procedures, including heart, kidney, and bone marrow (90–95). However, the absolute incidence of acalculous cholecystitis in these conditions appears to be low. Of 771 patients who underwent bone marrow transplantation, Jardines et al. reported 5 (0.6%) cases of acute acalculous cholecystitis (92). The risk is highest in those who receive ABOincompatible bone marrow transplantation and who require exchange transfusion (92). The number of blood transfusions and total parenteral nutrition are also identified risk factors in this clinical setting. 4— Chemotherapy Hepatic arterial infusion of lipiodol and a number of chemotherapeutic agents are associated with acalculous cholecystitis secondary to a chemically induced arteritis (96). It is currently recommended that patients who receive hepatic arterial infusions of floxuridine undergo cholecystectomy prior to treatment (28–30). Acalculous disease has also occurred with immunomodulatory therapy, such as interleukin2 and lymphokine activated killer (LAK) cell therapy for malignancy (40,41).
Page 599
5— Iatrogenic Cholecystitis Acute obstruction of the cystic duct following instrumentation of the biliary system, either percutaneously or following ERCP, has also been reported to cause acute acalculous cholecystitis (97). Cholecystitis may result from either an ascending bacterial infection (97), occlusion of the cystic duct by a stent, or a combination of these. Stentinduced acalculous cholecystitis usually responds rapidly to removal of the stent. 6— Posttraumatic Cholecystitis A reduction in peripheral and splanchnic circulation as well as the systemic release of inflammatory mediators probably explains the association of burns (17,98–102) and trauma (8,32,103–114) with acalculous cholecystitis. For instance, acalculous cholecystitis was not an infrequent complication of trauma and surgery during the Vietnam war (18). The interval between trauma and the development of symptoms is often long, acalculous cholecystitis developing a mean of 20 days after the injury (range 9 to 40 days) (32,64). Factors associated with the development of acalculous cholecystitis are similar to those reported for surgical patients and includes mechanical ventilation, use of PEEP, high doses of narcotics, shock with a high transfusion requirement, parenteral nutrition and renal failure (64). More recently an association between spinal cord injury and acalculous cholecystitis has been described (104,107,115). In a series of 191 patients admitted to the intensive care unit with an acute traumatic spinal cord lesion, 7 (3.6%) developed acute acalculous cholecystitis (115). 7— Total Parenteral Nutrition Total parenteral nutrition (TPN) predisposes to the development of acute acalculous cholecystitis in both adults and children (67,77,82,116,117). About 4% of all patients receiving TPN for more than 3 months will develop acalculous cholecystitis (118), and the incidence correlates with the duration of parenteral feeding (80). Serial ultrasound studies performed prospectively on patients receiving TPN have shown that the percentage of patients with biliary sludge may be as high as 100% after 6 weeks of treatment (119). In a series of 14 patients, 6 developed gallstones and 3 of these required cholecystectomy. In a larger study the incidence of acalculous cholecystitis and development of gallstones was 4 and 19%, respectively (80). The increased incidence of acalculous cholecystitis observed in patients receiving TPN has largely been ascribed to failure to stimulate normal gallbladder contraction during prolonged fasting. However, acutely ill patients appear to have a defect in the contractile response of the gallbladder to enteral feeding such that feeding by this route may not be protective, but no prospective studies have been performed (72). 8— Outpatient Presentation The increasing incidence of acalculous cholecystitis has been attributed to the larger number of patients who survive severe trauma, are immunosuppressed, are receiving chemotherapy or have undergone complex surgery. However, it has also been noted that the number of patients presenting with de novo acute acalculous cholecystitis is also increasing (120). This has been attributed to a rising incidence in elderly males. In a review of 47 cases of acute acalculous cholecystitis, 36 (77%) presented as outpatients and 11 (23%) occurred in hospitalized patients (120). Of those patients presenting from home, a high proportion were male and had a history of significant ischemic vascular disease (hypertension, angina, peripheral vascular disease, congestive cardiac failure) and diabetes (121). There have been several case reports of acalculous cholecystitis due to gallbladder involvement by a vasculitis (12–14) including systemic lupus erythematosus (SLE) (122) and the antiphospholipid syndrome (123). However, in some of these, cholecystitis may have been a result of aggressive immuno and myelosuppressive therapy rather than a direct result of vasculitis (122).
Page 600
9— Infective Causes Infective causes of acalculous cholecystitis can occur and result from infection with organisms with a natural tropism for the gallbladder (e.g., Salmonella) or may be due to opportunistic infections in immunosuppressed individuals (e.g., AIDS). Salmonella cholecystitis has been reported with both typhoidal (37,124–127) and nontyphoidal species (128–131). Indeed the gallbladder is the most common site of focal salmonella infection (132), Salmonella typhi or paratyphi being the most commonly isolated species. Cholecystitis due to other species is much less common (131). Bacterial cholecytitis may progress despite appropriate antibiotic therapy; as a result, a significant proportion of patients will require cholecystectomy to prevent gallbladder gangrene and perforation. Cholecystitis secondary to a wide variety of other organisms has been well described. These include Leptospira (133–136), Staphylococcus (137), and Candida (90,138–142). Candidal cholecystitis is most often reported in diabetics, immunosuppressed patients or in severely ill patients receiving longterm antibiotics (140). In most cases there is associated candidal septicemia (140). There have been several reported cases of acalculous disease occurring as a result of biliary involvement with Echinococcus (143) and Ascaris (144,145) from regions of the world where infection with these organisms is common. In these cases cholecystitis results from biliary or cystic duct obstruction rather than by direct invasion of the gallbladder. 10— HIV Acalculous cholecystitis is described with significant frequency in AIDS patients. In a surgical review of all surgery performed in 904 HIVpositive patients, cholecystitis was the most common indication and carried a high mortality in those with advanced AIDS (33%) (146). HIVpositive patients can present with either acute or more chronic symptoms with or without features of AIDS cholangiopathy (147). An opportunistic infection will be identified in 50 to 75% of these (146,148– 150). The most commonly identified pathogens include cytomegalovirus (CMV) (151–159), Cryptosporidium (148,155) and Microspora (148,160,161). In a retrospective review of 107 AIDS patients who underwent cholecystectomy for both acute and chronic cholecystitis, pathological evidence of an opportunistic infection was found in 47% with acalculous disease as compared to 22% of those with calculous cholecystitis (148). Biliary colonization with Cryptosporidium is found in over 80% of those with intestinal infection at the time of cholecystectomy, but gallbladder disease may be the first presentation of both cryptosporidial and CMV disease. In CMV cholecystitis, the mucosa may be deeply ulcerated (153), supporting a primary pathogenic role for CMV in this condition. Whether Cryptosporidium is directly responsible for the development of acalculous cholecystitis continues to be debated; nevertheless, the association is strong. In about 80% there is biochemical evidence of bile duct involvement, but in the majority serum bilirubin concentrations are within the normal range (162, 163). The introduction of highly active antiretroviral therapy (HAART) has brought about a dramatic reduction in the incidence of opportunistic infections, and new cases of Cryptosporidium, Microspora, and CMV infection have become less common. A similar reduction in acalculous cholecytitis should be observed in adequately treated patients. 11— Pediatric Cholecystitis Acalculous cholecystitis accounts for a significantly higher proportion of cholecystitis in children than in adults (76). Calculous cholecystitis is almost completely confined to children with hemolytic diseases and cystic fibrosis; thus acalculous disease accounts for almost 70% of all cases of cholecystitis in children (84). In most children it occurs in association with a severe systemic infection, including typhoid, scarlet fever, measles and AIDS (125,126,164,165). Cholecystitis also occurs in severely ill infants (166–173). Recognized risk factors include severe
Page 601
diarrhea, respiratory tract infections (173), vasculitides (173,174), hepatitis, and previous cardiac surgery (175). In children without a clear precipitating cause congenital biliary tract abnormalities should be sought. B— Clinical Presentation The clinical presentation of patients with acalculous cholecystitis may be clinically indistinguishable from that of calculous disease. However the severely ill patient is a substantial clinical challenge because of the multitude of other clinical problems and the patient's inability to report abdominal tenderness on clinical examination. Approximately 50% present as outpatients (7,176). Most present with abdominal pain; about of half of these will localize the pain to the right upper quadrant, the remainder in the epigastrium, and a small proportion in the left upper quadrant. The pain is usually described as rapid in onset, rising to maximum severity in 30 min to 1 h and often lasting for several hours. The pain is noncolicky but has a constant character and may radiate to the back or shoulder. Extension of the inflammatory process to the gallbladder serosa results in localization of the pain to the gallbladder fossa, with associated rightupperquadrant tenderness, guarding, and a positive Murphy's sign. The gallbladder is palpable in about 25% of cases (7). Onethird of patients have an associated fever and most (70%) have an elevated white blood cell count (176). Patients often have abnormal liver function tests (see below) and jaundice is not uncommon (1,3,32,81,177,178). Kalliafas and colleagues reported elevation of bilirubin levels in 64%, alkaline phosphatase in 40%, ALT in 40%, and AST in 52% (176). The presence of jaundice probably reflects extension of the inflammatory process from the gallbladder to the common bile duct as well as obstruction of the common bile duct by biliary sludge. Unfortunately, in those patients with concomitant illnesses, there are often numerous reasons for jaundice and abnormal liver function tests, which limits the diagnostic value of these findings. The clinical presentation in children is very similar (130). Gallbladder gangrene and perforation often lead to rapid deterioration, with sepsis, shock, and features of generalized peritonitis. In many series the diagnosis is confirmed or established only at the time of an exploratory laparotomy or following percutaneous drainage of the gallbladder (77). In the severely ill unconscious patient, the only indication of acalculous cholecystitis may be further unexplained clinical deterioration. This may include a reduced level of responsiveness, increasing fever, progressive hypoxia, or hemodynamic instability. Evidence of biliary disease is supported by progressive elevation in liver enzymes and progressive hyperbilirubinemia. Gallbladder perforation is often heralded by the development of ileus, increasing abdominal tenderness, distention, and rigidity. The diagnosis of acute acalculous cholecystitis in severely ill patients is frequent enough that it should always be considered as a possible cause of clinical deterioration. 1— Clinical Presentation in HIVPositive Individuals Acalculous cholecystitis in HIVpositive patients presents most frequently in the terminal phases of HIV infection, when the CD4 count is less than 100 and is usually associated with an opportunistic infection of the biliary system. In these cases the presentation may be of either acute or chronic acalculous cholecystitis and the presence or absence of associated intestinal or common bile duct infection modifies the clinical presentation. Those presenting with acute acalculous cholecystitis usually present with fever, and rightupperquadrant pain; a positive Murphy's sign is detected in over 50% (153). In chronic cases the presentation is often associated with many weeks or months of right upper quadrant pain. Unlike patients with acalculous cholecystitis of other causes, AIDS patients with pure cholecystitis often have normal serum bilirubin and alkaline phosphatase estimations (149,150,155,162). However, approximately 40 to 80% will have associated biliary tract disease, either AIDS cholangiopathy or papillary stenosis; in these cases elevation of the cannal
Page 602
icular enzymes is almost always present and therapy will have to be directed at both the gallbladder and biliary tract. Highdose paramomycin will not reach the biliary tree and is not a suitable treatment for these patients. As a result, the biliary tree may act as a reservoir for intestinal reinfection. Longterm therapy should be aimed at controlling the HIV infection. D— Radiological Diagnosis 1— Ultrasound Radiological testing is often the cornerstone of diagnosis. In most cases ultrasound is used as the primary modality (7), due to its portability and ability to exclude gallstones and biliary duct dilatation. In the absence of gallstones, the following suggest the diagnosis of acute acalculous cholecystitis (179–181): gallbladder distention, wallthickening, pericholecystic fluid, and ultrasound probe—induced tenderness (ultrasound Murphy's sign). Thickening of the gallbladder wall to greater than 3 to 3.5 mm is normally considered significant but must be interpreted with great caution in severely ill patients. Falsepositive examinations are frequent in patients with gallbladder sludge, ascites, hypoalbuminemia, and conditions such as cholesterolosis of the gallbladder (182). Other useful ultrasonographic findings include intramural gas or gas bubbles within the gallbladder, the "Champagne sign." These indicate emphysematous cholecystitis due to infection with a gasforming organism, most commonly a clostridial infection. A sonographic Murphy's sign (i.e., pain over the gallbladder due to pressure applied with the ultrasound probe) increases the specificity of the above findings. The presence of sludge alone should not be taken as evidence of acute acalculous cholecystitis, and intraluminal blood or pus may also be misinterpreted as sludge (182). The diagnostic accuracy of ultrasonography has been assessed in a large number of studies with a reported accuracy ranging from 25 to 100% (3,120,183–185). However, ultra
Figure 1 Ultrasound of the gallbladder demonstrating a grossly thickened gallbladder wall (marked). The edematous submucosa can clearly be seen as a dark rim. Note that there is a small amount of echogenic material within the gallbladder, with acoustic shadowing.
Page 603
sound has many pitfalls and the prevalence of significant ultrasonographic abnormalities in severely ill patients is high, despite a low incidence of clinically significant disease. In a prospective sequential ultrasound study of 45 patients following major trauma, a high proportion showed gallbladder abnormalities (108). Sludge was identified in 26 patients, thickening of the gallbladder wall (>3.5 mm) in 17, and 17 had gallbladder distention. Eight patients had all three features and were diagnosed as having acalculous cholecystitis, but only one of these required cholecystectomy. Therefore, single ultrasonographic criteria should be interpreted with caution and correlation with clinical findings is essential. More recently Helbich et al. proposed a scoring system to aid the ultrasonographic diagnosis of acute acalculous cholecystitis in severely ill patients (186). They graded gallbladder distention, wall thickening, sludge, striated thickening of the gallbladder wall, and the presence of pericholecystic fluid. Patients with scores below 5 were never diagnosed with cholecystitis, but higher scores were less than 50% accurate for the diagnosis of acute disease. Ultrasound can thus be used to exclude acalculous cholecystitis, but a "positive" scan should be interpreted carefully, and further testing or therapy must be guided by the clinical situation. Dynamic ultrasonography, using the contractile response to cholecystokinin has been suggested as a method to improve diagnostic accuracy. Raduns et al. reported on cholecystokinin ultrasonography in 15 posttrauma patients in whom cholecystitis was excluded at laparotomy (187). In only 4 patients was a normal contractile response (>50% emptying) to cholecystokinin observed. In 4, no response to cholecystokinin was observed. This study demonstrates both the reduced responsiveness of the gallbladder to cholecystokinin stimulation as well as the limited value of functional studies in severely ill patients. As a result, dynamic ultrasonography is not widely used at the present time; where there are suggestive or equivocal findings, alternative radiological tests should be performed. 2— Nuclear Medicine Studies Radionuclide studies have often been used to help confirm or refute the diagnosis in patients with suspected acalculous cholecystitis (81,88,120,188). Failure of the gallbladder to concentrate technetium iminodiacetic acid (IDA) derivatives due to cystic duct obstruction has been shown to be highly predictive of acute calculous cholecystitis, but its accuracy in acalculous disease is less good. In acalculous disease, failure of gallbladder filling may be due to poor gallbladder function or the accumulation of gallbladder sludge in the absence of acute gallbladder inflammation. Other features that are sometimes observed include bowing of the common bile duct due to a gallbladder distention or leakage of the radionuclide into the peritoneum or subhepatic space due to perforation. Many series assessing nuclear medicine scans have reported excellent sensitivity for the demonstration of acalculous cholecystitis (3,188–191). Unfortunately, many of these have been hampered by poor methodology; they are usually retrospective, including only patients with confirmed disease; and they have included no negative controls (189). When critically analyzed, radionuclide studies are hampered by a significant rate of falsepositive tests (190,192,193). In acalculous as opposed to calculous disease, the cystic duct is less often occluded (194,195), but poor hepatic excretory function and a poorly functioning gallbladder lead to falsepositive studies. Up to 30% of asymptomatic fasting patients who are receiving TPN will have a falsepositive examination, significantly affecting the specificity of a positive test result (193). Mirvis et al. reported a 54% rate of falsepositive tests but a low falsenegative rate of 5.2% (184). Falsenegative studies can occur as a result of focal acalculous cholecystitis (196), though dimethyl iminodiacetic acid (HIDA) scanning is particularly sensitive for the detection of gallbladder perforation (3). Despite these reservations, HIDA scintigraphy has been shown to have the best diagnostic accuracy when compared to ultrasonography and CT scanning (176). 3— MorphineAugmented HIDA Scintigraphy As already stated, the use of radionuclide scanning is hampered by the high rate of falsepositive studies, which is particularly true in severely ill patients (190,197,198). To improve
Page 604
the specificity of this test, several investigators have performed the IDA scan with the administration of morphine (3,176,190,199,200). Morphine enhances gallbladder filling by increasing sphincter of Oddi pressure and should reduce the number of falsepositive tests. While several studies have reported excellent results using this technique (3,176,190,199,200), other reports have been less encouraging (198). Fig et al. reported a sensitivity of 94%, but the specificity was only 69%, with a falsepositive rate of 40% (198). It therefore appears that while morphine administration improves the performance of radionuclide studies, one must still interpret a positive scan with caution (188). Morphineaugmented IDA scans are also of limited additional benefit in HIVpositive individuals (201). 4— Leukocyte Scintigraphy Radiolabeled leukocyte scintigraphy has also been used to diagnose cholecystitis (188,202,203). In a small series, TcHMPAO—labeled leukocyte scanning correctly diagnosed acute cholecystitis in 16 of 17 patients, with no falsepositive scans (202). Only two of the patients in this series had acute acalculous cholecystitis, both of who were correctly diagnosed. A scanning time of 4 h limits the clinical utility of leukocyte scintigraphy. 5— CT Scanning CT diagnosis of acute acalculous cholecystitis is thought to be as accurate as ultrasonography (184). Features sought are similar to those described for ultrasound examinations; thickening of the gallbladder wall, pericholecystic fluid, and intramural edema (the ''halo sign") (65,183,184,204). CT also appears to be an excellent method for detecting emphysematous
Figure 2 Computed tomography scan of the abdomen enhanced with intravenous contrast. The grossly edematous gallbladder can be seen with enhancement of the gallbladder wall, consistent with an acute inflammatory process.
Page 605
cholecystitis (205–207). However, despite some reports of increased sensitivity (183,176), it is not thought to be superior to ultrasonography and there is also a significant rate of falsenegative tests (208). Based on the cost and portability of ultrasound, ultrasound remains the test of first choice for the initial evaluation of these patients. E— Treatment 1— General Measures The patient should be resuscitated with fluids and electrolytes as necessary. Antibiotics are normally administered after the necessary blood, urine, and respiratory cultures are obtained. They should cover the most commonly identified organisms, including E. coli, Klebsiella, enterococci, and anaerobes, remembering that polymicrobial infection is not uncommon. Therapy will have to be modified to take into account special clinical circumstances—for instance, where Pseudomonas or fungal infection is suspected. One approach is to use triple therapy with ampicillin, gentamicin, and metronidazole, which is time honored and effective in most cases. Alternatively, a thirdgeneration cephalosporin plus metronidazole may be used, particularly in patients with borderline or impaired renal function. It should be noted that ceftriaxone has been associated with the formation of biliary sludge and is probably best avoided in these patients. An alternative would be to substitute the aminoglycoside with a fluoroquinolone, which achieves excellent biliary and tissue concentrations and is particularly valuable in patients with impaired renal function. The use of the newer fluoroquinolones with better grampositive coverage has not yet been reported, but these may offer an excellent alternative.
Figure 3 T2weighted magnetic resonance image showing a dilated gallbladder (GB) of high signal intensity. Also note the significant pericholecystic fluid consistent with an acute inflammatory process. MRI is a very sensitive technique for the demonstration of pericholecystic fluid.
Page 606
Alternative regimens include mezlocillin with an aminoglycoside and metronidazole, imipenem, or ampicillin/sulbactam. Standard antipseudomonal regimens—e.g., ceftazidime plus an aminoglycoside—may be required where pseudomonas is a likely pathogen. 2— Specific Therapy Once the diagnosis of acalculous cholecystitis has been made, the gallbladder should be either removed or drained. Early laparotomy has the advantage of removing the gallbladder and also facilitating the identification of other intraabdominal problems should the cause of the clinical deterioration or abdominal symptoms remain obscure (e.g., missed perforated ulcer, pancreatitis, etc.) With the advent of better radiological studies the need for laparotomy to identify other intraabdominal pathology has diminished. The definitive management of acute acalculous cholecystitis is cholecystectomy, and this should be the treatment of choice in otherwise healthy patients with significant cholecystitis. The laparoscopic approach is now most commonly used, with conversion to an open cholecystectomy in selected patients. In some cases the gallbladder is encased in a dense inflammatory mass, precluding laparoscopic surgery. There are no studies comparing the efficacy and safety of the laparoscopic approach to open cholecystectomy in highrisk patients with acalculous cholecystitis. Where perforation with generalized peritonitis is present, a laparotomy should always be performed. It has been suggested that laparoscopic cholecystectomy be performed in HIVpositive patients because of a lower complication rate (201). The incidence of postoperative complications, abscess formation, biliary leaks, and intraabdominal bleeding is higher in patients undergoing surgery for acalculous as compared to calculous cholecystitis, even in experienced hands (32). In the severely ill patient, or where other conditions preclude open surgery, placement of a cholecystostomy tube either surgically (76,209) or under radiological (34,35,87,178,210–214) control has given excellent results (178) and is probably the method of choice (35,76,178). The surgical approach requires a small subcostal incision, which can be done under local anesthesia. In some cases the presence of a large inflammatory mass may make this approach very difficult. Radiologically guided drainage is performed transhepatically, which limits the risk of peritoneal leakage (35,178,215). Successful drainage should result in a rapid clinical improvement (35,178,216), and the complication rate from this procedure should be low, with a high success rate (211). Complications include bile peritonitis, bleeding, vagally induced bradycardia, respiratory distress, and catheter dislodgement (211). In some cases a cholecystostomy tube may have to be placed as a therapeutic trial where acalculous cholecystitis is suspected but it remains uncertain; in these cases the response rate is high (63 to 94%) (35,217,218). Experience with this technique is increasing, and it is clear that it can be performed safely in the majority of cases even in the face of advanced disease (34). ERCPguided cannulation of the gallbladder with drainage has been reported, but experience is limited and it is probably neither safer nor more efficacious than the percutaneous approach (219). In patients with associated cholangitis, cholecystostomy does not result in adequate biliary drainage and a second procedure (ERCP or percutaneous drainage) may be required. Cholecystectomy should be performed at a later time in all patients with gallstones. Patients without stones may have the tube removed without cholecystectomy (35,220). At least one case of complete resorption of the gallbladder following drainage has been reported. F— Complications The rate of complications appears to correlate closely with the underlying condition of the patient. Gangrene, perforation, abscess, and death is manyfold higher in patients who develop
Page 607
acute acalculous cholecystitis as a complication of another illness as compared to those who present de novo (176). 1— Gangrene Gangrene is a common complication of untreated acalculous cholecystitis; up to 50% of severely ill patients will have evidence of gangrene (1,3,7,81,88), which always precedes gallbladder perforation. A particularly high incidence of gangrene and perforation has been reported in emphysematous cholecystitis (221). 2— Emphysematous Cholecystitis Emphysematous cholecystitis is a relatively rare complication of acalculous cholecystitis. It is more common in acalculous than calculous cholecystitis and is defined by the presence of gas within the gallbladder lumen, the wall, or the tissues adjacent to the gallbladder. It is seen more often in men than women; about 70% of cases are men (210) and up to 40% occur in patients with diabetes mellitus (221). Gasforming organisms, particularly clostridial species, have been isolated in about 45%, in most cases, it is thought that this is due to secondary infection of necrotic tissue rather than a primary event. The incidence of gangrene and perforation in association with emphysematous cholecystitis is about 75 and 20%, respectively (222). The reported mortality for emphysematous cholecystitis is approximately 15% (222). Because of the high incidence of these complications, these complications should be managed by cholecystectomy, with complete removal of gallbladder and all necrotic tissue (84). Successful cholecystectomy using the laparoscopic approach has recently been reported (223).
Figure 4 Emphysematous cholecystitis. Ultrasound of the gallbladder showing sludge and air within the gallbladder lumen.
Page 608
3— Gallbladder Perforation Perforation of the gallbladder complicates 10 to 20% of all cases of acalculous cholecystitis (8,33,64,88) and is more common in men than women. Gallbladder perforation may present fulminantly, within 48 h of the diagnosis of acute acalculous cholecystitis, or late (1,2,60). Niemeier has proposed a classification of gallbladder perforation (224): type I, acute free perforation with bile stained peritoneal fluid; type II, subacute perforation with inflammatory reaction surrounding the gallbladder or associated with a rightupperquadrant abscess; and type III, chronic perforation with cholecystoenteric or cholecystocutanteous fistula. Perforation more commonly complicates acalculous than calculous cholecystitis (225). The majority of patients present with rightupperquadrant pain, fever, nausea, and vomiting. In about 20% a mass is palpable in the right upper quadrant. With free perforation there is often an associated ileus. The incidence of hyperbilirubinemia is high, affecting about 50% of cases, though only a small proportion are frankly jaundiced (225). Not infrequently the presentation is subacute, such that a preoperative diagnosis is not made (225). Organisms are identified in about 75% of patients at the time of surgery and demonstrate the typical biliary pattern, with E. coli, Klebsiella, and other gramnegative bacilli. 4— Empyema of the Gallbladder While classically described as a complication of calculous disease, in 15 to 20% there are no gallstones present (226). Patients with empyema are often acutely ill and have a palpable and tender gallbladder. Treatment should be immediate percutaneous drainage (227) or cholecystectomy. Unfortunately, even with optimal treatment, there is a high incidence of complications, primarily infective in nature, including wound and intraabdominal infections (226). G— Prognosis The mortality rate of 5 to 20% is significantly greater than that observed in calculous cholecystitis, though in patients who present as outpatients the observed mortality is only slightly higher than for those with calculous cholecystitis. However in highrisk groups, such as those who are immunosuppressed or are recovering from major trauma or surgery, mortality rates as high as 67% have been reported (1,7,19,21,23,88). Furthermore, there has been no significant reduction in the mortality rate despite improvements in diagnostic studies and therapy (10,11). The high incidence in elderly patients and patients who are receiving intensive medical care probably explains the failure to achieve a better outcome and also reflects the changing epidemiology of this disease. II— Chronic Acalculous Cholecystitis A— Introduction Chronic acalculous choloecystitis may be defined both histologically and clinically. Histologically, a chronic inflammatory infiltrate of the gallbladder—which may of may not be associated with other pathological findings, including adenomyomatosis, cholesterolosis, or evidence of cystic duct stenosis (see below)—defines the disease. Clinically, the condition characterized by recurrent or chronic biliary pain. While a large proportion of these cases will have chronic cholecystitis histologically, this is not universal. Radiological investigations may be helpful in identifying patients with abnormal gallbladder morphology or function, but such findings do not always confirm that the gallbladder is the source of the patient's symptoms. As a result, the diagnosis depends as much on the clinical presentation and prior probability that the patient has acalculous cholecystitis as it does on the ability to perform "confirmatory tests." Recent data have suggested that chronic acalculous cholecystitis has increased as a
Page 609
postoperative diagnosis in those patients undergoing cholecystectomy (228). This probably reflects a lower threshold to perform cholecystectomies following the introduction of laparoscopic surgery rather than to an increased burden of disease. B— Pathology Macroscopic examination of the gallbladder usually shows wall thickening and fibrosis, and there may be associated musosal diverticula (RokitanskyAschoff sinuses). Histologically there is pronounced fibrosis of the gallbladder wall and muscular hypertrophy; these findings explain the reduced contractility and diminished gallbladder ejection fraction found in these patients. The relationship of these findings to the clinical presentation remains somewhat unclear. While the majority of patients with biliary symptoms characteristic of chronic acalculous cholecystitis have histological findings (229), this is not universal. Furthermore, a significant proportion of asymptomatic patients have chronic cholecystitis, as has been demonstrated in postmortem studies (230). Hence the correlation between histological findings and clinical features is not strong (230–232). C— Partial Cystic Duct Obstruction It has been suggested that stenosis or tortuosity of the cystic duct may lead to impaired gallbladder emptying and gallbladder inflammation (233,234). The presence of partial obstruction of the cystic duct can be associated with a normal oral cholecystogram, ultrasound, and HIDA scan, making the diagnosis especially difficult. It has been suggested that increased resistance to gallbladder emptying may result in pain following administration of cholecystokinin (CCK) with incomplete gallbladder emptying. As a result, CCK with reproduction of symptoms or inadequate gallbladder emptying has been used as a diagnostic aid (see below). D— Cholesterolosis Cholesterolosis of the gallbladder is a condition of unknown etiology characterized by the deposition of triglycerides and cholesterol in gallbladder epithelial cells, lamina propria, and macrophages. It is thought to result from increased absorption of cholesterol from supersaturated bile. Examination of the mucosal surface reveals numerous yellow specks due to small "cholesterol polyps" on a background of mildly inflamed gastric mucosa. This appearance is commonly referred to as a "strawberry gallbladder." It is thought that these small cholesterol polyps may become dislodged and form the nidus for subsequent cholesterol gallstone disease. Approximately 10 to 15% of patients with cholesterolosis are found to have gallstones. Interestingly, cholesterolosis does not reverse with bile acid therapy. E— Adenomyomatosis Adenomyomatosis involves hyperplasia of the mucosa and muscularis with the formation of intramural crypts called RokitanskyAschoff sinuses. The condition usually comprises the entire gallbladder but may be localized such that an area of hyperplasia can appear as a gallbladder mass. Alternatively, a ring of adenomyomatosis may separate the gallbladder into two separate compartments. The etiology of this condition is poorly understood but is thought to be due to increased intraluminal pressures either because of a gallbladder dysmotility or chronic obstruction to gallbladder emptying. Adenomyomatosis can be detected on ultrasound as a thickened and irregularappearing gallbladder, or the characteristic sinuses can be seen on oral cholecystography. Detection of this condition in association with typical symptoms of acalculous cholecystitis is an indication for cholecystectomy (235).
Page 610
F— Neoplasms Both benign and malignant neoplasms of the gallbladder can present with acalculous cholecystitis as a result of persistent or intermittent obstruction to gallbladder emptying. Biopsy is required to distinguish between a variety of gallbladder masses, including pseudotumours such as inflammatory polyps, cholesterol polyps, heterotopic nests, and adenomyomas. True neoplasms include adenomas and carcinomas; very rarely a variety of other neoplasms are found, including fibromas, lipomas, myomas, myxomas, carcinoids, and hemangiomas. Some 90% of adenomas are solitary and about 10% have evidence of carcinoma in situ. The risk of malignant change correlates with polyp size and may be associated with intestinal metaplasia. Both benign and malignant neoplasms of the gallbladder are often associated with gallstones. However, the frequency with which gallstones are found in association with gallbladder carcinoma varies widely. In Europe and the United States, gallstones are present in 60 to 90% of cases. However, in the Far East, where infectious diseases of the biliary tree are more common, the prevalence of gallstones is significantly lower (24). Secondary tumors of the gallbladder have also been reported; primary sites include pancreas, lung, kidney, ovary, colon, liver, and breast. Usually these are serosal implants, but they may involve the intraluminal portion of the gallbladder and can present with biliary symptoms. G— Clinical Features A review of patients who have undergone surgery for chronic acalculous cholecystitis suggests that these patients are often young, with a significant female preponderance (4,6,230,236). Because of their young age, it has been suggested that chronic cholecystitis with gallbladder dysmotility may be a prelude to the development of gallstones. Indeed, in the evaluation of Brugge et al. of 36 patients with biliary symptoms and a normal gallbladder examination, 16 were found to have cholesterol crystals (237). Furthermore, those patients with cholesterol crystals had a markedly reduced gallbladder ejection fraction, suggesting that defects in both gallbladder contractility and mucosal function are responsible for both the symptoms and the development of cholesterol crystals. Chronic acalculous cholecystitis may present with a variety of clinical complaints. Most commonly patients present with rightupperquadrant or epigastric pain related to meals. The pain may radiate to the back or shoulder and is most often precipitated by eating fatty foods. In some cases the diagnosis is straightforward, particularly where there is characteristic pain and radiological findings suggestive of a diseased gallbladder (see below). Nonspecific symptoms such as increased flatulence, dyspepsia, and poor tolerance of fatty foods are unlikely to be related to acalculous cholecystitis and are therefore much more difficult to interpret, especially when there are equivocal or even suggestive radiological findings. As a result, these symptoms should not prompt gallbladder investigations. On physical examination, there may be right hypochondrial tenderness with a positive Murphy's sign (230). More frequently, the physical signs are unhelpful and other diagnostic studies are necessary. Furthermore the diagnosis may depend upon the exclusion of other causes of abdominal pain, including peptic ulcer disease, chronic pancreatitis, renal calculi, chronic pyelonephritis, and functional bowel disorders. H— Investigations Almost all patients will undergo ultrasonography to exclude gallstones. In patients with clearly defined biliary colic, further workup may not be necessary, as symptoms alone may be sufficient to dictate the need for surgery (230). Those with equivocal symptoms usually undergo radiological evaluation even though the exact role for these tests continues to be debated (230,238–240). Occasionally ultrasonography may suggest a diseased gallbladder by demonstrating the presence of a thickened gallbladder wall, gallbladder polyps, or the presence of Rokitansky
Page 611
Aschoff sinuses. While nondiagnostic, these findings do support the diagnosis and are an indication for surgery in the setting of typical symptoms. In some cases it is necessary to exclude other diseases that may be presenting atypically, including urinary tract disease, pancreatitis, and other biliary diseases including sphincter of Oddi dysfunction. This may require urinalysis, CT scans, and/or ERCP with biliary manometry, bile analysis for crystals, and/or sphincterotomy when indicated. It is therefore not unusual for patients to undergo numerous investigations prior to surgery (232). Since currently available investigations have limited clinical utility, a careful history and examination may help obviate the need for multiple expensive and uncomfortable investigations. Where chronic acalculous cholecystitis is more strongly suspected, further evaluation should be directed at gallbladder studies, including oral cholecystography, ultrasound, or biliary scintigraphy with cholecystokinin (CCK) stimulation (see below). I— Administration of Cholecystokinin Administration of CCK in an attempt to provoke the patient's symptoms has been proposed as a diagnostic test (241–243). CCK is administered as a short infusion and the patient is monitored for the development of abdominal pain. The precipitated pain should have the same characteristics as the presenting symptoms or the test is considered nondiagnostic. Studies with adequate followup have shown that only a small proportion of those patients who have a good response to cholecystectomy have pain with CCK infusion; therefore this test is of limited clinical utility (231,232,234). J— Radiology The value of radiological studies for the diagnosis of chronic acalculous cholecystitis remains a source of controversy (244). Radiological tests aim to demonstrate either abnormalities of structure—for instance, a thickened gallbladder wall) or abnormality of function (e.g., decreased gallbladder contractility or failure to concentrate a contrast agent or tracer). Approaches include evaluation of the gallbladder's ability to concentrate a contrast material or an inadequate contractile response to an appropriate stimulus, such as CCK or a fatty meal (229,245,246). 1— Oral Cholecystography Oral cholecystography has long been used as a mean of confirming normal gallbladder structure and function. RokitanskyAschoff sinuses may be seen, confirming a structurally abnormal gallbladder. Failure of the gallbladder to concentrate oral contrast or decreased emptying in response to a fatty meal or CCK is evidence of abnormal function and is considered a relative indication for surgery (236,247,248). However the measurement of gallbladder emptying using oral cholecystography is thought to be less reliable and more subjective than the results achieved using radionuclide studies (234). As a result, this test has largely been supplanted by the more reproducible and less operatordependent CCK cholescintigraphy. 4— Ultrasound with CCK Stimulation Ultrasound is a particularly valuable test in the evaluation of the gallbladder. It is the most accurate means of excluding gallstones and can often identify adenomyomatosis, polyps, and tumors of the gallbladder. Gallbladder function in response to both a fatty meal and CCK infusion can be assessed by ultrasound (229,245). In a recent study by Barr et al., an ejection fraction of less than 60% was considered abnormal when compared to normal controls (229). Using these criteria, they determined that CCK ultrasonography had a sensitivity and specificity of 75 and 100%, respectively, for the detection of histologically confirmed acalculous cholecystitis. Unfortunately, the authors presented no longterm symptom followup in this cohort.
Page 612
5— Gallbladder Scintigraphy with CCK Stimulation Biliary scintigraphy with CCK stimulation to measure gallbladder ejection fraction has been used as a means of quantifying gallbladder function and to aid in the selection of patients for surgery (73,234,249–253). Measurement of gallbladder ejection fraction is both a measure of gallbladder contraction and the resistance to gallbladder emptying produced by the cystic and common bile ducts. A gallbladder ejection fraction of less than 35 to 50% is usually considered abnormal (234,238), but the correlation between gallbladder emptying and the histology is poor (254). The value of CCK scintigraphy has been assessed in several large studies. Misra et al. reported on the longterm followup of 187 patients with suspected chronic acalculous cholecystitis (238). At a mean of 22 months, patients were contacted and interviewed with regard to persistence or resolution of their symptoms. Of 69 patients with an abnormal CCK HIDA who underwent cholecystectomy, 58 (84%) had complete symptom resolution, 9 (13%) had partial symptom relief, and 2 (3%) had no improvement in symptoms. Histological examination demonstrated that 62% of these patients had abnormal gallbladder histology, predominantly chronic cholecystitis or cholesterolosis of the gallbladder. Some 95% of those with histological findings had complete symptom relief. In contrast, of the 41 patients who had had an abnormal CCK HIDA but did not undergo cholecystectomy, 41% had symptom improvement and 59% had no improvement. Of 44 patients with a normal CCK HIDA, 80% had spontaneous symptomatic improvement and 20% had continued symptoms. These data support the utility of CCK HIDA in the evaluation of patients with a normal ultrasound and symptoms compatible with biliary disease and are supported by other, smaller studies (234). In summary, the bulk of available data would support the use of biliary scintigraphy with CCK stimulation in the evaluation of patients with suspected chronic acalculous cholecystitis. However, the test should be used selectively in patients with pain and not used to evaluate those with nonspecific symptoms. K— Treatment Cholecystectomy for chronic acalculous cholecystitis accounts for 1.3 to 8% of all cholecystectomies (4,6,233). In patients with acalculous disease and welldefined biliary colic, the rate of symptom relief appears to approach that achieved in patients with calculous disease (4,6). Careful patient selection is vital, and it is clear that patients with true biliary colic benefit most from surgery (230,232). Some 80 to 90% of carefully selected patients will have complete symptom relief following surgery (4,232,249,250,255–258). References 1. Fox MS, Wilk PJ, Weissmann HS, Freeman LM, Gliedman ML. Acute acalculous cholecystitis. Surg Gynecol Obstet 1984; 159:13–6. 2. Johnson LB. The importance of early diagnosis of acute acalculus cholecystitis. Surg Gynecol Obstet 1987; 164:197–203. 3. Swayne LC. Acute acalculous cholecystitis: sensitivity in detection using technetium99m iminodiacetic acid cholescintigraphy. Radiology 1986; 160:33–38. 4. Glenn F, Wantz, GE. Acute cholecystitis following the surgical treatment of unrelated disease. Surg Gynecol Obstet 1956; 102:145–153. 5. Howard RJ. Acute acalculous cholecystitis. Am J Surg 1981; 141:194–198. 6. Lee AW, Proudfoot WH, Griffen WO Jr. Acalculous cholecystitis. Surg Gynecol Obstet 1984; 159:33–35. 7. Coelho JC, Campos AC, Moreira M, Moss Junior AA, Artigas GV. Acute acalculous cholecystitis. Int Surg 1991; 76:146–148.
Page 613
8. DuPriest RW Jr, Khaneja SC, Cowley RA. Acute cholecystitis complicating trauma. Ann Surg 1979; 89:84–89. 9. Jonsson PE, Andersson A. Postoperative acute acalculous cholecystitis. Arch Surg 1976; 11:1097–1101. 10. Gutman H, Kott I, Haddad M, Reiss R. Changing trends in surgery for benign gallbladder disease. Am J Gastroenterol 1988; 83:545–548. 11. Reiss R, Nudelman I, Gutman C, Deutsch AA. Changing trends in surgery for acute cholecystitis (see comments). World J Surg 1990; 4:567–570; discussion, 570–571. 12. Imai H, Nakamoto Y, Nakajima Y, Sugawara T, Miura AB. Allergic granulomatosis and angiitis (ChurgStrauss syndrome) presenting as acute acalculous cholecystitis. J Rheumatol 1990; 17:247–249. 13. Papaioannou CC, Hunder GG, Lie JT. Vasculitis of the gallbladder in a 70yearold man with giant cell arteritis. J Rheumatol 1979; 6:71–75. 14. Swanepoel CR, Floyd A, Allison H, Learmonth GM, Cassidy MJ, Pascoe MD. Acute acalculous cholecystitis complicating systemic lupus erythematosus: case report and review. BMJ (Clin Res Ed) 1983; 286:251–252. 15. Moolenaar W, Kreuning J, Eulderink F, Lamers CB. Ischemic colitis and acalculous necrotizing cholecystitis as rare manifestations of cholesterol emboli in the same patient. Am J Gastroenterol 1989; 84:1421–1422. 16. Savoca PE, Longo WE, Pasternak B, Gusberg RJ. Does visceral ischemia play a role in the pathogenesis of acute acalculous cholecystitis? J Clin Gastroenterol 1990; 12:33–36. 17. Munster AM, Goodwin MN, Pruitt BA Jr. Acalculous cholecystitis in burned patients. Am J Surg 1971; 122:591–593. 18. Lindberg EF, Grinnan GL, Smith L. Acalculous cholecystitis in Viet Nam casualties. Ann Surg 1970; 171:152–157. 19. Ouriel K, Green RM, Ricotta JJ, DeWeese JA, Adams JT. Acute acalculous cholecystitis complicating abdominal aortic aneurysm resection. J Vasc Surg 1984; 1:646–648. 20. Parsson H, Swartbol P, Andersson R, Norgren L. The role of septic complications in aortic aneurysm surgery. Int Angiol 1994; 13:129–132. 21. Scher KS, Sarap MD, Jaggers RL. Acute acalculous cholecystitis complicating aortic aneurysm repair. Surg Gynecol Obstet 1986; 163:475–478. 22. Leitman IM, Paull DE, Barie PS, et al. Intraabdominal complications of cardiopulmonary bypass surgery. Surg Gynecol Obstet 1987; 165:251–254. 23. Orlando Rd, Gleason E, Drezner AD. Acute acalculous cholecystitis in the critically ill patient. Am J Surg 1983; 145:472–476. 24. Williamson RC. Acalculous disease of the gall bladder. But 1988; 29:860–872. 25. Warren BL. Small vessel occlusion in acute acalculous cholecystitis. Surgery 1992; 111:163–168. 26. Tracey KJ, Beutler B, Lowry SF, Merryweather J, Wolpe S, Milsark IW, Hariri RJ, Fahey TJd, Zentella A, Albert JD, et al. Shock and tissue injury induced by recombinant human cachectin. Science 1986; 234:470–474. 27. Becker CG, Dubin T, Glenn F. Induction of acute cholecystitis by activation of factor XII. J Exp Med 1980; 151:81–90. 28. Yeung E, Jackson J, Finn JP, Thomas MG, Benjamin IS, Adam A. Acalculous cholecystitis complicating hepatic intraarterial lipiodol: case report. Cardiovasc Intervent Radiol 1989; 12:80–82. 29. Hohn DC, Rayner AA, Economou JS, Ignoffo RJ, Lewis BJ, Stagg RJ. Toxicities and complications of implanted pump hepatic arterial and intravenous floxuridine infusion. Cancer 1986; 57:465–470. 30. Lafon PC, Reed K, Rosenthal D. Acute cholecystitis associated with hepatic arterial infusion of floxuridine. Am J Surg 1985; 150:687–689. 31. Sjodahl R, Tagesson C, Wetterfors J. On the pathogenesis of acute cholecystitis. Surg Gynecol Obstet 1978; 146:199–202.
Page 614
32. Herlin P, Ericsson M, Holmin T, Jonsson PE. Acute acalculous cholecystitis following trauma. Br J Surg 1982; 69:475–476. 33. Devine RM, Farnell MB, Mucha P, Jr. Acute cholecystitis as a complication in surgical patients. Arch Surg 1984; 119:1389–1393. 34. Hwang TL, Chen MF. Percutaneous gallbladder drainage for acute acalculous cholecystitis during total parenteral nutrition. Br J Surg 1992; 79:237–238. 35. Vauthey JN, Lerut J, Martini M, Becker C, Gertsch P, Blumgart LH. Indiciations and limitations of percutaneous cholecystostomy for acute cholecystitis. Surg Gynecol Obstet 1993; 176:49–54. 36. Lotveit T, Osnes M, Aune S. Bacteriological studies of common duct bile in patients with gallstone disease and juxtapapillary duodenal diverticula. Scand J Gastroenterol 1978; 13:93–95. 37. Avalos ME, Cerulli MA, Lee RS. Acalculous acute cholecystitis due to Salmonella typhi (see comments). Dig Dis Sci 1992; 37:1772–1775. 38. Kaminski DL, Andrus CH, German D, Deshpande YG. The role of prostanoids in the production of acute acalculous cholecystitis by plateletactivating factor. Ann Surg 1990; 212:455–461. 39. Kaminski DL, Amir G, Deshpande YG, Beck D, Li AP. Studies on the etiology of acute acalculous cholecystitis: the effect of lipopolysaccharide on human gallbladder mucosal cells. Prostaglandins 1994; 47:319–330. 40. ChungPark M KB, Marmolya G, Karlins N, Wojcik E. Acalculous lymphoeosinophilic cholecystitis associated with interleukin2 and lymphokine activated killer cell therapy. Arch Pathol Lab Med 1990; 14:1073–1075. 41. Powell FC, Spooner KM, Shawker TH, Premkumar A, Thakore KN, Vogel SE, Kovacs JA, Masur H, Feuerstein IM. Symptomatic interleukin2induced cholecystopathy in patients with HIV infection. AJR 1994; 163:117–1121. 42. Hayward AF, Freston JW, Bouchier IA. Changes in the ultrastructure of gall bladder epithelium in rabbits with experimental gallstones. Gut 1968; 9:550–556. 43. Roslyn JJ, DenBesten L, Thompson JE, Jr., Silverman BF. Roles of lithogenic bile and cystic duct occlusion in the pathogenesis of acute cholecystitis. Am J Surg 1980; 140:126–130. 44. Borgman RF HF. Ear lesions produced in rabbits by sterol injections. Am J Vet Res 1972;33:2309. 45. Mills PR, Rooney PJ, Watkinson G, MacSween RN. Hypercholesterolaemic arthropathy in primary biliary cirrhosis. Ann Rheum Dis 1979; 38:179–180. 46. Sjodahl R, Tagesson C, Wetterfors J. Lysolecithinmediated inflammatory reaction in rabbit gallbladder: permeability increase and enzyme liberation. Acta Chir Scand 1975; 141:403–408. 47. Gottfries A. Lysolecithin: a factor in the pathogenesis of acute cholecystitis? An experimental study on rabbits based on clinical findings in humans. Acta Chir Scand 1969; 135:213–217. 48. Kaminski DL, Deshpande YG, Li A, Dysart F, Nag M. Gallbladder mucosal protein secretion during development of experimental cholecystitis. Dig Dis Sci 1995; 40:1157–1164. 49. Gottfries A. Studies on acute cholecystitis. A clinical and experimental study in humans and in animals with special reference to liver function and pathogenesis. Acta Chir Scand Suppl 1968; 393:5–32. 50. Sjodahl R, Wetterfors J. Lysolecithin and lecithin in the gallbladder wall and bile; their possible roles in the pathogenesis of acute cholecystitis. Scand J Gastroenterol 1974; 9:519–525. 51. Niederhiser DH, Harmon CK. Lipid metabolism by the gallbladder: I. The in situ uptake and metabolism of lysophosphatidylcholine. Biochim Biophys Acta 1978, 530:208–216. 52. Tagesson C, Norrby S, Sjodahl R. The prerequisites for local lysolecithin formation in
Page 615
the human gallbladder: III. Demonstration of two different phospholipase A activities. Scand J Gastroenterol 1979; 14:379–384. 53. Neiderhiser D, Thornell E, Bjorck S, Svanvik J. The effect of lysophosphatidylcholine on gallbladder function in the cat. J Lab Clin Med 1983; 101:699–707. 54. Lee SP, LaMont JT, Carey MC. Role of gallbladder mucus hypersecretion in the evolution of cholesterol gallstones. J Clin Invest 1981; 67:1712–1723. 55. Davenport HW, Warner HA, Code CF. Functional significance of gastric mucosal barrier to sodium. Gastroenterology 1968; 54(suppl):709–711. 56. Feldman S, Gibaldi M. Bile saltinduced permeability changes in the isolated rat intestine. Proc Soc Exp Biol Med 1969; 132:1031–1033. 57. Ivey KJ, DenBesten L, Clifton JA. Effect of bile salts on ionic movement across the human gastric mucosa. Gastroenterology 1970; 59:683–690. 58. WaldronEdward D, Boutros MI, Himal HS. Effect of bile on lysosomal stability in the mucosa of the canine gastric antrum. Gastroenterology 1977; 73:980–984. 59. Heuman R, Norrby S, Sjodahl R, Tiselius HG, Tagesson C. Altered gallbladder bile composition in gallstone disease. Relation to gallbladder wall permeability. Scand J Gastroenterology 1980; 15:581–586. 60. Long TN, Heimbach DM, Carrico CJ. Acalculous cholecystitis in critically ill patients. Am J Surg 1978; 136:31–36. 61. Andry G, Turnbull AD, Botet J, Kurtz RC. Cholesonographic characteristics of cystic duct metastasis causing acute acalculous cholecystitis: case report. J Surg Oncol 1986; 31:178–181. 62. Lee SP. Pathogenesis of biliary sludge. Hepatology 1990; 12:200S–203S; discussion 203S–205S. 63. Hilton HD, Griffin WT. Acute acalculous cholecystitis. Surgery 1968; 64:1047–1048. 64. Flancbaum L, Majerus TC, Cox EF. Acute posttraumatic acalculous cholecystitis. Am J Surg 1985; 150:252–256. 65. Cornwell EEd, Rodriguez A, Mirvis SE, Shorr RM. Acute acalculous cholecystitis in critically injured patients: preoperative diagnostic imaging. Ann Surg 1989; 210:52–55. 66. Johnson EE, HedleyWhyte J. Continuous positivepressure ventilation and choledochoduodenal flow resistance. J Appl Physiol 1975; 39:937–942. 67. Petersen SR, Sheldon GF. Acute acalculous cholecystitis: a complication of hyperalimentation. Am J Surg 1979; 138:814–817. 68. Roslyn JJ, Pitt HA, Mann L, Fonkalsrud EW, DenBesten L. Parenteral nutritioninduced gallbladder disease: a reason for early cholecystectomy. Am J Surg 1984; 148:58–63. 69. Toursarkissian B, Kearney PA, Holley DT, Cheung R, Fried A, Strodel WE. Biliary sludging in critically ill trauma patients (see comments). South Med J 1995; 88:420–424. 70. Kurumado K, Nagai T, Abe H. Mucus production of choledochal epithelial cells due to bile stagnation in the rat. Dig Dis Sci 1996; 41:263–271. 71. Scott GW, Otto WJ. Resistance and sphincterlike properties of the cystic duct. Surg Gynecol Obstet 1979; 149:177–182. 72. Merrell RC, MillerCrotchett P, Lowry P. Gallbladder response to enteral lipids in injured patients. Arch Surg 1989; 124:301–302. 73. Janowitz P, Kratzer W, Zemmler T, Tudyka J, Wechsler JG. Gallbladder sludge: spontaneous course and incidence of complications in patients without stones (see comments). Hepatology 1994; 20:291–294. 74. Glenn F, Becker CG. Acute acalculous cholecystitis: an increasing entity. Ann Surg 1982; 195:131–136. 75. Dabbs DJ. Eosinophilic and lymphoeosinophilic cholecystitis (see comments). Am J Surg Pathol 1993; 17:497–501. 76. Glenn F. Acute acalculous cholecystitis. Ann Surg 1979; 189:458–465. 77. Anderson DL. Acalculous cholecystitis—a possible complication of parenteral hyper alimentation: report of a case. Med Ann Dist Columbia 1972; 41:448–450.
Page 616
78. Doty JE, Pitt HA, PorterFink V, Denbesten L. Cholecystokinin prophylaxis of parenteral nutritioninduced gallbladder disease. Ann Surg 1985; 201:76–80. 79. Hasse C, Zielke A, Nies C, alBazaz B, Gotzen L, Rothmund M. Influence of ceruletid on gallbladder contraction: a possible prophylaxis of acute acalculous cholecystitis in intensive care patients? Digestion 1995; 56:389–394. 80. Roslyn JJ, Pitt HA, Mann LL, Ament ME, DenBesten L. Gallbladder disease in patients on longterm parenteral nutrition. Gastroenterology 1983; 84:148–154. 81. Shapiro MJ, Luchtefeld WB, Kurzweil S, Kaminski DL, Durham RM, Mazuski JE. Acute acalculous cholecystitis in the critically ill. Am Surg 1994; 60:335–339. 82. Tucker RA, Jenkins HL. Acalculous cholecystitis and fever related to total parenteral nutrition. Drug Intell Clin Pharm 1984; 18:897–899. 83. Duncan J. Femoral hernia: gangrene of the gallbladder: extravasation of bile; peritonitis; death. North Med J 1844; 2:151. 84. Barie PS, Fischer E. Acute acalculous cholecystitis. J Am Coll Surg 1995; 180:232–344. 85. Gallbladder Survey Committee OCACoS. 28,621 cholecystectomies in Ohio. Am J Surg 1970; 119:714–717. 86. Barie PS. In: Barie PS, Shires, GT, eds. Acalculous and Postoperative Cholecystitis. Boston: Little, Brown, 1993. 87. Ishikawa S, Ohtaki A, Koyano T, Takahashi T, Sato Y, Nakamura S, Ohwada S, Morishita Y. Percutaneous transhepatic gallbladder drainage for acute acalculous cholecystitis following cardiovascular surgery. J Cardiovasc Surg (Torino) 1997; 38:513–515. 88. Sessions SC, Scoma RS, Sheikh FA, McGeehin WH, Smink RD Jr. Acute acalculous cholecystitis following open heart surgery. Am Surg 1993; 59:74–77. 89. van Marle J, Franz RC. Acute acalculous cholecystitis in critically ill patients: case reports. S Afr Med J 1987; 72:58–60. 90. Mandak JS, Pollack B, Fishman NO, Furth EE, Kochman ML, Acker MA, Lichtenstein GR. Acalculous candidal cholecystitis: a previously unrecognized complication after cardiac transplantation. Am J Gastroenterol 1995; 90:1333–1337. 91. FernandezCruz L, Targarona EM, Cugat E, Alcaraz A, Oppenheimer F. Acute pancreatitis after renal transplantation. Br J Surg 1989; 76:1132–1135. 92. Jardines LA, O'Donnell MR, Johnson DL, Terz JJ, Forman SJ. Acalculous cholecystitis in bone marrow transplant patients. Cancer 1993; 71:354–358. 93. Hurley R, Weisdorf DJ, Jessurun J, Vercellotti GM, Miller WJ. Relapse of acute leukemia presenting as acute cholecystitis following bone marrow transplantation. Bone Marrow Transplant 1992; 10:387–389. 94. Pitkaranta P, Haapiainen R, Taavitsainen M, Elonen E. Acalculous cholecystitis after bone marrow transplantation in adults with acute leukaemia: case report. Eur J Surg 1991; 157:361–364. 95. Wiboltt KS, Jeffrey RB Jr. Acalculous cholecystitis in patients undergoing bone marrow transplantation. Eur J Surg 1997; 163:519–524. 96. Marymont JV, Dakhil, SR, Travers H, Housholder DF. Chemical cholecystitis associated with hepatic arterial chemotherapy delivered by a permanently implanted pump. Hum Pathol 1985; 12:986–990. 97. Alvarez C, Hunt K, Ashley SW, Reber HA. Emphysematous cholecystitis after ERCP. Dig Dis Sci 1994; 39:1719–1723. 98. Bender JS, Talamini MA. Diagnostic laparoscopy in critically ill intensivecareunit patients. Surg Endosc 1992; 6:302–304. 99. Girotti MJ. Acalculous cholecystitis in burned patients (letter). Can J Surg 1986; 29:81–82. 100. McDermott MW, Scudamore CH, Boileau LO, Snelling CF, Kramer TA. Acalculous cholecystitis: its role as a complication of major burn injury. Can J Surg 1985; 28:529–533.
Page 617
101. Ross DC, Lee KC, Peters WJ, Douglas LG. Acalculous cholecystitis in association with major burns. Burns Incl Therm Inj 1987; 13:488–491. 102. Still J, Scheirer R, Law E. Acute cholecystectomy performed through cultured epithelial autografts in a patient with burn injuries: a case report. J Burn Care Rehabil 1996; 17:429–431. 103. Blatt JM, Floman Y. Posttraumatic acute acalculous cholecystitis. Int Surg 1972; 57:982–983. 104. Branch CL Jr, Albertson DA, Kelly DL. Posttraumatic acalculous cholecystitis on a neurosurgical service. Neurosurgery 1983; 12:98–101. 105. Buckley PM, Hunter JM. Acute acalculous cholecystitis following multiple skeletal trauma: a report of three cases. Anaesthesia 1985; 40:23–26. 106. Fabian TC, Hickerson WL, Mangiante EC. Posttraumatic and postoperative acute cholecystitis. Am Surg 1986; 52:188–192. 107. Heruti RJ, BarOn Z, Gofrit O, Weingarden HP, Ohry A. Acute acalculous cholecystitis as a complication of spinal cord injury. Arch Phys Med Rehabil 1994; 75:822–824. 108. Imhof M, Raunest J, Ohmann C, Roher HD. Acute acalculous cholecystitis complicating trauma: a prospective sonographic study. World J Surg 1992; 16:1160–1165; discussion 1166. 109. Okada Y, Tanabe R, Mukaida M. Posttraumatic acute cholecystitis: relationship to the initial trauma. Am J Forensic Med Pathol 1987; 8:164–168. 110. Rice J, Williams HC, Flint LM, Richardson JD. Posttraumatic acalculous cholecystitis. South Med J 1980; 73:14–17. 111. Rozycki GS, Akin HE, Nelson HS, Jr., Maull KI. Posttraumatic acalculous cholecystitis. J Tenn Med Assoc 1985: 78:639–640. 112. Shields MA. Acute acalculous cholecystitis: an important complication of trauma. J R Coll Surg Edinb 1973; 18:83–86. 113. Ullman M, Hasselgren PO, Tveit E. Posttraumatic and postoperative acute acalculous cholecystitis. Acta Chir Scand 1984; 150:507–509. 114. Wilson TH Jr. Acute acalculous cholecystitis after trauma and surgery. Med Ann Dist Columbia 1970; 39:560–562. 115. Romero Ganuza FJ, La Banda G, Montalvo R, Mazaira J. Acute acalculous cholecystitis in patients with acute traumatic spinal cord injury. Spinal Cord 1997; 35:124–128. 116. Lin KY. Acute acalculous cholecystitis: a limited review of the literature. Mt Sinai J Med 1986; 53:305–309. 117. Thurston WA, Kelly EN, Silver MM. Acute acalculous cholecystitis in a premature infant treated with parenteral nutrition [published erratum appears in Can Med Assoc J 1986 Oct 1;135(7):729]. Can Med Assoc J 1986; 135:332–334. 118. Fisher RL. Hepatobiliary abnormalities associated with total parenteral nutrition. Gas troenterol Clin North Am 1989; 18:645–666. 119. Messing B, Bories C, Kuntslinger F, et al. Does total parenteral nutrition induce gallbladder sludge formation and lithiasis? Gastroenterology 1983; 84:1012– 1019. 120. Savoca PE, Longo WE, Zucker KA, McMillen MM, Modlin IM. The increasing prevalence of acalculous cholecystitis in outpatients: results of a 7year study. Ann Surg 1990; 211:433–437. 121. Barie PS, Jacobson, IM. In: Zakim DB, ed. Gallbladder Disease. 2nd ed. Philadelphia: Saunders, 1990. 122. Raijman I, Schrager M. Hemorrhagic acalculous cholecystitis in systemic lupus erythematosus (letter). Am J Gastroenterol 1989; 84:445–447. 123. Date K, Shirai Y, Hatakeyama K. Antiphospholipid antibody syndrome presenting as acute acalculous cholecystitis. Am J Gastroenterol 1997; 92:2127–2128. 124. Thambidorai CR, Shyamala J, Sarala R, Vatsala RB, Tamizhisai S. Acute acalculous cholecystitis associated with enteric fever in children. Pediatr Infect Dis J 1995; 14:812–813.
Page 618
125. Winkler AP, Gleich S. Acute acalculous cholecystitis caused by Salmonella typhi in an 11yearold. Pediatr Infect Dis J 1988; 7:125–128. 126. Yulevich A, Cohen A, Maor E, Bryk T, Mares AJ. Acute acalculous cholecystitis caused by Salmonella typhi in a 6yearold child. Eur J Pediatr Surg 1992; 2:301–303. 127. Chowdhury A, Goenka MK, Kochhar R, Kochhar S, Mehta SK. Acalculous cholecystitis complicated by empyema in a patient with typhoid fever. J Clin Gastroenterol 1995; 20:344–345. 128. James RA, Morris G, Winter R, KestonJones M. Acalculous cholecystitis due to Salmonella virchow. Br J Clin Pract 1990; 44:767–768. 129. Beiler HA, Kuntz C, Eckstein TM, Daum R. Cholecystolithiasis and infection of the biliary tract with Salmonella Virchow—a very rare case in early childhood. Eur J Pediatr Surg 1995; 5:369–371. 130. Tsakayannis DE, Kozakewich HP, Lillehei CW. Acalculous cholecystitis in children. J Pediatr Surg 1996; 31:127–130; discussion 130–131. 131. McCarron B, Love WC. Acalculous nontyphoidal salmonellal cholecystitis requiring surgical intervention despite ciprofloxacin therapy: report of three cases (see comments). Clin Infect Dis 1997; 24:707–709. 132. Latlitha MK, John, R. Unusual manifestations of salmonellosis—a surgical problem. Q J Med 1994; 87:301–309. 133. Barton LL, Escobedo MB, Keating JP, Ternberg JL. Leptospirosis with acalculous cholecystitis. Am J Dis Child 1973; 126:350–351. 134. McKiernan J, O'Brien DJ, Dundon S. Leptospirosis and acalculous cholecystitis. Ir Med J 1976; 69:71–72. 135. Monno S, Mizushima Y. Leptospirosis with acute acalculous cholecystitis and pancreatitis. J Clin Gastroenterol 1993; 16:52–54. 136. Baelen E, Roustan J. Leptospirosis associated with acute acalculous cholecystitis. Surgical or medical treatment? J Clin Gastroenterol 1997; 25:704–706. 137. Thomas WE, Thornton JR, Thompson MH. Staphylococcal acalculous cholecystitis. Br J Surg 1981; 68:136. 138. Brooks BJ, Jr., Williams WL, Sanders CV, Marier RL. Apparent ketoconazole failure in candidal cholecystitis. Arch Intern Med 1982; 42:1934–1935. 139. Gips M, Halpern M, Wolloch Y. Acalculous Candida cholecystitis. Eur J Surg 1992; 58:251–252. 140. Hiatt JR, Kobayashi MR, Doty JE, Ramming KP. Acalculous Candida cholecystitis: a complication of critical surgical illness. Am Surg 1991; 57:825–829. 141. Miller DD. Postoperative acalculous cholecystitis due to Torulopsis glabrata. Arch Surg 1976; 111:1404–1405. 142. Peison B, Benisch B. Acute acalculous cholecystitis secondary to Candida albicans. N J Med 1996; 93:39–42. 143. AbouKhalil S, Smith BM, MacLean JD, Poenaru D, Fried GM, Bret P, Barkun AN. Acute cholecystitis and cholangitis caused by Echinococcus granulosus. Am J Gastroenterol 1996; 91:805–807. 144. Kuzu MA, Ozturk Y, Ozbek H, Soran A. Acalculous cholecystitis: ascariasis as an unusual cause. J Gastroenterol 1996; 1:747–749. 145. Khuroo MS, Zargar SA. Biliary ascariasis: a common cause of biliary and pancreatic disease in an endemic area. Gastroenterology 1985; 88:418–423. 146. LaRaja RD, Rotherberg RE, Odom JW, et al. The incidence of intraabdominal surgery in acquired immune deficiency syndrome: a statistical review of 904 patients. Surgery 1989; 105:175–179. 147. Chui DW, Owen RL. AIDS and the gut. J Gastroenterol Hepatol 1994; 9:291–303. 148. French AL, Beaudet LM, Benator DA, Levy CS, Kass M, Orenstein JM. Cholecystectomy in patients with AIDS: clinicopathologic correlations in 107 cases. Clin Infect Dis 1995; 21:852–858.
Page 619
149. Liu KJ, Atten MJ, Donahue PE. Cholestasis in patients with acquired immunodeficiency syndrome: a surgeon's perspective. Am Surg 1997; 63:519–524. 150. Radin DR, Cohen H, Halls JM. Acalculous inflammatory disease of the biliary tree in acquired immunodeficiency syndrome: CT demonstraton. J Comput Assist Tomogr 1987; 11:775–778. 151. Blumberg RS, Kelsey P, Perrone T, Dickersin R, Laquaglia M, Ferruci J. Cytomegalovirus and Cryptosporidiumassociated acalculous gangrenous cholecystitis. Am J Med 1984; 76:1118–1123. 152. Bigio EH, Haque AK. Disseminated cytomegalovirus infection presenting with acalculous cholecystitis and acute pancreatitis. Arch Pathol Lab Med 1989; 13:1287–1289. 153. Adolph MD, Bass SN, Lee SK, Blum JM, Schreiber H. Cytomegaloviral acalculous cholecystitis in acquired immunodeficiency syndrome patients. Am Surg 1993; 59:679–684. 154. Aaron JS, Wynter CD, Kirton OC, Simko V. Cytomegalovirus associated with acalculous cholecystitis in a patient with acquired immune deficiency syndrome. Am J Gastroenterol 1988; 83:879–881. 155. Hinnant K, Schwartz A, Rotterdam H, Rudski C. Cytomegaloviral and cryptosporidial cholecystitis in two patients with AIDS (see comments). Am J Surg Pathol 1989; 13:57–60. 156. Kavin H, Jonas RB, Chowdhury L, Kabins S. Acalculous cholecystitis and cytomegalovirus infection in the acquired immunodeficiency syndrome. Ann Intern Med 1986; 104:53–54. 157. Keshavjee SH, Magee LA, Mullen BJ, Baron DL, Brunton JL, Gallinger S. Acalculous cholecystitis associated with cytomegalovirus and sclerosing cholangitis in a patient with acquired immunodeficiency syndrome. Can J Surg 1993; 36:321–325. 158. Ong EL, Ellis ME, Tweedle DE, Ferguson G, Haboubi NY, Knox WF. Cytomegalovirus cholecystitis and colitis associated with the acquired immunodeficiency syndrome. J Infect 1989; 18:73–75. 159. Saraux JL, Lenoble L, Toublanc M, Smiejan JM, Dombret MC. Acalculous cholecystitis and cytomegalovirus infection in a patient with AIDS (letter). J Infect Dis 1987; 155:829. 160. Knapp PE, Saltzman JR, Fairchild P. Acalculous cholecystitis associated with microsporidial infection in a patient with AIDS (see comments). Clin Infect Dis 1996; 22:195–196. 161. RamonGarcia G, SadowinskiPine S. Acalculous cholecystitis and microsporidiosis in a patient with AIDS (letter; comment). Clin Infect Dis 1996; 23:664. 162. Wind P, Chevallier JM, Jones D, Frileux P, Cugnenc PH. Cholecystectomy for cholecystitis in patients with acquired immune deficiency syndrome. Am J Surg 1994; 168:244–246. 163. Iannuzzi C, Belghiti J, Erlinger S, Menu Y, Fekete F. Cholangitis associated with cholecystitis in patients with acquired immunodeficiency syndrome. Arch Surg 1990; 125:1211–1213. 164. Roca M, Sellier N, Mensire A, Kalifa G, Bennet J. Acute acalculous cholecystitis in Salmonella infection. Pediatr Radiol 1988; 18:421–423. 165. Horii Y, Sugimoto T, Sakamoto I, Sawada T. Acute acalculous cholecystitis complicating Mycoplasma pneumoniae infection (letter). Clin Pediatr (Phila) 1992; 31:376–378. 166. Holcomb GW, Jr., O'Neill JA, Jr., Holcomb GWd. Cholecystitis, cholelithiasis and common duct stenosis in children and adolescents. Ann Surg 1980; 191:626–635. 167. Lucas CE, Walt AJ. Acute gengrenous acalculous cholecystitis in infancy: report of a case. Surgery 1968;64:847–849. 168. Mukamel E, Zer M, Avidor I, Versano I, Rachmel A, Dintsman M. Acute acalculous cholecystitis in an infant: a case report. J Pediatr Surg 1981; 16:521–522. 169. Crystal RF, Fink RL, Acute acalculous cholecystitis in childhood. Clin Pediatr (Phila) 1971; 10:423–426.
Page 620
170. Pieretti R, Auldist AW, Stephens CA. Acute cholecystitis in children. Surg Gynecol Obstet 1975; 140:16–18. 171. Traynelis VC, Hrabovsky EE. Acalculous cholecystitis in the neonate. Am J Dis Child 1985; 139:893–895. 172. Washburn ME, Barcia PJ. Uncommon cause of a right upper quadrant abdominal mass in the newborn: acute cholecystitis. Am J Surg 1980; 140:704–705. 173. Ternberg JL, Keating JP. Acute acalculous cholecystitis: complication of other illnesses in childhood. Arch Surg 1975; 110:543–547. 174. Richard B, Nadal D, Meuli M, Braegger CP. Acute acalculous cholecystitis in infective endocarditis. J Pediatr Gastroenterol Nutr 1993; 17:215–216. 175. Paret G, Gilad E, Jonas A, Meyers JJ, Barzilay Z. Acute acalculous cholecystitis in an infant after cardiac surgery. J Pediatr Surg 1994; 29:1580–1581. 176. Kalliafas S, Ziegler DW, Flancbaum L, Choban PS. Acute acalculous cholecytitis: incidence, risk factors, diagnosis, and outcome. Am Surg 1998; 64:471–475. 177. Kurzweil SM, Shapiro MJ, Andrus CH, Wittgen CM, Herrmann VM, Kaminski DL. Hyperbilirubinemia without common bile duct abnormalities and hyperamylasemia without pancreatitis in patients with gallbladder disease. Arch Surg 1994; 129:829–833. 178. Eggermont AM, Lameris JS, Jeekel J. Ultrasoundguided percutaneous transhepatic cholecystostomy for acute acalculous cholecystitis. Arch Surg 1985; 120:1354–1356. 179. Greenberg M, Kangarloo H, Cochran ST, Sample WF. The ultrasonographic diagnosis of cholecystitis and cholelithiasis in children. Radiology 1980; 137:745– 749. 180. Deitch EA. Utility and accuracy of ultrasonically measured gallbladder wall as a diagnostic criteria in biliary tract disease. Dig Dis Sci 1981; 26:686–693. 181. Deitch EA, Engel JM. Acute acalculous cholecystitis. Ultrasonic diagnosis. Am J Surg 1981; 142:290–292. 182. Becker CD, Burckhardt B, Terrier F. Ultrasound in postoperative acalculous cholecystitis. Gastrointest Radiol 1986; 11:47–50. 183. Blankenberg F, Wirth R, Jeffrey RB, Jr., Mindelzun R, Francis I. Computed tomography as an adjunct to ultrasound in the diagnosis of acute acalculous cholecystitis. Gastrointest Radiol 1991; 16:149–153. 184. Mirvis SE, Vainright JR, Nelson AW, Johnston GS, Shorr R, Rodriguez A, Whitley NO. The diagnosis of acute acalculous cholecystitis: a comparison of sonography, scintigraphy, and CT. AJR 1986; 147:1171–1175. 185. Savino JA, Scalea TM, Del Guercio LR. Factors encouraging laparotomy in acalculous cholecystitis. Crit Care Med 1985; 13:377–380. 186. Helbich TH, Mallek R, Madl C, Wunderbaldinger P, Breitenseher M, Tscholakoff D, Mostbeck GH. Sonomorphology of the gallbladder in critically ill patients: value of a scoring system and followup examinations. Acta Radiol 1997; 38:129–134. 187. Raduns K, McGahan JP, Beal S. Cholecystokinin sonography: lack of utility in diagnosis of acute acalculous cholecystitis. Radiology 1990; 175:463–466. 188. FinkBennett D. Augmented cholescintigraphy: its role in detecting acute and chronic disorders of the hepatobiliary tree. Semin Nucl Med 1991; 21:128–139. 189. Weissmann HS, Berkowitz D, Fox MS, Gliedman ML, Rosenblatt R, Sugarman LA, Freeman LM. The role of technetium99m iminodiacetic acid (IDA) cholescintigraphy in acute acalculous cholecystitis. Radiology 1983; 146:177–180. 190. Shuman WP, Rogers JV, Rudd TG, Mack LA, Plumley T, Larson EB. Low sensitivity of sonography and cholescintigraphy in acalculous cholecystitis. AJR 1984; 142:531–534. 191. Ramanna L, Brachman MB, Tanasescu DE, Berman DS, Waxman AD. Cholescintigraphy in acute acalculous cholecystitis. Am J Gastroenterol 1984; 79:650– 653. 192. Kalff V, Froelich JW, Lloyd R, Thrall JH. Predictive value of an abnormal hepatobiliary scan in patients with severe intercurrent illness. Radiology 1983; 146:191–194. 193. Warner BW, Hamilton FN, Silberstein EB, et al. The value of hepatobiliary scans in fasted patients receiving total parenteral nutrition. Surgery 1987; 102:595– 601.
Page 621
194. Ohrt HJ, Posalaky IP, Shafer RB. Normal gallbladder scintigraphy in acute cholecystitis. Clin Nucl Med 1983; 8:97–100. 195. Schneider PB. Acalculous cholecystitis: a case with variable cholescintigram. J Nucl Med 1984; 25:64–65. 196. Warshauer D, Scott G, Gottschalk A. Focal acute acalculous cholecystitis. AJR 1987; 149:505–506. 197. Flancbaum L, Alden SM, Trooskin SZ. Use of cholescintigraphy with morphine in critically ill patients with suspected cholecystitis. Surgery 1989; 106:668–673; discussion 673–674. 198. Fig LM, Wahl RL, Stewart RE, et al. Morphine augmented hepatobiliary scintigraphy in the severely ill: caution is in order. Radiology 1990; 175:467–473. 199. Flancbaum L, Choban PS, Sinha R, Jonasson O. Morphine cholescintigraphy in the evaluation of hospitalized patients with suspected acute cholecystitis. Ann Surg 1994; 220:25–31. 200. Flancbaum L, Choban PS. Use of morphine cholescintigraphy in the diagnosis of acute cholecystitis in critically ill patients. Intens Care Med 1995; 21:120–124. 201. Cacciarelli AG, Naddaf SY, elZeftawy HA, Aziz M, Omar WS, Kumar M, Atay S, Abujudeh H, Gillooley J, AbdelDayem HM. Acute cholecystitis in AIDS patients: correlation of Tc99m hepatobiliary scintigraphy with histopathologic laboratory findings and CD4 counts. Clin Nucl Med 1998; 23:226–228. 202. Lantto E, Jarvi K, Laitinen R, Lantto T, Taavitsainen M, Talvitie I, Vorne M. Scintigraphy with 99mTcHMPAO labeled leukocytes in acute cholecystitis. Acta Radiol 1991; 32:359–362. 203. Pounds TR, Hattner RS. Normal cholescintigram in In111 WBC positive acute acalculous cholecystitis: the converse photopenic ''rim" sign. Clin Nucl Med 1994; 19:483–485. 204. Herlin P, Jonsson PE, Karp W. Postoperative acute acalculous cholecystitis—an assessment of diagnostic procedures. Gastrointest Radiol 1980; 5:147–149. 205. Ohtani Y, Tanaka Y, Tsukui M, Goto K, Moriya H, Tobita K, Sekka T, Saito Y, Makuuchi H, Tajima T, Mitomi T. Acute emphysematous cholecystitis associated with pneumobilia: a case report. Tokai J Exp Clin Med 1996; 21:33–36. 206. Gill KS, Chapman AH, Weston MJ. The changing face of emphysematous cholecystitis. Br J Radiol 1997; 70:986–991. 207. Poleynard GD, Harris RD. Diagnosis of emphysematous cholecystitis by computerized tomography. Gastrointest Radiol 1979; 4:153–155. 208. Mirvis SE, Whitley NO, Miller JW. CT diagnosis of acalculous cholecystitis. J Comput Assist Tomogr 1987; 11:83–87. 209. Jurkovich GJ, Dyess DL, Ferrara JJ. Cholecystostomy: expected outcome in primary and secondary biliary disorders. Am Surg 1988; 54:40–44. 210. Teplick SK, Brandon JC, Haskin PH, Pavlides CA, Huppert AS. Percutaneous cholecystostomy in patient at high risk: treatment of acute acalculous cholecystitis. Postgrad Med 1987; 81:209–211, 214. 211. vanSonnenberg E, D'Agostino HB, Goodacre BW, Sanchez RB, Casola G. Percutaneous gallbladder puncture and cholecystostomy: results, complications, and caveats for safety. Radiology 1992; 183:167–170. 212. Boland GW, Lee MJ, Dawson SL, Mueller PR. Percutaneous cholecystostomy for acute acalculous cholecystitis in a critically ill patient (clinical conference). AJR 1993; 160: 871–874. 213. Melin MM, Sarr MG, Bender CE, van Heerden JA. Percutaneous cholecystostomy: a valuable technique in highrisk patients with presumed acute cholecystitis (see comments). Br J Surg 1995; 82:1274–1277. 214. Shirai Y, Tsukada K, Kawaguchi H, Ohtani T, Muto T, Hatakeyama K. Percutaneous transhepatic cholecystostomy for acute acalculous cholecystitis. Br J Surg 1993; 80: 1440–1442.
Page 622
215. Frazee RC, Nagorney DM, Mucha P Jr. Acute acalculous cholecystitis. Mayo Clin Proc 1989; 64:163–167. 216. Boland GW, Lee MJ, Leung J, Mueller PR. Percutaneous cholecystostomy in critically ill patients: early response and final outcome in 82 patients. AJR 1994; 163:339–342. 217. Browning PD, McGahan, J.P., Gerscovich, E.O. Percutaneous cholecystostomy for suspected acute cholecystitis in the hospitalised patient. J Vasc Intervent Radiol 1993; 4: 531–537 218. Furlan F, Fugazzola C, Brunelli G, et al. The treatment of acute cholecystitis by percutaneous cholecystostomy. Radiol Med 1992; 84:247–251. 219. Johlin FC, Jr., Neil GA. Drainage of the gallbladder in patients with acute acalculous cholecystitis by transpapillary endoscopic cholecystotomy (see comments). Gastrointest Endosc 1993; 39:645–651. 220. Pearse DM, Hawkins IF, Shaver R, et al. Percutaneous cholecystostomy in acute cholecystitis and common duct obstruction. Radiology 1984; 152:365–367. 221. Mentzer RM Jr, Golden GT, Chandler JG, Horsley JSd. A comparative appraisal of emphysematous cholecystitis. Am J Surg 1975; 129:10–15. 222. Mentzer RM, Jr., Golden GT, Chandler JG, Horsley JSd. Emphysematous cholecystitis —an important clinical variant of acute cholecystitis. Rev Surg 1974; 31:454–456. 223. Banwell PE, Hill AD, MenziesGow N, Darzi A. Laparoscopic cholecystectomy: safe and feasible in emphysematous cholecystitis. Surg Laparosc Endosc 1994; 4:189–191. 224. Niemeier OW. Acute free perforation of the gallbladder. Ann Surg 1934; 99:922–924. 225. Felice PR, Trowbridge PE, Ferrara JJ. Evolving changes in the pathogenesis and treatment of the perforated gallbladder: a combined hospital study. Am J Surg 1985; 149: 466–473. 226. Fry DE, Cox RA, Harbrecht PJ. Empyema of the gallbladder: a complication in the natural history of acute cholecystitis. Am J Surg 1981; 141:366–369. 227. vanSonnenberg E, Wittich GR, Casola G, Princenthal RA, Hofmann AF, Keightley A, Wing VW. Diagnostic and therapeutic percutaneous gallbladder procedures. Radiology 1986; 160:23–26. 228. Schwesinger WH, Diehl AK. Changing indications for laparoscopic cholecystectomy: stones without symptoms and symptoms without stones. Surg Clin North Am 1996; 76: 493–504. 229. Barr RG, Agnesi JN, Schaub CR. Acalculous gallbladder disease: US evaluation after slowinfusion cholecystokinin stimulation in symptomatic and asymptomatic adults (see comments). Radiology 1997; 204:105–111. 230. Frykberg ER, Duong TC, LaRosa JJ, Etienne HB. Chronic acalculous gallbladder disease: a clinical variant. South Med J 1988; 81:1353–1357. 231. Frassinelli P, Werner M, Reed JR III, Scagliotti C. Laparoscopic cholecystectomy alleviates pain in patients with acalculous biliary disease. Surg Laparosc Endosc 1998; 8: 30–34. 232. Jones DB, Soper NJ, Brewer JD, Quasebarth MA, Swanson PE, Strasberg SM, Brunt LM. Chronic acalculous cholecystitis: laparoscopic treatment. Surg Laparosc Endosc 1996; 6:114–122. 233. Lygidakis NJ. Surgery for acalculous cholecystitis. An organic and not a functional disease. Am J Gastroenterol 1981; 76:27–31. 234. Yap L, Wycherley AG, Morphett AD, Toouli J. Acalculous biliary pain: cholecystectomy alleviates symptoms in patients with abnormal cholescintigraphy (see comments). Gastroenterology 1991; 101:786–793. 235. Ram M, Midha D. Adenomyomatosis of the gallbladder. Surgery 1975; 78:224–229. 236. Nora PF, McCarthy W, Sanez N. Proceedings: cholecystokinin cholecystography in acalculous gallbladder disease. Arch Surg 1974; 108:507–511. 237. Brugge WR, Brand DL, Atkins HL, Lane BP, Abel WG. Gallbladder dyskinesia in chronic acalculous cholecystitis. Dig Dis Sci 1986; 31:461–467.
Page 623
238. Misra DC Jr., Blossom GB, FinkBennett D, Glover JL. Results of surgical therapy for biliary dyskinesia. Arch Surg 1991; 126:957–960. 239. Davis GB, Berk RN, Scheible FW, Witztum KF, Gilmore IT, Strong RM, Hofmann AF. Cholecystokinin cholecystography, sonography, and scintigraphy: detection of chronic acalculous cholecystitis. AJR 1982; 139:1117–1121. 240. Raptopoulos V, Compton CC, Doherty P, Smith EH, D'Orsi CJ, Patwardhan NA, Goldberg R. Chronic acalculous gallbladder disease: multiimaging evaluation with clinicalpathologic correlation. AJR 1986; 147:721–724. 241. Nathan MH, Newman A, Murray DJ, Camponovo R. Cholecystokinin cholecystography: a four year evaluation. Am J Roentgenol Radium Ther Nucl Med 1970; 110:240–251. 242. Lennard TW, Farndon JR, Taylor RM. Acalculous biliary pain: diagnosis and selection for cholecystectomy using the cholecystokinin test for pain reproduction. Br J Surg 1984; 71:368–370. 243. Sunderland GT, Carter DC. Clinical application of the cholecystokinin provocation test (see comments). Br J Surg 1988; 75:444–449. 244. Westlake PJ, Hershfield NB, Kelly JK, Kloiber R, Lui R, Sutherland LR, Shaffer EA. Chronic right upper quadrant pain without gallstones: does HIDA scan predict outcome after cholecystectomy? Am J Gastroenterol 1990; 85:986–990. 245. Hederstrom E, Forsberg L, Herlin P, Holmin T. Fatty meal provocation monitored by ultrasonography: a method to diagnose ambiguous gallbladder disease. Acta Radiol 1988; 29:207–210. 246. Proudfoot R, Mattingly SS, Snodgrass S, Griffen WO Jr. Cholecystokinin cholecystography: is it a useful test? South Med J 1985; 78:1143–1446. 247. Goldberg HI. Cholecystokinin cholecystography. Semin Roentgenol 1976; 11:175–179. 248. Griffen WO Jr, Bivins BA, Rogers EL, Shearer GR, Liebschutz D, Lieber A. Cholecystokinin cholecystography in the diagnosis of gallbladder disease. Ann Surg 1980; 191:636–640. 249. Mishkind MT, Pruitt RF, Bambini DA, Hakenewerth AM, Thomason MH, Zuger JH, Novick T. Effectiveness of cholecystokininstimulated cholescintigraphy in the diagnosis and treatment of acalculous gallbladder disease. Am Surg 1997; 63:769–774. 250. Barron LG, Rubio PA. Importance of accurate preoperative diagnosis and role of advanced laparoscopic cholecystectomy in relieving chronic acalculous cholecystitis. J Laparoendosc Surg 1995; 5:357–361. 251. Kloiber R, Molnar CP, Shaffer EA. Chronic biliarytype pain in the absence of gallstones: the value of cholecystokinin cholescintigraphy. AJR 1992; 59:509– 513. 252. Zech ER, Simmons LB, Kendrick RR, Soballe PW, Olcese JA, Goff WBd, Lawrence DP, DeWeese RA. Cholecystokinin enhanced hepatobiliary scanning with ejection fraction calculation as an indicator of disease of the gallbladder. Surg Gynecol Obstet 1991; 172:21–24. 253. Berk RN. Cholecystokinin cholecystography in the diagnosis of chronic acalculous cholecystitis and biliary dyskinesia: a critical appraisal. Gastrointest Radiol 1977; 1:325–330. 254. DeCamp JR, Tabatowski K, Schauwecker DS, Siddiqui A, Mullinix FM. Comparison of gallbladder ejection fraction with histopathologic changes in acalculous biliary disease. Clin Nucl Med 1992; 17:784–786. 255. Goncalves RM, Harris JA, Rivera DE. Biliary dyskinesia: natural history and surgical results. Am Surg 1998; 64:493–497; discussion 497–498. 256. Khosla R, Singh A, Miedema BW, Marshall JB. Cholecystectomy alleviates acalculous biliary pain in patients with a reduced gallbladder ejection fraction. South Med J 1997; 90:1087–1090. 257. Fenster LF, Lonborg R, Thirlby RC, Traverso LW. What symptoms does cholecystectomy cure? Insights from an outcomes measurement project and review of the literature. Am J Surg 1995; 169:533–538.
Page 624
258. Reed DN Jr, Fernandez M, Hicks RD. Kinevacassisted cholescintigraphy as an accurate predictor of chronic acalculus gallbladder disease and the likelihood of symptom relief with cholecystectomy. Am Surg 1993; 59:273–277. 259. Gomez NA, Gutierrez J, Leon CJ. Acute acalculous cholecystitis due to Vibrio cholerae (letter). Lancet 1994; 343:1156–1157. 260. Gomez NA, Leon CJ, Gutierrez J. Acute acalculous cholecystitis due to Vibrio cholerae. Surg Endosc 1995; 9:730–732. 261. West BC, Silberman R, Otterson WN. Acalculous cholecystitis and septicemia caused by nonO1 Vibrio cholerae: first reported case and review of biliary infections with Vibrio cholerae. Diagn Microbiol Infect Dis 1998; 30:187–191. 262. Allerberger F LB, Hirsch O, Dierich MP, Seeliger HP. Listeria monocytogenes cholecystitis. Z Gastroenterol 1989; 27:145–147. 263. Benator DA, French AL, Beaudet LM, Levy CS, Orenstein JM. Isospora belli infection associated with acalculous cholecystitis in a patient with AIDS. Ann Intern Med 1994; 121:663–664. 264. Macho JR, Meyer AA. Management of sepsis following injury. Crit Care Clin 1986; 2: 869–876. 265. Rubio PA, Farrell EM, Vitzu M. Postoperative acalculous cholecystitis. Int Surg 1981; 66:167–168. 266. Welling RE, Rath R, Albers JE, Glaser RS. Gastrointestinal complications after cardiac surgery. Arch Surg 1986; 121:1178–1180. 267. Gately JF, Thomas EJ. Acute cholecystitis occurring as a complicaton of other diseases. Arch Surg 1983; 118:1137–1141. 268. Stevens PE, Harrison NA, Rainford DJ. Acute acalculous cholecystitis in acute renal failure. Intens Care Med 1988; 14:411–416. 269. Huilgol VR, Markus SL, Vakil NB. Antibioticinduced iatrogenic hemobilia. Am J Gastroenterol 1997; 92:706–707. 270. Parry SW, Pelias ME, Browder W. Acalculous hypersensitivity cholecystitis: hypothesis of a new clinicopathologic entity. Surgery 1988; 104:911–916. 271. Boland G, Lee MJ, Mueller PR. Acute cholecystitis in the intensive care unit. N Horiz 1993; 1:246–260. 272. Hopkinson GB, Crowson MC, Barnes AD. Perforation of the aculculous gallbladder following renal transplantation. Transplant Proc 1985; 17:2014–2015. 273. Cappell MS. Hepatobiliary manifestations of the acquired immune deficiency syndrome. Am J Gastroenterol 1991; 86:1–15. 274. Hinnant K, Rotterdam H. Acalculous cholecystitis in the acquired immunodeficiency syndrome. Prog AIDS Pathol 1990; 2:151–162. 275. Ikeda S, Kimura W, Futakawa N, Komuro Y, Ono M, Zhao B, Muto T. Acute acalculous cholecystitis with a decrease in CD4/CD8 ratio. J Gastroenterol 1997; 32:268–272. 276. Nash JA, Cohen SA. Gallbladder and biliary tract disease in AIDS. Gastroenterol Clin North Am 1997; 26:323–335. 277. Feltis BA, Lee DA, Gruessner RW. Acute acalculous cholecystitis (AAC) resulting in gallbladder perforation in a solid organ transplant recipient: a case report. Clin Transplant 1998; 12:278–280.
Page 625
29— Gallbladder Cancer R. Montague Beazley Boston University School of Medicine, Boston, Massachusetts I— Introduction Although it is a rare gastrointestinal malignancy, gallbladder carcinoma ranks among the most intractable. Its initial presentation is usually subtle, often mimicking other hepatobiliary and pancreatic lesions, and its treatment is technically challenging, with the results in symptomatic patients being almost uniformly poor. The clinical course of gallbladder carcinoma has been aptly described as "characterized by initial periods of silent progression and a subsequent rapid deterioration" (1). The desperate problem of this disease is largely a function of the anatomy of the gallbladder and pathophysiology of the cancer. However, in spite of many decades of clinical disappointment with the diagnosis and management of gallbladder carcinoma, scattered reports are beginning to show that early diagnosis and aggressive treatment can lead to longterm survival (2). II— Epidemiology In general gallbladder cancer is a disease of the older patient, reaching its maximum incidence in the seventh decade. The incidence increases steadily with age in both sexes (3). Gallbladder cancer is one of the few malignancies with a female predominance, occurring between two and six times more frequently in females than in males. According to the American Cancer Society, there will be 7200 new cases of gallbladder cancer in 1999 (4). However, incidence data are difficult to interpret because gallbladder cancer may easily be confused and reported with other hepatobiliary and pancreatic lesions. Reported to constitute 0.76 to 1.2% of all cancers, it ranks as the fifth most common gastrointestinal malignancy and is the most common malignancy of the biliary tree. There are considerable geographic and ethnic variances in the incidence of gallbladder cancer, with comparisons of institutional registry data indicating a 25fold difference. The highest rates of gallbladder cancer occur in northeastern Europe, Poland, and the previous East Germany in particular; rates are also high among Israelis (especially Jews of European origin), American Indians, and Americans of Mexican origin (5). A number of South American countries have high rates of gallbladder cancer—including Chile, Mexico, and Bolivia— while Rhodesia, Spain, and Bombay, India, have low rates of gallbladder cancer (6,7). According to the U.S. Third National Cancer Survey, the rates among both sexes are 50% higher for whites than for blacks (8). Japan reports an incidence rate intermediate between that of American Indians and whites (9).
Page 626
III— Causation Although no definite genetic risk for gallbladder cancer has been defined in large studies, familial clustering of the disease has been observed. The strong relationship between gallbladder cancer and inflammation—i.e., chronic cholecystitis and cholelithiasis—has been recognized for many years. Indeed, in many series, upwards of 90 to 98% of patients with gallbladder cancer have concomitant gallstones (10). Nervi et al., using autopsy data, calculated that patients with gallstones have a risk of carcinoma seven times higher than that of patients without stones. The exact nature of the linkage between gallstones and gallbladder cancer is obscured by the fact that the frequency of gallstones greatly exceeds the incidence rate of gallbladder cancer. On the other hand, it is recognized that there is a 10 to 25% association between gallbladder cancer and the calcified or "porcelain gallbladder" (11). An association between hepatobiliary cancer, including gallbladder cancer, and typhoid carrier state has also been documented (12). It has been suggested that chronic inflammation of a variety of causes may be a predisposing factor in gallbladder cancer. Another interesting association is the recent observation of a high frequency of anomalous pancreaticobiliary ductal junction (APBDJ) in individuals with gallbladder cancer (13). Defined as the junction between the choledochus and the pancreatic duct occurring outside the duodenal wall and beyond the influence of the sphincter of Oddi, APBDJ is recognized as being related to choledochal cyst, which is predisposed to the development of biliary tract cancer. According to Chijiiwa et al., the normal incidence of: APBDJ approximates 1.5%. In a group of 53 consecutive patients with gallbladder cancer of whom 37 had had endoscopic retrograde cholangiopancreaticography (ERCP), 11% were observed to have APBDJ, a significantly high occurrence of this rare anomaly. None of those patients with APBDJ had gallstones, and they were 10 years younger on average than the stone/cancer patients (13). Chao et al. recently reported similar observations in a group of Taiwanese patients (14). Shimada et al. reported an increase in lysophosphatidylcholine and pancreatic enzymes in the bile of patients with APBDJ (15). Occasionally, gallbladder cancer has been observed in association with polyposis coli, Gardner's syndrome (16), PeutzJeghers syndrome (17), ulcerative colitis (18), and inflammatory bowel disease (19). IV— Pathology Between 75 and 90% of malignant gallbladder tumors are adenocarcinomas; 5 to 10% are squamous cell and 5% small(oat) cell cancers. Very rarely, lymphoma, melanoma, carcinoid, and carcinosarcomas have been reported. In addition, mesenchymal tumors have been described, including rhabdomyosarcomas, leiomyosarcomas, Kaposi's sarcoma, angiosarcomas, malignant fibrohistiocytomas, osteosarcomas, and chondrosarcomas. The gallbladder may also be the site of metastatic tumors such as lung and melanoma, while it may be involved by peritoneal implants or carcinomatosis arising from stomach, breast, ovary, pancreas, colonrectum, and lymphoma. Some 60% of gallbladder cancers arise from the fundus, 30% from the body, and 10% from the neck. About 70% extend into adjacent liver, ultimately encasing or obliterating the gallbladder. The thin gallbladder wall facilitates early tumor spread to the peritoneal cavity as well as tumoral access to draining veins, with subsequent hematogenous spread, particularly by way of veins in the gallbladder fossa entering the portal venous system of segments IV and V. In approximately 70% of cases, direct tumor invasion of these liver segments occurs. In many patients, direct invasion of the porta hepatis also occurs, with infiltration of the common hepatic duct and hilum of the liver. The latter scenario occurs most commonly in the 10% of tumors arising in the gallbladder neck or Hartman's pouch. Jaundice from extrahepatic bile duct invasion may account for gallbladder malignancy occasionally masquerading as Mirrizi's syn
Page 627
drome or a Klatskintype tumor. Tumors gaining access to major ductal structures may also extend intrahepatically along the Glissionian sheath, which surround the bile ducts, portal vein, and hepatic artery. Dye studies of gallbladder lymphatic drainage by Shirai et al. reveal pathways that descend through the copious lymphatics of the porta and by way of firstechelon nodes, the cystic duct, and pericholedochal lymph nodes into secondechelon nodes posterior to the pancreatic head, the portal vein, the common hepatic artery, and into interaortocaval nodes (20) (Fig. 1). The authors assert that it is imperative that the duodenum and pancreas be widely mobilized to fully expose and dissect the peripancreatic and interaortocaval areas. Shirai et al. suggest that by demonstrating internodal connections between the pericholedochal nodes and the intraaortocaval group, the latter group should not be considered as distant metastasis, as in the TMN staging system, but as regional disease (Fig. 2). A progression from atypia to dysplasia to carcinoma in situ may be observed in gallbladders harboring invasive adenocarcinoma. With widespread use of gallbladder ultrasonography, polypoid lesions have been increasingly diagnosed. While most polypoid lesions are benign, polypoid pseudotumors (i.e., cholesterolosis, hyperplastic or inflammatory polyps), there is some evidence for an adenomacarcinoma sequence. In one series, only 7.6% of polypoid lesions were true neoplasms; 43% were benign adenomas and 57% carcinomas (21).
Figure 1 Schematic representation of the nomenclature and location of lymph nodes associated with the gallbladder. The head of the pancreas is raised medially to expose posteriorly located lymph nodes. Arabic numerals indicate each group of lymph nodes: 1, cystic (duct) node(s); 2, pericholedochal nodes; 3, posterosuperior pancreaticoduodenal nodes; 4, retroportal nodes; 5, right coeliac nodes; 6, superior mesenteric nodes; 7, interaortocaval nodes. Ao, aorta; IVC, inferior vena cava; SMA, superior mesenteric artery; IMA, inferior mesenteric artery; LRV, left renal vein; RRV, right renal vein.
Page 628
Figure 2 Diagrammatic representation of the regional lymph nodes of the gallbladder. The arrows indicate the principal routes and the directions of lymph flow from the gallbladder. The broken arrows indicate probable routes, although they were not observed in this study because of the presence of adipose tissue surrounding them.
Single lesions greater than 1 cm in diameter or smaller sessile lesions occurring in patients over the age of 50 years who have gallstones should be strongly suspected of being malignant and treated as such. V— Histology Macroscopically, gallbladder cancer appears as a graywhite mass that may infiltrate the adjacent liver parenchyma. In some cases, the gallbladder will be completely infiltrated or even obliterated by the tumor, although typically papillary tumors may have an intraluminal polypoid component. Microscopically, approximately half the lesions will be classified as well differentiated, but not infrequently, while the superficial portion is well differentiated, the deeper invasive regions will be less well differentiated. Between 4 and 20% of tumors will be classified as papillary, which carries the more favorable prognosis, especially if the lesion is limited to the mucosa. Some 4 to 7% will be termed colloid, 3% will be signetcell type, and 13% will be the pleomorphic giantcell type, which is poorly differentiated. There is a very rare variant resembling renal cell carcinoma called clear cell adenocarcinoma. Some 5 to 10% will generally be classified as squamous cell and approximately 5% as smallcell or oatcell tumors. Problems that pathologists encounter in dealing with gallbladder carcinoma include: 1. Fibrosis of the gallbladder wall with obliteration of the normal anatomic layers. 2. Distinction of dysplastic changes and RokitanskyAschoff sinuses from invasive carcinoma. 3. Inadequate sectioning; frequently, the entire gallbladder must be sectioned. The Japanese Society of Biliary Surgery has classified gallbladder carcinoma microscopically; briefly, into a papillary form or a pedunculated tumor and the nodular form, which is a
Page 629
nonpedunculated type. Papillary, pedunculated, or nodular tumors showing infiltration are classified as infiltrating types. Combinations of the types—such as a papillary tumor that is infiltrating the liver—would be classified as papillary infiltrating (22). VI— Staging The pathological observations described above have been useful in the development of a staging system for gallbladder cancer. In 1976, Nevin and colleagues introduced a fourstage system that used extent of involvement of gallbladder wall, presence or absence of tumor in the liver or other organs, and histological tumor grading (23). Subsequently, this system was modified to include tumors with contiguous liver invasion as stage III and noncontiguous hepatic invasion as stage V (24). However, this modification failed to distinguish patients with invasion through the muscularis from those with minimal liver invasion or those with significant liver invasion (greater than 2 cm.). A third staging system has been proposed by the Japanese Biliary Surgical Society. It consists of four stages based on the presence or absence of lymph node metastasis, serosal invasion, peritoneal dissemination, hepatic invasion, and bile duct infiltration. This system, however, makes no accommodation for mucosal or muscularis invasion (T1) or transmural invasion (T2) (24). The most recent and currently accepted staging system is the AJCC/UICC TMN system, which consists of four stages, with stage III being liver invasion less than 2 cm; or lymph node metastasis (T3N1M0) and stage IV defined as (a) liver invasion greater than 2 cm (T4N0M0 or TxN1M0) or (b) distant metastasis (TxN2M0 or TxNxM1) (26) (Table 1). In a recent review of the Memorial SloanKettering experience, Bartlett el al. concluded nodal status to be the most powerful predictor of patient outcome and have proposed that T4N0 (liver invasion greater than 2 cm) should be included with stage III (27) (Fig. 3). VII— Natural History Between 77 and 97% of patients will present with pain, usually suggestive of cholecystitis. Other less constant symptoms and signs may include weight loss, nausea and vomiting, hepatomegaly and palpable abdominal mass, jaundice, and ascites (1,28). In the French Surgical Association Survey, of 724 patients who underwent surgery, 43% had exploratory laparotomy, but with a 30day mortality of 66%. The authors concluded that 77% of patients were beyond any possibility of curative treatment on initial presentation. These data are confirmed by the Surveillance, Epidemiology and End Results (SEER) Program of the National Cancer Institute (29). The median survival time for patients in this multiinstitutional survey was 3 months, with overall survival rates at 1 and 5 years of 14 and 5% respectively. Table 2 presents relative survival rates as observed from the National Cancer Database (30). VIII— Clinical Presentation The nonspecific signs and symptoms of gallbladder carcinoma frequently mimic those of benign biliary tract disease. Subtle differences, however, may exist to help make the clinical distinction of gallbladder carcinoma. These include a more diffuse nature of pain with a continuous pattern rather than the colicky rightupper quadrant pain typical of gallstone disease. Cancer patients may also have noted weight loss and are possibly less likely to have fever. Signs of jaundice are most commonly associated with unresectable disease. Patients presenting with ascites and a palpable abdominal mass are usually found to be unresectable; likewise, patients presenting
Page 630 Table 1 American Joint Committee on Cancer Staging for Cancer of the Gallbladdera Definition of TNM Primary tumor (T) TX
Primary tumor cannot be assessed
T0
No evidence of primary tumor
Tis
Carcinoma in situ
T1
Tumor invades lamina propria or muscle layer
T1a Tumor invades lamina propria
T1b Tumor invades muscle layer T2
Tumor invades perimuscular connective tissue; no extension beyond serosa or into liver
T3
Tumor perforates the serosa (visceral peritoneum) or directly invades one adjacent organ, or both (extension 2 cm or less into liver)
T4
Tumor extends more than 2 cm into liver, and/or into two or more adjacent organs (stomach, duodenum, colon, pancreas, omentum, extrahepatic bile ducts, any involvement of liver)
Regional lymph nodes (N) NX
Regional lymph nodes cannot be assessed
N0
No regional lymph node metastasis
N1
Metastasis in cystic duct, pericholedochal, and/or hilar lymph nodes (i.e., in the hepatoduodenal ligament)
N2
Metastasis in peripancreatic (head only), periduodenal, periportal, celiac, and/or superior mesenteric lymph nodes
Distant metastasis (M) MX
Distant metastasis cannot be assessed
M0
No distant metastasis
M1
Distant metastasis
Stage grouping Stage Tis 0
N0
M0
Stage T1 I
N0
M0
T2
N0
M0
Stage T1 III
N1
M0
T2
N1
M0
T3
N0
M0
T3
N1
M0
Stage T4 IVA
N0
M0
T4
N1
M0
N2
M0
Any N
M1
Stage Any T IVB
Any T
a
Carcinoid tumors and sarcomas are not included.
with nausea and vomiting secondary to duodenal or gastric outlet obstruction have advanced local disease. A minority of patients, those with early gallbladder carcinoma, are diagnosed "incidentally," being thought to have benign disease, in contrast to those with a preoperative diagnosis of gallbladder carcinoma. Table 3 summarizes the symptoms of gallbladder carcinoma as reflected by recent surgical reports. Early diagnosis of gallbladder carcinoma is problematic, since nonspecific signs and symptoms of this disease usually occur only after a tumor has spread beyond the gallbladder wall. Before the era of ultrasound (US) and computed tomography (CT), the rate of preoper
Page 631
Figure 3 Algorithm for management of gallbladder cancer based on revised staging system. LND, lymph node dissection; CBD, common bile duct. Table 2 Rates of Cumulative Relative Survival Rates 5 Years After Diagnosis, 1989–1990a Survival rate Treatment modalityb
AJCC stage
99% CTc
Cases
Nonradical surgery alone
0
60
58.0
28.8–87.2
I
137
46.7
27.9–65.5
II
129
14.4
1.8–27.0
III
137
8.2
0.0–17.7
IV
180
1.2
0.0–4.7
Radical surgery alone
IV
65
2.3 A
0.0–10.1
Surgery—NOS alone
IV
33
0.0 A
0.0–0.0
Nonradical surgery and adjuvant
I
32
42.0
6.3–77.7
II
50
22.3
1.3–43.3
III
103
7.9
0.0–16.8
IV
99
2.4
0.0–7.4
Radical surgery and adjuvant
IV
34
Nonsurgical treatment
IV
165
0.7
0.0–2.7
No treatment
IV
246
1.4
0.0–4.1
7.6 B
0.0–22.7
Key: AJCC, American Joint Committee on Cancer; NOS, not otherwise specified; CI, confidence interval; A, % survival 4 years after diagnosis, incomplete followup data to allow for 5year calculation; B, % survival 3 years after diagnosis, incomplete followup data to allow for 5year calculation. a Treatment and stage combinations with an insufficient number of cases available for analysis are not reported. b Nonradical surgery: simple surgical removal of primary site with or without lymph node dissection; Radical surgery: total removal of primary site plus partial or total removal of other organs: adjuvant radiotherapy, chemotherapy, or a combination of the two. c 99% CI: ±3 standard errors either side of the calculated survival rate.
Page 632 Table 3 Presenting Symptoms of Gallbladder Carcinoma Pain Jaundice Anorexia Nausea and vomiting Weight loss Fatigue Pruritus
ative diagnosis gallbladder carcinoma was 9.0% (31). With the development of US and CT, rates of preoperative diagnosis have risen to the range of 75 to 88% (32). US is usually the initial diagnostic procedure performed because of its effectiveness in diagnosing gallbladder disease. Gallstone disease can be distinguished from gallbladder carcinoma based on the mobility of calculi when the patient changes position during examination. US will demonstrate thickening of the gallbladder wall and/or polypoid or fungating processes protruding into the gallbladder lumen. More extensive disease with contiguous liver extension or enlargement of lymph nodes can also be documented by US. Iida et al. concluded that preoperative US was as accurate as intraoperative US and that both were superior to CT in detecting intrahepatic invasion. CT is useful in the detection of metastatic tumor or more extensive disease (33). Kumar and Aggarwal, in their review of 50 patients with gallbladder carcinoma, showed a diffuse thickening of the gallbladder in 6% of cancer patients, a polypoid mass within the gallbladder itself in 26% of patients, and a mass filling the entire gallbladder in 42% of the patients, whereas onequarter of the patients had a mass in the gallbladder fossa without a recognizable gallbladder. Lymph nodes greater than 10 mm in size showing heterogeneous enhancement, consistent with malignancy, were correctly diagnosed in 89% of patients with metastatic tumor (34). Surgical approaches of gallbladder carcinoma may be categorized into prophylactic or therapeutic types. Prophylactic surgery should be performed in those individuals who have benign lesions suspected of harboring malignancy—i.e., patients with porcelain gallbladders (25% risk) or gallbladder polyps, particularly lesions in patients over the age of 50 years that are solitary and 1 cm or greater in size (12.1%) (21). Some have proposed aggressive surgical treatment for smaller polyps, especially three in number regardless of size (35). Gallbladder wall thickening in itself is nonspecific and not an absolute surgical indication; however, such patients warrant close followup observation with repeat studies if surgery is not undertaken. Asymptomatic gallstones likewise are not indications for surgery. Laparoscopic surgery should not be used in the prophylactic setting when there is a higherthanaverage risk of gallbladder cancer, as diffuse peritoneal and portsite recurrences are more frequent following laparoscopic surgery (36,37). If, during a laparoscopic procedure, any suspicion of gallbladder cancer is raised, the procedure should be converted to an open one. If cancer is found incidentally in the gallbladder specimen (1 to 2% of all cholecystectomies), it is likely to be an early T1 or T2 lesion. For T1 lesions, most authors do not recommend reoperation. However, if the tumor involves the muscle (T1b), reoperation should be considered to remove the cystic duct and pericholedochal nodes as well as laparoscopic ports. Perforation of the gallbladder and/or bile leakage during resection increases the risk of peritoneal seeding of malignancy and is to be avoided at all costs. IX— Therapeutic Surgery Largely because of the poor prognosis of gallbladder carcinoma and the frequency of late presentation, there is no uniform opinion regarding surgical management. Gagner and Rossi
Page 633
recently surveyed 76 North American surgeons' operative preferences for various stages of gallbladder cancer (38). For lesions with mucosal invasion only, the majority of surgeons (63%) preferred cholecystectomy alone. A small group, 21%, recommended additional node dissection. For submuscosal invasion, 26% opted for cholecystectomy alone; 26% added node dissection, and 30% recommended adding wedge resection of the liver. When the serosa was clearly involved, 12% preferred simple cholecystectomy, while 26% added a wedge resection and 42% recommended node dissection and cholecystectomy. For microscopic or macroscopic liver invasion and positive nodes, half the surgeons would advocate wedge resection combined with cholecystectomy and node dissection. An extended hepatic lobectomy was favored by 16%. After reviewing four published series of gallbladder cancer based on the Nevin's staging (273 patients), the authors pointed out four problems in arriving at a treatment consensus: 1. No uniform pattern of treatment for each stage of disease 2. Radical surgery cannot be assessed according to disease stage 3. The rarity of gallbladder cancer in the United States and the fact that the minority of patients in most reports are candidates for resection 4. The lack of randomized studies comparing treatment modalities according to Nevin's classification X— Stage I Clinically unrecognized gallbladder carcinoma, a lesion confined to the mucosa (T1N0M0), offers the best opportunity for surgical cure. These patients are generally treated by simple cholecystectomy. However, if the diagnosis is made or suspected intraoperatively, cystic and pericholedochal lymph nodes should be sampled. Ouchi and colleagues found that extended cholecystectomy (cholecystectomy, wedge of the liver, and regional lymphadenectomy) was statistically superior to simple cholecystectomy at 5 years (100 versus 60% survival) in 19 patients with lesions involving the mucosa or muscularis extending to but not infiltrating the subserosa (39). In a French multicenter retrospective view, Benoist and coworkers found that 13 patients who had tumor confined to the mucosa (T1a) treated by cholecystectomy alone had a 45% survival at 5 years and 23 individuals with tumors invading the muscularis (T1b) who were similarly treated had a 44% 5year survival (40). XI— Stage II Stage II (T2) consists of tumor invading perimuscular connective tissue; no extension beyond the serosa or into the liver (N0 and M0). For stage II in Benoist's review, actuarial survivals were 61% at 1 year, 30% at 3 years, and 22% at 5 years. Of 26 stage II patients, 23 were treated by simple cholecystectomy and 3 by bile duct resection and resection of liver segments IV and V; 2 of whom these were alive at 5 years without evidence of disease (40). Cholecystectomy alone can be performed for T2 tumors when the tumor is not located in a portion of the gallbladder covered by peritoneum, when there is no tumor contact with other organs, and when regional nodes are negative. Otherwise, a firstlevel regional lymphadenectomy (cystic duct, bile duct, and nodes of the hilum of the liver) with resection of liver segments IV and V should be performed. Patients determined to have T2 lesions not recognized at the time of cholecystectomy should have relaparotomy with lymphadenectomy and liver resection (41). This is most likely to be the case after laparoscopic cholecystectomy. In utilizing the subserosal plane of dissection, it is easy to inadvertently leave a positive tumor margin. Some authors will find that resection of the extrahepaticsuprapancreatic bile duct facilitates en bloc regional node dissection (42). Bartlett et al. recommend duct resection when lymphadenectomy is difficult due to inflammation or a fatty hepatoduodenal ligament or when gross
Page 634
nodal enlargement will result in a close surgical margin (27). The more radical second operative approach was shown by Shirai et al. to be statistically more effective than cholecystectomy alone for T2 tumors (43). The authors conclude that indications for radical second operation for inapparent gallbladder carcinoma are: 1. T2 or more advanced carcinoma 2. Positive surgical margins 3. Positive cystic duct nodes (if examined) XII— Stage III and Stage IV Gallbladder Carcinoma Considerable disagreement exists with regard to the surgical management of patients with stages III and IV gallbladder carcinoma. While an increased incidence of nodal metastasis is frequently associated with increasing ''T" stage, a small percentage of T3 and T4 patients will be found to be pathologically N0. Bartlett et al., in their review of 58 gallbladder cancer patients, documented, by multivariate analysis, that nodal status was the only predictor of survival. The occasional patients who were T3 or T4 but N0 were candidates for aggressive resection. The authors found that out of 30 patients staged as T4, three were N0. Two of the three were found to be alive 4 years postoperatively, whereas no patient with nodal metastasis lived beyond 18 months. The authors postulated that nodal metastasis may be indicative of more aggressive tumor than that which invades the liver parenchyma only locally. For this reason they suggested a modification to the AJCC system, classifying T4, N0, and Tx,N1 tumor as stage IIIB and Tx and N2 tumors as stage IV (27). Benoist et al. concluded that in stages II to IV radical resection should only be considered in the absence of regional lymph nodes (40). They reported a 22% at 5 years survival for 9 stage III patients treated by radical resection and 0 survival for 6 stage III patients who were treated by cholecystectomy alone. Recently, Shirai et al. reported on 17 patients who underwent pancreaticoduodenectomy and hepatectomy with radical lymphadenectomy for gallbladder cancer. Indications for the extended procedure were direct invasion of adjacent organs, stomach, duodenum, or pancreas and/or peripancreatic lymph node metastasis. Five patients survived 5 years (29%); four of these had stage IVb disease with positive peripancreatic nodes. The 5year survival was 50% in those individuals who were considered to have had curative resections, whereas it was 0% (median survival 8 months) in those with incompletely resected tumor. Obviously, careful patient selection is mandatory before embarking upon on an extended procedure of this type, but satisfactory results are obtainable in some individuals with advanced lesions (43). Xl— Adjuvant Therapy Adjuvant therapy is an attractive option in gallbladder cancer because of the low cure rates associated with all but the earliest lesions. Unfortunately, the relative infrequency of the disease has precluded prospective randomized adjuvant studies. Most reports relate to single institutions treating patients with mixed surgical stages; as a result, any conclusions that may be drawn are suspect at best. In general, gallbladder cancer appears to be refractory to chemotherapy, and field limitations restrict radiation dosages. The combined approach of surgery followed by postoperative radiation may be effective in microscopic or subclinical residual disease. However, this approach is less successful in managing unresected macroscopic disease. Intraoperative radiation therapy (IORT) offers the advantage of dose delivery directly to the tumor bed or unresected residual tumor with exclusion of adjacent sensitive normal tissues. A major concern is the limitation of dose to adjacent tissues that are possibly cancer involved, such as liver parenchyma, bile ducts, vessels, and bilioenteric anastomoses. Thus IORT is frequently
Page 635 Table 4 Survival Versus Stage (TNM) in Gallbladder Cancer Survival rates (%) TNM stage
No. of patients
1year
3year
5year
Median survival month ± SEa
I
2
100
100
100
II
3
100
50
50
30 ± 23.3
III
29
44
39.6
IV
74
28.5
39.6
2.0
12 ± 1.5 0
7 ± 0.8
a
SE, standard error; differences in survival were statistically significant (p < 0.05).
followed by externalbeam radiation therapy (EBRT). By means of this approach, positive postsurgical microscopic disease may be controlled (44). Todoroki reviewed 57 patients reported to have had EBRT after resection in which microscopic residual disease was suspected (45). Of these, 45 underwent externalbeam radiation (51.8 Gy) and 12 received IORT (20 Gy), with 10 receiving additional EBRT (total dose, 40.4 Gy). Survival with EBRT alone or in combination with IORT exceeded that for patients who had macroscopic residual or unresectable disease. For stages I and II, 4 of 5 patients were alive 28 to 84 months posttreatment, with a single death occurring at 33 months. Stage III mean survival was 20.6 months (5.5 to 60 months), while in stage IV it was 14.7 months (4.4 to 36 months). Encouragingly, radiation was safely tolerated even by stage IV patients who had had aggressive surgical resections with multiple anastomoses. Todoroki collected data from the literature relating survival to stage of disease as well as tumor remaining after resection and survival. These data are presented in Tables 4 and 5. The benefits of radiation in unresectable gallbladder cancer are problematic and, according to Todoroki's extensive review, survivals average about 6.3 months. Unresectable gallbladder cancer is frequently surrounded by or involves radiosensitive tissues—including the liver, duodenum, stomach, and colon—which limit the therapeutic dose to approximately 60 Gy; this is inadequate to control gross disease. The National Cancer Database shows that palliative nonsurgical treatment was used in 8.9% of cases registered and no therapy was delivered in 17 patients, presumably because of advanced stage or debilitation (30). According to the National Cancer Database, which reviewed 2900 gallbladder cancer patients registered from U.S. hospitals between 1994 and 1995, approximately 4.7% of patients received treatment by chemotherapy alone. Perhaps the reason for the infrequent use of chemotherapy in gallbladder cancers is a widely held opinion that this tumor is chemoresistant. Table 5 Effects of Radiotherapy on Survival in Relation to Tumor Size After Surgery Survival rate (%) Residual tumor size
1year
2year
3year
5year
Median survival months ± SEa
Unresectable
22.5
0
0
0
5.5 ± 1.0
Macroscopic residue
27.2
5.2
0
0
7.5 ± 1.3
Microscopic residue
45.5
36.4
12.1
0
a
SE, standard error; differences in survival were statistically significant (p < 0.05).
12.0 ± 2.9
Page 636
References 1. MD Carmo, MO Perpetuo, M Valdivieso, LK Heilbrun, RS Nelson, T Connor, GP Bodey. Natural history study of gallbladder cancer: A review of 36 years experience at M.D. Anderson Hospital and Tumor Institute. Cancer 1978; 42:330–335. 2. C Bloechle, JR Izbicki, B Passlick, K Gawad, C Passow, X Rogiers, HW Schreiber, CE Broelsch. Is radical surgery in locally advanced gallbladder carcinoma justified? Am J Gastoenterol 1995; 90:2195–2200. 3. GO Strauch. Primary carcinoma of the gallbladder. Surgery 1960; 47:368–383. 4. SA Landis, T Murra, S Bolden, PA Wingo. Cancer statistics. CA 1999; 49:8–31. 5. WC Black, CR Key, TB Carmany, D Herman. Carcinoma of gallbladder in population of southwestern American Indians. Cancer 1977; 39:1267–1279. 6. J RiosDalenz, P Correa, W Haenszel. Morbidity from cancer in La Paz, Bolivia. Int J Cancer 1981; 28:307–314. 7. AK Diehl. Epidemiology of gallbladder cancer: a synthesis of recent data. J Natl Cancer Inst 1980; 65:1209–1213. 8. SJ Cutler, JL Young, eds. Third National Cancer Survey: Incidence Data. Natl Cancer Inst Monogr 1975; 41:22–24. 9. S Tominaga, T Kuroishi, H Ogawa, H Shimizu. Epidemiologic aspects of biliary tract cancer in Japan. Natl Cancer Instit Monogr 1979; 53:25–34. 10. F Nervi, I Duarte, G Gomez, G Rodriquez, G Depino, G Ferrerio, C Covarrubias, V Valdivieso, MI Torres, A Urzuo. Frequency of gallbladder cancer in Chile. Int J Cancer 1988; 41:657–659. 11. HC Polk. Carcinoma and the calcified gallbladder. Gastroenterology 1966; 50:582–585. 12. JC Welton, JS Marr, SM Friedman. Association between hepotobiliary cancer and typhoid carrier state. Lancet 1979; 791–794. 13. K Chijiiwa, M Tanaka, F Nakayama. Adenocarcarcinoma of the gallbladder associated with anomalous pancreaticobiliary ductal junction. Am Surg 1993; 59:430–434. 14. T Chao, VY Jan, M Chen. Primary carcinoma of the gallbladder associated with anomalous pancreatic biliary ductal junction. J Clin Gastroenterol 1995; 21:306– 308. 15. K Shimada, J Yanagisawa, F Nakayama. Increased lysopophospatidylcholine and pancreatic enzyme content in bile of patients with anomalous pancreaticobiliary ductal junction. Hepatology 1991; 13:438–444. 16. N Walsh, A Qizilbash, R Banerjee, GA Waugh. Biliary neoplasia in Gardner's syndrome. Arch Pathol Lab Med 1987; 111:76–77. 17. K Wada, M Tanaka, K Yamaguchi, K Wada. Carcinoma and polyps of the gallbladder associated with PeutzJeghers syndrome. Dig Dis Sci 1987; 32:943– 946. 18. RO Connor, B Harding, D Greene, J Coolican. Primary carcinoma of the gallbladder associated with ulcerative colitis. Postgrad Med J 1986; 62:871–872. 19. N Joffe, D Antonioli. Primary carcinoma of the gallbladder associated with chronic inflammatory bowel disease. Clin Radiol 1981; 32:319–324. 20. Y Shirai, K Yoshida, K Tsukada, T Ohtani, T Muto. Identification of the regional lymphatic system of the gallbladder by vital staining. Br J Surg 1992; 79:659– 662. 21. HL Yang, YG Sun, Z Wang. Polypoid lesions of the gallbladder: diagnosis and indications for surgery. Br J Surg 1992; 79:227–229. 22. K Sumiyoshi, E Nagai, K Chijiiwa, F Nakayama. Pathology of carcinoma of the gallbladder. World J Surg 1991; 15:315–321. 23. JE Nevin, TJ Morgan, S Kay, R King. Carcinoma of the gallbladder: staging, treatment and prognosis. Cancer 1976; 37:141–148. 24. JH Donohue, DM Nagorney, CS Grant, K Tsushima, DM Ilstrup, MA Adson. Carcinoma of the gallbladder: does radical resection improve outcome? Arch Surg 1990; 125:237–241. 25. H Onoyama, M Yamamoto, A Tseng, T Ajiki, Y Saitoh. Extended cholecystectomy for carcinoma of the gallbladder. World J Surg 1995; 19:758–763.
Page 637
26. O Beahrs, DE Henderson, RVP Hutter, MH Meyers, American Joint Committee on Cancer: Gallbladder. In: Manual for Staging of Cancer. Philadelphia: Lippincott 1988, pp 93–98. 27. DL Bartlett, Y Fong, JG Fortner, MF Brennan, LH Blumgart. Longterm results after resection for gallbladder cancer: indication for staging and management. Ann Surg 1996; 224:639–646. 28. P Cubertafond, A Gainant, G Cucchiaro. Surgical treatment of 724 carcinomas of the gallbladder: results of the French Surgical Association survey. Ann Surg 1994; 219:275–280. 29. DE Henson, JA Saavedra, D Corle. Carcinoma of the gallbladder: histologic types, stages of disease, grade, and survival rates. Cancer 1992; 70:1493–1497. 30. JH Donohue, AK Stewart, HR Menck. The National Cancer Database Report on Carcinoma of the gallbladder, 1989–1995. Cancer 1998; 83:2618–2628. 31. JM Piehler, RW Crichlow. Primary carcinoma of the gallbladder. Surg Gynecol Obstet 1978; 147:929–942. 32. K Chijiiwa, K Sumiyoshi, F Nakayama. Impact of recent advances in hepatobiliary imaging techniques on the preoperative diagnosis of carcinoma of the gallbladder. W J Surg 1991; 15:322–327. 33. F Iida, S Kajikawa, N Horigome. Evaluation of imaging examination for hepatic invasion of carcinoma of the gallbladder and postoperative patient outcome. J Am Coll Surg 1995; 180:72–76. 34. A Kumar, S Aggarwal. Carcinoma of the gallbladder: CT findings in 50 cases. Abdom Imaging 1994; 19:304–308. 35. H Shinaki, W Kimura, T Muto. Surgical indications for small polypoid lesions of the gallbladder. Am J Surg 1998; 175:114–117. 36. K Kubota, Y Bandai, T Noie, Y Ishizaki, M Teruya, M Makuuchi. How should polypoid lesions of the gallbladder be treated in the era of laparoscopic cholecystectomy? Surgery 1995; 117:481–487. 37. LA Wibbenmeyer, TP Wade, RC Chen, RC Meyer, RP Turgeon, CH Andrus. Laparoscopic cholecystectomy can disseminate in situ carcinoma of the gallbladder. J Am Coll Surg 1995; 181:504–510. 38. M Gagner, RL Rossi. Radical operations for carcinoma of the gallbladder: present status in North America. World J Surg 1991; 15:344–347. 39. K Ouchi, M Suzuki, T Tominaga, S Saijo, S Matsuno. Survival after surgery for cancer of the gallbladder. Br J Surg 1994; 81:1655–1657. 40. S Benoist, Y Panis, PL Fagniez. Longterm results after curative resection for carcinoma of the gallbladder. Am J Surg 1998; 175:118–122. 41. CE Morrow, DER Sutherland, G Florack, MM Eisenberg, TB Grage. Primary gallbladder carcinoma: significance of subserosal lesions and results of aggressive surgical treatment and adjuvant chemotherapy. Surgery 1983; 94:709–714. 42. Y Shirai, K Yoshida, K Tsukada, T Muto. Inapparent carcinoma of the gallbladder: an appraisal of a radical second operation simple cholecystectomy. Ann Surg 1992; 215:326–331. 43. Y Shirai, T Ohtani, K Tsukada, K Hatakeyama. Combined pancreaticoduodenectomy and hepatectomy for patients with locally advanced gallbladder carcinoma. Cancer 1997; 80:1904–1907. 44. M Mahe, C Stampfli, P Romestaing. Primary carcinoma of the gallbladder: potential for external radiation therapy. Radiother Oncol 1994; 33:204–208. 45. T Todoroki. Radiation therapy for primary gallbladder cancer. Hepatogastroenterology 1997; 44:1229–1239.
Page 639
30— Congenital and Cystic Diseases of the Biliary Tree David McAneny and James M. Becker Boston University School of Medicine, Boston, Massachusetts I— Introduction A variety of congenital diseases of the biliary tract occur due to aberrant development of the intrahepatic and extrahepatic bile ducts. Biliary atresia and choledochal cysts constitute the vast majority of these rare disorders. Although the mechanisms of these conditions are not fully elucidated, their respective managements have steadily evolved. II— Biliary Tract Embryology Hepatoblasts are bipotential liver precursor cells from which arise cholangiocytes or hepatocytes. The intrahepatic bile duct system begins to develop during the eighth week of embryonic life, when some hepatoblasts align in a sleeve surrounding the contiguous major portal vein branches. This primordial structure has been described as the ductal plate (1). A second layer of the ductal plate cells eventually accumulates. The portal layer abuts the mesenchyme about the portal vein branches, whereas the lobular layer is adjacent to the hepatic parenchyma. In time, a lumen develops between these two epithelial layers (2). The ductal plate formation propagates from the liver hilum to the peripheral portal vein branches. A similar pattern of ductal plate remodeling begins about the 12th week and results in the creation of tubular bile ducts within the periportal mesenchyme (3). Certain portions of the duplicated ductal plates dilate and later become bile ducts. The superfluous, nondilated epithelial cells apparently involute (4). The critical elements of ductal plate remodeling seem to involve the modulation of the epithelial layers, the periportal mesenchyme, and the actual portal veins (3). The intrahepatic bile duct cells convey immunoreactivity to transforming growth factor alpha throughout fetal life, and this factor may impart an autocrine effect upon duct formation (5). Mesenchymal components such as laminin, collagen type IV, and tenascin influence the differentiation of the ductal hepatoblasts (6). Finally, the portal vein system seems to provide a sort of scaffold for the embryonic ductal plates, whereas this phenomenon is not observed about the hepatic veins. The extrahepatic bile ducts and gallbladder derive from the endodermal epithelium of the distal foregut (7). This hepatic diverticulum evolves during the third week of embryonic life. When the diverticulum penetrates the septum transversum and detaches from the foregut,
Page 640
two sections arise. The pars hepatica becomes the liver and the more caudal pars cystica is the precursor of the gallbladder and the extrahepatic biliary tract. The pancreas derives from dorsal and ventral anlagen, the latter of which develops from and drains into the primordial distal common bile duct. After the clockwise rotation of the ventral anlage about the duodenum, the dorsal and ventral buds fuse, as do their respective ducts. The major duct of Wirsung is created by an anastomosis between the ventral duct and the left aspect of the dorsal duct. As a result of the origin of the ventral anlage, the distal bile duct and the duct of Wirsung share a common channel that is normally 0.2 to 1 cm in length (8). III— Biliary Atresia A— Overview Biliary atresia is a progressive obliterative process that affects the perinatal bile ducts in approximately 1 of 10,000 to 13,000 live births (9). This incidence yields about 400 to 600 cases each year in the United States (10). Although no racial or genetic association has been determined with biliary atresia, there is a distinct femaletomale predilection in a ratio of 1.4:1 (11). Most cases are isolated disorders, but as many as 20 to 30% of neonates have concomitant developmental malformations, including polysplenia or asplenia, anomalies of the portal vein and hepatic artery, abdominal situs inversus, intestinal malrotation, and cardiovascular or urinary system defects (12,13). B— Pathophysiology Biliary atresia is an idiopathic fibroobliterative condition that affects the bile ducts at any point from the hilar bifurcation to the duodenum. It is characterized by a sclerosing inflammation, fibrosis, and destruction of the ducts that results in progressive cholestasis, hepatic fibrosis, and eventual cirrhosis. Perinatal leakage of bile (with hydrophobic, hepatotoxic bile acids) from the ductal plates may provoke the intense inflammatory response. Two traditional theories of the development of biliary atresia have been recently disputed (4,13). One premise was based upon errant recanalization of a purported "solid phase" of bile duct formation. However, this solid stage of the bile duct lumen likely does not occur. The other theory proposed a failed fusion of the intrahepatic ducts of the ductal plate with the extrahepatic ducts of the pars cystica. Despite this attractive concept, it seems that there is no moment of embryonic development during which the two ductal systems are dissociated. Current evidence suggests that a disruption of the delicate balance between the bile duct epithelium and the investing mesenchyme arrests the remodeling process and creates ductal plate malformations (14), which have been identified in up to twothirds of infants with biliary atresia (15). This association implies that atresia is instigated in early fetal life, when the ductal plates are forming. Five mechanisms of pathogenesis have been proposed. These include a viral etiology, a toxic exposure, a defect in morphogenesis, an immune or inflammatory reaction, and an ischemic insult. With regard to viruses, isolated cases of atresia with reovirus type 3 seroreactivity have been described (16), and biliary atresia has resulted from reovirus inoculation in a murine model (17). Nevertheless, more recent data do not corroborate such an association in infants (18). Group A rotavirus has also been shown to cause biliary obstruction in mice (19), but polymerase chain reaction immunoassay techniques have not identified this virus as a common finding in biliary atresia (20). Finally, the impact of cytomegalovirus as a causative factor is not established. Although the timespace clustering of atresia cases has suggested viral causes, it also raises the possibility of a provocative toxin. However, no specific toxin has been declared a source of biliary atresia.
Page 641
A variety of defects in the morphogenesis of the extrahepatic biliary tree have been postulated. Just as a considerable number of infants with biliary atresia also have anomalies of visceral organ symmetry (12), transgenic mice with a recessive mutation in chromosome 4 have similar anomalies and die with severe jaundice in less than a week (21). The mutated gene is yet to be isolated. Another defect in morphogenesis could involve hepatocyte growth factor and the cmet oncogene, at least in a tissuespecific manner or perhaps at a critical moment of ductal plate formation and remodeling (22). Various intracellular adhesion molecules might also be abnormally expressed, again disturbing the ductal plate organization (23). Several features suggest an altered immune response in biliary atresia. The HLAB12 allele is significantly associated with these infants versus normal neonates, particularly in the absence of other congenital anomalies (24). Furthermore, abnormal expressions of certain molecules on the surface of bile duct epithelial cells possibly incite an inflammatory reaction to and destruction of the ductal plate. These include HLA class I molecules, intracellular adhesion molecule 1, and vascular cell adhesion molecule (25,26). Although multiple cases of biliary atresia have been identified within some families, there are also reports of identical twins discordant for this disorder (27). The immunology and genetics of atresia remain a challenge. Arterial pathology has also been recognized in tissues affected by biliary atresia (28). The histology suggests that some cases could be related to a fetal vascular insult. C— Clinical Presentation Infants with biliary atresia generally appear healthy and are of normal weight. This condition should be suspected when jaundice, typically manifested with acholic stools, persists 2 weeks after birth, although the differential diagnosis of neonatal conjugated hyperbilirubinemia is extensive (Table 1). Upon physical examination, ascites and hepatosplenomegaly may be evident. Two clinical patterns of atresia have been outlined (Table 2) (29). The fetal type includes an early onset of jaundice, and it is occasionally associated with other congenital anomalies. This variant is most likely initiated in utero. Conversely, the more common perinatal type frequently has a jaundicefree interval immediately after birth and other anomalies are not present. Notably, bile duct remnants are identified in the hepatoduodenal ligament in the perinatal pattern, in contrast to the fetal type. It is conceivable that the fetal and perinatal phenotypes have different etiologies. Kasai has classified the anatomic extent of biliary atresia (30). This process obliterates all or a limited portion of the extrahepatic biliary tract, as demonstrated in Fig. 1. In three of the four variations, ductal patency is maintained at the hilum. However, in type III, a fibrous cone extends throughout the biliary tract, including the hilar ducts, and no patent ducts are available for a surgical anastomosis. A precise mapping of the atresia is critical to its treatment. D— Evaluation The successful management of biliary atresia depends on the correction of the condition within the first 2 to 3 months of life. As a result, neonatal jaundice that persists beyond 2 weeks should trigger an expeditious evaluation to differentiate between biliary pathology and hepatocellular disease. In the United Kingdom, a "Yellow Alert" campaign was begun in 1993 to educate family doctors, midwives, and health visitors about the urgency of recognizing and evaluating jaundice in infants. An analysis of this public health effort suggests that an aggressive investigation after a 2week interval of jaundice is costeffective, especially considering the added benefits of determining other hepatobiliary diseases and of preventing major intracranial hemorrhages from vitamin K malabsorption (31). The differential diagnosis of conjugated hyperbilirubinemia in the infant is broad (Table 1), but neonatal hepatitis and biliary atresia account for 90% of cases in which conjugated hyperbilirubinemia exceeds 4 weeks (32). No single test short of laparotomy is diagnostic of biliary atresia. Important laboratory investigations include serum bilirubin (total and conjugated
Page 642 Table 1 Differential Diagnosis of Conjugated Hyperbilirubinemia in the Newborn Infectious
Metabolic
Coxsackievirus
Antitrypsin deficiency
1
Cytomegalovirus
Cystic fibrosis
Echovirus
Fructose intolerance
Hepatitis B virus
Galactosemia
Herpes simplex
Gaucher disease
Listeria
Hypothyroidism
Parvovirus B19
NeimannPick disease
Rubella virus
Tyrosinemia
Syphilis
Intrahepatic
Toxoplasmosis
Alagille syndrome
Tuberculosis
Byler disease
Varicella zoster virus
Caroli disease
Toxic
Congenital hepatic fibrosis
Parenteral nutrition related
Idiopathic neonatal hepatitis
Gramnegative sepsis Drugrelated
Nonsyndromatic bile duct paucity Extrahepatic
Genetic
Bile duct stenosis
Donahue syndrome
Biliary atresia
Trisomy 21
Choledochal cyst
Trisomy E
Extrinsic biliary tree compression
Miscellaneous
Inspissated bile syndrome
Histiocytosis X
Sclerosing cholangitis
Neonatal lupus erythematosus
Myeloproliferative disease
Source: From Ref. 7.
fraction), assays for congenital infections, thyroxine and 1antitrypsin levels, cultures, and a sweat chloride test (7). Several imaging studies are valuable in the assessment of neonatal jaundice. Ultrasonography examines the hepatic parenchyma, detects biliary structural anomalies such as choledochal cysts, and reveals findings of portal hypertension. Nevertheless, biliary atresia and neonatal hepatitis are often sonographically indistinguishable. For instance, as with hepatitis, dilated bile Table 2 Clinical Patterns of Biliary Atresia Embryonic or fetal type (35%) 1. Early onset of neonatal cholestasis 2. No jaundicefree period after physiological jaundice 3. No bile duct remnants in hepatoduodenal ligament 4. Associated congenital anomalies (10–20% of cases) Perinatal type (65%) 1. "Later onset" of neonatal cholestasis 2. Jaundicefree interval may be present after physiological jaundice 3. Remnants of bile duct structures found in hepatoduodenal ligament 4. No associated congenital anomalies Source: From Ref. 29.
Page 643
Figure 1 Patterns of extrahepatic biliary atresia. Type I, the "correctable" pattern, describes a patent common hepatic duct and an occluded distal bile duct. Type IIa refers to a patent gallbladder that is contiguous with a patent common bile duct. The gallbladder may serve as a conduit between the ductules of the hilum and the distal bile duct. In Type IIb, the most common variant, the entire extrahepatic biliary tract is obliterated. Type III portrays a "noncorrectable" pattern in which no ductules are present at the hilum of the liver and the entire extrahepatic biliary tract is occluded. (From Ref. 37.)
ducts are not evident in biliary atresia, likely because of reduced bile flow (33). Also, while evidence of a normal gallbladder suggests neonatal hepatitis (34), some infants with atresia have normalsized gallbladders by ultrasound (35). Therefore, this modality provides useful information but it is not necessarily diagnostic. Scintigraphy with 99mTcIDA is a particularly good test because the appearance of radionuclide in the small bowel effectively rules out biliary atresia (33). However, the value of this study is limited in severe neonatal hepatitis if the hepatocytes cannot extract and excrete the radionuclide. In this situation, the administration of oral phenobarbital may enhance the hepatic metabolism of 99mTcIDA. Computed tomography and magnetic resonance imaging are not ordinarily employed in the evaluation of biliary atresia, although magnetic resonance cholangiography offers some promise as it is refined. Percutaneous transhepatic cholangiography, generally through the gallbladder in infants, and endoscopic retrograde cholangiopancreatography are selectively applied invasive techniques. The combination of scintigraphy and percutaneous biopsy constitutes the diagnostic routine in many centers (29). When five to seven portal spaces are identified upon microscopic examination, a percutaneous liver biopsy conveys a 95% accuracy in the diagnosis of biliary atresia (36). Common histology characteristics include marginal proliferation of bile ductules, canalicular and cellular bile stasis, portal tract or perilobular edema or fibrosis, extramedullary hematopoiesis, infiltrates of neutrophils and lymphocytes in the portal tracts, and swelling, sacualization, and sloughing of biliary epithelium into the ductal lumen (37,38). Laparoscopy is currently not part of the routine evaluation of biliary atresia. However, when the diagnosis remains in doubt despite the above studies, an empirical laparotomy through a limited rightupperquadrant incision is indicated. Biliary atresia is established if the gallbladder is replaced by a fibrous structure. On the other hand, a cholangiogram is projected through the gallbladder if the organ appears to be normal. If cholangiography demonstrates a patent biliary tree from the liver to the duodenum, the diagnosis of atresia is excluded. It is
Page 644
essential to monitor coagulation factors perioperatively because a vitamin K responsive hemorrhagic diathesis can develop after 3 to 8 weeks of jaundice (31). E— Treatment Thomson first described biliary atresia in 1891 (39). Although Holmes demonstrated in 1916 that a distinct minority of infants had patent hilar ducts that would be amenable to surgical anastomosis (40), it was not until 1928 that Ladd performed such a reconstruction. Kasai and Suzuki reported the original portoenterostomy for biliary atresia with obliterated hilar ducts in 1959 (41). This landmark effort has become known as the Kasai procedure. The development of the Kasai portoenterostomy was predicated on the recognition that microscopic bile ducts are present in the fibrotic porta hepatis of infants with biliary atresia, at least early in the neonatal period. A laparotomy is performed through a rightupperquadrant incision and the liver and biliary tract are examined. A liver biopsy is obtained and the gallbladder is cannulated for a cholangiogram if it seems to have a patent lumen. In the most common variant of biliary atresia, Kasai's type IIb (Fig. 1), the entire extrahepatic biliary tract is involved (30). At surgery, the bile duct remnant is dissected to its bifurcation in the liver hilum, where the typically fibrotic "cone" is transected flush with the liver parenchyma. The divided duct is examined by frozen section analysis to determine the patency and diameter of ductules. If ducts are observed, a defunctionalized Roux limb of jejunum is sutured to the liver edge without mucosal apposition. If no substantial ductules are apparent, a more proximal dissection is conducted and the above process is repeated. About 5 to 10% of infants with biliary atresia have the "correctable" pattern with a patent common hepatic duct (type I, Fig. 1) (42). Although the hepatic duct is often ectatic in type I, this could be attributed to a nonepithelialized pseudocyst. Therefore, the duct is still divided at the liver hilum and an anastomosis is again created with a RouxenY loop of jejunum. Approximately 10 to 15% of patients have a patent gallbladder in continuity with a patent common bile duct (type IIa, Fig. 1) (42). These infants are managed with a portocholecystostomy, wherein the gallbladder serves as the conduit between distal bile duct and the microscopic ductules of the hilum. Type III atresia describes a "noncorrectable" pattern in which no ductules are microscopically present at the hilum and liver transplantation may be necessary. A variety of modifications have been applied to the original Kasai portoenterostomy in attempts to avoid complications, primarily cholangitis. Operations to prevent reflux of gut contents into the liver have included the creation of an intussusception valve in the distal efferent limb and the exteriorization of the efferent limb of the portoenterostomy. In the Sawaguchi modification, an isolated segment of jejunum diverts all bile to a cutaneous stoma (43). While these alternatives to the Kasai procedure have occasionally succeeded in some regards, none has consistently demonstrated a substantial improvement in outcome or mortality in comparison to the standard retrocolic RouxenY portoenterostomy that most centers currently practice (44,45). Disadvantages of the modifications include the need for additional operations (e.g., stoma closure or revision), stoma complications such as variceal hemorrhage, fluid and electrolyte depletion, and the potential to confound a later transplantation (44). Interestingly, a previous Kasai portoenterostomy did not adversely affect transplantation results in at least one series that specifically examined survival, operative blood loss, infections, and biliary complications (46). Some infants have inadequate portal ductules to support a portoenterostomy. Others initially respond to the Kasai procedure but later develop liver failure as a result of progressive duct sclerosis, usually from cholangitis. About twothirds of patients with biliary atresia will ultimately undergo liver transplantation (7). In fact, this has become the most common indication for pediatric liver transplantation in some centers (45). Recent advances in reducedsize, splitliver, and livingrelated transplant techniques have provided more options for small recipients (47). The limited availability of donor organs, especially of a matching size, has
Page 645
generated a philosophy of attempting a Kasai portoenterostomy as the primary therapy and reserving transplantation for subsequent liver failure. Transplantation is recommended for infants who never achieve sufficient bile drainage with a portoenterostomy and for those in whom the Kasai procedure is highly unlikely to succeed due to advanced liver disease, as manifest by cirrhosis and portal hypertension (48). Children whose livers fail over time due to progressive cholangitis and sclerosis will also require transplants. Liver transplantation currently cannot replace the portoenterostomy as the primary operation for all infants with biliary atresia for at least two practical reasons. First, a significant number of patients would die of liver failure before a suitable donor presented. Second, perhaps onethird of infants on whom portoenterostomy is performed will experience longterm, jaundicefree survival and will not require immunosuppression. Therefore, the Kasai portoenterostomy and liver transplantation are not at all competitive or mutually exclusive procedures. Instead, they are complementary components in the management of biliary atresia. F— Outcomes Without surgical intervention, less than 10% of children with biliary atresia survive 3 years (11). Various prognostic factors for effective bile drainage and survival following portoenterostomy have been identified and many are related to age. Therefore, time is critical during the evaluation and treatment of these patients. Long term biliary drainage is established in about half of the infants, and 10year survival is about 20% if the Kasai procedure is performed within 60 to 90 days of life (30,49,50). Moreover, if the surgery is conducted before 60 days, adequate biliary drainage is achieved in about 80% of cases and almost threequarters of the children survive 10 years (30,49,50). Age clearly influences factors such as the diameter of the bile duct remnant and hepatic fibrosis, indicating the dynamic nature of atresia. Ducts with diameters greater than 150 m have been reported to promote vastly better bile drainage than when the lumen is less than 50 m or the ducts are not epithelialized (51). However, others dispute whether duct diameter alters survival (52). The amount of hepatic fibrosis and cirrhosis also impacts outcome negatively, although the degree of inflammation along the extrahepatic ducts does not seem to be a significant factor (11). As would be expected, changes of portal hypertension—such as varices and ascites—are not favorable (11). A recent study used histology criteria of hepatitic changes and of cholangitis, predicting the likelihood of a successful portoenterostomy with an 86% sensitivity and an 88% specificity (53). The patterns of biliary atresia also have a bearing on outcome. Fiveyear survival among infants with a ''correctable" (type I) pattern was 72% after the Kasai operation, in contrast to only 38% for infants with totally obliterated extrahepatic ducts (type IIb) (11). A patent distal duct (type IIa), amenable to a portocholecystostomy, conveyed a 5year survival of 62%. In that the pattern of this disease determines the nature of surgical drainage, the type of operation does not directly influence survival, although the experience of the surgeon is critical (54). Just as the histology characteristics of livers do not differ between patients with or without congenital anomalies (29), the Biliary Atresia Registry revealed that associated anomalies do not necessarily diminish survival (11). The registry data do suggest that Caucasian race adversely affects survival, but the mechanism of this is unclear. Some investigators have recently identified prognostic laboratory tests. One group used regression analysis to design an equation that included serum parameters such as copper/zinc weight ratio, zinc sulfide turbidity, total bilirubin, cholinesterase, and gammaglutamyl transpeptidase (55). This formula appears to be valuable in predicting the risk of developing cirrhosis. Another project revealed greater expression of transforming growth factor alpha and proliferating cell nuclear antigen among infants in whom jaundice persisted after the Kasai procedure (56). These findings might eventually be commonly applied in the clinical arena. In the Biliary Atresia Registry, 816 patients underwent biliary drainage procedures with an operative mortality of just 0.4% (11). Early postoperative complications such as intraab
Page 646
dominal abscess, bowel obstruction, and hemorrhage were also uncommon. Effective bile drainage is initially accomplished in over threefourths of patients. The most common late complications of portoenterostomy are cholangitis and sequelae of portal hypertension. As mentioned earlier, technical modifications of the Kasai procedure have not diminished the incidence of recurrent cholangitis, which contributes to progressive duct injury, sclerosis, and reobstruction (57). Portocholecystostomy is associated with less frequent cholangitis than the standard Kasai procedure (11), but the gallbladder is a suitable conduit only when the distal bile duct is patent (type IIa). Antibiotics and hydration are the mainstays of the management of acute cholangitis, though there is no support for prophylactic therapy (58). Portal hypertension is present in most infants at the time of their original Kasai portoenterostomies (58), and it is evident in as many as twothirds of longterm survivors (59). It often manifests with variceal hemorrhage, ascites, and hypersplenism. Despite the debilities posed by both the repetitive sepsis of cholangitis and the effects of portal hypertension, most 10year survivors grow and develop reasonably well (59). About onequarter to onethird of infants with biliary atresia survive over 10 years after portoenterostomy (11,58,59). However, most longterm survivors have some degree of hepatic dysfunction and portal hypertension (58). In fact, liver failure, the related sepsis, and gastrointestinal hemorrhage account for most deaths (11). If jaundice resolves following portoenterostomy, 10year survival rates have been reported to be as high as 73 to 92% (29). Rather than replace portoenterostomy, liver transplantation is a wonderful adjunct for those patients who succumb to liver failure after portoenterostomy. Furthermore, even though the Kasai procedure is not necessarily curative, it can extend survival until such an age that the child is a better candidate for transplantation. About one third of patients with biliary atresia currently require transplantation at the age of 1 year, and another onethird will need a liver transplant by the teen years (29). Survival after transplantation ranges from 70 to 90%, with an excellent quality of life (29,58). IV— Choledochal Cysts A— Overview A choledochal cyst is a congenital ductal ectasia that involves a localized portion or a diffuse distribution of the biliary tract. It has been estimated to occur in 1 of 13,000 to 15,000 births in western countries (60), but it is perhaps 13 times as prevalent in Japan and other Asian nations (61). The recognition of cysts is actually rising with the increased usage of ultrasonography for fetal monitoring (61). The female sex is affected with a fourfold greater incidence than are males. While most choledochal cysts are diagnosed by the age of 10 years, adults present with these lesions as well and describe biliary symptoms. Frequent anatomic characteristics include an anomalous junction between the common bile duct and the pancreatic duct, intrahepatic ductal dilatation, and hepatic fibrosis (61). B— Pathophysiology The etiology of choledochal cysts is not well established. Potential causes might be a congenital weakness of the bile duct wall, errant ductal proliferation during fetal life, and a congenital obstruction of the distal duct (61). The current prevailing theory indicts an anomalous junction of the common bile duct with the duct of Wirsung, resulting in an unusually long common channel. This aberrant anatomy allows reflux of pancreatic juice into the biliary tract, where it incites intense inflammation, leading to mucosal destruction, a weakened duct wall, and fibrosis. However, an acquired variant of biliary cystic dilatation can evidently develop in adults (62). It could be that the choledochal cyst is the product of a congenital anomaly promoting an acquired biliary ectasia.
Page 647
A peculiar connection between the common bile duct and the pancreatic bulb was proposed as early as 1916 (63). Babbitt suggested the relationship between choledochal cysts and an anomalous pancreaticobiliary ductal junction (APBDJ) in 1969 (64). Although an APBDJ was found in only 2% of all cholangiograms in one series, almost threequarters of these patients had choledochal cysts (65). The embryological theory of the APBDJ proposes that the ventral anlage of pancreas arises from the primordial bile duct closer to the liver than normal and outside the duodenal wall. The length of the resulting common channel is 1.5 to 4.5 cm, with an average length of 2.8 cm, about five times greater than normal. Another important element of the APBDJ is the acuity of the angle of the communication between the bile duct and the duct of Wirsung. The APBDJ is conducive to the reflux of pancreatic fluid into the common bile duct, particularly considering the two to threefold greater maximum pressure in the pancreatic duct (66). In a rodent model of pancreaticobiliary malunion, the pancreatic proenzyme of phospholipaseA2 became activated upon exposure to bile, resulting in foci of pancreatic necrosis (67). Canine experiments have demonstrated a dilatation of the common bile duct following prolonged exposure to pancreatic fluid (68). High levels of amylase have also been measured in some choledochal cysts (68). Despite this strong supportive evidence, the embryological theory does not account for the few choledochal cysts that are not associated with an APBDJ. Furthermore, pancreatic acinar function is essentially absent during early fetal life, when some choledochal cysts have been identified by ultrasound (61). Finally, the bile in young children's cysts is usually not amylaserich. Two other, less popular, theories of pathogenesis have been espoused (7). One theory involves the concept of errant canalization of the bile duct with resultant distal obstruction and increased biliary pressure. The pressure presumably disrupts the integrity of the bile duct wall. The other theory relates to altered autonomic innervation in the distal bile duct. Laboratory models support both antenatal and acquired factors in the etiology of different choledochal cysts. Two histology characteristics of choledochal cysts are acknowledged (7). In the glandular type, portions of the cuboidal celllined epithelium are denuded, with evidence of acute and chronic inflammation. In the fibrotic type, acute inflammation is notably absent, although mural fibrosis is prominent. These features may relate to the duration of exposure of refluxed pancreatic enzymes to the biliary tract (69). Similarly, the degree of hepatic periportal fibrosis is a function of the extent and chronicity of biliary obstruction and the repetitive insults of cholangitis. C— Clinical Presentation Published reports observe the presence of the classic triad of a rightupperquadrant mass, jaundice, and pain in 13 to 63% of patients with choledochal cysts (70). Nevertheless, probably twothirds of patients do not exhibit the full triad (66). Other presentations include acute pancreatitis, cholangitis, variceal hemorrhage from portal hypertension, and spontaneous cyst rupture with bile peritonitis or ascites. When cysts degenerate into a malignancy, the presenting symptoms can be indistinguishable from the benign disease, although weight loss might be a component. Some cysts are discovered fortuitously, even in utero, by imaging techniques. The clinical manifestations seem to be related to a number of factors, including the type or morphology of the cyst, the age of the patient, and the acuity of the angle between the common bile duct and the duct of Wirsung. The Todani modification (71) of the heralded AlonsoLej classification (72) is the most commonly applied stratification of choledochal cyst morphology (Fig. 2). The type I cyst describes a diffuse, isolated segmental, or fusiform dilatation of the extrahepatic bile duct. Type I lesions are the most frequently reported variant in most series and account for 40 to 85% of all choledochal cysts (7). Type IVA disease describes multiple intrahepatic and extrahepatic biliary dilatations, and it is the next most common pattern. The type II cyst, a true diverticulum
Page 648
Figure 2 Todani modification of the AlonsoLej classification of choledochal cysts.
of the common bile duct, and type IVB lesions, multiple extrahepatic cysts, are uncommon forms. Choledochoceles (type III) are ectasias of the distal bile duct within the duodenal wall, and they are separately reviewed below. Caroli's disease (type V) pertains to multiple dilatations of the intrahepatic ducts, and it is also considered below as a distinct entity. The frequency of each of these variants is fairly constant among adults and children in most series (7). The spectrum of biliary dilatation typically influences the clinical presentation. For example, fusiform dilatation is associated with abdominal pain (66). On the other hand, one anticipates a cystic lesion when a patient exhibits a palpable mass or jaundice (66). Presentations also differ with age. Jaundice is common among infants (73), whereas most children have an abdominal mass with jaundice (7). The majority of adults relate a history of pain, and many experience sporadic attacks of pancreatitis or cholangitis (7,66). Moreover, an abdominal mass is rarely palpated in an adult or adolescent. Symptoms can be so nonspecific among adults that as many as half will have undergone a previous cholecystectomy for biliary pain (7). The delay in the diagnosis of an adult poses a much greater risk of portal hypertension, cirrhosis, biliary lithiasis, hepatic pyogenic abscess, and malignancy (61). Another determinant of symptoms is the angle of the junction between the duct of Wirsung and the distal common bile duct. Three main patterns of angles of anomalous pancreaticobiliary ductal union are recognized: a rightangled junction without an accessory duct of
Page 649
Santorini, an acutely angled junction without an accessory duct, and a patent duct of Santorini with intricate combinations of junctions (74). Further categorization is based upon the shape of the common channel. The angle of pancreaticobiliary ductal junction possibly influences both the shape of the choledochal cyst and the clinical presentation. For example, a rightangled junction corresponds to cystic lesions and promotes the development of a palpable mass and intermittent jaundice (66). Conversely, an acutely angled union is associated with a fusiform ductal dilatation and pain. A dilated common channel or a patent duct of Santorini might impart relapsing pancreatitis and ultimately lead to chronic inflammation of the gland (74). D— Evaluation Valuable laboratory tests in the evaluation of choledochal cysts include liver chemistries and amylase levels, especially when patients present with jaundice or pancreatitis. However, hyperamylasemia should not necessarily be construed as indicative of active pancreatitis because amylaserich pancreatic fluid can be absorbed through the denuded cyst wall (75). Therefore, an elevated serum amylase level in the absence of clinical pancreatitis should not delay prompt surgical intervention. Ultrasonography is ordinarily the first imaging study obtained to assess obstructive jaundice, and it is similarly useful for rightupperquadrant pain and masses. A capable ultrasonographer defines the gallbladder, the pancreas, the shape and size of the cyst, and the nature of the intrahepatic and extrahepatic ducts. In fact, ultrasonography has discovered choledochal cysts in utero as early as 15 weeks of gestation (73). Computed tomography provides additional data regarding the liver parenchyma, intrahepatic bile ducts, the porta hepatis, the distal bile duct, the pancreas, changes of portal hypertension, and possible malignant degeneration of the cyst. These two imaging modalities are complementary and often exclude solidorgan cystic lesions, pancreatic pseudocysts, gastrointestinal duplications, and hydronephrosis (66). When the biliary origin of a choledochal cyst cannot be ascertained, cholangiography is useful. This is accomplished by endoscopic retrograde cholangiopancreatography (ERCP) or with a percutaneous, transhepatic approach. The former is particularly good at demonstrating distal ductal anatomy, whereas the antegrade method offers a finer depiction of the proximal biliary tract. Cholangiography is indicated if the entire biliary tract has not been visualized, especially because of the hazards posed by residual protein plugs in the common channel or by a stenotic common hepatic duct. Magnetic resonance cholangiopancreatography (MRCP) is a novel diagnostic study that could supplant invasive cholangiograms as its software is refined (Fig. 3). The MRCP might become especially valuable for patients in whom active pancreatitis precludes ERCP (61). Scintigraphy with 99mTcIDA occasionally proves that a cyst is contiguous with the biliary tract, although it may be limited by weak concentration of the radionuclide (70). Scintigraphy is also handy in the assessment of postoperative anastomotic patency and integrity. Clinical presentation, the age of the patient, and the adequacy of previous studies dictate the number and sequence of imaging tests. E— Treatment Vater reported a fusiform dilatation of the common bile duct over 275 years ago (76). Douglas proffered the first legitimate description of a choledochal cyst in 1852 and recommended aspirations by trochar (77). For much of the twentieth century, internal drainage of bile duct cysts was the standard practice, despite the first resection of a biliary cyst occurring in 1924 (78). However, therapeutic failures of internal drainage, including malignant degeneration, have generated widespread support for complete cyst excision. The safety of this more elaborate operation has been established for quite a while (79). When untreated, choledochal cysts develop devastating complications, such as severe cholangitis, profound pancreatitis, variceal hemorrhage from portal hypertension due to biliary
Page 650
Figure 3 Magnetic resonance cholangiopancreatogram (MRCP) of a choledochal cyst. Note that there is not an obvious anomalous pancreaticobiliary ductal junction in this case. (Courtesy of Joseph T. Ferrucci, MD, Boston University Department of Radiology.)
cirrhosis, and bile peritonitis following spontaneous cyst rupture (70). Moreover, patients with these cysts have a propensity to develop biliary tract malignancies. Therefore, the natural history of choledochal cysts is that they are ultimately fatal if not corrected. Choledochocystojejunostomy and choledochocystoduodenostomy were formerly employed as standard therapy for choledochal cysts. However, internal drainage has been associated with an inordinately high morbidity. Anastomotic strictures are common, and one series related a 73% reoperation rate for this complication (80). It is likely that strictures are a consequence of pancreatic fluid and gut flora refluxing into the cyst and resulting in recurrent cholangitis, activation of enzymes, and the formation of stones. These same factors may play a role in malignant degeneration, for which the biliary cyst seems to have an even greater propensity following internal drainage (81). The standard of care for a choledochal cyst is formal excision with a RouxenY hepaticojejunostomy reconstruction. Preoperative care includes the correction of sepsis and the reversal of any coagulopathy that might have resulted from biliary obstruction and from antibiotics. Some surgeons favor the usage of percutaneous transhepatic stents to identify scarred ducts or to manage cholangitis (7). Proper stent placement is critical to avoid occluding the aberrant pancreatic duct and precipitating pancreatitis. In cases of acute sepsis, cysts occasionally require percutaneous transhepatic drainage or even direct percutaneous drainage, just as Douglas advocated in the nineteenth century. Ruptured cysts with peritonitis are generally treated with a temporizing external drainage by an operatively placed tube (61). In some respects, cyst excision is tantamount to a resection of a cholangiocarcinoma. The gallbladder is mobilized from the liver and used as a handle to maneuver the bile duct. The distal common bile duct is divided at a level where it has a fairly normal caliber, although the surgeon must appreciate the entrance of the aberrant pancreatic duct. The distal duct stump
Page 651
is ligated to prevent the leakage of pancreatic fluid from the long common channel. The proximal portion of the transected duct is elevated and a plane is dissected between the cyst, the hepatic artery, and the portal vein. Decompression of the cyst often enhances the dissection. The mobilization proceeds proximally to the hilar bifurcation, where the hepatic duct is divided. The hepatic duct should not be dissected substantially proximal to the site of division so as to minimize the chances of an ischemic stricture. When intense inflammation precludes the separation of the cyst from the portal vein or hepatic artery, the back wall of the cyst is left in situ and dissection continues in the submucosal plane (82). A mucosatomucosa hepaticojejunostomy is fashioned with a RouxenY loop of jejunum to restore biliary drainage. In selected cases, the efferent limb can be sutured to the abdominal wall and marked with titanium clips to facilitate later percutaneous access to the biliary anastomosis. Intraoperative cyst endoscopy is performed with a tiny fiberoptic device, such as a pediatric cystoscope. This technique has been instrumental in the intraoperative assessment of the hepatic ducts and the distal common bile duct for stenoses, the determination of the appropriate level for transection of the hepatic duct, the identification of the pancreatic duct to minimize the length of the distal common bile duct remnant, and the evacuation of protein plugs and stones from the common channel and from the intrahepatic ducts (83). The management of choledochal cysts is essentially the same for children as it is for adults. Of course, longstanding cholangitis, obstructive jaundice, cirrhosis, pancreatitis, and the possibility of a malignancy make surgery more complex in an adult patient. Pediatric surgeons have devised various technical modifications, including jejunal interposition conduits with intussusception valves, to avert postoperative complications such as cholangitis and stricture formation. However, large pediatric series report the practice of the traditional RouxenY hepaticojejunostomy (83). The discovery of a choledochal cyst by prenatal ultrasonography poses another pediatric clinical challenge—namely, the timing of surgery. Some surgeons support close surveillance of the infant for either biochemical evidence of liver deterioration or ultrasonographic growth of the cyst (73). If the cyst remains stable in size and the infant does not exhibit jaundice, surgery is delayed until the infant develops further and can better tolerate surgery and anesthesia. Conversely, progression of the cyst, dilatation of the intrahepatic ducts, or the manifestation of jaundice mandate a prompt cyst resection. F— Outcomes Operative mortality following choledochal cyst excision is rare (80,81,83). Morbidity correlates with the age of the patient and the extent of preexisting cholangitis, porta hepatis inflammation, hepatic fibrosis, and pancreatitis. For example, in a series of 200 children who had undergone cyst excision, no stricture formation or biliary lithiasis occurred among those under 5 years of age (83). However, 18 children in the overall group developed complications, including cholangitis, intrahepatic bile duct stone formation, residual stones in the remnant of the common channel, pancreatitis, and bowel obstruction. Reports among adults and older children indicate an array of complications similar to those seen in young children, although abdominal sepsis and sequelae of portal hypertension manifest as well (80). Furthermore, the incidence of major morbidity is greater among adults than among children (83). When internal drainages of choledochal cysts were common practice, anastomotic strictures were frequent late complications. In one report of adult patients, 11 had undergone cystenterostomies and required 32 additional hospitalizations for complications related to strictures (80). Of these individuals, 8 had some type of anastomosis revision. In that same series, only one stricture developed among the 15 patients who had had their cysts excised. Notably, no strictures, cholangitis, or biliary lithiasis occurred in a group of 70 children on whom intraoperative cyst endoscopy was performed (83). Perhaps this maneuver will become the standard of care. Estimates of the incidence of biliary tract malignancies in conjunction with choledochal cysts range from 3 to 26% in the United States and from 8 to 17% in Japan (84). This translates
Page 652
to a 1000 to 2000fold greater likelihood of development than in the general population (81). The chance of longterm survival following resection of a malignant cyst is best when the cancer is occult. Otherwise, most patients succumb to a clinically evident malignancy, with a 5% survival at 2 years (81,84,85). Tumor growth is likely promoted by the repetitive insults of cholangitis, because this cancer is quite rare among children (61,81,84). Interestingly, in one collection of cases of cysts with associated biliary neoplasms, only onethird of the tumors developed in the cysts (Fig. 4). Of the remainder, cholangiocarcinomas constituted 43% of cases; gallbladder tumors, 12%; and pancreatic tumors, 3% (84). Therefore, a field defect for malignancy may be present in the extrahepatic biliary tracts of these patients. Even after having had a cyst excised, patients retain a risk of developing a tumor (81,84). G— Choledochocele A choledochocele (type III cyst) affects the intramural portion of the common bile duct, causing a protrusion into the duodenal lumen. These lesions account for less than 5% of choledochal cysts in major series (86,87), with no apparent sex predilection (88). A variety of congenital and acquired etiologies have been proposed, including duodenal duplication, diverticular prominence of the terminal bile duct, and dilatation due to sphincter of Oddi dysfunction or papillary stenosis (88). Choledochoceles are generally regarded as larger than 1 cm in diameter, although
Figure 4 A 25yearold Caribbean woman presented with obstructive jaundice and back pain. Some portions of her choledochal cyst demonstrated typical inflammatory changes, whereas other sections revealed malignant degeneration. Top Left: The extrahepatic choledochal cyst is present in the left hepatic duct near the hilar bifurcation. The solid arrow indicates the common bile duct. Top Right: One segment of the choledochal cyst is lined by hyperplastic biliary epithelium, but not by malignancy. Bottom Left: Another portion of the cyst wall contains scattered malignant epithelial cells. Bottom Right: Immunohistochemical staining (for cytokeratin) confirms the presence of malignant epithelial cells in the cyst wall. (Courtesy of Niall Mulligan, MD, Boston University Mallory Institute of Pathology, and of Robert Beazley, MD, Boston University Section of Surgical Oncology.)
Page 653
the smaller ectasia seen in the "dilated common channel syndrome" suggests a spectrum of this anatomic variant (89). Over 90% of patients with choledochoceles present with upper abdominal pain (88), whereas jaundice, acute pancreatitis, or biliary lithiasis each occur in about one third of patients (88). A similar distribution of presenting symptoms is seen in the dilated common channel syndrome (89). The widespread application of ERCP has led to the greater recognition of choledochal cysts, with a sensitivity of 97% (88). Diagnostic features include an effaced, smoothsurfaced papilla that significantly protrudes into the duodenal lumen, a compressible and distensible ectasia of the distal bile duct with normal overlying duodenal mucosa, and no impacted ampullary stone. Stones are occasionally identified within the contrastenhanced cyst. The dilated common channel syndrome was detected in 3.5% of a large number of patients undergoing ERCP (89). The endoscopic hallmarks of this syndrome are similar to those of choledochoceles but are more subtle with respect to the dimensions of the cyst. Separate orifices of the pancreatic duct and the common bile duct within the common channel also distinguish these conditions. The management of choledochoceles has evolved from surgery to endoscopy. Malignant degeneration of type III cysts is quite rare, having been described in only three cases (88), so excision of the cyst seems to be superfluous. Instead, endoscopic sphincterotomy suffices to evacuate the static and presumably harmful contents of the cyst, but longterm efficacy is not yet established. Transduodenal sphincteroplasty and resection are best reserved for complex cysts that are not amenable to endoscopic techniques, while choledochoceles that are discovered fortuitously may be observed (88). Endoscopic sphincterotomy effectively resolves the presenting symptoms in the vast majority of patients with the dilated common channel syndrome (89). H— Caroli's Disease Caroli's disease is a rare congenital saccular dilatation of the intrahepatic bile ducts, of which two variants have been defined (3). The pure version of Caroli's disease is nonhereditary and it is not associated with congenital hepatic fibrosis or renal malformations. In addition, the cystic lesions involve either a segmental distribution of the intrahepatic ducts or a diffuse pattern. Conversely, Caroli's syndrome affects all of the liver and includes congenital hepatic fibrosis. The syndrome can be an autosomal recessive or dominant trait and may be accompanied by autosomal recessive polycystic kidney disease, adult polycystic kidney disease, and renal tubular ectasia. Although earlier reports contended that the latter form was more common, more recent data suggest that the pure disease is as common as the syndrome (90). Caroli's disease is currently considered to be a distinct entity that does not necessarily fall within the realm of choledochal cysts (33). Furthermore, it seems to have a predilection for the male sex (60). Desmet has attributed Caroli's disease to a ductal plate malformation (3). In the pure form of the disease, the arrest in the remodeling of the ductal plate affects the larger intrahepatic bile ducts. This errant remodeling persists into the late phases of embryological development in Caroli's syndrome, affecting the peripheral ducts as well. The histology findings include acute and chronic cholangitis with portal tract edema and fibrosis (60). The cysts are lined by ulcerated and hyperplastic ductal epithelium and contain pus, bilestained concretions, and stones. The clinical manifestations of Caroli's disease are a function of the variant. In the pure disease, the intrahepatic obstruction of bile flow results in recurrent attacks of cholangitis with fevers, intermittent jaundice, and pain. Chronic sequelae include hepatolithiasis, hepatic pyogenic abscesses, sepsis, pancreatitis, cholangiocarcinoma, and secondary amyloidosis (90). Hepatomegaly is common and liver chemistries reflect a cholestatic pattern. On the other hand, Caroli's syndrome often presents with complications of portal hypertension, such as variceal bleeding, due to the associated congenital hepatic fibrosis. Enlargement of the liver and spleen
Page 654
is typical in this variant. Caroli's disease is likely present at birth, but it usually is not clinically manifested until late childhood or adulthood (90). The differential diagnosis of Caroli's disease includes multiple liver cysts, polycystic liver disease, primary sclerosing cholangitis, and intrahepatic ductal dilatation due to extrahepatic bile duct obstruction (91). The diagnosis is secured by demonstrating that the cystic ectasias are contiguous with the biliary tract. Ultrasonography and computed tomography scanning reveal the intrahepatic cystic ectasias of Caroli's disease. The radiographic "central dot sign" is due to dilated ducts surrounding portal vein radicles and suggests the diagnosis (33). In addition, findings of portal hypertension, cirrhosis, and renal disease are evident with these noninvasive studies. Some investigators dispute concerns about precipitating cholangitis with cholangiography (92). Both diagnostic ERCP and percutaneous transhepatic cholangiography can be carefully conducted, especially to exclude diagnoses such as sclerosing cholangitis or choledocholithiasis. Scintigraphy and magnetic resonance cholangiopancreatography are noninvasive methods of proving the ductal relationship of the ectasias (93). Antibiotics constitute the primary management of acute cholangitis. However, the longterm goals of therapy are the establishment of adequate biliary drainage, the relief of symptoms, and the eradication of the potentially malignant disease when possible (94). Biliaryenteric bypasses do not address the intrahepatic nature of the disease and have been largely unsuccessful (91). Liver resection is indicated for disease that is confined to one lobe. The incidence of hepatobiliary malignancies occurring in Caroli's disease is about 7 to 14%, but no tumors have yet been reported to have developed following resection of the affected segment (90). Orthotopic liver transplantation has been successfully performed and is recommended when other treatments have failed to manage the complications of repetitive cholangitis and of portal hypertension (95). An intriguing philosophy of care employs the endoscopic clearance of intrahepatic stones, followed by dissolution therapy (92). Endoscopic sphincterotomy is performed to permit cholangioscopy, intraductal electrohydraulic lithotripsy, stent placement, and stone retrieval. Extracorporeal shock wave lithotripsy is another therapeutic option. Once the stones are cleared from the biliary tract, ursodeoxycholic acid is used to alter the bile acid pool. This approach is predicated on the concept that impacted intrahepatic stones are responsible for the recurrent episodes of cholangitis. The regimen was safely accomplished in six patients and eliminated all stones in four of them (92). No cholangitis occurred among these four patients during a mean interval of 6 years. Prophylactic endoscopic surveillance might be indicated, depending upon the dynamics of the disease. In general, the longterm prognosis of Caroli's disease remains discouraging due to relapsing sepsis, chronic liver failure, and portal hypertension (90). Nevertheless, the prospects of more sophisticated endoscopic capabilities and of liver transplantation offer promise. References 1. Van Eyken P, Sciot R, Callea F, Van der Steen K, Moerman P, Desmet VJ. The development of the intrahepatic bile ducts in man: a keratinimmunohistochemical study. Hepatology 1988; 8:1586–1595. 2. Blankenberg TA, Lund JK, Reubner BH. Normal and abnormal development of human intrahepatic bile ducts: an immunohistochemical perspective. Perspect Pediatr Pathol 1991; 14:143–167. 3. Desmet VJ. Pathogenesis of ductal plate abnormalities. Mayo Clin Proc 1998; 73:80–89. 4. Tan CE, Moscoso GJ. The developing human biliary system at the porta hepatis level between 11 and 25 weeks of gestation: a way to understanding biliary atresia: Part 2. Pathol Int 1994; 44:600–610. 5. Terada T, Ohta T, Nakanuma Y. Expression of transforming growth factoralpha and its receptor during human liver development and maturation. Virchows Arch 1994; 424:669–675.
Page 655
6. Terada T, Nakanuma Y. Expression of tenascin, type IV collagen and laminin during human intrahepatic bile duct development and in intrahepatic cholangiocarcinoma. Histopathology 1994; 25:143–150. 7. Lipset PA, Segev DL, Colombani PM. Biliary atresia and biliary cysts. Baillieres Clin Gastroenterol 1997; 11:619–641. 8. Kimura K, Ohoto M, Ono T, Tsuchiya Y. Congenital cystic dilatation of the common bile duct: relationship to anomalous pancreaticobiliary ductal union. Am J Roentgenol 1977; 128:571–577. 9. Ohi R, Ibrahim M. Biliary atresia. Semin Pediatr Surg 1992; 1:115–124. 10. Whitington PF, Alonso EM, Piper JB. Pediatric liver transplantation. Semin Liver Dis 1994; 14:303–317. 11. Karrer FM, Lilly JR, Stewart BA, Hall RJ. Biliary atresia registry. 1976–1989. J Pediatr Surg 1990; 25:1076–1080. 12. Carmi R, Magee CA, Neill CA, Karrer FM. Extrahepatic biliary atresia and associated anomalies: etiologic heterogeneity suggested by distinctive patterns of associations. Am J Med Genet 1993; 45:683–693. 13. Tan CE, Moscoso GJ. The developing human biliary system at the porta hepatis level between 29 days and 8 weeks of gestation: a way to understanding biliary atresia: Part 1. Pathol Int 1994; 44:587–599. 14. Desmet VJ. Congenital diseases of intrahepatic bile ducts: variation on the theme ''ductal plate malformation." Hepatology 1992; 16:1069–1083. 15. Terracciano LM, Cathomas G, Vecchione R, Tornillo L, Gudat F, Bianchi L. Extrahepatic bile duct atresia associated with hyperplasia of the intrahepatic bile ducts ("early severe form"): high incidence in a south Italian population. Pathol Res Pract 1995; 191–192. 16. Morecki R, Glaser JH, Cho S, Balistreri WF, Horwitz MS. Biliary atresia and reovirus type 3 infection. N Engl J Med 1982; 307:481. 17. Phillips PA, Keast D, Papadimitiau JM, Walters MNI, Stanley NF. Chronic obstructive jaundice induced by reovirus type 3 in weanling mice. Pathology 1969; 1:193–203. 18. Steele MI, Marshall CM, Lloyd RE, Randolph VE. Reovirus 3 not detected by reverse transcriptase—mediated polymerase chain reaction analysis of preserved tissue from infants with cholestatic liver disease. Hepatology 1995; 21:697–702. 19. RiepenhoffTalty M, Shaekel K, Clark HF, Mueller W, Uhnoo I, Rossi T, Fisher J, Ogra PL. Group A rotaviruses produce extrahepatic biliary obstruction in orally inoculated newborn mice. Pediatr Res 1993; 33:394–399. 20. Bobo L, Ojeh C, Chiu D, Machado A, Colombani P, Schwarz K. Lack of evidence for rotavirus by polymerase chain reaction/enzyme immunoassay of hepatobiliary samples from children with biliary atresia. Pediatr Res 1997; 41:229–234. 21. Yokoyama T, Copeland NG, Jenkins NA, Montgomery CA, Elder FFB, Overbeek PA. Reversal of leftright asymmetry: a situs inversus mutation. Science 1993; 260:679–682. 22. Sonnenberg E, Meyer D, Weidner KM, Birchmeier C. Scatter factor/hepatocyte growth factor and its receptor, the cMet tyrosine kinase, can mediate a signal exchange between mesenchyme and epithelia during mouse development. J Cell Biol 1993; 123:223–235. 23. Stamatoglou SC, Enrich C, Manson MM, Hughes RC. Temporal changes in the expression and distribution of adhesion molecules during liver development and regeneration. J Cell Biol 1992; 116:1507–1515. 24. Silveira TR, Salzano FM, Donaldson PT, MieliVergani G, Howard ER, Mowat AP. Association between HLA and extrahepatic biliary atresia. J Pediatr Gastroenterol Nutr 1993; 16:114–117. 25. Seidman SL, Duquesnoy RJ, Zeevi A, Fung JJ, Starzl TE, Demetris AJ. Recognition of major histocompatibility complex antigens on cultured human biliary epithelial cells by alloreactive lymphocytes. Hepatology 1991; 13:239–246. 26. Dillon P, Belchis D, Tracy T, Cilley R, Hafey L, Krummel T. Increased expression of intracellular adhesion molecules in biliary atresia. Am J Pathol 1994; 145:263–267.
Page 656
27. Moore TC, Hyman PE. Extrahepatic biliary atresia in one human leukocyte antigen identical twin. Pediatrics 1985; 76:604–605. 28. Ho CW, Shioda K, Shirasaki K, Takahashi S, Tokimatsu S, Maedia K. The pathogenesis of biliary atresia: a morphological study of the hepatobiliary system and the hepatic artery. J Pediatr Gastroenterol Nutr 1993; 16:53–60. 29. Balistreri WF, Grand R, Hoofnagle JH, Suchy FJ, Ryckman FC, Perlmutter DH, Sokol RJ. Biliary atresia: current concepts and research directions: Summary of a symposium. Hepatology 1996; 23:1682–1692. 30. Kasai M. Treatment of biliary atresia with special reference to hepatic portoenterostomy and its modifications. Prog Pediatr Surg 1974; 6:5–52. 31. Mowat AP, Davidson LL, Dick MC. Earlier identification of biliary atresia and hepatobiliary disease: selective screening in the third week of life. Arch Dis Child 1995; 72:90–92. 32. Thaler M, Gellis S. Studies in neonatal hepatitis and biliary atresia. Am J Dis Child 1968; 116:280–284. 33. Nicotra JJ, Kramer SS, Bellah RD, Redd DCB. Congenital and acquired biliary disorders in children. Semin Roentgenol 1997; 32:215–227. 34. Abramson SJ, Salvador T, Teele RL. The infant with possible biliary atresia: evaluation by ultrasound. Pediatr Radiol 1982; 12:1–5. 35. Kirks DR, Coleman RE, Filston HC, Rosenberg ER, Merten DF. An imaging approach to persistent neonatal jaundice. Am J Roentgenol 1984; 142:461–465. 36. Balistreri WF. Neonatal cholestasis—medical progress. J Pediatr 1985; 106:171–184. 37. Lefkowitch JH. Biliary atresia. Mayo Clin Proc 1998; 73:90–95. 38. Middlesworth W, Altman RP. Biliary atresia. Curr Opin Pediatr 1997; 9:265–269. 39. Thomson J. On congenital obliteration of bile ducts. Edinb Med J 1891; 37:523–531. 40. Holmes JB. Congenital obliteration of the bile ducts: diagnosis and suggestions for treatment. Am J Dis Child 1916; 11:405–431. 41. Kasai M, Suzuki S. A new operation for noncorrectable biliary atresia—hepatic portoenterostomy. Shujutsu 1959; 13:733–739. 42. Karrer FM, Hall RJ, Stewart BA, Lilly JR. Congenital biliary tract disease. Surg Clin North Am 1990; 70:1403–1418. 43. Sawaguchi S, Nakajo T, Hori T, et al. Reconstruction of the biliary tract in biliary atresia using jejunal conduit. J Jpn Surg Soc 1968; 69:1317. 44. Tagge DU, Tagge EP, Drongowski RA, Oldham KT, Coran AG. A longterm experience with biliary atresia. Ann Surg 1991; 214:590–598. 45. Otte JB, de Ville de Goyet J, Reding R, Hausleithner V, Sokal E, Chardot C, Debande B. Sequential treatment of biliary atresia with Kasai portoenterostomy and liver transplantation: a review. Hepatology 1994; 20:41S–48S. 46. Millis JM, Brems JJ, Hiatt JR, Klein AS, Ashizawa T. Ramming KP, QuinonesBaldrich WF, Busuttil RW. Orthotopic liver transplantation for biliary atresia. Evolution of management. Arch Surg 1988; 123:1237–1239. 47. Broelsch CE, Emond JC, Whitington PF. Application of reducedsize liver transplants as split grafts, auxiliary orthotopic grafts, and living related segmental transplants. Ann Surg 1990; 212:368–377. 48. Wood RP, Langnas AN, Stratta RJ, Pillen TJ, Williams L, Lindsay S, Meiergerd D, Shaw BW. Optimal therapy for patients with biliary atresia: portoenterostomy ("Kasai" procedures) versus primary transplantation. J Pediatr Surg 1990; 25:153–162. 49. Howard ER. Extrahepatic biliary atresia—a review of current management. Br J Surg 1983; 70:193–197. 50. Kasai M, Mochizuki I, Ohkohchi N, Chiba T, Ohi R. Surgical limitations for biliary atresia: indication for liver transplantation. J Pediatr Surg 1989; 24:851–854. 51. Chandra RS, Altman RP. Ductal remnants in extrahepatic biliary atresia: a histopathologic study with clinical correlation. J Pediatr 1978; 93:196–200.
Page 657
52. Tan CE, Davenport M, Driver M, Howard ER. Does the morphology of the extrahepatic biliary remnants in biliary atresia influence survival? A review of 205 cases. J Pediatr Surg 1994; 29:1459–1464. 53. Azarow KS, Phillips MJ, Sandler AD, Hagerstrand I, Superina RA. Biliary atresia: should all patients undergo a portoenterostomy? J Pediatr Surg 1997; 32:168– 174. 54. McClement JW, Howard ER, Mowat AP. Results of surgical treatment of extrahepatic biliary atresia in the United Kingdom 1980–82. Br Med J 1985; 290:345– 347. 55. Endo M, Masuyama H, Watanabe K, Hagane K, Ikawa H, Yokoyama J, Kiajima M. Calculation of biliary atresia prognostic index using a multivariate linear model. J Pediatr Surg 1995; 30:1575–1579. 56. Hossain M, Murahashi O, Ando H, Kaneko K, Ito K. Proliferation of intrahepatic bileduct epithelium in biliary atresia: a useful predictor of clinical outcome. Pediatr Surg Int 1996; 11:126–129. 57. Sartorelli KH, Holland RM, Allshouse MJ, Karrer FM, Lilly JR. The intussusception antireflux valve is ineffective in preventing cholangitis in biliary atresia. J Pediatr Surg 1996; 31:403–406. 58. Howard ER, Davenport M. The treatment of biliary atresia in Europe 19691995. Tohoku J Exp Med 1997; 181:75–83. 59. Karrer FM, Price MR, Bensard DD, Sokol RJ, Narkewicz MR, Smith DJ, Lilly JR. Longterm results with the Kasai operation for biliary atresia. Arch Surg 1996; 131:493–496. 60. McEvoy CF, Suchy FJ. Biliary tract disease in children. Pediatr Clin North Am 1996; 43:75–98. 61. Miyano T, Yamataka A. Choledochal cysts. Curr Opin Pediatr 1997; 9:283–288. 62. Schmid C, Meyer HJ, Ringe B, Scheumann GF, Pichlmayr R. Cystic enlargement of extrahepatic bile ducts. Surgery 1993; 114:65–70. 63. Kizumi K, Kodama T. A case of cystic dilatation of the common bile duct and etiology of the disease. Tokyo Med J 1916; 30:1413–1423. 64. Babbitt DP. Congenital choledochal cysts: new etiological concept based on anomalous relationships of common bile duct and pancreatic bulb. Ann Radiol (Paris) 1969; 12:231–240. 65. Chijiiwa K, Kimura H, Tanaka M. Malignant potential of the gallbladder in patients with anomalous pancreaticobiliary ductal junction: the difference in risk between patients with and without choledochal cyst. Int Surg 1995; 80:61–64. 66. Kim OH, Chung HJ, Choi BG. Imaging of the choledochal cyst. Radiographics 1995; 15:69–88. 67. Nakamura T, Okada A, Higaki J, Tojo H, Okamoto M. Pancreaticobiliary maljunctionassociated pancreatitis: an experimental study on the activation of pancreatic phospholipase A2. World J Surg 1996; 20:543–550. 68. Oguchi Y, Okada A, Nakamura T, Okumura K, Miyata M, Nakao K, Kawashima Y. Histopathologic studies of congenital dilatation of the bile duct as related to an anomalous junction of the pancreaticobiliary ductal systems: clinical and experimental studies. Surgery 1988; 103:168–173. 69. Okada A, Nakamura T, Higaki J, Okumura K, Kamata S, Oguchi Y. Congenital dilatation of the bile duct in 100 instances and its relationship with anomalous junction. Surg Gynecol Obstet 1990; 171:291–298. 70. Tan KC, Howard ER. Choledochal cyst: a 14year surgical experience with 36 patients. Br J Surg 1988; 75:892–895. 71. Todani T, Watanabe Y, Narusue M, Tabuchi K, Okajima K. Congenital bile duct cysts: classification, operative procedures, and review of thirtyseven cases including cancer arising from choledochal cyst. Am J Surg 1977; 134:263–269. 72. AlonsoLej F, Rever WB Jr, Pessagno DJ. Congenital choledochal cysts, with a report of 2, and an analysis of 94 cases. Surg Gynecol Obstet 1959; 108:1–30. 73. LugoVicente HL. Prenatally diagnosed choledochal cysts: Observation or early surgery? J Pediatr Surg 1995; 30:1288–1290.
Page 658
74. Komi N, Takchara H, Kunitomo K, Miyoshi Y, Yagi T. Does the type of anomalous arrangement of pancreaticobiliary ducts influence the surgery and prognosis of choledochal cyst? J Pediatr Surg 1992; 27:728–731. 75. Stringel G, Filler RM. Fictitious pancreatitis in choledochal cyst. J Pediatr Surg 1982; 17:359–361. 76. Vater A. Dissertation in auguralis medica, poes diss. qua Scirrhis viscerum disser, e.s. ezlerus. Vol 70. Edinburgh: University Library 1723; 19. (quoted by Stain et al, Ref 80.) 77. Douglas AH. Case of dilatation of the common bile duct. Monthly J Med Sci 1852; 14:97–100. 78. McWhorter GL. Congenital cystic dilatation of the common bile duct. Ann Surg 1924; 8:604–626. 79. Kasai M, Asakura Y, Taira Y. Surgical treatment of choledochal cyst. Ann Surg 1970; 172:844–851. 80. Stain SC, Guthrie CR, Yellin AE, Donovan AJ. Choledochal cyst in the adult. Ann Surg 1995; 222:128–133. 81. Ishibashi T, Kasahara K, Yasuda Y, Nagai H, Makino S, Kanazawa K. Malignant change in the biliary tract after excision of choledochal cyst. Br J Surg 1997; 84:1687–1691. 82. Lilly JR. Total excision of choledochal cyst. Surg Gynecol Obstet 1978; 146:254–256. 83. Yamataka A, Ohshiro K, Okada Y, Hosoda Y, Fujiwara T, Kohno S, Sunagawa M, Futagawa S, Sakakibara N, Miyano T. Complications after cyst excision with hepaticoenterostomy for choledochal cysts and their surgical management in children and adults. J Pediatr Surg 1997; 32:1097–1102. 84. Fieber SS, Nance FC. Choledochal cyst and neoplasm: a comprehensive review of 106 cases and presentation of two original cases. Am Surg 1997; 63:982– 987. 85. Rossi RL, Silverman ML, Braasch JW, Munson JL, ReMine SG. Carcinomas arising in cystic conditions of the bile ducts: a clinical and pathologic study. Ann Surg 1987; 205:377–384. 86. Flanigan DP. Biliary cysts. Ann Surg 1975; 182:635–643. 87. Yamaguchi M. Congenital choledochal cyst: analysis of 1,433 patients in the Japanese literature. Am J Surg 1980; 140:653–657. 88. Masetti R, Antinori A, Coppola R, Coco C, Mattana C, Crucitti A, La Greca A, Fadda G, Magistrelli P, Picciocchi A. Choledochocele: changing trends in diagnosis and management. Surg Today 1996; 26:281–285. 89. Elton E, Hanson BL, Biber BP, Howell DA. Dilated common channel syndrome: endoscopic diagnosis, treatment, and relationship to choledochocele formation. Gastrointest Endosc 1998; 47:471–478. 90. Taylor ACF, Palmer KR. Caroli's disease. Eur J Gastroenterol Hepatol 1998; 10:105–108. 91. Pinto RB, Lima JP, da Silveira TR, Scholl JG, de Mello ED, Silva G. Caroli's disease: report of 10 cases in children and adolescents in southern Brazil. J Pediatr Surg 1998; 33:1531–1535. 92. CaroliBosc FX, Demarquay JF, Conio M, Peters EP, Buckley MJM, Paolini O, ArmengolMiro JR, Delmont JP, Dumas R. The role of therapeutic endoscopy associated with extracorporeal shockwave lithotripsy and bile acid treatment in the management of Caroli's disease. Endoscopy 1998; 30:559–563. 93. Asselah T, Ernst O, Sergent G, L'hermine C, Paris JC. Caroli's disease: A magnetic resonance cholangiopancreatography diagnosis. Am J Gastroenterol 1998; 93:109–110. 94. Dagli U, Atalay F, Sasmaz N, Bostanoglu S, Temucin G, Sahin B. Caroli's disease: 1977–1995 experiences. Eur J Gastroenterol Hepatol 1998; 10:109–112. 95. Schiano TD, Fiel MI, Miller CM, Bodenheimer HC, Min AD. Adult presentation of Caroli's syndrome treated with orthotopic liver transplantation. Am J Gastroenterol 1997; 92:1938–1940.
Page 659
31— Primary Sclerosing Cholangitis John M. Vierling and Thomas D. Amankonah CedarsSinai Medical Center and UCLA School of Medicine, Los Angeles, California I— Introduction Primary sclerosing cholangitis (PSC) is a chronic cholestatic liver disease of children and adults characterized by segmental fibrosing inflammation of the intra and/or extrahepatic bile ducts (1–6). Neither the etiology nor the pathogenetic mechanisms of PSC have been defined (7,8). The association of PSC with inflammatory bowel disease (IBD), HLA haplotypes, autoantibodies, and immunological abnormalities provides circumstantial support for the hypothesis that PSC is an autoimmune disease (7,9). Fibrosing inflammation of either the small ducts (microscopic disease) and/or medium to large intrahepatic or extrahepatic ducts (macroscopic disease) leads to progressive narrowing or obliteration of their lumens, biochemical and clinical evidence of cholestasis, and ultimately secondary biliary cirrhosis (10,11). Cirrhotic patients are at risk for hepatic failure and complications of portal hypertension (12–17). In addition, both precirrhotic and cirrhotic patients are at risk for development of cholangiocarcinoma (18). Thus, PSC should be regarded as a premalignant condition of the biliary tract. Despite increased knowledge about the natural history, clinical, radiological, and histopathological features of PSC, therapeutic options are limited. No medical therapy has been shown to improve the natural history of PSC. Thus, patients with advanced PSC require orthotopic liver transplantation (OLT) to survive (14). II— Disease Associations and Risk Factors A— Inflammatory Bowel Disease PSC is strongly associated with inflammatory bowel disease (IBD) (Table 1). The vast majority of PSC patients (87 to 98.7%) with IBD have ulcerative colitis (UC) (19), while 1.3 to 13% have Crohn's disease of the colon (ileocolitis or colitis) (4,20–23). PSC is not associated with Crohn's disease restricted to the small bowel (1,4). The prevalence of UC in PSC varies geographically, ranging from 21 to 23% in Japan (24,25), to 44% in Spain (26), to 50% in India (27), to 71% in the United States (28), and to 98% in Norway (29). To determine the true prevalence of IBD in PSC, all patients must undergo colonoscopy with biopsy. One such study in Norway showed that 98% of PSC patients had colitis (29), while a similar study in the United States identified colitis in 71% (28). IBD is most often diagnosed several years before PSC, but PSC may occur at any time in the course of IBD (19,30–32). IBD may be detected colonoscopically as a presymptomatic process after PSC is diagnosed, or it may occur years after the diagnosis of PSC. The onset of PSC may also occur after protocolectomy for
Page 660 Table 1 Diseases Associated with Sclerosing Cholangitis Primary sclerosing cholangitis
Secondary sclerosing cholangitis
Ulcerative colitis
Choledocholithiasis
Crohn's colitis or ileocolitis
Infections in immunocompromised patients:
Type 1 autoimmune hepatitis
(Cryptosporidium, Trichosporon, cytomegalovirus, Cryptococcus visceral protothecosis)
HTLV1—associated myelopathy
Ischemic injury to hepatic artery or arterioles
Trauma
Neoplasia
Toxic injury
Floxuridine (hepatic artery injection) Formalin injection of ecchinococcal cysts
Congenital abnormalities
Celiac sprue
Miscellaneous conditions Alopecia universalis
Angioblastic lymphadenopathy
Autoimmune hemolytic anemia
Biliary atresia
Bronchiectasis
Cavernous transformation of portal vein
Cyctic fibrosis
Dermatitis herpetiformis
Diabetes mellitus
Glomerulonephritis
Histiocytosis X
Hodgkin's disease
Hypereosinophilia
Hyperimmunoglobulin M immunodeficiency
Hypogammaglobulinemia
IgA deficiency
Immune thrombocytopenic purpura
Intraabdominal adenopathy
Mast cell cholangiopathy
Mediastinal fibrosis
Metastatic prostate carcinoma
Pancreatitis (acute or chronic)
Perforating folliculitis
Peyronie's disease
Polymyositis
Porphyria cutanea tarda
Pseudotumor of orbit
Pyoderma gangrenosum
Retroperitoneal fibrosis
Rheumatoid arthritis
Thyroiditis (Riedel's struma)
Sarcoidosis
Sjögren's syndrome
Systemic lupus erythematosus
System sclerosis
Thymoma
Vasculitis
Page 661
UC (4,20,23,31); conversely, IBD may also develop following liver transplantation for PSC (17,33). Despite a strong association, there is no correlation between the severity of UC and PSC (4,20,31). B— Autoimmune Hepatitis PSC patients may have clinical, laboratory, and histopathological features resembling type 1 autoimmune hepatitis (AIH) (20,23,34), and both conditions appear to coexist in some patients (34–37). Application of an international diagnostic scoring system for AIH in 114 patients with PSC showed that 2% could be classified as "definite" and 33% as "probable" AIH (37). All five patients (four male) in another report met "definite" criteria for AIH and responded to therapy with corticosteroid and azathioprine (35). This finding emphasizes the importance of endoscopic retrograde cholangiography (ERC) in differentiating PSC and AIH. Distinction may be particularly difficult in PSC patients with only smallduct pathology and a normal cholangiogram (11). It is also important to note that type I AIH also occurs in patients with IBD (1,38,39). C— Other Associated Diseases Multiple other diseases (1,4,40,41) have also been reported in association with sclerosing cholangitis (Table 1). Many of the associations appear to be coincidental rather than true associations in which the frequency of the disease within patients with sclerosing cholangitis should be greater than its frequency in the general population. It is conceptually important to distinguish between PSC and secondary sclerosing cholangitis. D— Risk Factors PSC occurs in all races (1,4) but may be less common in people of African than in those of European origin (42). In contrast, a recent prospective study of patients with UC showed that women of African or Caribbean genetic origin were significantly more likely to develop PSC over a short period of time (odds ratio 119, p = 0.0002) than patients of European or Asian descent (42). Disease presentation varies in different parts of the world, but consistent features include predilection for middleaged males and an association with IBD (13,26) ranging from 50% in India (27) to 71% in the United States (28) and 100% in Norway (29,43). Recent studies indicate that PSC occurs more frequently in nonsmokers, and patients with UC who smoke have a decreased risk of PSC (44,45). Paradoxically, in a recent casecontrol study, PSC patients who smoked had an increased risk of cholangiocarcinoma (46). E— Pregnancy Pregnancy does not adversely affect the progression of PSC, but pruritus can become so intense as to result in premature delivery (47). Maternal and neonatal outcomes were excellent. III— Prevalence The exact prevalence of PSC is unknown. Prior to 1980 only about 100 cases had been reported (48). With the advent of ERC, more cases were identified, especially among patients with IBD (49). As noted above, the geographic prevalence of UC in PSC varies from 24 to 98%, and the prevalence of PSC in patients with IBD and persistently abnormal liver tests ranges from
Page 662
2.4 to 7.5% (12). Based on estimates of the U.S. prevalence of UC of 40 to 220 cases per 100,000, the assumption that 2.4 to 7.5% have or will develop PSC and that 71% of U.S. patients with PSC have IBD, the estimated U.S. prevalence of PSC is 1 to 6 per cases per 100,000 population (1,2,4). The prevalence of small duct sclerosing cholangitis (11) is unknown, but a recent epidemiological study in Sweden reported smallduct PSC in 7% of patients (49). IV— Clinical Features and Laboratory Tests A— Age and Gender PSC afflicts all age groups, having been reported in infancy, childhood, and throughout adult life (1,4). The majority of patients are diagnosed at a mean age 32 to 42 years, and the maletofemale ratio is 2 to 2.3:1 (13,20,23,29,50). In one longitudinal study, the average age at diagnosis was 10 years later for women than for men (31). Although PSC can occur in infancy, most children are diagnosed in the second decade (4,51–54). B— Signs and Symptoms At the time of diagnosis, patients with PSC may be asymptomatic or symptomatic with respect to their hepatobiliary disease (Table 2) (4,13,26,28,50,55,56). Currently, 15 to 45% of patients are asymptomatic at the time of diagnosis (28,55), while 20 years ago 67% of patients presented with symptoms of fatigue, malaise, jaundice, pruritus, abdominal pain, nausea, vomiting, fever, anorexia, and weight loss; only 7 to 10% were asymptomatic (20,23,57). Asymptomatic patients are now more frequently recognized due to increasing knowledge of PSC and of serum liver tests and diagnostic ERC. Signs of decompensated cirrhosis, including ascites and variceal bleeding, remain rare presenting features (4,20,43,56,58). Despite progressive cholestasis, hyperpigmentation, xanthelasma, and xanthomata are less frequent than in primary biliary cirrhosis (PBC) (59). Presentation with ascending cholangitis is also rare except in patients who have had prior surgical manipulation of the bile ducts (20,23,60,61). Table 2 Asymptomatic and Symptomatic Presentations of Primary Sclerosing Cholangitis Presentation
Frequency
Asymptomatic Symptomatic Abdominal pain Ascending cholangitis
16–37% 5–28%
Ascites
2–10%
Jaundice
30–69%
Weight loss
10–34%
Variceal bleeding Hepatomegalya a
15–44%
2–14% 34–62%
Hepatomegaly present in either asymptomatic or symptomatic patients. Sources: Data from Refs. 4, 13, 26, 28, 50, 55, and 56.
Page 663
Patients may remain asymptomatic despite advancing disease, and up to 17% of asymptomatic patients have cirrhosis, as compared with 50% of symptomatic patients (13). In a longitudinal study recording daily symptoms in 84 PSC patients enrolled in a therapeutic trial of colchicine, symptoms (primarily pruritus and abdominal pain) were intermittent, usually lasting for a few days (62). Other patients had symptoms for months or years before spontaneous resolution. Only pruritus was significantly correlated with levels of serum alkaline phosphatase. C— Comparison of PSC Patients with and without Inflammatory Bowel Disease A variable proportion of patients with PSC never develops IBD. Two comparative studies have indicated that histopathological (63) and cholangiographic features (64) did not differ among patients with and without IBD. In contrast, one study of patients with mostly advanced PSC reported differences in gender ratios and cholangiographic findings between patients with and without IBD (65). Those with UC were more frequently male (maletofemale ratio 2.9:1) compared to those with Crohn's colitis (ratio 1:1) and those without IBD (ratio 0.72:1). Only 59% of those with IBD presented with abnormal laboratory tests as the first manifestation of hepatobiliary disease, while 72% of patients without IBD presented symptomatically with fatigue, pruritus, and jaundice. Strictures of both intra and extrahepatic ducts were more frequent in patients with IBD (86%) than in those without (46%). Conversely, strictures confined solely to the extrahepatic ducts were less frequent in patients with IBD (7%) than in those without (38%). Despite these intriguing findings, it is unwarranted to conclude that PSC in association with IBD is an entity distinct from PSC occurring without IBD. D— Comparison of Ulcerative Colitis in Patients with and without Primary Sclerosing Cholangitis The prevalence of PSC is significantly lower among UC patients with distal colitis (0.05%) than in those with more extensive involvement of the colon (5.5%) (66). Over 95% of PSC patients with UC have extensive colitis and only 5% have distal colitis; both findings differ significantly from those in UC patients without PSC (66). In a 20year casecontrolled study, patients with PSC had a more quiescent course of UC than patients with UC alone (67). PSC patients with subclinical IBD can be detected with colonoscopic biopsies, and some may have dysplasia (30). During followup, such patients may develop overt IBD 1 to 7 years later. Colonoscopy with biopsies should be performed to diagnose UC in patients with PSC and to evaluate for dysplasia. Flexible sigmoidoscopy may be insufficient, since one study found rectal sparing in 3% of patients with PSC and UC (39). E— Laboratory Tests Most patients with PSC have a cholestatic liver test profile at diagnosis, with elevated alkaline phosphatase levels 1.5 to 3 times the upper limit of normal (ULN). In a large series, the degree of elevation was significantly higher in PSC than in other hepatobiliary diseases complicating IBD (68). Increased alkaline phosphatase, however, is not a prerequisite for diagnosis of PSC (69,70), since up to 8.5 to 10% of patients in two series had normal alkaline phosphatase levels at diagnosis (13,66). In addition, alkaline phosphatase levels may fluctuate in and out of the normal range during the course of disease. Decreased biliary secretion of copper leads to hepatic copper retention and abnormally high urinary excretion of copper in 90% of patients (71). Other liver tests are elevated to variable degrees (13,22,28,31,58,66,72–77). Over 90% of PSC patients have mild to moderate elevations of aminotransferase levels. Total bilirubin
Page 664
levels are normal in 60% at diagnosis but gradually increase with time (4,12,55,58). Fluctuating bilirubin levels may occur with passage of sludge or calculi or as a result of ascending cholangitis (4). Decreased synthetic function with hypoalbuminemia and prolonged prothrombin time is rare at the time of diagnosis and indicates advanced disease. A disproportionate decrease in albumin without comparable elevation of prothrombin time should suggest possible malnutrition or proteinlosing enteropathy. Increased immunoglobulin concentrations also occur in PSC. Hypergammaglobulinemia was present in 30% of patients, while selective increases of 1.5 × ULN in IgG concentrations was found in 44% (37), and 20 to 45% had increased levels of IgM (20,77). Eosinophilia occurs rarely in PSC. However, a hypereosinophilic syndrome has been reported in some patients (78,79). F— Autoantibodies Antinuclear or anti—smooth muscle antibodies were present in 22% of PSC patients in one large series (37). Both autoantibodies tend to occur with lesser frequency and lower titers than in type 1 AIH (20,37,77,80). Antimitochondrial antibodies are rare in PSC, and the antigenic epitopes differ from those recognized in PBC (7,23,37). Antineutrophil cytoplasmic antibodies (ANCA) that bind in a perinuclear pattern (pANCA) were detected in 26 to 90% of patients with PSC with or without IBD (81–84). Recent studies suggest that pANCA are found in approximately 80% of PSC patients and that the titers may fluctuate (85). Interestingly, pANCA were also found in 25% of relatives of PSC patients, indicating a possible immunogenetic predisposition (86). However, pANCA is neither sensitive nor specific for PSC, being found in UC without PSC (81,86), PBC (82), and type 1 AIH (82,87). In one comprehensive study, the frequency of pANCA detected by immunofluorescence was 87% in PSC, 17% in UC without PSC, 13% in PBC, and 16% in AIH (82). In contrast, other series reported pANCA in 65 to 83% of patients with UC without PSC (81,86) and in 92% of patients with type I AIH (87). Thus, pANCA should not be used as a diagnostic serological test to differentiate among these conditions. V— Pathology A— Histopathology and Staging Diseases of the biliary tract can be subdivided anatomically into smallduct (interlobular and septal) and largeduct (medium and large intra and extrahepatic) types. The former is diagnosed by examination of biopsies, the latter by cholangiography (10,11,88,89). PSC is now recognized as both a small and largeduct disease. The smallduct histopathology (Fig. 1A), originally referred to by the obsolete term pericholangitis, culminates in destruction of the ducts (ductopenia, Fig. 1C), which determines the progression to secondary biliary cirrhosis (10). Fibrous obliterative cholangitis (Fig. 1B) results in annular stenoses and thickening of the walls of intra and/or extrahepatic bile ducts of medium to large caliber. These changes, often identifiable on cholangiograms, are diagnostic of largeduct PSC. Progressive biliary obstruction and worsening cholestasis results in secondary biliary cirrhosis (Fig. 1D). A histomorphometric study of surgical biopsy specimens in PSC showed that destruction of ducts was confined to small and mediumsized ducts, and the reduced ratio of the bile duct volume to artery volume was consistent with bile duct atrophy (90). Abnormalities of peribiliary glands were also noted in PSC, consisting of proliferation, lymphocytic inflammation, dilatation, fibrosis, or destruction (91). A recent study of PSC livers, removed at liver transplantation, showed that histopathology varied with respect to the size of the bile ducts (92). Ductopenia was observed in portal tracts containing small interlobular bile or septal bile ducts, but concentric, peribiliary fibrosis was
Page 665
Figure 1 Histopathology of primary sclerosing cholangitis. A. Small duct lesion with mild inflammation of portal tract and abnormal biliary epithelium. B. Portal inflammation with periductular, concentric fibrosis and atrophy of the biliary epithelium. C. Ductopenia with fibrous scar at site of bile duct (inferior to portal vein). D. Biliary cirrhosis. (Photomicrographs courtesy of Lidjida Petrovic, M.D.)
inconspicuous. In contrast, the diagnostic histopathological lesion of PSC—concentric, periductal fibrosis and chronic nonsuppurative fibrousobliterative cholangitis— was observed exclusively in ducts of medium caliber. Inflammation, epithelial ulceration, and cholangioectasia characterized histopathology of the large intra and extrahepatic ducts. Porta hepatis lymph nodes are often enlarged due to inflammatory hyperplasia. Since percutaneous needle biopsies rarely retrieve samples containing mediumsized bile ducts, a variety of nondiagnostic histopathological features are most often observed. In order of frequency, biopsies from patients with PSC exhibit: (a) bile duct proliferation; (b) periductal fibrosis; (c) periductal inflammation; and (d) ductopenia (63). Progressive ductopenia leads to secondary biliary cirrhosis. Ludwig et al. subdivided the histopathology of PSC into four stages to encompass the wide variability of pathology observed in needle biopsy specimens and to permit stratification of patients for clinical trials. Stage I is characterized by inflammation, expansion of connective tissue, and cholangitis confined to the portal tracts. Stage II is characterized by interface hepatitis or portal fibrosis extending into the periportal parenchyma. Stage III is characterized by bridging necrosis or the presence of fibrous septa. Stage IV is biliary cirrhosis with fibrosis and regenerating nodules. B— Hepatic Atrophy Gross lobar atrophy has been observed in PSC and was associated with severe stenoses at the hilum (93).
Page 666
C— Perihepatic Lymphadenopathy Reactive lymphadenopathy is commonly found on imaging studies and characteristically is distributed in the porta hepatis, pancreaticoduodenal area, and the gastrohepatic ligament (94). The size of the lymph nodes is usually <1.5 cm, and they should not be misinterpreted as evidence of malignancy. Massive intraabdominal lymphadenopathy is rare (95). D— Gallbladder Histopathological changes of chronic cholecystitis occur in up 20%, although cholelithiasis is infrequent (96,97). Lymphoplasmacytic acalculous cholecystitis has been considered a distinctive form of chronic cholecysfitis in PSC (98). The volume of the gallbladder is increased in PSC (99,100), and the thickened, fibrosed wall is chronically inflamed (101). Occasionally, choledocholithiasis is present, presumably as a result of bile stasis at sites of stenosis. E— Cholangiocarcinoma The reported prevalence of cholangiocarcinoma in patients with PSC ranges from 7.1 to 13.8%, but the prevalence at autopsy was 27.3 to 41.7% in the same series (12,18,20,49,76). It occurs most frequently in cirrhotic patients and involves intra or extrahepatic ducts of medium to large caliber. However, cholangiocarcinoma, can also occur in precirrhotic patients. Tumors most often infiltrate the lamina propria of the ducts, causing sclerosis and narrowing that is difficult to distinguish from PSC (18). Occasionally cholangiocarcinoma will form a nodular mass (14,102). Immunohistochemical studies (103,104)indicate that tumors express carbohydrate antigen (CA) 199 (71%), carcinoembryonic antigen (CEA) (91%), and mutant p53 (78.5%). F— Pancreas Fibrosing inflammation of the pancreatic duct rarely results in stenosis (105), but acute or chronic pancreatitis may occur in up to 15% of patients with PSC (40,106– 112). Involvement of the pancreatic duct suggests that the pathogenetic mechanisms of PSC can target all derivatives of the embryonic foregut. VI— Diagnosis A— Diagnostic Criteria PSC was originally diagnosed only at laparotomy based on extrahepatic bile duct thickening and intraoperative cholangiography demonstrating irregular strictures. The original diagnostic criteria of Warren (113) required (a) absence of previous biliary surgery; (b) absence of cholelithiasis; (c) diffuse involvement of the extrahepatic biliary tree; and (d) exclusion of cholangiocarcinoma. The advent of ERC in the 1970s and recognition that PSC predisposes to biliary tract calculi (97,99,109) and cholangiocarcinoma (18) led to modification of these diagnostic criteria. Current diagnostic criteria (Table 3) are based on clinical, laboratory, cholangiographic, histopathological, and serological findings. Secondary causes of sclerosing cholangitis (Table 3) must be excluded. These secondary causes include previous biliary surgery, trauma, biliary calculi, ischemic injury of hepatic artery or arterioles, ischemic injury caused by floxuridine treatment, formalin treatment of ecchinococcal cysts communicating with the biliary tract, congenital abnormalities of the biliary tract, and infectious cholangiopathies in immunodeficiency syndromes.
Page 667 Table 3 Diagnostic Criteria for the Diagnosis of Primary Sclerosing Cholangitis Parameter
Inclusion criteria
Exclusion criteria
Clinical
Asymptomatic/symptomatic History of IBD Hepatomegaly
Immunodeficiency syndrome with infection Trauma Ischemia Floxuridine chemotherapy Formalin injection for ecchinococcal cysts
Laboratory tests
Normal or alkaline phosphastase Aminotransferases Normal or bilirubin
Markers of active viral hepatitis B or C
Histopathology
Portal inflammation Periductular inflammation/fibrosis Ductopenia Obliterative fibrous cholangitis Biliary cirrhosis
Viral hepatitis NSDC
Serology
ANA pANCA
AMA
Cholangiography
Normal (small duct histopathology) Sclerosis of extrahepatic biliary tract with or without sclerosis of intrahepatic ducts
Normal with histopathology incompatible with PSC Choledocholithiasis Congenital abnormalities
Key: IBD, inflammatory bowel disease; ANA, antinuclear antibodies; pANCA, perinuclear staining of antineutrophil cytoplasmic antibodies; AMA, antimitochondrial antibodies; NSDC, nonsuppurative destructive cholangitis. Source: From Ref. 1.
B— Cholangiography In the majority of patients, the diagnosis of PSC is made on the basis of typical cholangiographic findings using ERC or percutaneous transhepatic cholangiography (2,64). Diffuse, multifocal strictures separated by normalcaliber or ectatic segments result in a beaded appearance (Fig. 2). Strictures are typically short and annular, but longer confluent strictures, especially of the common bile duct, may be seen with advanced disease. Some large ectasias may have a diverticular appearance (64). Usually both intra and extrahepatic ducts are concurrently involved, although strictures can be confined to the intrahepatic ducts in up to 20% (58,114). Strictures of the extrahepatic biliary tract without intrahepatic involvement occur in approximately 10% of patients (20). In cirrhotics the proximal intrahepatic ducts may appear tortuous or stretched and attenuated (64). The gallbladder and cystic duct may be involved in up to 15% (2); abnormalities of the pancreatic duct are rarely observed (105). Recently, magnetic resonance cholangiography has been used to detect strictures in distal intra and extrahepatic ducts (115). C— SmallDuct Disease with Normal Cholangiography It is important to note that early stages of PSC may manifest only as smallduct (microscopic) disease without cholangiographic evidence of largeduct (macroscopic) disease (11). In such cases, the clinical diagnosis of smallduct PSC should be based on (a) typical liver biopsy
Page 668
Figure 2 Endoscopic retrograde cholangiogram in primary sclerosing cholangitis. Note the multiple, annular strictures and ectasias of the extrahepatic and intrahepatic bile ducts. (Cholangiogram courtesy of Carey Strom, M.D.)
findings; (b) cholestatic liver tests similar to those found in largeduct PSC; and (c) the presence of IBD (Fig. 3). D— Differentiation of Benign and Malignant Strictures PSC must be differentiated from secondary sclerosing cholangitis resulting from prior biliary surgery, choledocholithiasis, or cholangiocarcinoma. Distinguishing between benign strictures of PSC and those of cholangiocarcinoma, which develops in approximately 15% of patients with PSC (18), is not always possible with cholangiography. Cholangiographic features favoring a diagnosis of cholangiocarcinoma include (64,102,116,117) (a) excessive duct dilatation; (b) polypoid masses; (c) rapid progression of duct dilatation or stricturing observed on serial cholangiograms; and (d) formation of a short, dominant stricture of the extrahepatic ducts. VII— Natural History The natural history of PSC is difficult to define due to the variability of clinical disease associated with smallduct lesions and the location and severity of strictures in the intra and/or extrahepatic biliary tract (4,13,28). Thus, some patients with PSC have exacerbations and remissions, while others have a rapid progression and some have nonprogressive disease for decades. The typical clinical course, however, is characterized by complications of progressive
Page 669
Figure 3 KaplanMeier estimates of survival for asymptomatic and symptomatic patients with primary sclerosing cholangitis. Note the highly significant difference in survival (p < 0.001). (From Ref. 13.)
cholestasis, cirrhosis, portal venous hypertension, hepatic failure, or cholangiocarcinoma (13,28,7521,83,112bb). Current information, including evidence that PSC can occur without elevation of alkaline phosphatase (69,70), suggests that PSC progresses through four sequential phases: (a) preclinical, noncholestatic phase characterized by absence of symptoms and normal liver tests; (b) an asymptomatic, cholestatic phase with lack of symptoms but abnormal liver tests; (c) a symptomatic, cholestatic phase with abnormal liver tests with a cholestatic profile; and (d) a cirrhotic phase with risk or presence of complications of portal hypertension. In addition, it is important to note that histopathological and cholangiographic progression can occur without new symptoms or signs (20). Thus, up to 17% of asymptomatic patients were found to have cirrhosis as compared with 50% of symptomatic patients (13). The median survival from diagnosis to death or transplantation is approximately 12 years, but the range extends to 21 years (13,28,29,43,49,50). The wide range probably reflects uncertainty regarding the true duration of disease, especially among asymptomatic patients, and earlier performance of transplantation. Actuarial survival is significantly greater for asymptomatic patients than for symptomatic patients (Fig. 3); at 10 years, survival is 80% for asymptomatic patients and only 50% for symptomatic patients (13). In a cohort of asymptomatic patients without cirrhosis, 76% developed symptoms or signs of liver disease during a mean followup of 6 years and 31% of the asymptomatic patients died or received transplants (74). By 7 years, approximately 45% developed symptoms (Fig. 4). Initial cholangiographic findings may also be prognostic: severe intrahepatic strictures portend early jaundice, while short, severe extrahepatic strictures were associated with pruritus, abdominal pain, and fever (118). The natural history is adversely affected by the development of cholangiocarcinoma (18). Several prognostic models have been developed to calculate the survival free of death or transplantation (see below). VIII— Complications Patients with PSC are at risk for multiple complications (Table 4). They occur with variable frequency and clinical impact.
Page 670
Figure 4 KaplanMeier estimate of the time to onset of symptoms in asymptomatic patients with primary sclerosing cholangitis. Time of diagnosis is used as the starting point. (From Ref. 74.)
A— Cholangiocarcinoma Cholangiocarcinoma, the most dreaded and lifethreatening complication of PSC, has a reported prevalence of 7.1 to 13.8% in four large series (12,18,20,49,76). However, the prevalence at autopsy in these same series was substantially greater, varying from 27.3 to 41.7%. Similarly, the prevalence of unsuspected or undetected cholangiocarcinomas discovered at laparotomy or in explanted livers from PSC patients undergoing OLT was also high, averaging 17.8% (range 2.9 to 36.4%) in 10 reported series (13,16,50,60,119–122,123,124). In one longitudinal study, Table 4 Complications of Primary Sclerosing Cholangitis Complication
Comment Tends to occur with advanced disease
Dominant strictures
May occur at any stage of disease
Cholelithiasis
Cholesterol and pigmented stones
Acalculous cholecystitis
Infrequent
Hepatic osteodystrophy
Primarily osteoporosis
Steatorrhea
Associated with advanced cholestasis
Fatsoluble vitamin deficiency
Associated with advanced cholestasis
Pancreatitis
Rare
Peristomal varices after colectomy with ileosromy
Infrequent
Pruritus
Frequent from early to later stages
Ascending bacterial cholangitis
Infrequent prior to advanced strictures
Portal venous hypertension
Progressive in cirrhotic patients
Cholangiocarcinoma
Page 671
the cumulative actuarial incidence of cholangiocarcinoma. (Fig. 5) was 30% between 8 and 15 years of disease (125). These discrepancies between the estimated clinical prevalence and that observed longitudinally or at transplant or autopsy indicate that clinically occult cholangiocarcinomas are common in advanced PSC and that diagnostic tests and imaging fail to detect most tumors. The median survival of patients with cholangiocarcinoma in a large series was 5 months, and 63% had evidence of metastatic disease, primarily to regional lymph nodes (76). Virtually all tumors diagnosed prior to transplant recur lethally following orthotopic liver transplantation (122). In one large series of 305 patients with PSC, 26% died of cholangiocarcinomas that were diagnosed an average of 32.5 months after the diagnosis of PSC (13). A recent study showed that cholangiocarcinoma developed more frequently in PSC patients with colonic dysplasia or carcinoma, suggesting that colonoscopic screening with biopsies could be used to detect patients at high risk (30). Detection of cholangiocarcinoma is very difficult, since the tumor generally infiltrates along the lamina propria of the bile ducts and causes progressive stricturing that is often indistinguishable from the progression of PSC (18). Cholangiocarcinomas rarely form mass lesions detectable by imaging (14,102). The diagnostic sensitivities of ultrasonography (7%), computed tomography (29%), and ERC (58.3%) were disappointingly low (103). Comparative studies are needed to determine the diagnostic sensitivity of magnetic resonance imaging and cholangiography (126). Recently, positron emission tomography was found to detect ''hot spots" in patients with PSC and cholangiocarcinoma. (127). Clinical features—including the frequency of fever, jaundice, weight loss, abdominal pain, pruritus, hepatosplenomegaly, and ascites— did not differ significantly between PSC patients with and without cholangiocarcinoma (20,76,124). No significant differences in laboratory tests were detected in patients with PSC alone or PSC with cholangiocarcinoma (76,120,121,124). Serial ERC (102,128) may be useful in detecting cholangiocarcinoma, but suspicious findings (see above) may be absent or unreliable (103,128,129). Cytological evaluation of aspirated bile or mucosal brushings may aid in diagnosis. Cytology exhibited high specificities and positive predictive values in the diagnosis of cholangiocarcinomas in patients without PSC (130–132). Unfortunately, low sensitivities (11 to 30% for bile and 50 to 56% for brushings) and negative predictive values did not permit exclusion of cholangiocarcinoma on the basis of a negative cytology. In one study that included patients
Figure 5 Cumulative actuarial incidence of cholangiocarcinoma from time of onset of primary sclerosing cholangitis. (From Ref. 282.)
Page 672
with PSC (34%), diagnostic accuracy was increased when repeated samples were tested (133). Cytology of multiple samples of bile, brushings, and washings from occluded biliary stents had a combined sensitivity of only 75% (134), indicating that routine use of cytology in PSC strictures will rarely detect cholangiocarcinoma. Recently, the diagnostic value of carbohydrate antigen (CA) 199 and carcinoembryonic antigen (CEA) has been investigated (18). CA199 testing uses a monoclonal antibody with specificity for sialosylfucosyllactotetraose, a carbohydrate determinant of the Lewis sialylated blood group antigen. Biliary epithelial cells in PSC aberrantly express ABO blood group antigens, including Lewis antigen (135). One series reported elevations of serum CA199 levels in patients with cholangiocarcinoma, but overlapping elevations were also noted in patients with cirrhosis or bacterial cholangitis (136). Immunohistochemical analysis showed that 71% of cholangiocarcinomas stain positively for CA 199 and 91% for CEA (103), indicating that they are markers of the tumor rather than the strictured biliary tract. Whereas the circulating levels of either tumor marker alone were neither highly sensitive nor specific, an index combining them was derived (103) using the formula CA 199 + (CEA × 40). This index was used to evaluate cholangiocarcinoma in three PSC groups: (a) 15 patients with histologically confirmed tumor; (b) 22 transplanted patients without tumor in the explant; and (c) 37 patients with stable disease not requiring transplant and no clinical evidence of tumor (103). Using an arbitrary index value of >400 as a cutoff, both specificity and positive predictive value were 100%. However, the sensitivity was only 66%, due to values of <400 in the tumor group. Notably, none of the transplanted patients without tumor had values >400. However, the index was positive in only about half (54%) of transplanted patients whose occult tumors could be detected only in the explanted livers. More reliable and accurate tests are needed to detect early cholangiocarcinoma. B— Dominant Strictures Dominant strictures are defined as focal, highgrade narrowings of distal intra or extrahepatic bile ducts that mechanically intensify cholestasis. They are frequently manifest by progressive jaundice, pruritus, ascending cholangitis, and fat malabsorption. Dominant strictures occur in 15 to 20% of PSC patients during the course of disease (64) and must be differentiated from cholangiocarcinomas (18,128). Distinction between dominant strictures and cholangiocarcinoma may be impossible with currently available methods (see above). C— Cholelithiasis and Cholecystitis Cholelithiasis occurs in approximately 20% of patients with PSC (96,97), and approximately 33% of patients undergo cholecystectomy. Bacteria are present in the bile in up to 80% of PSC patients with cholelithiasis (137). Acalculous cholecystitis also occurs, and lymphoplasmacytic acalculous cholecystitis has been considered a distinctive form of chronic cholecystitis in PSC (98). Calculi of the biliary tract should not be considered an exclusionary criterion for the diagnosis of PSC (see above) and must be considered as a possible explanation for episodes of worsening cholestasis. Patients with or without calculi do not differ with respect to age, frequency of inflammatory bowel disease (IBD), extent of cholangiographic disease, immunogenetics, prevalence of cholangiocarcinoma, or need for transplantation (97). D— Hepatic Osteodystrophy Osteodystrophy, primarily osteoporosis, is a frequent complication of PSC (138–140). In one series, 50% of patients had vertebral bone mineral densities below the fracture threshold (140). In a larger series of patients in a randomized trial of ursodeoxycholic acid (UDCA) therapy in PSC, patients had significantly lower bone mineral density than control values adjusted for
Page 673
age, gender, and ethnic group (138). However, the rate of bone loss did not significantly differ between PSC patients (with or without UDCA therapy) and controls. In contrast to the earlier study (140), only 8.6% of patients had a bone mineral density below the fracture threshold. These patients were characterized by older age, longer duration of IBD, and more advanced PSC. Only age was significantly inversely related to baseline bone mineral density. Thus, osteoporosis should be suspected in older PSC patients with advanced disease and long duration of IBD. Bone mineral densitometry of the lumbar spine is the diagnostic test of choice. E— Steatorrhea and FatSoluble Vitamin Deficiency Progressive chronic cholestasis results in decreased concentrations of intestinal bile acids required for micellar absorption of fat and fatsoluble vitamins A, D, E, and K (141). Steatorrhea may be clinically inapparent. Initial lipid and fatsoluble vitamin levels were measured in 56 patients with PSC enrolled in a clinical therapeutic trial and 87 patients with advanced disease being evaluated for OLT (142). Among patients entering the trial, elevations were noted for cholesterol (41%) and high densitylipoprotein cholesterol (20%), but only 2% had elevated triglycerides. Cholesterol levels directly correlated with total bilirubin levels and histological stage of disease. Vitamin deficiencies were noted in 40% for vitamin A, 14% for vitamin D, and 2% for vitamin E. Among transplant candidates, cholesterol was elevated in only 29%, and 17% had increased triglycerides. Interestingly, cholesterol levels were inversely correlated with total bilirubin in this group. Vitamin deficiencies were more prevalent in this group: 82% for vitamin A, 57% for vitamin D, and 43% for vitamin E. Thus, hypercholesterolemia and deficiencies of fatsoluble vitamins are common, especially in advanced PSC, and vitamin supplementation should be provided. F— Pancreatitis Acute or chronic pancreatitis caused by sclerosis of pancreatic ducts is a rare complication of PSC (106,107) but should be considered in patients with chronic abdominal pain or evidence of pancreatic pathology on imaging. G— Peristomal Varices Following colectomy with ileostomy for UC, PSC patients with portal hypertension may develop bleeding peristomal varices (143–145). H— Pruritus Pruritus due to cholestasis can occur in any PSC patient with elevated alkaline phosphatase levels (146). Rapid onset or abrupt worsening may accompany episodes of intensified cholestasis associated with ascending cholangitis, cholelithiasis, dominant stricture, or cholangiocarcinoma. I— Ascending Cholangitis Ascending cholangitis is infrequent in the absence of prior manipulation of the bile ducts or development of highgrade obstruction (137). Recurrent episodes may be difficult to treat because of the emergence of antimicrobialresistant bacteria.
Page 674
J— Portal Venous Hypertension Biliary cirrhosis is associated with portal venous hypertension and its complications (56). These include edema, ascites, varices (with or without bleeding), hepatic encephalopathy, hypersplenism, hypoalbuminemia, coagulopathy, muscular wasting, and spontaneous bacterial peritonitis. IX— Colorectal Carcinoma with Concomitant Ulcerative Colitis Classically, the two major risk factors for development of colorectal carcinoma in UC patients were extensive duration of disease and pancolitis. More recently, the presence of PSC was also shown to be a risk factor (147–154). In one large study (30), the cumulative risk of colorectal carcinoma in patients with both PSC and UC was 9, 31, and 50% after 10, 20, and 25 years of disease, respectively. In contrast, the cumulative risk was significantly less at the same time points in patients with UC alone: 2, 5, 10%, respectively. These findings were substantiated in a casecontrol study showing a cumulative incidence of colorectal carcinoma in UC patients with PSC of 11% after 10 years and 31% after 20 years compared with 3% at 10 years and 8% at 20 years in patients with UC alone (155). In another large study of 178 patients with PSC (152), no increased risk of colorectal carcinoma was found in comparison to patients with UC (some of whom also had PSC.). However, among the subgroup of patients with concomitant PSC and UC, the cumulative incidence of colorectal carcinoma or dysplasia was 20% at 20 years and 67% at 30 years after the diagnosis of UC (152). These results indicate that PSC patients with UC have an increased risk of developing colorectal carcinoma. Indeed, these patients remain at risk even after undergoing successful OLT (156,157). X— Prognostic Models The prognosis of asymptomatic and symptomatic patients differs (13). The 10year actuarial survival for asymptomatic patients was 93%, while that for symptomatic patients was (47%). Models based on multivariate analyses (158) have been developed to predict the prognosis of patients with PSC (Table 5). Each of these models except one contained a histological parameter that necessitated a current liver biopsy for accurate prediction of survival. A recent comparison of the original Mayo Risk Score and the ChildTurcottePugh (CTP) score indicated that the ageadjusted CTP score was as accurate in predicting survival before transplantation as the Mayo model (156). The CTP score is designed to assess cirrhotic patients with portal venous hypertension and is calculated by assigning 1 to 3 points (total minimum 5, maximum 15) based on established criteria for (a) serum bilirubin; (b) serum albumin; (c) prothrombin time; (d) presence or absence of ascites; and (e) the grade of hepatic encephalopathy. It is advantageous because of its simplicity and accessibility of needed information. The Mayo Clinic Risk Score for PSC was recently revised for the third time (Table 5) to exclude need for a liver biopsy without compromising the accuracy of the survival estimate (158). The second refinement was made by collaboration with four international centers (Multicenter Study Group) to create a database on 426 PSC patients that included clinical, laboratory, serological, and histological detail collected during a 20year followup (43). The newest model was created and validated by randomly dividing the database in half and then developing the model using one half and validating it using the other half. In the absence of evidence that any therapy effectively alters the natural history of PSC, all patients were considered untreated. In the new model the independent variables included age, total serum bilirubin, serum albumin, AST, and the presence or absence of a history of variceal bleeding. The new Mayo model closely correlated with the earlier model and accurately predicted survival free of transplant across the disease spectrum in PSC.
Page 675 Table 5 Independent Clinical Variables in Prognostic Models for Primary Sclerosing Cholangitis Mayo Clinic Original model
King'sCambridge
Norwegian
Multicenter
Age
Age
Age
Age
Histological stage
Bilirubin
Histological stage
Histological stage
Hepatomegaly
Bilirubin
Bilirubin
Splenomegaly
Splenomegaly
Hemoglobin
Alkaline phosphatase
IBD
a
Age
Bilirubin
Albumin
AST
Variceal bleeding
New model
Key: IBD, inflammatory bowel disease; AST, aspartate aminotransferase. a New Mayo model equation: R = 0.03 (age in years) + 0.54 × loge (bilirubin in mg/dL) + 0.54 × loge (AST in U/L) + 1.24 (for history of variceal bleeding) 0.84 × (albumin in g/dL).
It is important to note that all PSC survival models and the CTP score have significant drawbacks. For example, none has been prospectively tested, none account for development of cholangiocarcinoma, and none are sensitive for risk factors for variceal bleeding or fluctuating levels of bilirubin. Since serum bilirubin is the most heavily weighted independent variable, calculations of risk scores during reversible episodes of increased biliary obstruction or ascending cholangitis may significantly underestimate future survival. Comparison of the actual survival of patients transplanted for PSC with the survival estimates of the new Mayo Risk Score model showed significantly increased survival following transplantation (158). Ongoing studies are evaluating the impact of PSC severity on the posttransplant cost and outcome. These analyses should also provide important information on the impact of unsuspected cholangiocarcinoma in the explanted recipient liver, which has been associated with decreased survival (159). XI— Differential Diagnosis The differential diagnosis of PSC includes common hepatobiliary diseases associated with cholestasis as well as diseases with cholangiographic features resembling PSC (Table 1). In addition, the strong association between PSC and IBD, particularly UC, often requires differential diagnosis in UC patients with abnormal liver tests. A— Cholestatic Diseases Classic diseases that require differentiation from PSC include PBC, autoimmune cholangitis, extrahepatic biliary obstruction without strictures, secondary sclerosing cholangitis (due to choledocholithiasis, surgical trauma, arterial ischemia, intraarterial chemotherapy or formaldehyde injection of ecchinococcal cysts), idiopathic cholangiohepatitis, druginduced cholestasis, and chronic viral, druginduced hepatitis.
Page 676
B— Autoimmune Hepatitis As discussed above, PSC patients may have features resembling type 1 AIH (20,23,34), and both may coexist in some patients (34–37). Using an international diagnostic scoring system for AIH, 2% of PSC patients could be classified as "definite" and 33% as "probable" for having AIH (37). This finding emphasizes the importance of ERC in differentiating PSC and AIH. Distinction may be particularly difficult in PSC patients with only smallduct pathology and a normal cholangiogram (11). Type 1 AIH also occurs in patients with IBD, and both PSC and AIH should be considered in patients with IBD and abnormal liver tests (1,38,39). C— Other Hepatobiliary Diseases in Inflammatory Bowel Disease Although PSC is the most frequent hepatobiliary disease associated with UC (see above), a variety of other hepatobiliary conditions may also afflict patients with UC (1). These include steatosis, nonspecific portal inflammation indistinguishable from smallduct PSC lesions (see above), AIH, amyloidosis, hepatic granulomatosis, and cholangiocarcinoma in the absence of PSC. D— Immunodeficiency and Infection Immunodeficiency due to HIV infection has been associated with cholangiographic findings reminiscent of PSC (41). In addition, patients infected with Cryptosporidium (160), Trichosporon (161), cytomegalovirus (162,163), and Cryptococcus (164) may also exhibit cholangiographic evidence of biliary sclerosis. XII— Etiology and Pathogenesis A— Autoimmune and Nonautoimmune Factors Although the etiology of PSC remains undefined, associations with HLA haplotypes, multiple autoantibodies and IBD suggested a possible immunopathogenesis (7,8). However, the hypothesis that PSC is an autoimmune disease (9,165) remains controversial due to differences between PSC and classic examples of autoimmunity. These differences include (a) a male predominance and absence of female predilection, (b) absence of diseasespecific autoantibodies, and (c) poor response to immunosuppressive medications. The strong association of PSC and IBD, especially UC, has been viewed as circumstantial support for an "autoimmune" pathogenesis in PSC. Indeed, both IBD and PSC have been considered examples of associated autoimmune diseases (7). Numerous immunological abnormalities have been detected in PSC; however, their role in pathogenesis remains speculative, and the possibility exists that many are secondary epiphenomena (7). The numerous abnormalities provide circumstantial support for the notion that altered immune function facilitates pathogenetic mechanisms in PSC. Reported immunologic abnormalities include (a) decreased proportions of circulating T cells and CD8 T cells (166,167), (b) increased proportions of circulating B cells (168), (c) decreased Tsuppressor cell function in vitro (169), (d) increased autologous mixed lymphocyte reactivity in vitro (170), (e) immune complexlike substances in blood and bile (171), (f) complement activation with increased levels of C3b and C4d (172), (g) C3d (terminal complex) deposition on walls of hepatic arteries but not bile ducts (173), and (h) decreased in vivo clearance of artificial immune complexes by hepatic macrophages (174). Nonimmune factors are also associated with sclerosis of the biliary tract and can be indistinguishable from PSC. Indeed, infection, toxicity, ischemia and neoplasia (Table 1), pro
Page 677
voke pathogenetic mechanisms resulting in PSClike lesions (41). The resulting biliary obstruction instigates mechanisms responsible for secondary sclerosing cholangitis and biliary cirrhosis (see below). In addition, the minority of PSC patients who do not have and never develop IBD are indistinguishable from PSC patients with IBD. Either distinctly different etiologies and mechanisms of pathogenesis may exist for PSC in the presence or absence of IBD or, alternatively, a shared pathogenetic mechanism may exist that is independent of IBD. In this context, it is important to note that colonoscopy and biopsies may reveal subclinical IBD in patients with established PSC (30). Recent evidence strongly suggests that bacterial constituents may play a primary role in etiopathogenesis of UC and possibly PSC. For example, in "knockout" mice prone to develop a deficiency of Tcell receptor alpha, the immune response to intestinal bacterial antigens is instrumental in both the pathogenesis of colitis and development of pANCA (175). Similarly, interleukin 10 (IL10) (/) knockout mice with spontaneous colitis also developed pANCA, and absorption of their sera with bacterial antigens significantly decreased specific perinuclear staining (176). Specific perinuclear staining of pANCA from patients with PSC was also diminished or abolished by absorption with bacterial antigens (176). These observations strongly indicate that immune responses to bacterial antigens are involved in the generation of colitis and pANCA. Immune reactions to bacterial cellwall elements in genetically susceptible rats result in PSC lesions (177,178). Thus, immune responses of immunogenetically susceptible individuals to bacterial antigens, possibly acting as molecular mimics for autoantigens, is an attractive alternative to the hypothesis that autoantigens induce PSC. B— Immunogenetic Susceptibility The immunogenetics of PSC patients has been extensively investigated, and a recent reanalysis has cast doubt on the validity of the original HLA associations. PSC was statistically associated with increased frequency of class I, II, and III HLA alleles (7,179); subsequent molecular genotyping indicated that susceptibility to PSC is determined by two haplotypes: (a) A1B8DRB3*0101 (DR52a)DRB1*0301 (DR3)DQA1*0501DQB1*0201 and (b)DRB3*0101DRB1*1301DQA1*0103 DQB1*0603. In contrast, resistance to PSC is determined by DRB1*04 (DR4) alleles. A model based on the shared amino acid motifs of DRB5*0101 [found in all DRB1*1501 (DR2) haplotypes] and DRB3*0101 alleles suggested that susceptibility was conferred by a leucine at position 38 of the DRb polypeptide (180). HLA DPB (centromeric) and Cw (telomeric) loci mark the boundaries of PSC susceptibility within the MHC. The recent reassessment of the published genotyping data (179), corrected for the number of a priori alleles tested, showed that PSC is not significantly associated with DRB1*0301 (DR3) but is negatively associated with DRB1*04 (DR4). Moreover, an association with DRB1*1301 was not confirmed in PSC. Increased susceptibility formerly attributed to the haplotype DRB3*0101 DRB1*1301DQA1*0103DQB1*0603 was reassigned to the DQA1*0103DQB1*0603 alleles. The negative association of DR4 was also extended to a haplotype including DQB1*0302. The strongest positive associations in recent analyses were with HLA B8 (which is in linkage disequilibrium with DRB3*0101 and DRB1*0301) and the class III TNF a promoter polymorphism at position —308 and the TNFAZ allele. Further work is necessary to determine if these class III associations are due to linkage disequilibrium with HLAB8. With respect to immunopathogenesis, it is noteworthy that HLA DR3 is associated with high TNF a production (181–183), possibly due to linkage disequilibrium with class III TNF alleles (184). It is also notable that UC is not associated with these HLA haplotypes. However, the HLA associations with PSC account for only 50 to 55% of cases based on any one allele or 75 to 80% of cases based on leucine at position 38 of DRb . Therefore, 20 to 50% of PSC cases cannot be accounted for by an immunogenetic model. Studies of other susceptibility loci in nonHLA immunogenes may ultimately identify additional associations.
Page 678
C— Autoantibodies and Anticolon Antibodies PSC is associated with pANCA in 26 to 90% of patients (81–84). However, pANCA is not specific for PSC, occurring in UC without PSC (185,186), type 1 AIH (87), and PBC (184). Whether diseasespecific ANCA antigens exist remains speculative. One study supporting possible antigenic differences showed that DNase digestion of neutrophils resulted in loss of binding by UCassociated pANCA and converted the pANCA pattern to cytoplasmic binding in the majority of patients with PSC and type 1 AIH (185). Another study used phage cloning techniques to show that the pANCA antigen(s) in UC are unique (187). Evidence that immune responses to bacterial antigens are essential for T cell—receptor alpha "knockout" mice to develop colitis and pANCA (175) suggests that bacteria may also induce pANCA in PSC. The possibility that pANCA represents a crossreactivity to enteric bacterial antigens was supported by a recent study showing that absorption of human pANCApositive sera greatly reduced or abolished the perinuclear reaction of the pANCA (176). Crossreactivity of pANCA with bacterial antigens is also consistent with the finding that 81% of PSC patients have antibodies against enterobacterial proteins (188). Other putative autoantibodies have been implicated in PSC pathogenesis. Anticolon antibodies were found in 62% of patients with PSC and UC, compared to only 17% of patients with UC alone (189). These anticolon antibodies did not crossreact with hepatobiliary tissue. In contrast, another anticolon antibody directed against a 40kDa molecule on colonic epithelial cells also crossreacted with undefined antigens in skin and biliary epithelia (190,191). This finding suggests, but does not prove, that immune responses may be directed against shared antigens in colonic and biliary epithelia. D— Biliary Epithelial and Endothelial Cells as Immunological Targets Biliary epithelial and endothelial cells (BECs) have been considered targets of the immune response in vanishing bile duct syndromes (7) but may not be the primary targets in PSC. In PSC, BECs express class I HLA, aberrant class II HLA, and ICAM1, the phenotype of either antigenpresenting cells (APCs) or susceptible target cells for CD4 or CD8 BECspecific cytotoxic T lymphocytes (CTL). Recent studies indicated that human BECs were not immunogenic for T cells and lacked B7 expression (192,193). In contrast, expression of B7 costimulatory molecules was observed in primary biliary cirrhosis (PBC) (194–196), suggesting that BECs might activate CD4 T cells under optimal conditions in PSC. However, expression of CD58 (LFA3), costimulatory B7 and Fas (CD95) in PSC was modest and intermittent in comparison to PBC (196). Moreover, BECs did not concomitantly express both class II HLA and ICAM1 in PSC, indicating that only a minority of cells might present antigen or serve as targets of cytolysis (197). Thus, the BECs in PSC lack the essential epitopes of target cells for cellmediated immunity. Both BECs and colonocytes exhibit inappropriate expression of ABO blood group antigens in PSC (135). ABO antigens that crossreacted with anti—bile duct antibodies were identified in the cytoplasm of BECs from small and large bile ducts. Involvement of ABO antigens in PSC pathogenesis is unlikely, since there is no evidence of antibodymediated bile duct injury or Tcell reactions against ABO blood group antigens in PSC. Inappropriate ABO expression could be an epiphenomenon induced by cytokines or be an early changes of dysplasia (135). The histopathology of PSC also argues against BECs as direct targets of T cells. Portal tract infiltrates are composed primarily of neutrophils and CD4 T cells with increased proportions of monocytes/macrophages and decreased proportions of natural killer (NK) cells by comparison with peripheral blood (167,196,198). CD4 Th1 cells predominated in the portal tracts of patients with PSC and PBCs (196). Peribiliary CD8 CTLs are infrequently observed in precirrhotic patients when BEC destruction should be prominent but are paradoxically increased in cirrhotic livers (167). Moreover, nonsuppurative destructive cholangitis (NSDC) lesions are rare in PSC (10). Even though BECs may have heterogeneous gene expression
Page 679
(199,200), the distribution of lesions varying from small ducts alone to intrahepatic and/or extrahepatic ducts is not explained by the hypothesis that BECs are target cells. Several studies suggest that portal tract T cells may be sensitized to epithelial antigens expressed by the gut. Three Tcell lines propagated from the common bile ducts of two PSC patients (201) had oligoclonal Tcell receptors (TCRs). Oligoclonality persisted in a second biopsy specimen taken from one patient more than a year later. The patients' Tcell lines proliferated in response to cultured enterocytes and were cytotoxic for enterocyte cell lines. T cells isolated from PSC livers also preferentially expressed Vb 3 TCRs (202), suggesting activation by a limited number of antigens. No correlation was noted between Vb 3 TCR expression and the histopathological stage of disease. Together, these findings indicate that T cells in hepatobiliary infiltrates may have been activated by limited antigen(s) expressed by enterocytes. Although bile duct injury could be secondary to Tcell cross reactions between enterocytes and BECs (derived from the embryonic foregut), the fibrous obliterative histopathology does not suggest that BECs are the targets of effector T cells in PSC (7,92,203). The hypothesis that endothelial cells of the peribiliary capillary plexi might be target cells in PSC (7) is intriguing, since interruption of arterial blood supply could cause ischemic strictures and atrophy of the biliary epithelia. However, recent immunohistochemical evidence indicates that the peribiliary capillary plexi remain intact but are pushed away from the bile duct by the deposition of concentric layers of peribiliary fibrosis (204). Although this argues against endothelial cells as targets of the immune reaction, separation of the capillary plexuses from the bile ducts could cause local ischemia and atrophy of the biliary epithelia. E— Role of Chemokines and Cytokines Chemokines and cytokines most likely play important roles in portal inflammation, periductular fibrosis, atrophy of the biliary epithelia, and portal fibrogenesis. BECs produce a variety of chemokines and cytokines (Table 6), and the expression of several is upregulated by inflammation. The mRNA of chemokine IL8 was present in cultures of normal human BECs and a cholangiocarcinoma cell line (205). Moreover, induction of IL8 secretion in vitro by recombinant IL1b , TNF a , or endotoxin (lipopolysaccharide) indicated that either cytokines or endotoxin could promote peribiliary localization of inflammation in vivo. Evidence that bile from PSC patients contained significantly increased levels of IL8 compared to bile from patients with PBC, alcoholic cirrhosis, or fulminant hepatitis (205) suggested possible disease specificity. BECs isolated from the PSC livers also contained IL8 mRNA, and intracellular IL8 protein was immunohistochemically detected in both isolated BECs and intact ducts. Concurrent secretion of the chemokine monocyte chemotactic protein1 (MCP1) expressed in BECs could attract and activate inflammatory cells with diverse pathogenetic effects in the peribiliary environment (Table 6). Biliary secretion of chemokines could also explain the composition of portal tract inflammatory infiltrates containing neutrophils, CD4 T cells, increased monocyte/macrophages, and decreased NK cells (167). F— Role of CD66a or Biliary Glycoprotein in Inflammation or Fibrogenesis Recent evidence that human bile contains CD66a, also known as biliary glycoprotein (BGP), suggests that it could be involved in the pathogenesis of inflammation and/or fibrosis in PSC. CD66a is a member of the CEA family and the human homologue of rat cell CAM (206). CD66 is expressed by neutrophils and monocytes; it promotes adhesion to Eselectin and serves as a receptor for lectins galectin 3 and bacterial type 1 fimbriae. Specific crosslinking of neutrophil CD66a with CD66b or nonspecific crossreacting antigen 90 (CD66c) increases b 2 integrin—mediated adhesion and oxidant activity. CD66a is also expressed on canalicular membranes of hepatocytes, ductular epithelia, enterocytes, and some nonhepatic endothelial cells. In the human breast, CD66a is expressed by epithelial cells and myoepithelial cells (207).
Page 680 Table 6 Effect of Expression of Chemokines and Cytokines by Biliary Epithelial Cells Molecules
Target cells a
Effects
IL8
Neutrophils
Neutrophils
Monocyte chemotactic protein1
Monocytes
Chemoattraction
Tcell blasts
Intracellular Ca2+
CD4 T cells
Exocytosis of granules
Fibroblasts
Upregulate integrin expression
Eosinophils
Basophils
Activation
Chemotaxis
Upregulate integrin expression
Stimulate T cells, fibroblasts, eosinophils, basophils
BEC
HLA class I expression
Aberrant HLA class II expression
ICAM1 expression
BEC?, fibroblasts, T cells
Fibrogenesis?
Suppression of Tcell responses
BEC
Cytotoxicity?
Macrophages
Activation
Hepatocytes
Acutephase reaction
CD4 T cells
IL4 favoring CD4 Th2 stimulation
Chemokines
Cytokines IFNg
TGFb
a
TNFa
IL6a
Respiratory burst Monocytes
Respiratory burst
Key: BEC, biliary epithelial cells; IL, interleukin; IFN, interferon; TGF, transforming growth factor; TNF, tumor necrosis factor. a Not constitutively expressed but upregulated by inflammation.
Epithelial expression decreased in breast carcinoma but remained intense in myoepithelial cells within infiltrative scars and sclerosing adenosis. CD66a expression has not been characterized in PSC livers, but its relationship to neutrophil function and myoepithelial cells in fibrotic lesions suggests a possible role in inflammation and periductal fibrosis. G— Pathophysiological Consequences of Biliary Obstruction and Cholestasis Bile duct obstruction results in a sequence of events (208) that is deleterious to the liver. The sequence includes (a) increase in LPS levels; (b) LPS activation of Kupffer cells and portal tract macrophages; (c) secretion of IL1b , TNF a , IL6, TGF a /b and leukotrienes; (d) TNF a —induced loosening of BEC tight junctions and regurgitation of bile; (e) LPS paralysis of BEC production; (f) interruption of cholehepatic cycling between BECs and peribiliary capillaries; (g) BEC chemokine and cytokine secretion with recruitment and activation of neutrophils, monocytes, and T cells; (h) enzymatic degradation of extracellular matrix; and (i) ductular proliferation. In experimental biliary obstruction, proliferating cholangiocytes secrete plateletderived growth factor (PDGF) (209,210), which contributes to the cytokine activation of adjacent hepatic stellate cells. This fibroductular reaction can progress to secondary biliary fibrosis and cirrhosis unless obstruction is relieved. It is notable that PDGF is also expressed to a lesser extent by periductular mesenchymal cells that could activate peribiliary fibroblasts.
Page 681
The pathogenetic mechanisms of biliary obstruction are pertinent to the pathogenesis of PSC. Both the microscopic and macroscopic lesions of PSC result in progressive obstruction and would be expected to trigger similar events. Thus, the key issue is the mechanism(s) that initiate the periductal inflammation and obliterative fibrosing cholangitis in PSC that secondarily stimulate the above cascade. H— Animal Models of PSC Studies of animal models strongly suggest that immune responses of immunogenetically susceptible hosts to bacterial products of the gut may be primary pathogenetic events in PSC. Two experimental models of colitis support this hypothesis: muramyl peptide—induced colitis in rabbits (211) and Escherichia coli chemotactic peptide—induced colitis in rats (212). Both produced histopathological lesions reminiscent of PSC. More compelling evidence comes from a rat model of small bowel bacterial overgrowth that results in portal inflammation, cholangiolar proliferation, and cholangiographic strictures of both intra and extrahepatic bile ducts (213,214). Only a genetically susceptible strain developed pathology, which is in accord with the concept of host immunogenetic susceptibility in PSC. The extent of hepatobiliary injury was correlated significantly with production of TNF by Kupffer cells. Pathology could not be prevented by UDCA, polymixin B (which binds intraluminal endotoxin), or immunosuppression with prednisone, methotrexate, or cyclosporin A. In contrast, both TNF production and hepatobiliary inflammation were prevented by treatment with (a) mutanolysin, an enzyme that selectively cleaves bacterial wall peptidoglycanpolysaccharide; (b) palmitate, which blocks phagocytosis by hepatic macrophages; or (c) pentoxyfylline, which inhibits TNF secretion by hepatic macrophages. The model indicates that hepatic macrophage phagocytosis of bacterial cellwall components delivered in portal venous blood is a critical event in the initiation of inflammation and sclerosis of the biliary tract. This model is also in concert with evidence that immunogenetic predisposition to PSC may be associated with the class III HLA TNF A2 allele (179). A recent study also demonstrated sclerosing cholangitis in a congenic model of murine chronic graftversushost disease (CGVHD) that normally produces only NSDC lesions (215). CGVHD in humans also results in NSDC, but sclerosing cholangitis following bone marrow transplantation has been reported (216). It is intriguing to speculate that variation in the bacterial flora of the gut of the host might lead to different pathology during CGVHD. I— Postulated Mechanism of PSC Pathogenesis The critical initial event in the pathogenesis of PSC may be the reaction of an immunogenetically susceptible host to bacterial cellwall products in the setting of increased concentrations of endotoxin, resulting in production of TNF by hepatic macrophages (Fig. 6). Whether an altered enteric barrier and/or flora is a prerequisite remains unknown. However, potential exposure to bacterial components would be increased in patients with IBD but might also occur result from episodes of gastroenteritis or bacterial overgrowth. Increased concentrations of TNF and endotoxin in the peribiliary lymphatic space of Mall could stimulate BEC secretion of chemokines and cytokines, promote regurgitation of bile, inhibit secretory functions of BECs, and interfere with the cholehepatic circulation. Peribiliary chemokines and cytokines would attract and activate neutrophils, monocyte/macrophages, T cells, and fibroblasts. This, coupled with secretion of PDGF, would promote enzymatic digestion of extracellular matrix and collagen synthesis by activated myofibroblasts, resulting in peribiliary fibrosis. Expanding layers of concentric fibrosis would push the peribiliary capillary plexi away from the bile duct, resulting in progressive focal ischemia and atrophy of BECs. Obliterative fibrous cholangitis would cause progressive biliary obstruction, triggering pathophysiological mechanisms observed in experimental biliary obstruction. Ongoing inflammation and fibrosis would cause ductopenia of small bile ducts, which, along with progressive stricturing of medium and large intra and extrahepatic ducts, would accelerate biliary obstruction. Proliferation of ductular
Page 682
Figure 6 Postulated pathogenesis of primary sclerosing cholangitis. (From Ref. 8.)
cholangiocytes would initiate a fibroductular reaction, with interface hepatitis and bridging fibrous septa, culminating in secondary biliary cirrhosis. The combination of ischemia, aberrant gene expression, and atrophy of BECs; exposure to cytokines; and interruption of cholehepatic cycling may contribute to the development of cholangiocarcinoma. XIII— Treatment Theoretical goals of therapy in PSC include (a) prevention or retardation of ductopenia and the resultant biliary cirrhosis; (b) prevention or retardation of complications, especially cholangiocarcinoma; and (c) symptomatic management of chronic cholestasis. None of these goals have been fully achieved. A— Therapy for Primary Disease Multiple medical and nontransplant surgical therapies have been investigated in PSC. No curative therapy has been identified for PSC except OLT. Recent studies suggest that PSC progression may be retarded, but these results have not been independently validated (85,217). Assessment of therapeutic efficacy is quite difficult in PSC due to the great variation in the severity of disease among patients; the unpredictable natural history, with fluctuating exacerbations and remissions; and the complications of dominant strictures and cholangiocarcinoma. 1— Biliary Tract Surgery to Alleviate Obstruction Nontransplant surgical attempts to relieve biliary obstruction did not retard progression of PSC to secondary biliary cirrhosis (20,23,50,60,65,218). Moreover, surgical intervention increases
Page 683
the risk of recurrent ascending cholangitis (60,65). Both internal and external biliary drainage has been performed in uncontrolled reports but these have not been shown to be beneficial. Currently, most surgeons advocate avoidance of hepatobiliary surgery in patients with PSC (219). 2— Proctocolectomy Proctocolectomy has had no effect on disease progression in patients with PSC and UC (61,220–222). Moreover, the onset of PSC may occur after proctocolectomy (19). Perioperative mortality was increased in PSC patients with cirrhosis who required colectomy for treatment of UC (221). As noted above, development of peristomal varices at the ileostomy site may occur in PSC patients with portal hypertension (143–145). Thus, ileoanal, ileorectal, or ileal pouchanal anastomoses are recommended because they are not associated with formation of varices and bleeding (223). However, the incidence of chronic pouchitis (60%) is increased in PSC patients compared to UC patients without PSC (15%) (224). The risk of pouchitis was unrelated to disease severity, but it was less frequent in small than in largeduct PSC (224). Interestingly, pouchitis has persisted in some patients following OLT, despite highdose immunosuppression (225). This observation suggests that antibiotics, rather than immunosuppression, represent the treatment of choice. 3— Antibiotics Uncontrolled trials of antibiotic therapy have been performed in PSC, based on the concept that PSC was caused or exacerbated by portal bacteremia or bacterial toxins (226–228). Antibiotics were ineffective for the primary disease and should be reserved for treatment of ascending cholangitis. 4— Oral Nicotine PSC occurs more frequently in nonsmokers, and patients with UC who smoke have a decreased risk of PSC (44,45). Based on these findings, a pilot study of oral nicotine was performed in eight patients with PSC (229). Three patients were intolerant of drug and were discontinued: two experienced reactivation of UC within a month, and one had dizziness and palpitations after 4 months. Of the five patients completing 1 year of therapy, three had to temporarily decrease the dose due to adverse effects. No significant changes in liver tests were noted, despite evidence of compliance with elevated plasma nicotine levels. 5— Immunosuppressive, Antifibrotic, and AntiInflammatory Therapy A variety of immunosuppressive, antifibrotic, and antiinflammatory therapeutic agents have been studied in PSC (2,230). As yet, none has improved the natural history of disease. Corticosteroids and Azathioprine Anecdotal reports of the efficacy of corticosteroids (231) never led to controlled clinical trials. Evidence that PSC both developed and progressed in UC patients treated with corticosteroids, azathioprine, or 6mercaptopurine curtailed enthusiasm for controlled studies. Moreover, it was feared that corticosteroids would accelerate osteopenia and promote spontaneous fractures. Experience with azathioprine therapy has been limited, and such treatment was found to be ineffective (232). Penicillamine
D
As a result of chronic cholestasis, hepatic copper concentrations progressively increase in PSC. A randomized, doubleblind, placebocontrolled trial of D penicillamine in PSC produced the expected cupriuresis but did not improve survival or clinical, laboratory, or histopathological features (233). Significant toxicity occurred in 21% of patients receiving Dpenicillamine. Colchicine A randomized, doubleblind, placebocontrolled trial demonstrated no beneficial effect on survival or clinical, laboratory, or histopathological features (62). The efficacy of colchicine and prednisone was tested and compared with the course of untreated, historic
Page 684
controls (234). Improved laboratory tests at 6 and 12 months were not sustained after 24 months, and serial liver biopsies showed no improvement. Methotrexate Based on the marked improvement of two men with PSC treated with methotrexate (235), an openlabel trial was begun in 10 patients, showing beneficial clinical, biochemical, and histological effects after 12 months (236). However, the results of a larger, randomized, doubleblind, placebocontrolled trial in 24 patients showed no significant benefit with respect to histopathological progression, cholangiographic findings, or clinical outcome despite a reduction of alkaline phosphatase in the treatment group (237). No significant toxicity was observed in these trials, but severe Pneumocystis carinii infection has been reported in a patient with PSC treated with methotrexate (238). In the absence of evidence that methotrexate improves the natural history of PSC and in view of potential toxicities, this drug should not be used empirically. Cyclosporine Cyclosporine (average dose 3.1 mg/kg/day) was evaluated in a doubleblind, placebocontrolled trial in 34 precirrhotic patients with PSC (239). Of these, 30 had concomitant UC and 3 had had colectomy. After 2 years, cyclosporine treatment significantly reduced alkaline phosphatase but did not improve symptoms of fatigue and pruritus or prevent histopathological progression or development of cirrhosis. However, cyclosporine significantly improved UC in those patients with concomitant disease. Tacrolimus In a 12month, openlabel trial of lowdose tacrolimus in 10 patients with PSC, bilirubin decreased by 75%, while alkaline phosphatase and aminotransferase levels fell by 70 and 83%, respectively (240). Trough levels of tacrolimus were maintained between 0.6 and 1.0 ng/mL, and no significant toxicities were noted. No controlled trial of tacrolimus therapy has been performed. Ursodeoxycholic Acid The efficacy of UDCA therapy in PSC remains controversial due to differences in the primary endpoints of reported studies. Openlabel, randomized, controlled trials have verified significant improvement of laboratory tests in PSC patients treated with UDCA (217). However, the impact on clinical and histological progression and development of complications requiring OLT is less clear. In three placebocontrolled trials, UDCA therapy was associated with significant improvement in laboratory tests (including decreased bilirubin in three of four studies) and histology in the three studies assessing this variable (241–243). However, clinical symptoms of pruritus and fatigue were not improved by UDCA. Portal inflammatory infiltrates were decreased, but other histopathological features remained unchanged. The dose of UDCA varied from 10 to 15 mg/kg/day, and it is notable that another controlled trial of UDCA 600 mg/day was ineffective (244). The largest randomized, doubleblind, placebocontrolled trial of UDCA (13 to 15 mg/kg/day) assessed time to treatment failure as the primary endpoint in 105 patients (243). Treatment failure was defined as (a) death; (b) OLT; (c) histopathological progression of two stages or to cirrhosis; (d) development of varices, ascites, or hepatic encephalopathy; (e) sustained quadrupling of bilirubin; (f) marked worsening of fatigue or pruritus; (g) drug intolerance; or (h) voluntary withdrawal from the study. After a median followup of 2.2 years, there was no significant difference between groups in the time to treatment failure, which occurred in 53 and 52% of placebo and UDCAtreated patients, respectively. As anticipated, alkaline phosphatase, AST, bilirubin, and albumin significantly improved with UDCA but not with placebo. In a smaller prospective, randomized, doubleblind, placebocontrolled trial of UDCA in 14 patients with PSC, 6 received UDCA (241). One patient withdrew from both the treatment and control groups. After a year of therapy, laboratory tests were significantly improved in the UDCA group. Histology was also improved, and the density of HLA class I antigen expression on hepatocytes was decreased. In another doubleblind, placebocontrolled trial, statistically significant improvements in alkaline phosphatase, gamma glutamyltransferase, and aminotransferases led to discontinuation of the placebo control arm and continuance as a prospective, nonrandomized study (242). In addition to UDCA, all patients had serial ERC and endoscopic dilatation of dominant strictures, which distinguishes this series from all others. Of 65 patients treated with UDCA (750 mg/
Page 685
day; 8.8 to 15.3 mg/kg/day; median 10.85 mg/kg/day) for up to 8 years, 10 (15.4%) developed dominant strictures requiring dilatation. One patient died due to cholangiocarcinoma and another required OLT. The actuarial KaplanMeier estimate of survival for the 65 patients was significantly greater than the predicted survival using the Mayo Risk Score model. Recently, the preliminary results of a randomized, doubleblind, placebocontrolled trial of highdose UDCA (20 mg/kg/day) were reported (245). Twentysix patients were randomized to UDCA or placebo after baseline liver biopsies and cholangiograms. Two patients withdrew: one on UDCA due to a dominant stricture requiring stenting and the other from the placebo group for variceal hemorrhage. One placebotreated patient died of a nonhepatobiliary cause. After 2 years, 22 patients underwent repeat biopsies and cholangiography. Symptoms did not improve in either group, but, as expected, liver biochemical test significantly improved in the UDCA group. The hepatic activity index of biopsies was unchanged or improved in 10 of 11 treated with UDCA and worsened in 7 of 10 treated with placebo. The histopathological stage of disease was unchanged or decreased in 8 of 11 patients in the UDCA group, while it increased in 7 of 11 in the placebo group. Cholangiographic severity in the UDCA group was unchanged in 7 of 11, improved in 2 of 11, and worse in 2 of 11. In contrast, cholangiography in the placebo group was unchanged in none, improved in 4 of 11, and worse in 7 of 11. These encouraging results should lead to larger randomized trials of highdose UDCA in PSC. Ursodeoxycholic Acid and Methotrexate. A pilot study evaluating UDCA (13 to 15 mg/kg/day) combined with methotrexate (0.25 mg/kg/week) versus UDCA alone did not demonstrate efficacy of the combination (246). Laboratory tests improved in both groups, but there was no additional benefit with the addition of methotrexate. Toxicities (alopecia and pulmonary) related to methotrexate led to its withdrawal in several patients. 6— Orthotopic Liver Transplantation OLT is the only potentially curative therapy for PSC patients with decompensated cirrhosis and complications of portal hypertension. Occasionally patients with less advanced PSC have been transplanted because of an intolerable quality of life due to debilitating pruritus, severe osteopenia with spontaneous fractures, or recurrent ascending cholangitis. Because of actual or potential involvement of the common bile duct, OLT is exclusively performed with a choledochojejunostomy biliary anastomosis. Optimal timing for OLT (see Sec. X, ''Prognostic Models," above) is difficult to achieve due to the variability in the natural history of the native disease, the impact of complications, and the progressive risk of cholangiocarcinoma. Although a recent report encouragingly indicated that the survival of patients with small cholangiocarcinomas discovered incidentally at the time of transplant is good (14), most centers continue to regard an established diagnosis of cholangiocarcinoma as an absolute contraindication to OLT (123). The Mayo Clinic prognostic model, now in its third iteration (Table 4), can be used to assess predictions of survival without transplant (158). By using independent variables of age, bilirubin, albumin, aspartate aminotransferase, and history of variceal bleeding and excluding the need for current liver histology, the new model achieves the required simplicity to redefine minimal listing criteria for OLT candidacy. Indeed, the United Network for Organ Sharing (UNOS) recently introduced the Mayo model for the determination of minimal listing criteria and will list patients with a predicted 1year survival of 95%. It is notable that medical therapies, such as UDCA, do not appear to mask either the need or indications for OLT (247). Two recent retrospective studies support the conclusion that OLT is effective therapy for advanced PSC (14,159). Among 216 patients transplanted for PSC at the University of Pittsburgh or Mayo Clinic between 1981 and 1990, the mean age was 42.1 ± 11.3 (standard deviation) years and pretransplant bilirubin was 13.3 ± 13 mg/dL (159). Cirrhosis was present in 97%, and 63% had splenomegaly. The mean followup postOLT was 34 months (range 1 to 104 months). The actual survival of transplanted patients was compared with predicted survival calculated using the Mayo Multicenter Study Group Index, which had been shown to
Page 686
accurately estimate survival of PSC patients with a wide spectrum of disease activity. As early as 6 months, actual survival after OLT (89%) already exceeded predicted survival (83%). After 5 years, the actual survival of patients undergoing OLT was 73%, a significant difference compared to the estimated survival of 28% without OLT. In the second series, 127 consecutive patients transplanted for PSC over a 12year period were assessed (14). The majority of patients had associated IBD (72%) and had undergone previous biliary surgery (62%). The 5year actuarial graft and patient survivals were 72 and 85%, respectively. Neither graft nor patient survival was adversely affected by a history of prior biliary surgery. Four patients with a known cholangiocarcinoma who were transplanted had recurrence within 6 months. In contrast, 10 patients with incidental cholangiocarcinomas (defined as <1 cm in size) detected in the explant had a 5year survival of 83%. The impact of IBD on the postOLT course of PSC was assessed in series of 31 patients with and 24 patients without IBD (248). Although patients with IBD had significantly more episodes of acute rejection, the incidence of ductopenic rejection did not differ between the groups, and the 5year survival was not adversely affected by the presence of IBD. According to proposed guidelines for the monitoring and selection of PSC patients for OLT (247), asymptomatic patients should undergo biannual evaluations. Symptomatic patients with diffuse sclerotic lesions should be monitored more closely and considered for enrollment in controlled therapeutic trials of medical therapy. Patients with dominant strictures should undergo ERC with brushings for cytology or, preferably, direct biopsy to diagnose potential cholangiocarcinoma. Benign dominant strictures should be balloondilated. Cirrhotic patients with dominant strictures will undoubtedly meet minimal UNOS listing criteria on the basis of elevated bilirubin and should be listed for OLT. Since cholangiocarcinoma. cannot be predicted or detected even with careful monitoring, it is rational to consider OLT in patients with PSC at earlier stages of disease. This should be facilitated by use of the Mayo model for calculation of minimal listing criteria based on an estimated 1 year survival of 95%. Recurrence of Primary Sclerosing Cholangitis after Orthotopic Liver Transplantation Whether the higherthanexpected incidence of intrahepatic biliary strictures in PSC patients following OLT is due to ischemic strictures or recurrent PSC remains controversial (14,249–251). Persistence of lower titers of pANCA after OLT was not found to correlate with histological evidence of disease recurrence (252). Ischemic, intrahepatic biliary strictures are a known complication of OLT for any indication. In two series, intrahepatic biliary strictures occurred with an incidence of 8.2% of 1590 allografts (253) and 15% of 687 allografts (254). Solitary strictures were found in 24% of patients, while 76% had multiple strictures (253,254). Significant risk factors predisposing to intrahepatic strictures included (a) hepatic artery occlusion, (b) PSC, (c) choledochojejunostomy biliary anastomosis, (d) use of EuroCollins preservation solution, (e) evidence of cholangitis on liver biopsy, and (f) young age. The increased incidence of intrahepatic strictures in PSC led to speculation of disease recurrence postOLT. One series compared the histopathology of patients transplanted with PSC and a control group without PSC who had choledochojejunostomy biliary anastomoses (250). Among 22 PSC patients, 33% had histological evidence of cholangitis and 14% had obliterative fibrous cholangirls. In contrast, on 3 of 22 controls had histological cholangitis (14%) and one (5%) had obliterative fibrous cholangitis. In a report of a larger cohort of 150 transplanted PSC patients, a probable recurrence of PSC was defined as the presence of typical biliary strictures on cholangiography >90 days postOLT and/or fibrous cholangitis on liver biopsy (255). Thirty patients were excluded because of strictures associated with hepatic artery thrombosis or stenosis, ductopenic rejection, biliary anastomotic strictures, or strictures evident <90 days postOLT. The findings among the remaining 120 PSC patients were compared with those of 30 nonPSC patients with choledochojejunostomy biliary anastomoses and 434 other patients with ducttoduct anastomoses. Twentyfour patients (20%) fulfilled criteria for PSC recurrence with a mean time to diagnosis of 421 days postOLT. Of these 24 patients, 22 (92%) met the cholangiographic criterion, while 11 (46%) had fibrous cholangitis and 9 (38%) met both criteria for recurrence. The incidence of strictures among PSC patients was significantly greater
Page 687
than that in either control group: 4 of 434 (1%) with ducttoduct anastomoses or 1 of 26 (4%) with choledochojejunostomy biliary anastomoses. These findings are consistent with the view that PSC can recur after OLT, but differentiation of recurrent disease and ischemic strictures remains difficult. Risk of Colon Cancer after Orthotopic Liver Transplantation Patients with UC have an increased risk of colorectal carcinoma associated with long duration of disease and pancolitis (19). Recent series indicated that the risk of colorectal carcinoma is also increased following OLT in patients with PSC and UC (123,152,156,157). In one series of 33 patients with PSC and IBD (32 UC, 1 Crohn's colitis) undergoing OLT, 2 had had prior protocolectomy and 4 died perioperatively (156). Despite negative screening colonoscopies prior to OLT in the remaining 27 patients, 3 (11%) developed colonic neoplasia within 9 to 13 months. Two had colon carcinoma, one had villous adenoma with dysplasia, and all had successful proctocolectomy. Importantly, there were no differences between the mean duration of IBD among those with and without neoplasia. In another series, 81 of 108 patients transplanted for PSC had IBD (80 with UC and one with Crohn's disease), and 24 (30%) had had proctocolectomy (152). After a mean followup of 4.2 years, 3 of 57 patients with intact colons developed colon carcinoma, and the cumulative incidence of dysplasia was 15 and 21% at 5 and 8 years, respectively. The risk of carcinoma after OLT was increased fourfold compared to that expected over a comparable period in nontransplant patients. However, there was no adverse impact on actuarial survival between patients with and without an intact colon. In contrast, none of 127 consecutive patients transplanted for PSC over a 12 year period developed colonic carcinoma in another series (14,256). The evidence indicates that the risk of colorectal dysplasia and carcinoma is clinically important in patients transplanted with concurrent PSC and IBD. Thus, most centers advocate screening colonoscopy with multiple biopsies every 6 months for the first 2 years and every year thereafter to detect rapidly developing colonic dysplasia/neoplasia in patients with PSC and preexisting IBD. Ulcerative Colitis after Orthotopic Liver Transplantation The impact of OLT and immunosuppression on ulcerative colitis after OLT varied among reported series (33,123,256–259). In one center, 7 of 14 patients with symptomatic UC preoperatively continued to have active disease, while 3 of 13 with asymptomatic UC preoperatively developed active disease (256). Intractable UC warranted proctocolectomy in four patients, one of whom refused. In contrast, another study of 23 PSC patients with UC showed that all 6 asymptomatic patients remained so after OLT, while 15 of 17 patients with symptomatic UC preoperatively had a marked reduction in the severity after OLT (257). OLT resulted in decreased frequency of stools, abdominal cramps, and flares in the majority. UC was more aggressive after OLT in another report of 18 patients transplanted for PSC and concomitant UC (33). During a mean followup of 38 months, the course of UC was worse in 50% and unchanged in 50%. Among 12 patients with quiescent UC before OLT, 4 worsened. Importantly, UC developed in 3 of 12 patients without preoperative disease. In another series of 25 patients transplanted for PSC with UC, 6 had undergone colectomy, 1 subtotal colectomy and 3 partial colectomy before OLT (258). After a mean followup of 37 months, 49% had quiescent disease, 14% had mild flares and 6% had severe flares. Mild flares responded to treatment with 5 aminosalicylates, while severe flares required increased doses of prednisone. Four PSC patients were reported who required an ileal pouch anal anastomosis for UC after OLT (259). The overall complication rate was high, including postoperative bleeding and one hepatic artery thrombosis. These reports indicate that intense immunosuppression after OLT does not prevent flares of UC colitis or prevent the de novo onset of disease. In patients with pouchitis before OLT, highdose immunosuppression may not improve it (225). B— Therapy for Complications 1— Cholangiocarcinoma There is no effective therapy for cholangiocarcinoma, and median survival after diagnosis is approximately 5 months. Lethal recurrences of cholangiocarcinoma in transplant recipients in
Page 688
24 months (14,123,260) led most centers to abandon transplantation of patients with proven tumors. However, a recent series indicated excellent longterm survival in transplant recipients with cholangiocarcinomas of <1 cm diameter discovered incidentally at transplant (14). 2— Dominant Strictures Benign dominant strictures should be endoscopically or percutaneously dilated and, if necessary, stented to prevent the pathophysiological consequences of biliary obstruction (217–219,261–266). Biliary diversion surgery should be avoided in PSC due to the risk of rapid stricturing, increased incidence of subsequent ascending bacterial cholangitis (60,61), and the adverse impact of prior biliary surgery on the outcome of OLT (16,125). 3— Cholelithiasis Asymptomatic cholelithiasis should be observed. Cholecystectomy is indicated only for patients with symptomatic cholecystitis and cholelithiasis. UDCA may prevent formation of and/or dissolve cholesterol gallstones in PSC. Pigmented stones made up of calcium bilirubinate may accumulate proximal to strictures and contribute to the degree of biliary obstruction. There is no specific therapy for pigmented stones. 4— Hepatic Osteodystrophy If low serum levels of 25hydroxy vitamin D indicate deficiency, patients should be supplemented with vitamin D derivatives and advised to increase sun exposure to generate vitamin D3 (141,267). Patients with osteoporosis should increase dietary calcium intake (1000 mg/day) and take a bisphosphonate and supplemental vitamin D (267). 5— Steatorrhea and FatSoluble Vitamin Deficiency Steatorrhea may be subclinical, and deficiencies of fatsoluble vitamins occur in patients with PSC (141). The differential diagnosis of steatorrhea in PSC includes celiac sprue (268–270), pancreatic exocrine insufficiency resulting from chronic pancreatitis (106), and small bowel bacterial overgrowth (271,272). Steatorrhea due to insufficient intraluminal concentrations of bile acids in the small bowel in patients with PSC may be improved by restriction of dietary fat. Patients should receive empiric supplements of vitamin D and have adequate dietary calcium and phosphorous. Vitamin E supplements of 400 IU/day are safe and effective in maintaining vitamin E levels. In patients with severe malabsorption, water soluble Dalphatocopherol or tocopheryl polyethylene glycol succinate are effective. The clinical significance of low serum levels of vitamin K in the absence of coagulopathy remains unclear. Parenteral vitamin K is indicated for patients with coagulopathy, and vitamin K1 is preferable to vitamin K2 (menadione2 methyl1; 4 naphthaquinone), which may be hepatotoxic. Zinc deficiency should be excluded when vitamin A replacement is contemplated. Oral replacement is preferable to parenteral administration. In patients with severe cholestatic malabsorption, oral absorption of all fat soluble vitamins can be increased by the coadministration of tocopheryl polyethylene glycol succinate. 6— Pruritus Nonabsorbable anionexchange resins, such as cholestyramine or colestipol, bind undefined pruritogenic factors and bile acids in the small intestine and increase their fecal excretion (146). Antihistamines have a modest impact on cholestatic pruritus, but their soporific effect may help patients sleep. If these measures are insufficient, phenobarbital can be added to the regimen at bedtime. Rifampicin is beneficial in PBC (273,274) and malignancy (275); it may be tried empirically for refractory pruritus in PSC. Opiate antagonists are currently under investigation
Page 689
for the treatment of cholestatic pruritus (276). For intractable pruritus, plasmapheresis, plasma exchange, or charcoal hemoperfusion has been anecdotally reported to be beneficial (277,278). 7— Ascending Cholangitis Episodes of ascending cholangitis should be treated with antibiotics administered intravenously (137). The probable causative organisms include Escherichia coli, Klebsiella species, and Enterococcus faecalis (279). Antibiotic choice should be refined on the basis of cultures of blood and/or bile, and more than one organism is often found (279–281). References 1. Vierling JM. Hepatobiliary complications of ulcerative colitis and Crohn's disease. In: Zakim D, Boyer TD, eds. Hepatology: A Textbook of Liver Disease, 3rd ed. Philadelphia: Saunders, 1996, pp 1366–1405. 2. Lee YM, Kaplan MM. Primary sclerosing cholangitis. N Engl J Med 1995; 332:924–933. 3. Martin M. Primary sclerosing cholangitis. Annu Rev Med 1993; 44:221–227. 4. Bergquist A, Broome U. Clinical features of primary sclerosing cholangitis. In: Lindor KD, Dickson ER, eds. PBC, PSC, and Adult Cholangiopathies. Philadelphia: Saunders, 1998, pp 283–301. 5. Ponsioen CI, Tytgat GN. Primary sclerosing cholangitis: a clinical review. Am J Gastroenterol 1998; 93:515–523. 6. Marotta PJ, LaRusso NF, Wiesner RH. Sclerosing cholangitis. Baillieres Clin Gastroenterol 1997; 11:781–800. 7. Vierling J, Hu KQ. Immunologic mechanism of hepatobiliary injury. In: Kaplowitz NE, ed. Liver and Biliary Disease, 2nd ed. Baltimore, MD: Williams & Wilkins, 1996, pp 55–87. 8. Vierling J. Aetiopathogenesis of primary sclerosis cholangitis. In: Mann M, Stiehl A, Chapman R, Weisner RH, eds. Primary Sclerosis Cholangitis. Freiburg, Germany: Kluwer, 1998, pp 37–45. 9. Chapman RW. Role of immune factors in the pathogenesis of primary sclerosing cholangitis. Semin Liver Dis 1991; 11:1–4. 10. Ludwig J. Histopathology of primary sclerosis cholangitis. In: Mann M, Stiehl A, Chapman R, Weisner RH, eds. Primary Sclerosis Cholangitis. Freiburg: Kluwer, 1998, pp 14–21. 11. Ludwig J. Smallduct primary sclerosing cholangitis. Semin Liver Dis 1991; 11:11–17. 12. Aadland E, Schrumpf E, Fausa O, Elgjo K, Heilo A, Aakhus T, et al. Primary sclerosing cholangitis: a longterm followup study. Scand J Gastroenterol 1987; 22:655–664. 13. Broome U, Olsson R, Loof L, Bodemar G, Hultcrantz R, Danielsson A, et al. Natural history and prognostic factors in 305 Swedish patients with primary sclerosing cholangitis. Gut 1996; 38:610–615. 14. Goss JA, Shackleton CR, Farmer DG, Arnaout WS, Seu P, Markowitz JS, et al. Orthotopic liver transplantation for primary sclerosing cholangitis: a 12year single center experience. Ann Surg 1997; 225:472–481. 15. Harnois DM, Gores GJ, Ludwig J, Steers JL, LaRusso NF, Wiesner RH. Are patients with cirrhotic stage primary sclerosing cholangitis at risk for the development of hepatocellular cancer? J Hepatol 1997; 27:512–516. 16. Narumi S, Roberts JP, Emond JC, Lake J, Ascher NL. Liver transplantation for sclerosing cholangitis. Hepatology 1995; 22:451–457. 17. Wiesner RH, Porayko MK, Hay JE, LaRusso NF, Steers JL, Krom RA, et al. Liver transplantation for primary sclerosing cholangitis: impact of risk factors on outcome. Liver Transplant Surg 1996; 2:99–108.
Page 690
18. Riordan SM, Williams R. Risk of cholangiocarcinoma in primary sclerosing cholangitis. In: Mann M, Stiehl A, Chapman R, Weisner RH, eds. Primary Sclerosis Cholangitis. Freiburg, Germany: Kluwer, 1998, pp 69–85. 19. Broome U. Primary sclerosing cholangitis—relationship to inflammatory bowel disease. In: Mann M, Stiehl A, Chapman R, Weisner RH, eds. Primary Sclerosis Cholangitis. Freiburg, Germany: Kluwer, 1998, pp 60–68. 20. Chapman RW, Arborgh BA, Rhodes JM, Summerfield JA, Dick R, Scheuer PJ, et al. Primary sclerosing cholangitis: a review of its clinical features, cholangiography, and hepatic histology. Gut 1980; 21:870–877. 21. Rasmussen HH, Fallingborg JF, Mortensen PB, Vyberg M, TageJensen U, Rasmussen SN. Hepatobiliary dysfunction and primary sclerosing cholangitis in patients with Crohn's disease. Scand J Gastroenterol 1997; 32:604–610. 22. Tobias R, Wright J, Kottler R. Primary sclerosing cholangitis associated with inflammatory bowel disease in Capetown, 1975–1981. S Afr Med J 1983; 63:229– 235. 23. Wiesner RH, LaRusso NF. Clinicopathologic features of the syndrome of primary sclerosing cholangitis. Gastroenterology 1980; 79:200–206. 24. Takikawa H, Manabe T. Primary sclerosing cholangitis in Japan—analysis of 192 cases. J Gastroenterol 1997; 32:134–137. 25. Okada H, Mizuno M, Yamamoto K, Tsuji T. Primary sclerosing cholangitis in Japanese patients: association with inflammatory bowel disease. Acta Med Okayama 1996; 50: 227–235. 26. Escorsell A, Pares A, Rodes J, SolisHerruzo JA, Miras M, de la Morena E. Epidemiology of primary sclerosing cholangitis in Spain: Spanish Association for the Study of the Liver. J Hepatol 1994; 21:787–791. 27. Kochhar R, Goenka MK, Das K, Nagi B, Bhasin DK, Chawla YK, et al. Primary sclerosing cholangitis: an experience from India. J Gastroenterol Hepatol 1996; 11:429–433. 28. Wiesner RH, Grambsch PM, Dickson ER, Ludwig J, MacCarty RL, Hunter EB, et al. Primary sclerosing cholangitis: natural history, prognostic factors and survival analysis. Hepatology 1989: 10:430–436. 29. Schrumpf E, Abdelnoor M, Fausa O, Elgjo K, Jenssen E, Kolmannskog F. Risk factors in primary sclerosing cholangitis. J Hepatol 1994; 21:1061–1066. 30. Broome U, Lofberg R, Lundqvist K, Veress B. Subclinical time span of inflammatory bowel disease in patients with primary sclerosing cholangitis. Dis Colon Rectum 1995; 38:1301–1305. 31. Fausa O, Schrumpf E, Elgjo K. Relationship of inflammatory bowel disease and primary sclerosing cholangitis. Semin Liver Dis 1991; 11:31–39. 32. Vajro P, Cucchiara S, Vegnente A, Iorio R, de Silva C, Cipolletta L, et al. Primary sclerosing cholangitis preceding Crohn's disease in a child with Down's syndrome. Dig Dis Sci 1998; 43:166–169. 33. Papatheodoridis GV, Hamilton M, Mistry PK, Davidson B, Rolles K, Burroughs AK. Ulcerative colitis has an aggressive course after orthotopic liver transplantation for primary sclerosing cholangitis (see comments). Gut 1998; 43:639–644. 34. Rabinovitz M, Demetris AJ, BouAbboud CF, Van Thiel DH. Simultaneous occurrence of primary sclerosing cholangitis and autoimmune chronic active hepatitis in a patient with ulcerative colitis. Dig Dis Sci 1992; 37:1606–1611. 35. McNair AN, Moloney M, Portmann BC, Williams R, McFarlane IG. Autoimmune hepatitis overlapping with primary sclerosing cholangitis in five cases. Am J Gastroenterol 1998; 93:777–784. 36. Gohlke F, Lohse AW, Dienes HP, Lohr H, MarkerHermann E, Gerken G, et al. Evidence for an overlap syndrome of autoimmune hepatitis and primary sclerosing cholangitis. J Hepatol 1996; 24:699–705. 37. Boberg KM, Fausa O, Haaland T, Holter E, Mellbye OJ, Spurkland A, et al. Features of autoimmune hepatitis in primary sclerosing cholangitis: an evaluation of 114 primary
Page 691
sclerosing cholangitis patients according to a scoring system for the diagnosis of autoimmune hepatitis. Hepatology 1996; 23:1369–376. 38. Seibold F, Weber P, Jenss H, Scheurlen M. Autoimmune hepatitis in inflammatory bowel disease: report of two unusual cases. Z Gastroenterol 1997; 35:29–32. 39. Perdigoto R, Carpenter HA, Czaja AJ. Frequency and significance of chronic ulcerative colitis in severe corticosteroidtreated autoimmune hepatitis. J Hepatol 1992; 14:325–331. 40. Neuberger J. Extrahepatic manifestations of primary sclerosing cholangitis. In: Mann M, Stiehl A, Chapman R, Weisner RH, eds. Primary Sclerosis Cholangitis. Freiburg, Germany: Kluwer, 1998, pp 86–98. 41. Sherlock S. Pathogenesis of sclerosing cholangitis: the role of nonimmune factors. Semin Liver Dis 1991; 11:5–10. 42. Kelly P, Patchett S, McCloskey D, Alstead E, Farthing M, Fairclough P. Sclerosing cholangitis, race and sex. Gut 1997; 41:688–689. 43. Dickson ER, Murtaugh PA, Wiesner RH, Grambsch PM, Fleming TR, Ludwig J, et al. Primary sclerosing cholangitis: refinement and validation of survival models. Gastroenterology 1992; 103:1893–1901. 44. Loftus EVJ, Sandborn WJ, Tremaine WJ, Mahoney DW, Zinsmeister AR, Offord KP, et al. Primary sclerosing cholangitis is associated with nonsmoking: a case control study (see comments). Gastroenterology 1996; 110:1496–1502. 45. van Erpecum KJ, Smits SJ, van de Meeberg PC, Linn FH, Wolfhagen FH, van BergeHenegouwen GP, et al. Risk of primary sclerosing cholangitis is associated with nonsmoking behavior (see comments). Gastroenterology 1996; 110:1503–1506. 46. Bergquist A, Glaumann H, Persson B, Broome U. Risk factors and clinical presentation of hepatobiliary carcinoma in patients with primary sclerosing cholangitis: a casecontrol study. Hepatology 1998; 27:311–316. 47. Janczewska I, Olsson R, Hultcrantz R, Broome U. Pregnancy in patients with primary sclerosing cholangitis. Liver 1996; 16:326–330. 48. Dickson ER, Larusso NF, Wiesner RH. Primary sclerosing cholangitis. Hepatology 1984; 4:33S–35S. 49. Broome U, Glaumann H, Hellers G, Nilsson B, Sorstad J, Hultcrantz R. Liver disease in ulcerative colitis: an epidemiological and follow up study in the county of Stockholm. Gut 1994; 35:84–89. 50. Farrant JM, Hayllar KM, Wilkinson ML, Karani J, Portmann BC, Westaby D, et al. Natural history and prognostic variables in primary sclerosing cholangitis. Gastroenterology 1991; 100:1710–1717. 51. elShabrawi M, Wilkinson ML, Portmann B, MieliVergani G, Chong SK, Williams R, et al. Primary sclerosing cholangitis in childhood. Gastroenterology 1987; 92:1226–1235. 52. Gregorio GV, Portmann B, Reid F, Donaldson PT, Doherty DG, McCartney M, et al. Autoimmune hepatitis in childhood: a 20year experience. Hepatology 1997; 25:541–547. 53. Jacquemin E, Hermans D, Myara A, Habes D, Debray D, Hadchouel M, et al. Ursodeoxycholic acid therapy in pediatric patients with progressive familial intrahepatic cholestasis. Hepatology 1997; 25:519–523. 54. Kozaiwa K, Tajiri H, Sawada A, Tada K, Etani Y, Miki K, et al. Three paediatric cases of primary sclerosing cholangitis treated with ursodeoxycholic acid and sulphasalazine. J Gastroenterol Hepatol 1998; 13:825–829. 55. Okolicsanyi L, Fabris L, Viaggi S, Carulli N, Podda M, Ricci G. Primary sclerosing cholangitis: clinical presentation, natural history and prognostic variables: an Italian multicentre study. The Italian PSC Study Group. Eur J Gastroenterol Hepatol 1996; 8: 685–691. 56. Shetty K, Rybicki L, Carey WD. The ChildPugh classification as a prognostic indicator for survival in primary sclerosing cholangitis. Hepatology 1997; 25:1049– 1053.
Page 692
57. Boberg KM, Schrumpf E. Primary sclerosis cholangitis: diagnosis and differential diagnosis. In: Mann M, Stiehl A, Chapman R, Weisner RH, eds. Primary Sclerosis Cholangitis. Freiburg, Germany: Kluwer, 1998, pp 3–13. 58. Helzberg JH, Petersen JM, Boyer JL. Improved survival with primary sclerosing cholangitis: a review of clinicopathologic features and comparison of symptomatic and asymptomatic patients. Gastroenterology 1987; 92:1869–1875. 59. Wiesner RH, LaRusso NF, Ludwig J, Dickson ER. Comparison of the clinicopathologic features of primary sclerosing cholangitis and primary biliary cirrhosis. Gastroenterology 1985; 88:108–114. 60. Ismail T, Angrisani L, Powell JE, Hubscher S, Buckels J, Neuberger J, et al. Primary sclerosing cholangitis: surgical options, prognostic variables and outcome. Br J Surg 1991; 78:564–567. 61. Martin FM, Rossi RL, Nugent FW, Scholz FJ, Jenkins RL, Lewis WD, et al. Surgical aspects of sclerosing cholangitis: results in 178 patients. Ann Surg 1990; 212:551–556. 62. Olsson R, Broome U, Danielsson A, Hagerstrand I, Jarnerot G, Loof L, et al. Colchicine treatment of primary sclerosing cholangitis. Gastroenterology 1995; 108:1199–1203. 63. Ludwig J, Barham SS, LaRusso NF, Elveback LR, Wiesner RH, McCall JT. Morphologic features of chronic hepatitis associated with primary sclerosing cholangitis and chronic ulcerative colitis. Hepatology 1981; 1:632–640. 64. MacCarty RL, LaRusso NF, Wiesner RH, Ludwig J. Primary sclerosing cholangitis: findings on cholangiography and pancreatography. Radiology 1983; 149:39– 44. 65. Rabinovitz M, Gavaler JS, Schade RR, Dindzans VJ, Chien MC, Van Thiel DH. Does primary sclerosing cholangitis occurring in association with inflammatory bowel disease differ from that occurring in the absence of inflammatory bowel disease? A study of sixtysix subjects. Hepatology 1990; 11:7–11. 66. Olsson R, Danielsson A, Jarnerot G, Lindstrom E, Loof L, Rolny P, et al. Prevalence of primary sclerosing cholangitis in patients with ulcerative colitis. Gastroenterology 1991; 100:1319–1323. 67. Lundqvist K, Broome U. Differences in colonic disease activity in patients with ulcerative colitis with and without primary sclerosing cholangitis: a case control study. Dis Colon Rectum 1997; 40:451–456. 68. Schrumpf E, Elgjo K, Fausa O, Gjone E, Kolmannskog F, Ritland S. Sclerosing cholangitis in ulcerative colitis. Scand J Gastroenterol 1980; 15:689–697. 69. Balasubramaniam K, Wiesner RH, LaRusso NF. Primary sclerosing cholangitis with normal serum alkaline phosphatase activity. Gastroenterology 1988; 95:1395–1398. 70. Clements D, Rhodes JM, Elias E. Severe bile duct lesions without biochemical evidence of cholestasis in a case of sclerosing cholangitis. J Hepatol 1986; 3:72– 74. 71. Gross JBJ, Ludwig J, Wiesner RH, McCall JT, LaRusso NF. Abnormalities in tests of copper metabolism in primary sclerosing cholangitis. Gastroenterology 1985; 89:272–278. 72. Shepherd HA, Selby WS, Chapman RW, Nolan D, Barbatis C, McGee JO, et al. Ulcerative colitis and persistent liver dysfunction. Q J Med 1983; 52:503–513. 73. LaRusso NF, Wiesner RH, Ludwig J, MacCarty RL. Current concepts: primary sclerosing cholangitis. N Engl J Med 1984; 310:899–903. 74. Porayko MK, Wiesner RH, LaRusso NF, Ludwig J, MacCarty RL, Steiner BL, et al. Patients with asymptomatic primary sclerosing cholangitis frequently have progressive disease (see comments). Gastroenterology 1990; 98:1594–1602. 75. Porayko MK, LaRusso NF, Wiesner RH. Primary sclerosing cholangitis: a progressive disease? Semin Liver Dis 1991; 11:18–25, 76. Rosen CB, Nagorney DM, Wiesner RH, Coffey RJJ, LaRusso NF. Cholangiocarcinoma complicating primary sclerosing cholangitis. Ann Surg 1991; 213:21–25. 77. Schrumpf E, Fausa O, Kolmannskog F, Elgjo K, Ritland S, Gjone E. Sclerosing cholangitis in ulcerative colitis: a followup study. Scand J Gastroenterol 1982; 17:33–39.
Page 693
78. MirMadjlessi SH, Sivak MVJ, Farmer RG. Hypereosinophilia, ulcerative colitis, sclerosing cholangitis, and bile duct carcinoma. Am J Gastroenterol 1986; 81:483–485. 79. Scheurlen M, Mork H, Weber P. Hypereosinophilic syndrome resembling chronic inflammatory bowel disease with primary sclerosing cholangitis. J Clin Gastroenterol 1992; 14:59–63. 80. Zauli D, Schrumpf E, Crespi C, Cassani F, Fausa O, Aadland E. An autoantibody profile in primary sclerosing cholangitis. J Hepatol 1987; 5:14–18. 81. Duerr RH, Targan SR, Landers CJ, LaRusso NF, Lindsay KL, Wiesner RH, et al. Neutrophil cytoplasmic antibodies: a link between primary sclerosing cholangitis and ulcerative colitis. Gastroenterology 1991; 100:1385–1391. 82. Klein R, Eisenburg J, Weber P, Seibold F, Berg PA. Significance and specificity of antibodies to neutrophils detected by western blotting for the serological diagnosis of primary sclerosing cholangitis. Hepatology 1991; 14:1147–1152. 83. Seibold F, Weber P, Schoning A, Mork H, Goppel S, Scheurlen M. Neutrophil antibodies (pANCA) in chronic liver disease and inflammatory bowel disease: do they react with different antigens? Eur J Gastroenterol Hepatol 1996; 8:1095–1100. 84. Snook JA, Chapman RW, Fleming K, Jewell DP. Antineutrophil nuclear antibody in ulcerative colitis, Crohn's disease and primary sclerosing cholangitis. Clin Exp Immunol 1989; 76:30–33. 85. Lo SK, Fleming KA, Chapman RW. A 2year followup study of antineutrophil antibody in primary sclerosing cholangitis: relationship to clinical activity, liver biochemistry and ursodeoxycholic acid treatment. J Hepatol 1994; 21:974–978. 86. Seibold F, Weber P, Klein R, Berg PA, Wiedmann KH. Clinical significance of antibodies against neutrophils in patients with inflammatory bowel disease and primary sclerosing cholangitis. Gut 1992; 33:657–662. 87. Targan SR, Landers C, Vidrich A, Czaja AJ. Hightiter antineutrophil cytoplasmic antibodies in type1 autoimmune hepatitis (see comments). Gastroenterology 1995; 108: 1159–1166. 88. Desmet VJ. Current problems in diagnosis of biliary disease and cholestasis. Semin Liver Dis 1986; 6:233–245. 89. Desmet VJ. Destructive intrahepatic bile duct diseases. Recenti Prog Med 1990; 81: 392–398. 90. Casali AM, Carbone G, Cavalli G. Intrahepatic bile duct loss in primary sclerosing cholangitis: a quantitative study. Histopathology 1998; 32:449–453. 91. Terasaki S, Nakanuma Y, Unoura M, Kaneko S, Kobayashi K. Involvement of peribiliary glands in primary sclerosing cholangitis: a histopathologic study. Intern Med 1997; 36: 766–770. 92. Harrison RF, Hubscher SG. The spectrum of bile duct lesions in endstage primary sclerosing cholangitis. Histopathology 1991; 19:321–327. 93. Hadjis NS, Adam A, Blenkharn I, Hatzis G, Benjamin IS, Blumgart LH. Primary sclerosing cholangitis associated with liver atrophy. Am J Surg 1989; 158:43– 47. 94. Outwater E, Kaplan MM, Bankoff MS. Lymphadenopathy in sclerosing cholangitis: pitfall in the diagnosis of malignant biliary obstruction. Gastrointest Radiol 1992; 17: 157–160. 95. AlbertiFlor JJ, Kalemeris G, Dunn GD, Avant GR. Primary sclerosing cholangitis associated with massive intraabdominal lymphadenopathy. Am J Gastroenterol 1986; 81: 55–60. 96. Thorpe ME, Scheuer PJ, Sherlock S. Primary sclerosing cholangitis, the biliary tree, and ulcerative colitis. Gut 1967; 8:435–448. 97. Pokorny CS, McCaughan GW, Gallagher ND, Selby WS. Sclerosing cholangitis and biliary tract calculi—primary or secondary? Gut 1992; 33:1376–1380. 98. Jessurun J, BolioSolis A, Manivel JC. Diffuse lymphoplasmacytic acalculous cholecystitis: a distinctive form of chronic cholecystitis associated with primary sclerosing cholangitis. Hum Pathol 1998; 29:512–517.
Page 694
99. Brandt DJ, MacCarty RL, Charboneau JW, LaRusso NF, Wiesner RH, Ludwig J. Gallbladder disease in patients with primary sclerosing cholangitis. AJR 1988; 150:571–574. 100. van de Meeberg PC, Portincasa P, Wolfhagen FH, van Erpecum KJ, VanBergeHenegouwen GP. Increased gallbladder volume in primary sclerosing cholangitis. Gut 1996; 39:594–599. 101. Jeffrey GP, Reed WD, Carrello S, Shilken KB. Histological and immunohistochemical study of the gall bladder lesion in primary sclerosing cholangitis. Gut 1991; 32:424–429. 102. MacCarty RL, LaRusso NF, May GR, Bender CE, Wiesner RH, King JE, et al. Cholangiocarcinoma complicating primary sclerosing cholangitis: cholangiographic appearances. Radiology 1985; 156:43–46. 103. Ramage JK, Donaghy A, Farrant JM, Iorns R, Williams R. Serum tumor markers for the diagnosis of cholangiocarcinoma in primary sclerosing cholangitis. Gastroenterology 1995; 108:865–869. 104. Rizzi PM, Ryder SD, Portmann B, Ramage JK, Naoumov NV, Williams R. p53 Protein overexpression in cholangiocarcinoma arising in primary sclerosing cholangitis. Gut 1996; 38:265–268. 105. Epstein O, Chapman RW, LakeBakaar G, Foo AY, Rosalki SB, Sherlock S. The pancreas in primary biliary cirrhosis and primary sclerosing cholangitis. Gastroenterology 1982; 83:1177–1182. 106. Borkje B, Vetvik K, Odegaard S, Schrumpf E, Larssen TB, Kolmannskog F. Chronic pancreatitis in patients with sclerosing cholangitis and ulcerative colitis. Scand J Gastroenterol 1985; 20:539–542. 107. Imrie CW, Brombacher GD. Sclerosing cholangitis: a rare etiology for acute pancreatitis. Int J Pancreatol 1998; 23:71–75. 108. Johnson CD. Two cases of acute pancreatitis associated with sclerosing cholangitis (letter). Int J Pancreatol 1998; 24:141. 109. Kaw M, Silverman WB, Rabinovitz M, Schade RR. Biliary tract calculi in primary sclerosing cholangitis (see comments). Am J Gastroenterol 1995; 90:72–75. 110. Nadir A, Nadir F, Hassanein T, Gurakar A, Wright HI, Van Thiel DH. Acute relapsing pancreatitis induced with ursodeoxycholic acid therapy. J Okla State Med Assoc 1995; 88:295–298. 111. Pineau BC, Pattee LP, McGuire S, Sekar A, Scully LJ. Unusual presentation of primary sclerosing cholangitis. Can J Gastroenterol 1997; 11:45–48. 112. Schimanski U, Stiehl A, Stremmel W, Theilmann L. Low prevalence of alterations in the pancreatic duct system in patients with primary sclerosing cholangitis. Endoscopy 1996; 28:346–349. 113. Warren KW, Athanassiades S, Monge JI. Primary sclerosing cholangitis: a study of fortytwo cases. Am J Surg 1966; 111:23–38. 114. Bhathal PS, Powell LW. Primary intrahepatic obliterating cholangitis: a possible variant of "sclerosing cholangitis." Gut 1969; 10:886–893. 115. Ernst O, Asselah T, Sergent G, Calvo M, Talbodec N, Paris JC, et al. MR cholangiography in primary sclerosing cholangitis. AJR 1998; 171:1027–1030. 116. Campbell WL, Ferris JV, Holbert BL, Thaete FL, Baron RL. Biliary tract carcinoma complicating primary sclerosing cholangitis: evaluation with CT, cholangiography, US, and MR imaging. Radiology 1998; 207:41–50. 117. Lumsden AB, Alspaugh JP. Cholangiocarcinoma complicating primary sclerosing cholangitis: cholangiographic appearances (letter). Radiology 1986; 158:856. 118. Olsson RG, Asztely MS. Prognostic value of cholangiography in primary sclerosing cholangitis. Eur J Gastroenterol Hepatol 1995; 7:251–254. 119. Muiesan P, Shanmugam RP, Devlin J, Rela M, Heaton ND, Saxena R, et al. Orthotopic liver transplantation for primary sclerosing cholangitis. Transplant Proc 1994; 26:3574–3576.
Page 695
120. AbuElmagd KM, Selby R, Iwatsuki S, Fung J, Tzakis A, Todo S, et al. Cholangiocarcinoma and sclerosing cholangitis: clinical characteristics and effect on survival after liver transplantation. Transplant Proc 1993; 25:1124–1125. 121. Miros M, Kerlin P, Walker N, Harper J, Lynch S, Strong R. Predicting cholangiocarcinoma in patients with primary sclerosing cholangitis before transplantation. Gut 1991; 32:1369–1373. 122. Marsh JWJ, Iwatsuki S, Makowka L, Esquivel CO, Gordon RD, Todo S, et al. Orthotopic liver transplantation for primary sclerosing cholangitis. Ann Surg 1988; 207:21–25. 123. Knechtle SJ, D'Alessandro AM, Harms BA, Pirsch JD, Belzer FO, Kalayoglu M. Relationships between sclerosing cholangitis, inflammatory bowel disease, and cancer in patients undergoing liver transplantation. Surgery 1995; 118:615–619. 124. Broome U, Eriksson LS. Assessment for liver transplantation in patients with primary sclerosing cholangitis. J Hepatol 1994; 20:654–659. 125. Farges O, Malassagne B, Sebagh M, Bismuth H. Primary sclerosing cholangitis: liver transplantation or biliary surgery. Surgery 1995; 117:146–155. 126. Ernst O, Asselah T, Talbodec N, Sergent G. MR cholangiopancreatography: a promising new tool for diagnosing primary sclerosing cholangitis (letter; comment). AJR 1997; 168:1115–1116. 127. Keiding S, Hansen SB, Rasmussen HH, Gee A, Kruse A, Roelsgaard K, et al. Detection of cholangiocarcinoma in primary sclerosing cholangitis by positron emission tomography. Hepatology 1998; 28:700–706. 128. Van Laethem JL, Deviere J, Bourgeois N, Love J, Gelin M, Cremer M, et al. Cholangiographic findings in deteriorating primary sclerosing cholangitis. Endoscopy 1995; 27: 223–228. 129. Martins EB, Fleming KA, Garrido MC, Hine KR, Chapman RW. Superficial thrombophlebitis, dysplasia, and cholangiocarcinoma in primary sclerosing cholangitis. Gastroenterology 1994; 107:537–542. 130. Ferrari JA, Lichtenstein DR, Slivka A, Chang C, CarrLocke DL. Brush cytology during ERCP for the diagnosis of biliary and pancreatic malignancies (see comments). Gastrointest Endosc 1994; 40:140–145. 131. Desa LA, Akosa AB, Lazzara S, Domizio P, Krausz T, Benjamin IS. Cytodiagnosis in the management of extrahepatic biliary stricture. Gut 1991; 32:1188– 1191. 132. Kurzawinski T, Deery A, Dooley J, Dick R, Hobbs K, Davidson B. A prospective controlled study comparing brush and bile exfoliative cytology for diagnosing bile duct strictures. Gut 1992; 33:1675–1677. 133. Rabinovits M, Zajko AB, Hassanein T, Shetty B, Bron KM, Schade RR, et al. Diagnostic value of brush cytology in the diagnosis of bile duct carcinoma: a study in 65 patients with bile duct strictures. Hepatology 1990; 12:747–752. 134. Mansfield JC, Griffin SM, Wadehra V, Matthewson K. A prospective evaluation of cytology from biliary strictures. Gut 1997; 40:671–677. 135. Bloom S, Heryet A, Fleming K, Chapman RW. Inappropriate expression of blood group antigens on biliary and colonic epithelia in primary sclerosing cholangitis. Gut 1993; 34:977–983. 136. Rogers SA, Podolsky DK. Predicting cholangiocarcinoma in patients with primary sclerosing cholangitis: an analysis of the serological marker CA 19–9. Hepatology 1994; 19: 543–545. 137. Ponsioen CI, Huibregtse K. Treatment of complications of primary sclerosing cholangitis. In: Mann M, Stiehl A, Chapman R, Weisner RH, eds. Primary Sclerosis Cholangitis. Freiburg, Germany: Kluwer, 1998, pp 115–123. 138. Angulo P, Therneau TM, Jorgensen A, DeSotel CK, Egan KS, Dickson ER, et al. Bone disease in patients with primary sclerosing cholangitis: prevalence, severity and prediction of progression. J Hepatol 1998; 29:729–735.
Page 696
139. Hay JE. Bone disease in cholestatic liver disease. Gastroenterology 1995; 108:276–283. 140. Hay JE, Lindor KD, Wiesner RH, Dickson ER, Krom RA, LaRusso NF. The metabolic bone disease of primary sclerosing cholangitis. Hepatology 1991; 14:257–261. 141. Kowdley KV. Lipids and lipidactivated vitamins in chronic cholestatic diseases. In: Lindor KD, Dickson ER, eds. PBC, PSC, and Adult Cholangiopathies. Philadelphia: Saunders, 1998, pp 373–389. 142. Jorgensen RA, Lindor KD, Sartin JS, Larusso NF, Wiesner RH. Serum lipid and fatsoluble vitamin levels in primary sclerosing cholangitis. J Clin Gastroenterol 1995; 20: 215–219. 143. Wiesner RH, LaRusso NF, Dozois RR, Beaver SJ. Peristomal varices after proctocolectomy in patients with primary sclerosing cholangitis. Gastroenterology 1986; 90:316–322. 144. Fucini C, Wolff BG, Dozois RR. Bleeding from peristomal varices: perspectives on prevention and treatment. Dis Colon Rectum 1991; 34:1073–1078. 145. Peck JJ, Boyden AM. Exigent ileostomy hemorrhage: a complication of proctocolectomy in patients with chronic ulcerative colitis and primary sclerosing cholangitis. Am J Surg 1985; 150:153–158. 146. Bergassa NV, Jones EA. The pruritus of cholestasis. In: Lindor KD, Dickson ER, eds. PBC, PSC, and Adult Cholangiopathies. Philadelphia: Saunders, 1998, pp 391–405. 147. Broome U, Lindberg G, Lofberg R. Primary sclerosing cholangitis in ulcerative colitis a risk factor for the development of dysplasia and DNA aneuploidy? (see comments). Gastroenterology 1992; 102:1877–1880. 148. Brentnall TA, Haggitt RC, Rabinovitch PS, Kimmey MB, Bronner MP, Levine DS, et al. Risk and natural history of colonic neoplasia in patients with primary sclerosing cholangitis and ulcerative colitis (see comments). Gastroenterology 1996; 110:331–338. 149. D'Haens GR, Lashner BA, Hanauer SB. Pericholangitis and sclerosing cholangitis are risk factors for dysplasia and cancer in ulcerative colitis (see comments). Am J Gastroenterol 1993; 88:1174–1178. 150. Kornfeld D, Ekbom A, Ihre T. Is there an excess risk for colorectal cancer in patients with ulcerative colitis and concomitant primary sclerosing cholangitis? A population based study (see comments). Gut 1997; 41:522–525. 151. Leidenius M. Ulcerative colitis—association with primary sclerosing cholangitis and colorectal neoplasia. Ann Chir Gynaecol 1998; 87:72–73. 152. Loftus EVJ, Aguilar HI, Sandborn WJ, Tremaine WJ, Krom RA, Zinsmeister AR, et al. Risk of colorectal neoplasia in patients with primary sclerosing cholangitis and ulcerative colitis following orthotopic liver transplantation. Hepatology 1998; 27:685–690. 153. Marchesa P, Lashner BA, Lavery IC, Milsom J, Hull TL, Strong SA, et al. The risk of cancer and dysplasia among ulcerative colitis patients with primary sclerosing cholangitis. Am J Gastroenterol 1997; 92:1285–1288. 154. Nuako KW, Ahlquist DA, Sandborn WJ, Mahoney DW, Siems DM, Zinsmeister AR. Primary sclerosing cholangitis and colorectal carcinoma in patients with chronic ulcerative colitis: a casecontrol study. Cancer 1998; 82:822–826. 155. Leidenius MH, Farkkila MA, Karkkainen P, Taskinen EI, Kellokumpu IH, Hockerstedt KA. Colorectal dysplasia and carcinoma in patients with ulcerative colitis and primary sclerosing cholangitis. Scand J Gastroenterol 1997; 32:706–711. 156. Bleday R, Lee E, Jessurun J, Heine J, Wong WD. Increased risk of early colorectal neoplasms after hepatic transplant in patients with inflammatory bowel disease. Dis Colon Rectum 1993; 36:908–912. 157. Higashi H, Yanaga K, Marsh JW, Tzakis A, Kakizoe S, Starzl TE. Development of colon cancer after liver transplantation for primary sclerosing cholangitis associated with ulcerative colitis. Hepatology 1990; 11:477–480. 158. Wiesner RH. Liver transplantation for primary biliary cirrhosis and primary sclerosing cholangitis: predicting outcomes with natural history models. Mayo Clin Proc 1998; 73: 575–588.
Page 697
159. AbuElmagd KM, Malinchoc M, Dickson ER, Fung JJ, Murtaugh PA, Langworthy AL, et al. Efficacy of hepatic transplantation in patients with primary sclerosing cholangitis. Surg Gynecol Obstet 1993; 177:335–344. 160. Hamour AA, Bonnington A, Hawthorne B, Wilkins EG. Successful treatment of AIDSrelated cryptosporidial sclerosing cholangitis. AIDS 1993; 7:1449–1451. 161. Patel SA, Borges MC, Batt MD, Rosenblate HJ. Trichosporon cholangitis associated with hyperbilirubinemia, and findings suggesting primary sclerosing cholangitis on endoscopic retrograde cholangiopancreatography. Am J Gastroenterol 1990; 85:84–87. 162. Hayward AR, Levy J, Facchetti F, Notarangelo L, Ochs HD, Etzioni A, et al. Cholangiopathy and tumors of the pancreas, liver, and biliary tree in boys with X linked immunodeficiency with hyperIgM. J Immunol 1997; 158:977–983. 163. Mehal WZ, Hattersley AT, Chapman RW, Fleming KA. A survey of cytomegalovirus (CMV) DNA in primary sclerosing cholangitis (PSC) liver tissues using a sensitive polymerase chain reaction (PCR) based assay. J Hepatol 1992; 15:396–399. 164. Bucuvalas JC, Bove KE, Kaufman RA, Gilchrist MJ, Oldham KT, Balistreri WF. Cholangitis associated with Cryptococcus neoformans. Gastroenterology 1985; 88:1055–1059. 165. Chapman RW, Jewell DP. Primary sclerosing cholangitis—an immunologically mediated disease? West J Med 1985; 143:193–195. 166. Si L, Whiteside TL, Schade RR, Starzl TE, Van Thiel DH. Tlymphocyte subsets in liver tissues of patients with primary biliary cirrhosis (PBC), patients with primary sclerosing cholangitis (PSC), and normal controls. J Clin Immunol 1984; 4:262–272. 167. Whiteside TL, Lasky S, Si L, Van Thiel DH. Immunologic analysis of mononuclear cells in liver tissues and blood of patients with primary sclerosing cholangitis. Hepatology 1985; 5:468–474. 168. Valenski WR, Herrod HG, Williams JW. In vitro evidence for B cell dysfunction in patients with chronic liver disease. J Clin Lab Immunol 1989; 28:169–172. 169. Kilby AE, Krawitt EL, Albertini RJ, Chastenay BF, John A. Suppressor Tcell deficiency in primary sclerosing cholangitis: case and family study. Dig Dis Sci 1991; 36:1213–1216. 170. Lindor KD, Wiesner RH, LaRusso NF, Homburger HA. Enhanced autoreactivity of Tlymphocytes in primary sclerosing cholangitis. Hepatology 1987; 7:884– 888. 171. Bodenheimer HCJ, Larusso NF, Thayer WRJ, Charland C, Staples PJ, Ludwig J. Elevated circulating immune complexes in primary sclerosing cholangitis. Hepatology 1983; 3:150–154. 172. Senaldi G, Donaldson PT, Magrin S, Farrant JM, Alexander GJ, Vergani D, et al. Activation of the complement system in primary sclerosing cholangitis. Gastroenterology 1989; 97:1430–1434. 173. Garred P, Lyon H, Christoffersen P, Mollnes TE, TranumJensen J. Deposition of C3, the terminal complement complex and vitronectin in primary biliary cirrhosis and primary sclerosing cholangitis. Liver 1993; 13:305–310. 174. Minuk GY, Angus M, Brickman CM, Lawley TJ, Frank MM, Hoofnagle JH, et al. Abnormal clearance of immune complexes from the circulation of patients with primary sclerosing cholangitis. Gastroenterology 1985; 88:166–170. 175. Mizoguchi E, Mizoguchi A, Chiba C, Niles JL, Bhan AK. Antineutrophil cytoplasmic antibodies in Tcell receptor alpha—deficient mice with chronic colitis. Gastroenterology 1997; 113:1828–1835. 176. Seibold F, Brandwein S, Simpson S, Terhorst C, Elson CO. pANCA represents a crossreactivity to enteric bacterial antigens. J Clin Immunol 1998; 18:153– 160. 177. Lichtman SN, Keku J, Clark RL, Schwab JH, Sartor RB. Biliary tract disease in rats with experimental small bowel bacterial overgrowth. Hepatology 1991; 13:766–772. 178. Lichtman SN, Keku J, Schwab JH, Sartor RB. Hepatic injury associated with small bowel bacterial overgrowth in rats is prevented by metronidazole and tetracycline. Gastroenterology 1991; 100:513–519.
Page 698
179. Donaldson P. Immunogenetics and epidemiology of primary sclerosis cholangitis. In: Mann M, Stiehl A, Chapman R, Weisner RH, eds. Primary Sclerosis Cholangitis. Freiburg, Germany: Kluwer, 1998, pp 22–36. 180. Farrant JM, Doherty DG, Donaldson PT, Vaughan RW, Hayllar KM, Welsh KI, et al. Amino acid substitutions at position 38 of the DR beta polypeptide confer susceptibility to and protection from primary sclerosing cholangitis. Hepatology 1992; 16:390–395. 181. Freund YR, Sgarlato G, Jacob CO, Suzuki Y, Remington JS. Polymorphisms in the tumor necrosis factor alpha (TNFalpha) gene correlate with murine resistance to development of toxoplasmic encephalitis and with levels of TNFalpha mRNA in infected brain tissue. J Exp Med 1992; 175:683–688. 182. Jacob CO. Genetic variability in tumor necrosis factor production: relevance to predisposition to autoimmune disease. Reg Immunol 1992; 4:298–304. 183. Jacob CO. Studies on the role of tumor necrosis factor in murine and human autoimmunity. J Autoimmun 1992; 5(suppl A): 133–143. 184. Bansi D, Chapman R, Fleming K. Antineutrophil cytoplasmic antibodies in chronic liver diseases: prevalence, titre, specificity and IgG subclass. J Hepatol 1996; 24:581–586. 185. Vidrich A, Lee J, James E, Cobb L, Targan S. Segregation of pANCA antigenic recognition by DNase treatment of neutrophils: ulcerative colitis, type 1 autoimmune hepatitis, and primary sclerosing cholangitis. J Clin Immunol 1995; 15:293–299. 186. Ruemmele FM, Targan SR, Levy G, Dubinsky M, Braun J, Seidman EG. Diagnostic accuracy of serological assays in pediatric inflammatory bowel disease (see comments). Gastroenterology 1998; 115:822–829. 187. Eggena M, Targan SR, Iwanczyk L, Vidrich A, Gordon LK, Braun J. Phage display cloning and characterization of an immunogenetic marker (perinuclear anti neutrophil cytoplasmic antibody) in ulcerative colitis. J Immunol 1996; 156:4005–4011. 188. Hopf U, Berg T, Korber J, Klein R, Berg PA. Autoimmune markers in primary sclerosing cholangitis. In: Mann M, Stiehl A, Chapman R, Weisner RH, eds. Primary Sclerosis Cholangitis. Freiburg, Germany: Kluwer, 1998, pp 46–54. 189. Mandal A, Dasgupta A, Jeffers L, Squillante L, Hyder S, Reddy R, et al. Autoantibodies in sclerosing cholangitis against a shared peptide in biliary and colon epithelium. Gastroenterology 1994; 106:185–192. 190. Das KM. Relationship of extraintestinal involvements in inflammatory bowel disease: new insights into autoimmune pathogenesis. Dig Dis Sci 1999; 44:1–13. 191. Das KM, Vecchi M, Sakamaki S. A shared and unique epitope(s) on human colon, skin, and biliary epithelium detected by a monoclonal antibody. Gastroenterology 1990; 98: 464–469. 192. Leon MP, Bassendine MF, Gibbs P, Thick M, Kirby JA. Immunogenicity of biliary epithelium: study of the adhesive interaction with lymphocytes. Gastroenterology 1997; 112:968–977. 193. Leon MP, Kirby JA, Gibbs P, Burt AD, Bassendine MF. Immunogenicity of biliary epithelial cells: study of the expression of B7 molecules. J Hepatol 1995; 22:591–595. 194. Tsuneyama K, Harada K, Yasoshima M, Kaji K, Gershwin ME, Nakanuma Y. Expression of costimulatory factor B72 on the intrahepatic bile ducts in primary biliary cirrhosis and primary sclerosing cholangitis: an immunohistochemical study. J Pathol 1998; 186: 126–130. 195. Spengler U, Leifeld L, Braunschweiger I, Dumoulin FL, Lechmann M, Sauerbruch T. Anomalous expression of costimulatory molecules B71, B72 and CD28 in primary biliary cirrhosis. J Hepatol 1997; 26:31–36. 196. Dienes HP, Lohse AW, Gerken G, Schirmacher P, Gallati H, Lohr HF, et al. Bile duct epithelia as target cells in primary biliary cirrhosis and primary sclerosing cholangitis. Virchows Arch 1997; 431:119–124. 197. Broome U, Hultcrantz R, Scheynius A. Lack of concomitant expression of ICAM1 and HLADR on bile duct cells from patients with primary sclerosing cholangitis and primary biliary cirrhosis. Scand J Gastroenterol 1993; 28:126–130.
Page 699
198. Hashimoto E, Lindor KD, Homburger HA, Dickson ER, Czaja AJ, Wiesner RH, et al. Immunohistochemical characterization of hepatic lymphocytes in primary biliary cirrhosis in comparison with primary sclerosing cholangitis and autoimmune chronic active hepatitis (see comments). Mayo Clin Proc 1993; 68:1049–1055. 199. Roberts SK, Ludwig J, Larusso NF. The pathobiology of biliary epithelia. Gastroenterology 1997; 112:269–279. 200. Charlotte F, L'Hermine A, Martin N, Geleyn Y, Nollet M, Gaulard P, et al. Immunohistochemical detection of bcl2 protein in normal and pathological human liver. Am J Pathol 1994; 144:460–465. 201. Probert CS, Christ AD, Saubermann LJ, Turner JR, Chott A, CarrLocke D, et al. Analysis of human common bile ductassociated T cells: evidence for oligoclonality, T cell clonal persistence, and epithelial cell recognition. J Immunol 1997; 158:1941–1948. 202. Broome U, Grunewald J, Scheynius A, Olerup O, Hultcrantz R. Preferential V beta3 usage by hepatic T lymphocytes in patients with primary sclerosing cholangitis. J Hepatol 1997; 26:527–534. 203. Woolf GM, Vierling JM. Disappearing intrahepatic bile ducts: the syndromes and their mechanisms. Semin Liver Dis 1993; 13:261–275. 204. Washington K, Clavien PA, Killenberg P. Peribiliary vascular plexus in primary sclerosing cholangitis and primary biliary cirrhosis. Hum Pathol 1997; 28:791– 795. 205. Hiroki F, Matsumoto K, Melhem Meal. Participation of bile duct cells in biliary inflammation: production of IL8 in response to irritant stimuli. Submitted, 1999. 206. Stocks SC, RuchaudSparagano MH, Kerr MA, Grunert F, Haslett C, Dransfield I. CD66: role in the regulation of neutrophil effector function. Eur J Immunol 1996; 26: 2924–2932. 207. Riethdorf L, Lisboa BW, Henkel U, Naumann M, Wagener C, Loning T. Differential expression of CD66a (BGP), a cell adhesion molecule of the carcinoembryonic antigen family, in benign, premalignant, and malignant lesions of the human mammary gland. J Histochem Cytochem 1997; 45:957–963. 208. Desmet V, Roskams T, Van Eyken P. Ductular reaction in the liver. Pathol Res Pract 1995; 191:513–524. 209. Pinzani M, Milani S, Grappone C, Weber FLJ, Gentilini P, Abboud HE. Expression of plateletderived growth factor in a model of acute liver injury. Hepatology 1994; 19: 701–707. 210. Pinzani M, Milani S, Herbst H, DeFanco R, Grappone C, Gentilini A, et al. Expression of plateletderived growth factor and its receptors in normal human liver and during active hepatic fibrogenesis. Am J Pathol 1996; 148:785–800. 211. Kuroe K, Haga Y, Funakoshi O, Mizuki I, Kanazawa K, Yoshida Y. Extraintestinal manifestations of granulomatous enterocolitis induced in rabbits by long term submucosal administration of muramyl dipeptide emulsified with Freund's incomplete adjuvant. J Gastroenterol 1996; 31:199–206. 212. Yamada S, Ishii M, Liang LS, Yamamoto T, Toyota T. Small duct cholangitis induced by Nformyl Lmethionine Lleucine Ltyrosine in rats. J Gastroenterol 1994; 29:631–636. 213. Lichtman SN, Bachmann S, Munoz SR, Schwab JH, Bender DE, Sartor RB, et al. Bacterial cell wall polymers (peptidoglycanpolysaccharide) cause reactivation of arthritis. Infect Immun 1993; 61:4645–4653. 214. Lichtman SN, Okoruwa EE, Keku J, Schwab JH, Sartor RB. Degradation of endogenous bacterial cell wall polymers by the muralytic enzyme mutanolysin prevents hepatobiliary injury in genetically susceptible rats with experimental intestinal bacterial overgrowth. J Clin Invest 1992; 90:1313–1322. 215. Nonomura A, Kono N, Minato H, Nakanuma Y. Diffuse biliary tract involvement mimicking primary sclerosing cholangitis in an experimental model of chronic graftversushost disease in mice. Pathol Int 1998; 48:421–427.
Page 700
216. Geubel AP, Cnudde A, Ferrant A, Latinne D, Rahier J. Diffuse biliary tract involvement mimicking primary sclerosing cholangitis after bone marrow transplantation. J Hepatol 1990; 10:23–28. 217. Stiehl A, Benz C, Sauer P, Theilmann L. Treatment of primary sclerosing cholangitis: the role of ursodeoxycholic acid and endoscopy. In: Mann M, Stiehl A, Chapman R, Weisner RH, eds. Primary Sclerosis Cholangitis. Freiburg, Germany: Kluwer, 1998, pp 99–102. 218. Ahrendt SA, Pitt HA, Kalloo AN, Venbrux AC, Klein AS, Herlong HF, et al. Primary sclerosing cholangitis: resect, dilate, or transplant? Ann Surg 1998; 227:412–423. 219. Eckhauser FE, Colleti LM, Knol JA. The changing role of surgery for sclerosing cholangitis. Dig Dis 1996; 14:180–191. 220. Cangemi JR, Wiesner RH, Beaver SJ, Ludwig J, MacCarty RL, Dozois RR, et al. Effect of proctocolectomy for chronic ulcerative colitis on the natural history of primary sclerosing cholangitis. Gastroenterology 1989; 96:790–794 221. Post AB, Bozdech JM, Lavery I, Barnes DS. Colectomy in patients with inflammatory bowel disease and primary sclerosing cholangitis. Dis Colon Rectum 1994; 37:175–178. 222. Mikkola K, Kiviluoto T, Riihela M, Taavitsainen M, Jarvinen HJ. Liver involvement and its course in patients operated on for ulcerative colitis. Hepatogastroenterology 1995; 42:68–72. 223. Kartheuser AH, Dozois RR, LaRusso NF, Wiesner RH, Ilstrup DM, Schleck CD. Comparison of surgical treatment of ulcerative colitis associated with primary sclerosing cholangitis: ileal pouchanal anastomosis versus Brooke ileostomy. Mayo Clin Proc 1996; 71:748–756. 224. Penna C, Dozois R, Tremaine W, Sandborn W, LaRusso N, Schleck C, et al. Pouchitis after ileal pouchanal anastomosis for ulcerative colitis occurs with increased frequency in patients with associated primary sclerosing cholangitis. Gut 1996; 38:234–239. 225. Zins BJ, Sandborn WJ, Penna CR, Landers CJ, Targan SR, Tremaine WJ, et al. Pouchitis disease course after orthotopic liver transplantation in patients with primary sclerosing cholangitis and an ileal pouchanal anastomosis. Am J Gastroenterol 1995; 90:2177–2181. 226. Cox KL, Cox KM. Oral vancomycin: treatment of primary sclerosing cholangitis in children with inflammatory bowel disease. J Pediatr Gastroenterol Nutr 1998; 27:580–583. 227. Mistilis SP, Skyring AP, Goulston SJ. Effect of longterm tetracycline therapy, steroid therapy and colectomy in pericholangitis associated with ulcerative colitis. Aust Ann Med 1965; 14:286–294. 228. Wiesner RH, Ludwig J, LaRusso NF, MacCarty RL. Diagnosis and treatment of primary sclerosing cholangitis. Semin Liver Dis 1985; 5:241–253. 229. Angulo P, Bharucha AE, Jorgensen RA, DeSotel CK, Sandborn WJ, Larusso NF, et al. Oral nicotine in treatment of primary sclerosing cholangitis: a pilot study. Dig Dis Sci 1999; 44:602–607. 230. Kaplan MM. Immunosuppressive therapy in primary sclerosing cholangitis. In: Mann M, Stiehl A, Chapman R, Weisner RH, eds. Primary Sclerosis Cholangitis. Freiburg, Germany: Kluwer, 1998, pp 103–107. 231. Myers RN, Cooper JH, Padis N. Primary sclerosing cholangitis: complete gross and histologic reversal after longterm steroid therapy. Am J Gastroenterol 1970; 53:527–538. 232. Wagner A. Azathioprine treatment in primary sclerosing cholangitis. Lancet 1971; 2: 663–664. 233. LaRusso NF, Wiesner RH, Ludwig J, MacCarty RL, Beaver SJ, Zinsmeister AR. Prospective trial of penicillamine in primary sclerosing cholangitis. Gastroenterology 1988; 95:1036–1042.
Page 701
234. Lindor KD, Wiesner RH, Colwell LJ, Steiner B, Beaver S, LaRusso NF. The combination of prednisone and colchicine in patients with primary sclerosing cholangitis. Am J Gastroenterol 1991; 86:57–61. 235. Kaplan MM, Arora S, Pincus SH. Primary sclerosing cholangitis and lowdose oral pulse methotrexate therapy: clinical and histologic response. Ann Intern Med 1987; 106:231–235. 236. Knox TA, Kaplan MM. A doubleblind controlled trial of oralpulse methotrexate therapy in the treatment of primary sclerosing cholangitis (see comments). Gastroenterology 1994; 106:494–499. 237. Knox TA, Kaplan MM. Treatment of primary sclerosing cholangitis with oral methotrexate. Am J Gastroenterol 1991; 86:546–552. 238. Duerksen DR, BlondelHill E, Bailey RJ. Pneumocystis carinii pneumonia complicating methotrexate treatment of primary sclerosing cholangitis. Am J Gastroenterol 1995; 90:1886–1887. 239. Wiesner RH, Steiner B, LaRusso N, Lindor K, Baldus W. A controlled clinical trial evaluating cycolsporin in the treatment of primary sclerosing cholangitis (abstr). Hepatology 1991; 14:63A. 240. Van Thiel DH, Carroll P, AbuElmagd K, RodriguezRilo H, Irish W, McMichael J, et al. Tacrolimus (FK 506), a treatment for primary sclerosing cholangitis: results of an openlabel preliminary trial. Am J Gastroenterol 1995; 90:455–459. 241. Beuers U, Spengler U, Kruis W, Aydemir U, Wiebecke B, Heldwein W, et al. Ursodeoxycholic acid for treatment of primary sclerosing cholangitis: a placebo controlled trial. Hepatology 1992; 16:707–714. 242. Stiehl A, Walker S, Stiehl L, Rudolph G, Hofmann WJ, Theilmann L. Effect of ursodeoxycholic acid on liver and bile duct disease in primary sclerosing cholangitis: a 3year pilot study with a placebocontrolled study period. J Hepatol 1994; 20:57–64. 243. Lindor KD. Ursodiol for primary sclerosing cholangitis. Mayo Primary Sclerosing CholangitisUrsodeoxycholic Acid Study Group (see comments). N Engl J Med 1997; 336:691–695. 244. De Maria N, Colantoni A, Rosenbloom E, Van Thiel DH. Ursodeoxycholic acid does not improve the clinical course of primary sclerosing cholangitis over a 2 year period. Hepatogastroenterology 1996; 43:1472–1479. 245. Mitchell S, Bansi D, Hunt NFK, Chapman R. High dose ursodeoxycholic acid (UDCA) in primary sclerosis cholangitis (PSC): results after two years of randomised doubleblind, placebocontrolled trial (abstr). Gut 1997; 40(suppl):TH115. 246. Lindor KD, Jorgensen RA, Anderson ML, Gores GJ, Hofmann AF, LaRusso NF. Ursodeoxycholic acid and methotrexate for primary sclerosing cholangitis: a pilot study. Am J Gastroenterol 1996; 91:511–515. 247. Hay JE. Liver transplantation for primary biliary cirrhosis and primary sclerosing cholangitis: does medical treatment alter timing and selection? Liver Transplant Surg 1998; 4:S9–S17. 248. Miki C, Harrison JD, Gunson BK, Buckels JA, McMaster P, Mayer AD. Inflammatory bowel disease in primary sclerosing cholangitis: an analysis of patients undergoing liver transplantation. Br J Surg 1995; 82:1114–1117. 249. Sebagh M, Farges O, Kalil A, Samuel D, Bismuth H, Reynes M. Sclerosing cholangitis following human orthotopic liver transplantation. Am J Surg Pathol 1995; 19:81–90. 250. Harrison RF, Davies MH, Neuberger JM, Hubscher SG. Fibrous and obliterative cholangitis in liver allografts: evidence of recurrent primary sclerosing cholangitis? Hepatology 1994; 20:356–361. 251. Boyer TD. Does primary sclerosing cholangitis recur after liver transplantation? No! Liver Transplant Surg 1997; 3:S24–S25. 252. Haagsma EB, Mulder AH, Gouw AS, Horst G, Meerman L, Slooff MJ, et al. Neutrophil cyctoplasmic autoantibodies after liver transplantation in patients with primary sclerosing cholangitis. J Hepatol 1993; 19:8–14.
Page 702
253. Campbell WL, Sheng R, Zajko AB, AbuElmagd K, Demetris AJ. Intrahepatic biliary strictures after liver transplantation. Radiology 1994; 191:735–740. 254. Sheng R, Campbell WL, Zajko AB, Baron RL. Cholangiographic features of biliary strictures after liver transplantation for primary sclerosing cholangitis: evidence of recurrent disease. AJR 1996; 166:1109–1113. 255. Graziadei IW, Weisner RH, Batts KP, Marotta PJ, LaRusso NF, Porayko MK, Hay JE, Gores GJ, Charlton MR, Ludwig J, Poterucha JJ, Steers JL, Krom RA. Recurrence of primary sclerosing cholangitis following liver transplantation. Hepatology 1999; 29:1050–1056. 256. Shaked A, Colonna JO, Goldstein L, Busuttil RW. The interrelation between sclerosing cholangitis and ulcerative colitis in patients undergoing liver transplantation. Ann Surg 1992; 215:598–603. 257. Gavaler JS, Delemos B, Belle SH, Heyl AE, Tarter RE, Starzl TE, et al. Ulcerative colitis disease activity as subjectively assessed by patientcompleted questionnaires following orthotopic liver transplantation for sclerosing cholangitis. Dig Dis Sci 1991; 36:321–328. 258. Befeler AS, Lissoos TW, Schiano TD, Conjeevaram H, Dasgupta KA, Millis JM, et al. Clinical course and management of inflammatory bowel disease after liver transplantation. Transplantation 1998; 65:393–396. 259. Rowley S, Candinas D, Mayer AD, Buckels JA, McMaster P, Keighley MR. Restorative proctocolectomy and pouch anal anastomosis for ulcerative colitis following orthotopic liver transplantation. Gut 1995; 37:845–847. 260. Stieber AC, Marino IR, Iwatsuki S, Starzl TE. Cholangiocarcinoma in sclerosing cholangitis: the role of liver transplantation. Int Surg 1989; 74:1–3. 261. Cotton PB, Nickl N. Endoscopic and radiologic approaches to therapy in primary sclerosing cholangitis. Semin Liver Dis 1991; 11:40–48 262. Lombard M, Farrant M, Karani J, Westaby D, Williams R. Improving biliaryenteric drainage in primary sclerosing cholangitis: experience with endoscopic methods. Gut 1991; 32:1364–1368. 263. Silvis SE, Nelson DB, Meier PB. Tenyear response to stenting in a patient with primary sclerosing cholangitis. Gastrointest Endosc 1998; 47:83–87. 264. Springer DJ, Gaing AA, Siegel JH. Radiologic regression of primary sclerosing cholangitis following combination therapy with an endoprosthesis and ursodeoxycholic acid. Am J Gastroenterol 1993; 88:1957–1959. 265. van Milligen de Wit AW, Rauws EA, van Bracht J, Mulder CJ, Jones EA, Tytgat GN, et al. Lack of complications following shortterm stent therapy for extrahepatic bile duct strictures in primary sclerosing cholangitis. Gastrointest Endosc 1997; 46:344–347. 266. van Milligen de Wit AW, van Bracht J, Rauws EA, Jones EA, Tytgat GN, Huibregtse K. Endoscopic stent therapy for dominant extrahepatic bile duct strictures in primary sclerosing cholangitis. Gastrointest Endosc 1996; 44:293–299. 267. Hay JE. Osteoporosis. In: Lindor KD, Dickson ER, eds. PBC, PSC, and Adult Cholangiopathies. Philadelphia: Saunders, 1998, pp 407–419. 268. Schrumpf E. Association of primary sclerosing cholangitis and celiac disease: fact or fancy? Hepatology 1989; 10:1020–1021. 269. Venturini I, Cosenza R, Miglioli L, Borghi A, Bagni A, Gandolfo M, et al. Adult celiac disease and primary sclerosing cholangitis: two case reports. Hepatogastroenterology 1998; 45:2344–2347. 270. Volta U, De Franceschi L, Molinaro N, Cassani F, Muratori L, Lenzi M, et al. Frequency and significance of antigliadin and antiendomysial antibodies in autoimmune hepatitis. Dig Dis Sci 1998; 43:2190–2195. 271. Donaldson RMJ. Small bowel bacterial overgrowth. Adv Intern Med 1970; 16:191–212. 272. Drude RBJ, Hines CJ. The pathophysiology of intestinal bacterial overgrowth syndromes. Arch Intern Med 1980; 140:1349–1352. 273. Ghent CN, Carruthers SG. Treatment of pruritus in primary biliary cirrhosis with rifam
Page 703
pin: results of a doubleblind, crossover, randomized trial. Gastroenterology 1988; 94:488–493. 274. Podesta A, Lopez P, Terg R, Villamil F, Flores D, Mastai R, et al. Treatment of pruritus of primary biliary cirrhosis with rifampin. Dig Dis Sci 1991; 36:216– 220. 275. Price TJ, Patterson WK, Olver IN. Rifampicin as treatment for pruritus in malignant cholestasis. Support Care Cancer 1998; 6:533–535. 276. Terra SG, Tsunoda SM. Opioid antagonists in the treatment of pruritus from cholestatic liver disease. Ann Pharmacother 1998; 32:1228–1230. 277. Hoek FJ, Grijm R, Sanders GT, Tytgat GN, Wilmink JM. Removal of bile acids from the blood by hemodialysis with a polyacrylonitril membrane: treatment of pruritus of cholestatic disease. Digestion 1982; 23:135–140. 278. Lauterburg BH, Pineda AA, Dickson ER, Baldus WP, Taswell HF. Plasmaperfusion for the treatment of intractable pruritus of cholestasis. Mayo Clin Proc 1978; 53:403–407. 279. Pitt HA, Postier RG, Cameron JL. Biliary bacteria: significance and alterations after antibiotic therapy. Arch Surg 1982; 117:445–449. 280. Csendes A, Mitru N, Maluenda F, Diaz JC, Burdiles P, Csendes P, et al. Counts of bacteria and pyocites of choledochal bile in controls and in patients with gallstones or common bile duct stones with or without acute cholangitis. Hepatogastroenterology 1996; 43:800–806. 281. Maluenda F, Csendes A, Burdiles P, Diaz J. Bacteriological study of choledochal bile in patients with common bile duct stones, with or without acute suppurative cholangitis. Hepatogastroenterology 1989; 36:132–135. 282. Tateno C, Yoshizato K. Growth and differentiation in culture of clonogenic hepatocytes that express both phenotypes of hepatocytes and biliary epithelial cells. Am J Pathol 1996; 149:1593–1605.
Page 705
32— Vanishing Bile Duct Syndrome P. Aiden McCormick and Niamh Nolan St. Vincent's University Hospital, Dublin, Ireland I— Development of Bile Ducts The intrahepatic bile ducts arise from liver cell plates and, at first, tubular structures known as the ductal plate are formed. These structures surround the mesenchyme around the vessels and, with time, the primitive tube buds off into a series of connecting ducts that form the biliary system. The bile ducts develop from hilum to periphery. The smallest ones are the cholangioles or ductules, which drain the lobules and traverse portal tracts without blood vessels. Interlobular bile ducts (diameter < 100 m) are larger, lined by cuboidal epithelium, and accompanied by hepatic artery and portal vein radicles. Septal bile ducts are greater than 100 m in diameter and most are lined by columnar epithelium. Segmental bile ducts are 400 to 800 m in diameter and lead into the right and left hepatic bile ducts (1,2). The normal number of bile ducts is expressed as a ratio to the associated artery (e.g., 1:1). However, this will vary from tract to tract in the normal liver, with occasionally two bile ducts, or, on occasion, none present. Quantitative reference standards have recently been published (3). In this study there were, on average, 2.3 interlobular bile ducts per portal tract, compared to 2.6 hepatic arteries and 0.7 portal veins. Some 7% of portal tracts did not contain a bile duct and 9% did not contain a hepatic artery (Fig. 1). Bile ducts express antigens, which have a role in the inflammatory process. These include class 1 major histocompatibility complex (MHC) antigens, heatshock proteins VLA2, 3, and 6 (4). The expression of additional antigens may be induced. These molecules present antigen to CD8+ and CD4+ T lymphocytes; they mediate adhesion of inflammatory cells and intracellular reactions. This immunological profile of the bile ducts protects the liver against infection and also serves as a mechanism to induce immunological activity against a donated organ. II— Vanishing Bile Duct Syndrome/Ductopenia/Paucity of Bile Ducts Bile duct paucity refers to a reduction in the number of intrahepatic ducts to less than one per interlobular tract. However, as the number can vary from tract to tract, as shown above, it is important to evaluate a sufficient number of portal tracts. A good rule of thumb is to expect a duct of a diameter comparable to that of the arteriole to be present. Thus, tracts not containing an arteriole are excluded; but if an arteriole is present and the duct is not, this tract may be interpreted as lacking the duct (5). Duct loss is determined by calculating the ratio of the number of bile ducts to hepatic arterioles within each portal tract. It is important to count portal
Page 706
Figure 1 Normal portal tract containing portal vein radicle, hepatic arteriole branch, and a bile duct of equivalent size.
tracts and not the total number of ducts in tracts because ductular proliferation may appear to increase the number of ducts (6). However, The diagnosis of ductopenia may be made on a small biopsy containing as few as four portal tracts provided that all lack a bile duct (7). Special stains may help in the identification of bile ducts (5). Trichrome stain highlights the biliary epithelium and periodic acid—Schiff (PAS) will identify the basement membrane surrounding the duct. Immunohistochemical methods for staining cytokeratin will identify the biliary epithelium, although distinguishing these from hepatocytes depends on use of the appropriateweight keratins (4,8). In practice, the trichrome stain and cytokeratin markers are the most useful stains for identifying bile duct loss (Fig. 2). The effects of bile duct damage or loss may be reflected elsewhere in the biopsy (1,4). Bile pigment accumulates (bilirubinostasis) and is a feature in the perivenular zone. This is identified by green pigment accumulation in hepatocytes or bile canaliculi and highlighted by van Gieson's stain. Bile acid excretion is impaired, resulting in ''cholate stasis" in the periportal zone. Cholate is soluble and results in swollen hepatocytes, which accumulate copper. This is easily identified by the Shikataorcein stain for copperassociated protein or methods that show copper itself (9). Identification of cholate stasis is a useful diagnostic feature in chronic cholestatic disorders of any etiology (10). Ductular proliferation or bile duct reduplication occurs at the periphery of the portal tracts as a response to damage to the native bile ducts. The ductules appear to originate from preexisting cholangioles and proliferation of stem cells at the margin of the tract (11). This ductular reaction is associated with a polymorphonuclear cell response and fibrosis leading to piecemeal biliary necrosis. Fibrosis originates both from basement membrane surrounding the ductules and from activated mesenchymal cells around the ducts. Such a reaction and fibrosis progress to link portal tracts and biliary cirrhosis eventually occurs (Figs. 3, 4).
Page 707
Figure 2 Cytokeratin immunohistochemical marker Cam 5.2 (Becton Dickinson) highlights biliary epithelium and ductular reaction but also stains hepatocytes.
Figure 3 Prominent ductular reaction in an expanded portal tract with associated fibrosis. Note apparent metaplasia of peripheral cells of hepatocyte parenchyma.
Page 708
Figure 4 Primary biliary cirrhosis. There are irregularly shaped hepatocyte nodules forming a "jigsaw" pattern. Note the peripheral pallor caused by swelling of the hepatocytes due to cholate stasis.
III— Vanishing Bile Duct Syndrome in Liver Transplantation The biliary apparatus is a frequent site of injury in patients following liver transplantation. The bile canaliculus is one of the liver structures most susceptible to ischemia reperfusion injury. Electron microscopy demonstrates dilatation of the biliary canaliculus and marked loss of biliary microvilli in the first 2 weeks after transplantation (12). These changes are associated with biochemical cholestasis and are reversible. In contrast, later immunological injury and loss of bile ducts is usually irreversible and is a serious problem in liver transplantation. This syndrome of chronic (ductopenic) rejection is a frequent cause of graft loss and retransplantation. It usually occurs more than 60 days after transplantation. The pathological hallmarks are loss of bile ducts (Fig. 5) and foamy cell or obliterative arteriopathy (13) (Fig. 6). For practical purposes, the diagnosis is usually made on the basis of loss of bile ducts, as arterioles of sufficient size to manifest arteriopathy are rarely seen in needle liver biopsies. Ductopenia in this setting is defined as loss of bile ducts involving more than 50% of portal tracts. Vanishing bile duct syndrome may be defined as absence of bile ducts associated with severe canalicular cholestasis in a chronically cholestatic patient on adequate immunosuppression (14). Ideally, 20 portal tracts should be assessed, and each tract should contain a hepatic arteriole with an accompanying bile duct. It is unusual to have 20 portal tracts in a needle biopsy; therefore sequential liver biopsies may have to be assessed. If ductopenia is profound, the diagnosis may be made after examination of fewer portal tracts (7). In the early stages, ductopenia may be mild or evolving and further liver biopsies after an appropriate interval may be required to clarify matters. Although large arterioles are rarely seen, the effects of arteriopathy may be seen in the perivenular zone with ischemic cell dropout, hemorrhage, and later hemosiderin deposition. Cholestasis is also seen in this zone. Hepatocytes may be ballooned and sinusoidal foamy macrophages may be present. However, protracted disease does not result in either a ductular reaction or biliary cirrhosis, in contrast to chronic primary biliary cirrhosis or primary sclerosing cholangitis (4,14) (Figs. 7 and 8).
Page 709
Figure 5 Portal tract without a bile duct. There is an obvious hepatic arteriole and a normal portal vein radicle. Note the characteristic "empty," noninflamed appearance of the tract, which contrasts with the cellrich appearance of a tract in acute cellular rejection.
Figure 6 Foamcell arteriopathy of chronic rejection. The lumen of the arteriole is almost completely occluded by the foamy cells. The bile duct cannot be identified in this tract. The finding of such an affected arteriole on needle biopsy is unusual.
Page 710
Figure 7 Perivenular congestion, hemorrhage, and hepatic cell dropout. These features all point to an ischemic etiology, in this case due to foamcell arteriopathy in larger vessels.
Figure 8 Sinusoidal macrophages, associated with foam cell arteriopathy. If these cells are seen in the correct clinical setting with supportive histological features, chronic rejection is likely.
Page 711
A— Clinical Course Chronic ductopenic rejection is usually preceded by acute rejection. Typically a patient may have a number of episodes of acute cellular rejection that respond incompletely to standard immunosuppression (15). Two histological and clinical patterns have been described. In one there is evidence of ductopenia with minimal portal tract inflammation followed by progressive cholestasis. A second pattern involves progressive ischemic injury of the centrilobular hepatocytes with fibrosis and a more rapid evolution to liver failure (16). Most patients develop features of chronic rejection within a year of transplantation. In one series of 22 patients with chronic rejection, the onset was less than 4 weeks in 4; 6 to 26 weeks in 13; and greater than 6 months in 5 (17). B— Reversible Ductopenia Occasionally ductopenic rejection may be reversible. Freese et al. described two children whose loss of 55 and 75% of bile ducts, respectively, resolved at 12 and 9 months (16). The return of interlobular bile ducts was preceded by bile ductular proliferation and some mild fibrosis. Hubscher et al. also described reversible ductopenia following liver transplantation (18). Blakolmer et al. described 10 patients with a diagnosis of chronic rejection who recovered to normal histology or biliary tests (19). A number of these patients had less than 50% bile duct loss. The investigators suggested that greater than 50% bile duct loss, severe central fibrosis, and the presence of foamcell clusters were associated with irreversible chronic rejection. C— Mechanism of Chronic Ductopenic Rejection The precise immunological mechanisms underlying ductopenia rejection are not clear. A central role has been ascribed to a cellular immune response to MHC donor antigens. In the liver, these antigens are expressed mainly on biliary epithelium and vascular endothelium, which are the principal sites of injury in chronic rejection (20). A role for a humoral component has also been suggested by the finding of antitissue antibodies in 71% of patients developing chronic rejection. These antibodies were directed against smooth muscle or the nucleus (21). D— Risk Factors for Chronic Rejection The incidence of chronic ductopenic rejection (Table 1) after liver transplantation varies between 5 and 20%. Patients retransplanted for chronic rejection have an increased risk of chronic rejection in the second graft, 21 versus 5.2% in one series (22). Whether the primary liver disease influences the chance of chronic rejection is controversial. In a series of 423 consecutive primary liver transplants in the United Kingdom, chronic rejection was more common in patients transplanted for primary biliary cirrhosis or autoimmune chronic active hepatitis (22). However a North American series of 768 primary liver transplantations found that the risk of chronic rejection was similar for patients with primary biliary cirrhosis, primary sclerosing cholangitis, alcoholic liver disease, and hepatitis C (17). Another North American study suggested that primary sclerosing cholangitis was a risk factor for chronic rejection (15). Racial mismatch between donor and recipient is an important factor in chronic rejection. In a study from the United Kingdom, nonEuropean recipients developed chronic rejection at over twice the rate of European recipients (12.6 versus 5.9%) (23). Similar findings have been noted in patients who received renal transplants. Sex mismatch is more controversial. In one study, transplantation of a male liver into a female recipient was associated with an increased risk of chronic rejection (22). On the other hand, a large study of 1138 liver transplants found no gender bias in the incidence of chronic rejection but reported a decreased graft survival rate in male recipients of female livers (24).
Page 712 Table 1 Possible Risk Factors for Chronic Ductopenic Rejection Recurrent or poorly responsive acute rejection Retransplantation for chronic rejection Cytomegalovirus infection Racial mismatch Centrilobular necrosis on biopsy Etiology PBC or autoimmune chronic active hepatitis Sex mismatch
It is intuitively obvious that antigenic mismatch between donor and recipient must have a central role in development of chronic ductopenic rejection. However, it has proven very difficult to define the clinically relevant antigens. Currently liver grafts are matched for ABO blood group types. ABO mismatch usually produces rapid graft loss due to primary humoral rejection with hemorrhagic infiltration of the liver rather than chronic rejection (25). A number of studies have looked at the role of HLA mismatch, but clearcut associations have not been found. It was suggested that cytomegalovirus (CMV) infection in patients with HLA DR matching was associated with a higher incidence of chronic rejection (26), but this has not been confirmed by subsequent studies (22). There does appear to be some relationship between CMV and chronic rejection. In one study of 200 consecutive liver transplants, all of the 10 patients who developed chronic rejection had a history of CMV infection. Nine grafts were examined, and persistent CMV DNA was found in remaining bile ducts and moderately expressed in endothelial cells of vascular structures at the time of retransplantation (27). If CMV infection is important, the successful prophylaxis of CMV infection with oral ganciclovir should reduce the incidence of chronic rejection. Acute rejection is probably a risk factor for the development of chronic ductopenic rejection. This particularly applies to the entity described as late cellular rejection. If this is defined as acute cellular rejection occurring more than 30 days after liver transplantation, then 27% of such patients may progress to chronic ductopenic rejection (28). The associated histological features of centrilobular necrosis or moderate ductopenia carry 50 and 80% risks of progressing to chronic rejection when seen in the context of late acute rejection. Centrilobular necrosis has similarly ominous implications in pediatric liver transplantation (29). The incidence of chronic rejection appears to be falling. This may be due to better immunosuppressive regimens and more effective treatment of acute rejection episodes. The addition of azathioprine to cyclosporine and steroids appears to reduce the incidence of ductopenic rejection in cyclosporinebased regimens (15). Maintaining adequate levels of immunosuppression in the early posttransplant period may be important, particularly for cyclosporine (30). The increasing use of tacrolimus as primary or rescue therapy is also probably contributing to a reduction in chronic rejection. E— Treatment of Chronic Rejection Tacrolimus appears to be more efficacious in preventing chronic ductopenic rejection than cyclosporine. In the European multicenter study, 1.4% of patients treated with tacrolimus developed chronic rejection, compared to 6.4% treated with cyclosporine (31). Side effects requiring withdrawal of treatment are more common in patients treated with tacrolimus, 14 versus 5% (32). For this reason many physicians treat initially with a cyclosporinebased regimen and change to tacrolimus if there is a suspicion of chronic rejection. Tacrolimus may be particularly effective in patients with acute persistent rejection with less than 50% bile duct loss (33). It would appear prudent to change to tacrolimus rescue therapy early rather than later. In a large
Page 713
series from 17 transplant centers in the United States, 91 patients were converted to tacrolimus for chronic rejection. A bilirubin level greater than 10 mg/dL was an adverse prognostic factor. Patients converted later than 90 days after transplant had a better survival that those converted earlier, with a 1year survival of 88 versus 66% (34). Established chronic rejection with greater than 50% bile duct loss is believed to be irreversible in most cases, but occasional reversibility is described (18). IV— Ischemic Cholangiopathy Interruption of the hepatic arterial blood supply may occur because of surgical injury, hepatic artery thrombosis, or chemotherapeutic embolization and result in the development of biliary strictures. These may be seen radiologically as multiple segmental strictures and dilatations. The main pathological findings are in the extrahepatic and larger intrahepatic ducts with segmental necrosis and inflammation. The portal tracts show standard features of largeduct obstruction, with edema, inflammation, ductular reaction, and cholestasis. Bile duct damage and ductopenia may also be seen (35,36). Nonanastomotic biliary strictures may also be seen in 2 to 19% patients following liver transplantation (37). They are believed to be due to ischemic damage to the biliary tree and may also be seen in patients with hepatic artery thrombosis or ABOincompatible liver grafts (38). It has been suggested that surgical techniques to reduce the period of warm ischemia experienced by the biliary apparatus may reduce the incidence of this complication (39). Ischemic cholangitis may occur in patients with vasculitis, polyarteritis nodosa, and giantcell temporal arteritis, with the development of biliary ductopenia on liver biopsy (35). V— Graft versus Host Disease This condition is frequent after bone marrow transplantation but rare in liver transplantation. It may also occur occasionally in immunosuppressed patients following blood transfusion. Acute graftversushost disease (GVHD) is manifest by a maculopapular skin rash, abdominal pain, and abnormal liver function tests. The mechanism is the presence of immunologically competent donor T cells reacting against host epithelium. In liver transplantation, passenger T lymphocytes in the donor organ are believed to be responsible. Histologically, there is damage to biliary epithelium with vacuolation, eosinophilia of cytoplasm, and eventual destruction and loss of the bile ducts (40). In chronic GVHD, the development of extensive portal fibrosis and cirrhosis has been described (41). Chronic GVHD has many similarities with primary biliary cirrhosis. Mononuclear portal inflammation with destruction of interlobular and septal bile ducts is seen in both conditions (42) (Fig. 9). Hallmarks of primary biliary cirrhosis, such as noncaseating granulomas and antimitochondrial antibodies, may also be seen in chronic GVHD. Ursodeoxycholic acid may be useful in chronic GVHD involving the liver (43). GVHD has been described following liver transplantation, but typically the liver is spared in this situation (44). However, the prognosis is poor, with most patients dying of the disease or septic complications. Successful treatment with steroids and antithymocyte globulin has been described (45). VI— Adult Idiopathic Ductopenia Adult idiopathic ductopenia was first described as a distinct entity by Ludwig et al. in 1988 (46). They reported three adult patients with chronic cholestatic liver disease and loss of interlobular bile ducts. All patients met the criteria for ductopenia, with bile ducts identifiable in
Page 714
Figure 9 Chronic graftversushost disease in a bone marrow transplant recipient. Note bile duct loss and fibrosis of the portal tract.
less than 50% of portal tracts. Other causes of bile duct loss were excluded and cholangiography was normal. This was a severe progressive disease and resulted in liver transplantation in two of the patients. Age of onset was the principal feature separating this condition from neonatal or infantile nonsyndromic paucity of bile ducts. The criteria for diagnosis included onset of cholestasis in late adolescence or later, ductopenia, normal cholangiogram, and no other known cause for cholestasis. The authors also suggested that the diagnosis should not be made in patients with inflammatory bowel disease because of the difficulty in excluding smallduct primary sclerosing cholangitis. Other reports soon followed, suggesting that idiopathic adulthood ductopenia (IAD) was a distinct syndrome (47,48). Brugera and colleagues reviewed all cases of chronic cholestasis presenting to their liver unit in Barcelona over an 8year period (48). Of 179 patients, 160 were found to have primary biliary cirrhosis, 8 had primary sclerosing cholangitis, 3 had Alagille's syndrome, and 8 were diagnosed as having adult idiopathic ductopenia. Thus it is a relatively rare condition, representing approximately 5% of patients with chronic cholestasis sent to a referral unit. The most common presenting symptoms of idiopathic adulthood ductopenia are jaundice and pruritus (48). Some patients are picked up on the basis of abnormal liver function tests. Approximately 30% of the cases described have had evidence of portal hypertension at presentation, and bleeding from esophageal varices was the initial manifestation in some cases. There appears to be a familial element in approximately 20% of cases (48). Progression to hepatic failure, death, or liver transplantation appears to be common. Individual patients have been treated with penicillamine or ursodeoxycholic acid but without proven efficacy. A mild, nonprogressive form of idiopathic biliary ductopenia has also been described in one report of 24 patients (49). These patients were asymptomatic and were picked up on the basis of abnormal liver function tests. All had increased serum gammaglutamyltransferase levels; alanine aminotransferase and alkaline phosphatase levels were increased in 75 and 54%, respectively. Ductopenia was mild, with bile ducts being present in 62 ± 7% of portal tracts. Thus these patients would not fulfill the standard criteria for diagnosis of bile duct paucity.
Page 715
None of these patients had symptoms of liver disease, such as pruritus. In addition, they were older than most patients with idiopathic ductopenia, with a mean age at diagnosis of 41 years (range 27 to 57). Repeat liver biopsies were available in three patients 3, 6, and 9 years after the first, which showed no evidence of progression. Five patients were treated with ursodeoxycholic acid and liver function tests returned to normal in four. Thus there may be a spectrum of severity, with some patients having mild, nonprogressive bile duct paucity. The degree of bile duct paucity appears to have major prognostic implications. It is recommended that at least 20 portal tracts be examined before a definite diagnosis of ductopenia is made. This may require repeated liver biopsy. VII— Hepatic Sarcoidosis Sarcoidosis is a multisystem granulomatous disease of unknown etiology. Noncaseating granulomas are found in the liver in approximately twothirds of patients with sarcoidosis and frequently have characteristic inclusions such as Schaumann and asteroid bodies (50). They rarely cause symptoms, although there may be minor abnormalities in liver function tests. Devaney and colleagues described the patterns of liver injury in 100 patients with hepatic sarcoidosis collected over 15 years at the Armed Forces Institute of Pathology (51). Noncaseating granulomas were present in all cases. Fibrosis was seen in 21% and frank cirrhosis in 6%. Cholestasis was noted in 58 patients, and 37 of these had evidence of biliary ductopenia. The histological features resembled primary biliary cirrhosis in 19 and primary sclerosing cholangitis in 13. Sarcoidosis may also cause noncirrhotic portal hypertension (52). Cholestatic sarcoidosis appears to be more common in males than females and in West Indian and North American black patients (53). Anecdotal reports suggest that steroid therapy does not prevent progression to cirrhosis. On occasion it may be difficult to distinguish sarcoidosis from primary biliary cirrhosis (53), particularly as a lowlevel alveolitis resembling sarcoidosis has recently been described in patients with primary biliary cirrhosis (54). Antimitochondrial antibodies and a Kveim test may help in distinguishing these two idiopathic granulomatous diseases (53) (Fig. 10a and b). VIII— Ductopenia in Infancy and Childhood Two syndromes of idiopathic ductopenia have been described in infancy and childhood—syndromic and nonsyndromic biliary hypoplasia. Syndromic biliary hypoplasia is also known as Alagille's syndrome of arteriohepatic dysplasia (55,56) (Fig. 11). It is an autosomaldominant disorder caused by mutations on the human Jagged 1 gene on chromosome 20p12 (57). Most patients present with jaundice in the neonatal period, followed by symptomatic pruritus. Fat malabsorption and failure to thrive may be a problem. Extrahepatic manifestations—including cardiac, skeletal and ocular abnormalities—are helpful in making the diagnosis. The most common cardiac abnormality is peripheral pulmonary artery stenosis. Fusion of the vertebral bodies may give rise to the appearance of "butterfly vertebrae." Arteriohepatic dysplasia appears to be a relatively benign condition. Jaundice usually remits after 5 years of age, although the liver function tests remain cholestatic. Approximately 20% of these children ultimately develop cirrhosis. Nonsyndromic biliary hypoplasia is less common. It has been associated with a number of other conditions, including alpha1antitrypsin deficiency, Down's syndrome, and cystic fibrosis. The prognosis may be worse than that for Alagille's syndrome, particularly if there is associated alpha1antitrypsin deficiency or a familial component. It is suggested that the histological features may differ with established ductopenia, portal fibrosis, and expanded portal tracts present in the nonsyndromic form before 90 days of age (58). Electron microscopy
Page 716
Figure 10 Sarcoidosis of liver on needle biopsy showing ductopenia (a) and noncaseating granuloma (b).
features may also differ, with canalicular dilatation and reduction in numbers of microvilli in the nonsyndromic form and nondilated canaliculi in the syndromic form. Langerhans cell histiocytosis or histiocytosis X is a rare immunological disorder that can cause liver infiltration and biliary complications in children. The appearances can mimic sclerosing cholangitis and may progress to fibrosis and cirrhosis. Liver transplantation has been successful in patients with endstage liver disease in this condition, and survival for up to 7 years without recurrence has been recorded (59).
Page 717
Figure 11 Alagille's syndrome. Note lack of bile duct, several vascular channels, and mild portal tract fibrosis.
IX— DrugInduced Ductopenia Many drugs have been reported to cause cholestatic hepatitis (60). In most cases this is a selflimiting condition with rapid resolution. Occasional patients may develop chronic cholestasis; of these, some have histological features of vanishing bile duct syndrome. The degree of ductopenia may be directly related to the degree of acute cholangitis in the early episode. About 7% of cases of chlorpromazinerelated cholestasis persist for more than 3 months. The clinical picture may resemble that of primary biliary cirrhosis, although antimitochondrial antibodies are usually absent. Progression to severe fibrosis and cirrhosis has also been reported with chlorpromazine, tolbutamide, thiabendazole, methyltestosterone, and flucloxacillin (61,62). Vanishing bile duct syndrome has also been reported as a rare complication of treatment with a number of antibiotics, including ampicillin, amoxicillin, flucloxacillin, erythromycin, tetracycline, doxycycline, and cotrimoxazole (61). Flucloxacillin may cause a particularly prolonged and severe cholestatic syndrome. Ductopenia has been reported within 3 weeks of the start of the illness. An immunopathological mechanism is likely, as circulating antibodies to flucloxacillinaltered liver proteins have recently been described in affected patients. The authors have seen a patient with cotrimoxazoleinduced ductopenia. The patient declined a second liver biopsy, but abnormalities on liver function tests improved over 2 years. This appears to be a common experience with druginduced ductopenia, with eventual resolution in most cases. Druginduced vanishing bile duct syndrome is rarely seen in children. There is one case report of a 9yearold girl who developed StevensJohnson syndrome and subsequent vanishing bile duct syndrome after ingesting ibuprofen (63). X— Paraneoplastic Bile Duct Paucity Biochemical cholestasis is frequently seen in patients with Hodgkin's disease. A number of patients with severe cholestasis and paucity of bile ducts have recently been described (64,65).
Page 718
Figure 12 Vanishing bile duct syndrome associated with Hodgkin's disease. In this case cholestasis and ductopenia preceded the diagnosis and treatment of Hodgkin's disease.
In three out of four patients, jaundice was the presenting feature. In one case, successful chemotherapy for the Hodgkin's disease was associated with resolution of cholestasis and reappearance of bile ducts (65) (Fig. 12). XI— Primary Biliary Cirrhosis Primary biliary cirrhosis (PBC) is a disease of presumed autoimmune etiology, where the interlobular and septal bile ducts are progressively destroyed (66). Some 90% of patients are female and the disease usually presents in middle life with pruritus, abnormal liver function tests, and progressive cholestasis. Circulating antimitochondrial antibodies are found in most patients. The M2 subtype antimitochondrial antibody is very specific for primary biliary cirrhosis and is useful in diagnosis. M2 antibodies react with components of the pyruvate dehydrogenase complex of mitochondrial enzymes. It is not clear how antibodies to an intracellular antigen are related to the pathogenesis of this condition. However, expression of the E2 component of the pyruvate dehydrogenase complex has been demonstrated on the luminal surface of biliary epithelium in patients with primary biliary cirrhosis. This finding is specific for patients with primary biliary cirrhosis and may have pathophysiological relevance (67). Patients with primary biliary cirrhosis frequently have other presumed autoimmune conditions, including thyroid disease, sicca syndrome, CRST syndrome, renal tubular acidosis, etc. A large number of drug treatments have been tried in PBC. The only one that has been clearly shown to improve prognosis is ursodeoxycholic acid (68). Trials of ursodeoxycholic acid in combination with other therapies are currently under way. Liver transplantation is effective for patients with endstage liver disease. Because of the progressive nature of PBC, predictive models have been developed to aid in choosing the optimal time for liver transplantation. The best known is the Mayo Model, which is also predictive in patients taking ursodeoxycholic acid
Page 719
(69). Following liver transplantation, antimitochondrial antibodies usually remain positive, although the titers may fall. Recurrent primary biliary cirrhosis has been described in the new liver in up to 8.7% of patients (21). There is a suggestion that recurrent PBC may be more frequent in patients receiving tacrolimus compared to cyclosporine (70). However, it may be difficult to distinguish recurrent PBC from other transplantrelated pathologies—such as chronic rejection, hepatitis C, or nonspecific hepatitis—and the true incidence of recurrent disease is debated. Most studies to date suggest that recurrent PBC rarely causes severe liver dysfunction or graft loss. XII— Autoimmune Cholangitis Approximately 5 to 10% of patients with otherwise typical primary biliary cirrhosis do not have circulating antimitochondrial antibodies. It has been suggested that some of these patients may have a distinct clinical condition termed autoimmune cholangitis. This condition is characterized by cholestatic liver function tests, antinuclear or anti—smooth muscle antibodies and a favorable response to steroids (71). However, further studies suggest that patients with and without antimitochondrial antibodies have a similar clinical course and response to treatment and do not support the concept of a distinct clinical entity (72). XIII— Primary Sclerosing Cholangitis Primary sclerosing cholangitis affects the large bile ducts and is usually diagnosed by cholangiography. Histological changes of biliary ductopenia may be seen on biopsy. This condition is discussed in detail elsewhere in this book. References 1. MacSween RNM, Burt AD. Pathology of the intrahepatic bile ducts. In: Anthony PP, MacSween RNM, eds. Recent Advances in Pathology 14. New York: Churchill Livingstone, 1989, pp 161–183. 2. Gerber MA, Thung SN. Histology of the liver. Am J Surg Pathol 1987; 11:709–722. 3. Crawford AR, Lin X, Crawford JM. The normal adult human liver biopsy: a quantitative reference standard. Hepatology 1998; 28:323–331. 4. Demetris AJ. Immune cholangitis: liver allograft rejection and graftversushost disease. Mayo Clin Proc 1998; 73:367–379. 5. Snover D. Biopsy in the evaluation of neonatal cholastasis. In: Snover D, ed. Biopsy of Liver Disease. Baltimore: Williams & Wilkins, 1992, pp 38–51. 6. Ludwig J, Wiesner RH, Batts KP, Perkins JD, Krom RAF. The acute vanishing bile duct syndrome (acute irreversible rejection) after orthotopic liver transplantation. Hepatology 1987; 7:476. 7. Goldin RD. Diagnosis of biliary ductopenia. Histopathology 1997; 31:397–399. 8. Von Eyken P, Desmet VJ. Cytokeratins and the liver. Liver 1993; 13:113–122. 9. Jain S, Scheuer PJ, Archer B, Newman SP, Sherlock S. Histological demonstration of copper and copperassociated protein in chronic liver diseases. J Clin Pathol 1978; 31:784–790. 10. Harrison RF, Hubscher SC. The spectrum of bile duct lesions in evolving primary sclerosing cholangitis. Histopathology 1991; 19:321–327. 11. Burt AD, MacSween RNM. Bile duct proliferation—its true significance? Histopathology 1993; 23:599–602.
Page 720
12. Cutrin JC, Cantino D, Biasi F, Chiarpotto E, Salizzoni M, Andorno E, Massano G, Lanfranco G, Rizzetto M, Boveris A, Poli G. Reperfusion damage to the bile canaliculi in transplanted human liver. Hepatology 1996; 24:1053–1057. 13. International Working Party. Terminology for hepatic allograft rejection. Hepatology 1995; 22:648–654. 14. Snover D. The liver biopsy in transplantation. In: Snover D, ed. Biopsy of Liver Disease. Baltimore: Williams & Wilkins, 1992:217–231. 15. Van Hoek B, Wiesner RH, Krom RAF, Ludwig J, Moore SB. Severe ductopenic rejection following liver transplantation: incidence, time of onset, risk factors, treatment, and outcome. Semin Liver Dis 1992; 12:41. 16. Freese D, Snover DC, Sharp H, Gross CR, Savick SK, Payne WD. Chronic rejection after liver transplant: a study of the clinical, histopathologic and immunologic features. Hepatology 1991; 13:882–891. 17. Farges O, Saliba F, Farhamant H, Samuel D, Bismuth A, Reynes M, Bismuth H. Incidence of rejection and infection after liver transplantation as a function of the primary disease: possible influence of alcohol and polyclonal immunoglobulins. Hepatology 1996; 23:240–248. 18. Hubscher SG, Neuberger JM, Buckels JAC, Elias E, McMaster P. Vanishing bile duct syndrome after liver transplantation—is it reversible? Transplantation 1991; 51:1004–1110. 19. Blakolmer K, Seaberg EC, Batts K, Ferrell L, Markin R, Wiesner RH, Demetris AJ. Reversible and irreversible chronic allograft rejection of the human liver. Hepatology 1998; 28:353A. 20. Oguma S, Belle S, Starzl TE, Demetris AJ. A histometric analysis of chronically rejected human liver allografts: insights into the mechanisms of bile duct loss: direct immunologic and ischemic factors. Hepatology 1989; 9:204–209. 21. Dubel L, Farges O, Johanet C, Sebagh M, Bismuth H. High incidence of antitissue antibodies in patients experiencing chronic liver allograft rejection. Transplantation 1998; 65:1072–1075. 22. Candinas D, Gunson BK, Nightingale P, Hubscher S, McMaster P, Neuberger JM. Sex mismatch as a risk factor for chronic rejection of liver allografts. Lancet 1995; 346:1117–1121. 23. Devlin JJ, O'Grady JG, Tan KC, Calne RY, Williams R. Ethnic variations in patient and graft survival after liver transplantation: identification of a new risk factor for chronic allograft rejection. Transplantation 1993; 56:1381–1384. 24. Brooks BK, Levy MF, Jennings LW, Abbasoglu O, Vodapally M, Goldstein RM, Husberg BH, Gonwa TA, Klintmalm GB. Influence of donor and recipient gender on the outcome of liver transplantation. Transplantation 1996; 62:1784–1787. 25. Gugenheim J, Samuel D, Reynes M, Bismuth H. Liver transplantation across ABO blood group barriers. Lancet 1990; 336:519–523. 26. Donaldson PT, Alexander GJM, O'Grady J, Neuberger J, Portman B, Thick M, Davis H, et al. Evidence of an immune response to HLA class 1 antigens in the vanishing bile duct syndrome after liver transplantation. Lancet 1987; 1:945–948. 27. Lauterschlager I, Hockerstedt K, Jalanko H, Loginov R, Salmela K, Taskinen E, Ahonen J. Persistent cytomegalovirus in liver allografts with chronic rejection. Hepatology 1997; 25:190–194. 28. Anand AC, Hubscher SG, Gunson BK, McMaster P, Neuberger JM. Timing, significance, and prognosis of late acute liver allograft rejection. Transplantation 1995; 60:1098–1103. 29. Allen KJ, Rand LB, Hart J, Whitington PF. Prognostic implications of centrilobular necrosis in pediatric liver transplant recipients. Transplantation 1998; 65:692– 698. 30. Soin AS, Rasmusen A, Jamieson NV, et al. Cyclosporine levels in the early posttransplant period: predictive of chronic rejection in liver transplantation? Transplant Proc 1995; 27:1129–1130.
Page 721
31. European FK506 Multicentre Liver Study Group. Randomised trial comparing tacrolimus (FK506) and cyclosporin in prevention of liver allograft rejection. Lancet 1994; 344: 423–428. 32. The U.S. Multicenter FK506 Liver Study Group. A comparison of tacrolimus (FK506) and cyclosporine for immunosuppression in liver transplantation. N Engl J Med 1994; 331:1110–1115. 33. Samuel D, Ladouzi M, Reynes M, Bismuth H. Tacrolimus rescue therapy for rejection in liver transplantation: experience in 104 adult patients. Hepatology 1997; 26:235A. 34. Sher LS, Cosenza CA, Michel J, Makowka L, Miller CM, Schwartz ME, Busuttil R, et al. Efficacy of tacrolimus as rescue therapy for chronic rejection in orthotopic liver transplantation: a report of the US multicenter liver study group. Transplantation 1997; 64: 258–263. 35. Batts K. Ischemic cholangitis. Mayo Clin Proc 1998; 73:380–385. 36. Ludwig J, Kin CH, Wiesner RH, Krom RAF. Floxuridineinduced sclerosing cholangitis: an ischemic cholangiopathy? Hepatology 1989; 9:215–218. 37. Fisher A, Miller CH. Ischemictype biliary strictures in allografts; the Achilles heel revisited? Hepatology 1995; 21:589–591. 38. SanchezUrdazpal L, Gores GJ, Ward EM, Maus TP, Wahlstrom E, Moore SB, Wiesner RH, Krom RAF. Ischemictype biliary complications after orthotopic liver transplantation. Hepatology 1992; 16:49–53. 39. Sankary HN, McChesney L, Frye E, Cohn S, Foster P, Williams J. A simple modification in operative technique can reduce the incidence of nonanastomotic biliary strictures after orthotopic liver transplant. Hepatology 1995; 21:63–69. 40. Shulman HM, Sharma P, Amos D, Fenster LF, McDonald GB. A coded histologic study of hepatic graftversushost disease after human bone marrow transplantation. Hepatology 1988; 8:463–470. 41. Knapp AB, Crawford JM, Rappeport JM, Gollan JL. Cirrhosis as a consequence of graftversushost disease. Gastroenterology 1987; 92:513–519. 42. Czaja AJ. Chronic graftversushost disease and primary biliary cirrhosis: sorting the puzzle pieces. Lab Invest 1994; 70:589–592. 43. Fried RH, Murakami CS, Fisher LD, Wilson RA, Sullivan KM, McDonald GB. Ursodeoxycholic acid treatment of refractory chronic graftversushost disease of the liver. Ann Intern Med 1992; 116:624–629. 44. Roberts JP, Ascher NL, Lake J, Capper J, Purohit S, Garovoy M, Lynch R, Ferrell L, Wright T. Graft vs host disease after liver transplantation in humans: a report of four cases. Hepatology 1991; 14:274–281. 45. Burdick JF, Vogelsang GB, Smith WJ, Farmer ER, Bias WB, Kaufmann SH, Horn J, Colombani PM, Pitt HA, Perler BA, Merritt WT, Williams GM, Boitnott JK, Herlong HF. Severe graftversushost disease in a livertransplant recipient. N Engl J Med 1988; 318:689–691. 46. Ludwig J, Wiesner RH, La Russo NF. Idiopathic adulthood ductopenia; a cause of cholestatic liver disease and biliary cirrhosis. J Hepatol 1988; 7:193–199. 47. Zafrani ES, Metreau JM, Douvin C, Larrey D, Massari R, Reynes M, Benhamou JP, Dhumeaux D. Idiopathic biliary ductopenia in adults: a report of five cases. Gastroenterology 1990; 99:1823–1828. 48. Bruguera M, Llach J, Rodes J. Nonsyndromic paucity of intrahepatic bile ducte in infancy and idiopathic ductopenia in adulthood: the same syndrome? Hepatology 1992; 15:830–834. 49. Moreno A, Carreno V, Cano A, Gonzalez C. Idiopathic biliary ductopenia in adults without symptoms of liver disease. N Engl J Med 1997; 336:835–838. 50. Ishak KG. Sarcoidosis of the liver and bile ducts. Mayo Clin Proc 1998; 73:467–472. 51. Devaney K, Goodman ZD, Epstein MS, Zimmerman HJ, Ishak KG. Hepatic sarcoidosis; clinicopathologic features in 100 patients. Am J Surg Pathol 1993; 17:1272–1280.
Page 722
52. Valla D, PessegueiroMiranda H, Degott C, Lebrec D, Rueff B, Benhamou JP. Hepatic sarcoidosis with portal hypertension; a report of seven cases with a review of the literature. Q J Med 1987; 63:531–544. 53. Bass NM, Burroughs AK, Scheuer PJ, James DG. Chronic intrahepatic cholestasis due to sarcoidosis. Gut 1982; 23:417–421. 54. Spiteri MA, Johnson M, Epstein O, Sherlock S, Clarke SW, Poulter LW. Immunological features of lung lavage cells from patients with primary biliary cirrhosis may reflect those seen in pulmonary sarcoidosis. Gut 1990; 31:208–212. 55. Alagille D, Odievre M, Gautier M, Dommergues JP. Hepatic ductular hypoplasia associated with characteristic facies, vertebral malformations, retarded physical, mental and sexual development, and cardiac murmur. J Pediatr 1975; 86:63–71. 56. Riely CA, Cotlier E, Jensen PS, Klatskin G. Arteriohepatic dysplasia: a benign syndrome of intrahepatic cholestasis with multiple organ involvement. Ann Intern Med 1979; 91: 520–527. 57. Oda T, Elkahloun AG, Pike BL, Okajima K, Krantz ID, Genin A, Piccoli DA, Meltzer PS, Spinner NB, Collins FS, Chandrasekharappa SC. Mutations in the human Jaggedl gene are responsible for alagille syndrome. Nature Genet 1997; 16:235–242. 58. Kahn E, Daum F, Markowitz J, Teichberg S, Duffy L, Harper R, Aiges H. Nonsyndromic paucity of interlobular bile ducts: light and electron microscopic evaluation of sequential liver biopsies in early childhood. Hepatology 1986; 6:890–901. 59. Zandi P, Panis Y, Debray D, Bernard O, Houssin D. Pediatric liver transplantation for Langerhans' cell histiocytosis. Hepatology 1995; 21:129–133. 60. Farrell GC. Drug induced cholestasis. In: Farrell GC, ed. Druginduced Liver Disease. Edinburgh: Churchill Livingstone, 1994, pp 319–369. 61. Vial T, Biour M, Descotes J, Trepo C. Antibioticassociated hepatitis: update from 1990. Ann Pharmacother 1997; 31:204–220. 62. Degott C, Feldmann G, Larrey D, DurandSchneider A, Grange D, Machayekhi J, Moreau A, Potet F, Benhamou J. Druginduced prolonged cholestasis in adults: a histological semiquantitative study demonstrating progressive ductopenia. Hepatology 1992; 15:224–251. 63. Srivastava M, PerezAtayde A, Jonas MA. Drugassociated acuteonset vanishing bile duct and StevensJohnson syndromes in a child. Gastroenterology 1998; 115:743–746. 64. Hubscher SG, Lumley MA, Elias E. Vanishing bile duct syndrome: a possible mechanism for intrahepatic cholestasis in Hodgkin's lymphoma. Hepatology 1993; 17:70–77. 65. Crosbie OM, Crown JP, Nolan NPM, Murray R, Hegarty JE. Resolution of paraneoplastic bile duct paucity following successful treatment of Hodgkin's disease. Hepatology 1997; 26:5–8. 66. Neuberger J. Primary biliary cirrhosis. Lancet 1997; 350:375–879. 67. Tsuneyama K, Van de Water J, Leung PSC, Cha S, Nakanuma Y, Kaplan M, DeLellis R, Coppel R, Ansari A, Gershwin ME. Abnormal expression of the E2 component of the pyruvate dehydrogenase complex on the luminal surface of biliary epithelium occurs before major histocompatibility complex class II and BB1/B7 expression. Hepatology 1995; 21:1031–1037. 68. Heathcote EJ, Lindor K, Poupon R, et al. Combined analysis of French, American and Canadian randomized trial of ursodeoxycholic acid therapy in primary biliary cirrhosis (abstr). Gastroenterology 1996; 110:A1082. 69. Kilmurry MR, Heathcote EJ, CauchDudek K, O'Rourke K, Bailey RJ, Blendis LM, Ghent CN, Minuk GY, Pappas C, Scully LJ, Steinbrecher UP, Sutherland LR, Williams CN, Worobetz LJ. Is the Mayo model for predicting survival useful after the introduction of ursodeoxycholic acid treatment for primary biliary cirrhosis? Hepatology 1996; 23: 1148–1153. 70. Dmitrewski J, Hubscher SG, Mayer AD, Neuberger JM. Recurrence of primary biliary cirrhosis in the liver allograft: the effect of immunosuppression. J Hepatol 1996; 24:253–257.
Page 723
71. BenAri Z, Dhillon AP, Sherlock S. Autoimmune cholangiopathy; part of the spectrum of autoimmune chronic active hepatitis. Hepatology 1993; 18:10–15. 72. Invernizzi P, Crosignani A, Battezzati PM, Covini G, De Valle G, Larghi A, Zuin M, Podda M. Comparison of the clinical features and clinical course of anfimitochondrial antibodypositive and negative primary biliary cirrhosis. Hepatology 1997; 25:1090–1095.
Page 725
33— Cholangiocarcinoma Steven A. Ahrendt and Henry A. Pitt Medical College of Wisconsin, Milwaukee, Wisconsin I— Introduction Cholangiocarcinoma is an uncommon tumor that may occur anywhere along the intrahepatic or extrahepatic biliary tree. The risk of cholangiocarcinoma is increased in conditions in which bile is stagnant, infected, or both. These clinical situations include primary sclerosing cholangitis, choledochal cysts, and hepatolithiasis. Cholangiocarcinoma almost always presents with painless jaundice, and this diagnosis should be considered in every case of obstructive jaundice. The diagnostic evaluation, clinical management, and prognosis of a bile duct cancer are in large part determined by the anatomic location; and therefore, cholangiocarcinomas are best classified into three broad anatomic groups: (a) intrahepatic, (b) perihilar, and (c) distal (1). The hepatic duct bifurcation is the most frequently involved site, and approximately 60 to 80% of cholangiocarcinomas encountered at tertiary referral centers are found in the perihilar region (2–3). Altemeier and coworkers first reported three cases of primary adenocarcinoma of the perihilar region in 1957 (4). This initial study emphasized the difficulties involved in the early diagnosis and surgical resection of these lesions. In 1965, Klatskin reported 13 patients with cancers involving the perihilar region. This latter report stimulated interest in this uncommon malignancy, which subsequently has been called Klatskin's tumor (5). Perihilar cholangiocarcinomas also involve major portal vascular structures, making resection difficult. Distal tumors are the second most common and are usually treated like pancreatic cancer with pancreatoduodenectomy. Purely intrahepatic cholangiocarcinomas occur with the lowest frequency and are managed like hepatocellular carcinomas with liver resection. Surgical resection with negative margins is the goal of therapy and, when possible, does offer a chance for longterm diseasefree survival. Unfortunately, many patients will only be candidates for palliative bypass or operative or nonoperative intubation aimed to provide biliary drainage and prevent cholangitis and hepatic failure. The role of chemotherapy and radiation as adjuvants to surgical resection or for palliation remains controversial. II— Incidence Approximately 20,600 new cases of liver and biliary tract cancer are diagnosed annually in the United States (6). Liver and biliary tract malignancies account for more than 16,500 deaths per year in the United States. Between 2500 and 3000 of these cancers are bile duct tumors or cholangiocarcinomas. The incidence of cholangiocarcinoma increases with age, and these tumors occur with similar frequency in men and women. Overall, the incidence of cholangio
Page 726
carcinoma in the United States is approximately 1.0 per 100,000 people per year (2). The annual frequency per 100,000 population is 7.3 in Israelis, 6.5 in American Indians, and 5.5 among the Japanese. In autopsy series, the incidence of cholangiocarcinoma varies from 0.01 to 0.46% (2,7). III— Etiology and Associated Diseases A number of etiological factors have been linked to cholangiocarcinoma (Table 1). Factors common to a number of these etiological factors include stones, biliary stasis, and infection (2,7). One of the more impressive associations between a carcinogen and the development of bile duct malignancies has been documented exposure to the radiocontrast agent Thorotrast. Thorotrast was widely used in the 1930s and 1940s and is retained in the reticuloendothelial system as thorium, which has a biological halflife of 200 to 400 years (2,8). Thoriumrelated cholangiocarcinomas have been diagnosed after a mean latency of 35 years and tend to occur peripherally, often within the intrahepatic biliary tree (8). Chronic exposure to low levels of radionuclides may also be responsible for the high incidence of cholangiocarcinoma observed recently in West Virginia (2). In addition to radionuclides, a number of chemical carcinogens—including asbestos, dioxin, nitrosamines, and polychlorinated biphenyls—have been implicated in the development of cholangiocarcinoma (2). Chronic hepatitis C infection has been linked with the nodular minute form of intrahepatic cholangiocarcinoma (9). Cholangiocarcinoma is also associated with several biliary tract diseases, including primary sclerosing cholangitis (10), choledochal cysts (11), and hepatolithiasis (12). Despite these many associations, the vast majority of American patients with cholangiocarcinoma do not have any obvious risk factors. Patients with sclerosing cholangitis are at increased risk of developing cholangiocarcinoma. Between 10 and 30% of patients undergoing liver transplant have unsuspected cholangiocarcinoma in the hepatectomy specimen (13–14). With liver transplantation and a decrease in the mortality associated with hepatic failure in patients with primary sclerosing cholangitis, cholangiocarcinoma. has become the leading cause of death in patients with this disease (10). A strong association exists between chronic ulcerative colitis and primary sclerosing cholangitis. Between 60 and 80% of all patients with sclerosing cholangitis have coexisting ulcerative colitis (15–16), and the incidence of cholangiocarcinoma may be higher in primary sclerosing cholangitis associated with ulcerative colitis (10). The prevalence of cholangiocarcinoma in ulcerative colitis is 0.2 to 1.4%, which is significantly greater than the risk for the general population. The vast majority if not all patients with ulcerative colitis and cholangiocarcinoma have associated primary sclerosing cholangitis (17). Patients with ulcerative colitis who develop cholangiocarcinoma typically have the cholangiocarcinoma diagnosed in the fifth decade of life, which is approximately 20 years earlier than the mean age at diagnosis in cholangiocarcinoma patients without ulcerative colitis (10). The medical and/or surgical treatment of ul Table 1 Etiological Risk Factors for Cholangiocarcinoma Primary sclerosing cholangitis
Thorium dioxide (Thorotrast)
Ulcerative colitis
Radionuclides
Choledochal cysts
Nitrosamines
Hepatolithiasis
Dioxin
Opisthorchis viverrini
Polychlorinated biphenyls
Clonorchis sinensis
Hepatitis C
Source: Adapted from Ref. 2.
Page 727
cerative colitis does not influence the subsequent development of cholangiocarcinoma, because many patients have had bile duct carcinoma develop years after a total protocolectomy. In a recent series of 25 patients with cholangiocarcinoma complicating sclerosing cholangitis, only 30% of tumors were resected (10). The median survival following the diagnosis of cholangiocarcinoma was only 7 months. However, surgical resection was associated with an improvement in survival. The occurrence of cholangiocarcinoma was not related to the duration or the histological stage of the sclerosing cholangitis. In fact, 48% of patients presented with both diseases simultaneously, and 80% of the cases of cholangiocarcinoma occurred before the progression of the sclerosing cholangitis to cirrhosis (10). These cancers were most often extrahepatic and commonly occurred near the hepatic duct bifurcation. This site is also a common position for dominant benign strictures in primary sclerosing cholangitis, making the cholangiographic differentiation of benign and malignant lesions difficult (17,18). An increased incidence of cholangiocarcinoma has also been observed in patients with choledochal cysts (11). Approximately 2.5 to 28% of patients with cystic abnormalities of the bile duct will develop cholangiocarcinoma or gallbladder cancer. Choledochal cysts present either in infancy with jaundice, an abdominal mass, or abdominal pain, or in adult life with abdominal pain. The risk of cholangiocarcinoma increases with age (19). In a series of 42 patients reported recently from The Johns Hopkins Hospital, 11 were diagnosed with choledochal cysts before age 16, while 31 patients were diagnosed in adult life (11). The three patients (7%) with cholangiocarcinoma. had their choledochal cysts diagnosed as adults. Over 80% of patients with biliary cysts have an anomalous high entry of the pancreatic duct into the bile duct (APBDJ), which is also associated with an increased incidence of gallbladder cancer (20). This finding suggests that reflux of pancreatic secretions into the bile duct may lead to malignant transformation of the biliary epithelium. Gallstones are present in approximately onethird of the patients with cholangiocarcinoma, which is no different from the incidence of gallstones in the ageadjusted general population. In contrast, hepatolithiasis is a definite risk factor for cholangiocarcinoma. In certain parts of East Asia, intrahepatic stones are endemic. Cholangiocarcinoma will develop in between 5 and 10% of the patients with hepatolithiasis (2,12). Cholangiocarcinoma has been reported to develop a mean of 8 years following treatment for hepatolithiasis and may occur despite complete stone clearance from the intrahepatic biliary tree (21). The liver flukes Chlonorchis sinenis and Opisthorchis viverrini and a diet high in nitrosamines may be cofactors contributing to tumor formation in these patients (2). IV— Pathology More than 95% of bile duct cancers are adenocarcinomas (7). Most of these tumors are of the infiltrating nodular or diffusely infiltrating varieties. Papillary or purely nodular tumors are less frequent and are less commonly associated with extensive intra or extramural spread (22). The pathological determination of malignancy may be difficult, especially with biopsies or brushings performed in the setting of cholangitis, hepatolithiasis, biliary obstruction, or stenting. Positive staining for CEA, CA199, or CA50 supports the diagnosis of malignancy (1). The molecular events underlying the pathogenesis of cholangiocarcinoma are being increasingly defined (Table 2). Overexpression of the p53 tumor suppressor gene is present in between 19 and 66% of cholangiocarcinomas (23–25). The incidence of p53 overexpression is higher in distal bile cancers than in intrahepatic tumors, and p53 overexpression appears to occur later in neoplastic progression (24). Ohashi et al. reported Kras mutations in 67% of extrahepatic cholangiocarcinomas and also noted a higher frequency of mutations in distal bile duct cancers (26). Kras mutations have also been detected in atypical biliary epithelium associated with ABPDJ and in biliary cysts (27,28). Moreover, Hruban et al. have identified Kras mutations in 7% of benign bile duct proliferations identified on liver biopsy, suggesting that Kras mutation may be an early event in the progression of cholangiocarcinoma (29).
Page 728 Table 2 Molecular Events in the Pathogenesis of Cholangiocarcinoma p53 protein overexpression
19–66%
Kras gene mutations
67%
p16 gene mutations
64%
DPC4 gene mutations
16%
Mutations of the p16 gene have been identified in 64% of biliary tract cancers, whereas DPC4 mutations have been detected in just 16% of biliary tract cancers (30,31). Overexpression of the p53 gene or Kras gene mutations may have a future role in predicting patient survival. V— Diagnosis A— Clinical Presentation More than 90% of patients with perihilar or distal tumors present with jaundice. Less common presenting clinical features include pruritus, mild abdominal pain, fatigue, anorexia, and weight loss (2). Cholangitis is not commonly present at the time of diagnosis but develops after biliary manipulation. In comparison, patients with intrahepatic tumors usually present with pain and, by definition, are not jaundiced. Except for jaundice, the physical examination is usually normal in patients with cholangiocarcinoma. B— Laboratory Data At the time of presentation, most patients with perihilar and distal cholangiocarcinoma will have a total serum bilirubin level greater than 10 mg/dL (2). Marked elevations are also routinely observed in alkaline phosphatase and gamma glutamyl transferase. Alanine aminotransferase and aspartate aminotransferase are usually elevated only to a minor degree. Patients with longstanding biliary obstruction may have laboratory evidence of depressed hepatocyte synthetic function such as a low serum albumin or prolonged prothrombin time. An elevated serum alkaline phosphatase together with an elevated gamma glutamyl transferase should be further evaluated with computed tomography (CT) scanning and cholangiography to exclude an intrahepatic cholangiocarcinoma. The serum tumor markers carcinoembryonic antigen (CEA) and alpha fetoprotein (AFP) are typically normal. Serum CA 199 and CA 50 may be elevated in patients with cholangiocarcinoma and may be useful in screening patients in highrisk groups for developing cholangiocarcinoma. Serum CA199 levels may fall once biliary obstruction is relieved. Recently, the use of serum and bile tumor markers has been shown to improve the early detection of cholangiocarcinoma in patients with primary sclerosing cholangitis (32,33). The use of both serum CEA and CA 199 levels together has been shown to have a sensitivity of 60% and a specificity of 100% in diagnosing occult cholangiocarcinomas in patients with sclerosing cholangitis (32). C— Radiological Evaluation The goals of radiological evaluation in patients with cholangiocarcinoma include the delineation of the overall extent of the tumor, including involvement of the bile ducts, liver, portal vessels, and distant metastases. An orderly sequence of studies will usually achieve these goals. The initial radiographic studies consist of either abdominal ultrasound or CT scanning. Intrahepatic tumors are easily visualized on CT and appear as liver masses with or without biliary ductal
Page 729
dilation (Fig. 1). A perihilar cholangiocarcinoma will give a picture of a dilated intrahepatic biliary tree, a normal or collapsed gallbladder and extrahepatic biliary tree, and a normal pancreas. Distal bile duct tumors lead to dilation of both the intra and extrahepatic biliary tree and gallbladder with or without a pancreatic head mass (7). The primary tumor mass in patients with perihilar and distal tumors has been difficult to visualize on ultrasound and standard CT scan (Fig. 2). Improvements in these imaging tech
Figure 1 a. A MR scan demonstrates a large central intrahepatic cholangiocarcinoma. b. An endoscopic cholangiogram from the same patient demonstrates some distortion of the intrahepatic biliary tree but no definite obstructing lesion.
Page 730
Figure 2 a. A CT scan from a patient with a perihilar cholangiocarcinoma obtained following placement of an endoscopic stent into the right hepatic duct demonstrates dilation of the left intrahepatic biliary ducts and a nondilated right biliary ductal system. b. More caudal scan from same patient demonstrating endoscopic stent in place, a patent portal vein, and no evidence of a mass lesion. c. Leftsided percutaneous transhepatic cholangiogram from the same patient demonstrates dilated left hepatic ducts. d. Rightsided percutaneous transhepatic cholangiogram demonstrates a Bismuth type II perihilar cholangiocarcinoma. The patient was managed with a hilar resection with negative histological margins.
Page 731
Figure 2 Continued.
niques have increased their utility in defining the extent of biliary tract tumors. Duplex ultrasonography identified masses in 87% of 39 patients with perihilar cholangiocarcinoma (34). The extent of bile duct involvement was accurately determined in 86% of the tumors and portal vein involvement by tumor was identified with a sensitivity of 86%. Thinsection spiral CT has produced comparable results, with an overall sensitivity for detecting ductal thickening or a mass approaching 100%; however, identification of the level of biliary obstruction was possible in only in 63% of patients (35).
Page 732
Further delineation of biliary anatomy has been traditionally obtained cholangiographically in patients with perihilar or distal tumors through either the percutaneous transhepatic or the endoscopic retrograde routes. The most proximal extent of the tumor is the most important feature in determining resectability. In patients with tumors of the perihilar region, percutaneous transhepatic cholangiography is favored because it defines the proximal extent of rumor involvement most reliably (Fig. 2) (2). This approach also allows for the preoperative placement of percutaneous transhepatic catheters. Endoscopic cholangiography may provide more information in patients with distal tumors (7). Distal cholangiocarcinomas are often associated with obstruction of the distal common bile duct and a patent pancreatic duct, whereas both the common bile duct and pancreatic duct are usually obstructed in carcinoma of the head of the pancreas. The cholangiogram may be unremarkable in patients with intrahepatic cholangiocarcinoma (Fig. 1). The advantages of transhepatic catheter placement include (a) assistance in the technical aspects of hilar dissection by allowing palpation of the catheter within the biliary tree at the time of exploration and (b) facilitation of intraoperative silastic transhepatic stent placement. Currently available randomized studies do not support the practice of placing preoperative transhepatic catheters in an effort to reduce operative mortality (36). However, if a liver resection is contemplated, preoperative drainage may be justified. Recently, magnetic resonance cholangiopancreatography (MRCP) has documented diagnostic accuracy comparable to percutaneous and endoscopic cholangiography. In a series of 14 patients with cholangiocarcinoma, diagnosticquality MRCP images were better at visualizing intrahepatic biliary anatomy than ERCP (37). In recent series reported by Lee et al., MRCP was as accurate as ERCP at defining the level of biliary obstruction and in defining the nature of the obstructing lesion (malignant versus benign) (38). The overall diagnostic accuracy of MRCP and ERCP were 72 and 61%, respectively. MRCP also has the added advantage of obtaining twodimensional (2D) images to further define an obstructing lesion. Guthrie et al. demonstrated a mass or ductal thickening in all 24 patients with hilar cholangiocarcinoma using T2weighted spinecho MR imaging (39). Angiography has also been used to further define vascular involvement, especially in patients with perihilar tumors. Findings of hepatic arterial or portal venous encasement are present in onethird of patients with hilar cholangiocarcinoma on angiography. The use of angiography alone predicted resectability in 71% of patients with perihilar cholangiocarcinoma (40). MR imaging (MRI) can also provide information on vascular involvement. With the use of gadolinium enhancement in a series of 24 patients with perihilar tumors, occlucled portal vein branches were demonstrated in 10 patients and tumorencased branches in 2 patients (39). MRI may eventually provide all the necessary information to accurately stage patients with cholangiocarcinoma and avoid any invasive procedures in patients in whom preoperative biliary drainage is not indicated. D— Biopsy/Cytology In efforts to establish a tissue diagnosis, percutaneous fine needle aspiration biopsy, brushandscrape biopsy, and cytological examination of bile have all been used (41). If surgery is contemplated; a preoperative tissue diagnosis is not essential. Prolonged efforts to obtain a preoperative tissue diagnosis are not indicated unless the patient is not an operative candidate. Bile obtained from a percutaneous catheter will demonstrate malignant cells in approximately 30% of cases. This yield may be improved to approximately 40% by brush cytological techniques through transhepatic stents, or at the time of endoscopic procedures and to 67% by percutaneous fineneedle aspiration. Cholangioscopy through percutaneous tube tracts may be used to guide biopsies and determine the extent of the tumor. However, even with these efforts, up to onethird of patients with cholangiocarcinoma will have negative biopsy and/or cytological results. Intraoperative fineneedle aspiration is also very accurate; however, needle biopsy is warranted only if the lesion is unresectable (7). Punch biopsies of the lumen are also useful
Page 733
in this setting. If the tumor is localized and resectable, efforts to establish a tissue diagnosis before resection are usually unnecessary. VI— Staging, Classification, and Preoperative Assessment An appropriate management strategy can be devised only after defining the stage, anatomic extent, and operative risk of a patient with cholangiocarcinoma. Cholangiocarcinoma is staged according to the tumor, node, metastasis (TNM) classification of the American Joint Commission on Cancer (Table 3). Using this system, stage I tumors are limited to the bile duct mucosa or muscular layer, whereas stage II tumors invade periductal tissues. Stage III tumors have regional lymph node metastases, whereas stage IV tumors either invade adjacent structures (IVA) or have distant metastases (IVB) (42). Cholangiocarcinoma is best classified into three broad groups: (a) intrahepatic, (b) perihilar, and (c) distal (Fig. 3) (1). This classification correlates with anatomic distribution and implies the treatment for each site. Cancers of the hepatic duct bifurcation have also been classified further by Bismuth according to their anatomic location (43). In this system, type I tumors are confined to the common hepatic duct and type II tumors involve the hepatic duct bifurcation including both the main right and left hepatic ducts. Type IIIa and IIIb tumors extend into the right and left secondary intrahepatic ducts, respectively, and type IV tumors involve the secondary intrahepatic duct on both sides. A careful evaluation of the overall general medical condition of the patient as well as accurate staging are necessary prior to selecting the appropriate management for the patient with cholangiocarcinoma. The preoperative assessment should include the usual evaluation of cardiac risk factors, respiratory status, and renal function as well as overall performance status. Patients with obstructive jaundice often have decreased hepatic protein synthesis as well as altered hemostatic mechanisms and they are at an increased risk for infectious complications (2,44). Several studies have defined preoperative risk factors associated with an increase in morbidity and mortality in patients undergoing treatment for malignant biliary obstruction. In 1981 Pitt and colleagues identified eight risk factors predictive of mortality, including serum albumin less than 3.0 g/dL (45). Little also defined a mortality index predictive of procedurerelated mortality in a prospective analysis of patients with obstructive jaundice (46). The mor Table 3 Staging of Extrahepatic Cholangiocarcinoma Stage
Tumor
Node
Metastasis
I
T1
N0
M0
II
T2
N0
M0
III
T1–2
N1–2
M0
IVA
T3
Any N
M0
IVB
Any T
Any N
M1
Key: T1, tumor invades mucosa or muscle layer; T2, tumor invades perimuscular connective tissue; T3, tumor invades adjacent structures: liver, pancreas, duodenum, gallbladder, colon, stomach; N0, no regional lymph node metastasis; N1, metastasis in the cystic duct or the pericholedochal and/or hilar lymph nodes (i.e., hepatoduodenal ligament); N2, metastasis in the peripancreatic (head only), periduodenal, periportal, celiac, superior mesenteric, and/or posterior pancreaticoduodenal lymph nodes; M0, no distant metastasis; M1, distant metastasis. Source: Adapted from Ref. 42.
Page 734
Figure 3 Distribution of 294 cholangiocarcinoma into intrahepatic, perihilar, and distal subgroups. (From Ref. 1.)
tality index was derived from the preoperative serum creatinine, serum albumin, and severity of cholangitis. Several recent large series of patients undergoing resection for perihilar cholangiocarcinoma have also identified low preoperative serum albumin and perioperative sepsis as factors contributing to operative mortality (45,47). Control of sepsis and intensive nutritional support should be undertaken preoperatively in the malnourished patient with cholangiocarcinoma. Once the preoperative evaluation is completed, a determination of resectability is made (2,44). In the past the combination of an abdominal computed tomography (CT) scan, cholangiography, and visceral angiography were all useful in making this determination. CT or MRI scan findings signifying unresectable disease include peripheral hepatic metastases or extrahepatic disease. Extensive bilobar involvement in patients with intrahepatic tumors also precludes resection. Findings on traditional or MRI cholangiography suggestive of unresectable disease in patients with perihilar cholangiocarcinoma include proximal extension of tumor into second order bile ducts in both hepatic lobes. The angiographic or MRI findings of tumor encasement or occlusion of the proper hepatic artery, main portal vein, or both right and left portal venous branches or hepatic arterial branches are also considered contraindications to resection by most groups. The combination of angiography and cholangiography provides better data than cholangiography alone to stage the patient with perihilar cholangiocarcinoma (40). More recently, MRI may provide all the data previously obtained by these two studies and CT scanning (39,40). For patients with distal cholangiocarcinoma, a goodquality spiral CT scan can provide sufficient information to predict resectability. VII— Surgical Resection Curative treatment of patients with cholangiocarcinoma is only possible with complete resection. For patients with anatomically resectable intrahepatic cholangiocarcinoma and without advanced cirrhosis, partial hepatectomy is the procedure of choice (2,7). After careful explo
Page 735
ration of the peritoneal surfaces and regional lymph nodes for metastatic disease, the liver is mobilized and examined with intraoperative ultrasound. Approximately 40% of patients with resectable intrahepatic cholangiocarcinomas will have either multiple tumors and/or tumors involving both hepatic lobes (48). Hepatic resection is planned to completely remove all tumor with an adequate margin. Most commonly, this surgery involves a formal lobectomy or trisegmentectomy. In addition, care must be taken to achieve a negative bile duct margin if the tumor approaches the hilum (7). For patients with tumors involving the hepatic hilum or proximal common hepatic duct (Bismuth types I or II) that have no vascular invasion, removal is often possible by local tumor excision (1,2). Preoperatively, transhepatic Ring catheters are placed in the right and left hepatic ducts. These catheters aid in the intraoperative identification of the right and left hepatic ducts and are not placed solely to relieve jaundice preoperatively (44). Patients are explored through an upper midline or right subcostal incision. A careful examination of all peritoneal surfaces is undertaken to exclude any tumor dissemination before beginning with the hilar dissection. Similarly, the liver is carefully examined for evidence of metastatic disease. If a cholecystectomy has not been performed previously, the gallbladder is mobilized out of the liver bed and the cystic artery is ligated and divided. The cystic duct is likewise ligated to prevent spillage of gallbladder bile throughout the procedure. The common bile duct is dissected free and encircled just proximal to the suprapancreatic portion. The common bile duct is then divided, the distal end is oversewn with interrupted silk sutures, and a frozen section is obtained to confirm a negative distal bile duct margin. The common bile duct is then reflected cephalad, skeletonizing the portal vein and proper hepatic artery. Once the entire common hepatic duct has been elevated off the portal vasculature, the left and right hepatic ducts can be identified by palpating the diverging Ring catheters. In patients without significant extension of tumor into either the right or left hepatic ducts on preoperative cholangiogram, the left and right hepatic ducts are encircled. The left and right hepatic ducts are then divided above the extent of palpable tumor. Frozensection examination can be used to determine the adequacy of the surgical margin. The percutaneous transhepatic catheters are then used to guide progressively larger coudé catheters through both the left and right hepatic ducts. Finally, 16 Fr silastic catheters are pulled into position in the left and right hepatic ducts. A 60cm retrocolic RouxenY limb is then constructed, and bilateral hepaticojejunostomies are performed over the silastic stents. The free ends of the silastic catheters are brought out through separate stab wounds in the upper abdomen. Proximal extension of a hilar cholangiocarcinoma into the intrahepatic segments of either the right or left hepatic duct renders these tumors incurable by local tumor resection. Complete tumor resection in these patients is achievable only with combined resection of the extrahepatic biliary tree and hepatic resection. Improvements in operative morbidity and mortality as well as in longterm survival when negative surgical margins are achieved support the use of this approach when complete tumor resection is possible (2,44). The need for hepatic resection can usually be predicted on the basis of preoperative angiography, cholangiography, or MRI. When unilateral neoplastic involvement of the right or left portal vein or hepatic ducts is visualized radiographically, the initial hilar dissection is performed as described previously, with division of the uninvolved hepatic duct (Fig. 4). A frozen section should be obtained to confirm a negative hepatic duct margin on the uninvolved side. Next, the extrahepatic segments of the hepatic vein, portal vein, and hepatic artery to the involved lobe are divided or occluded with vascular clamps. The hepatic parenchyma is then divided, using the ultrasonic dissector or cautery. Caudate lobe involvement is common in perihilar cholangiocarcinoma, and caudate lobectomy is frequently necessary in order to achieve negative tumor margins (49). Biliary enteric continuity is restored to a RouxenY limb of jejunum as described above. Several more aggressive approaches have been used in patients with more widespread cholangiocarcinoma involving the hepatic hilum. Combined hepaticopancreaticoduodenectomy has been reported in patients with diffuse cholangiocarcinoma involving the hepatic hilum and
Page 736
Figure 4 a. Diagram illustrating left hepatic and hilar resection of Bismuth type IIIb cholangiocarcinoma with preoperatively placed transhepatic stents. b. Diagram demonstrates resected left hepatic lobe and hilum with perihilar cholangiocarcinoma (top) and right hepatic lobe with divided right hepatic duct prior to reconstruction (bottom). c. Silastic transhepatic stent is placed through a right RouxenY cholangiojejunostomy after left hepatic lobectomy. (From Ref. 94.)
entire extrahepatic biliary tree (50,51). Preoperative portal vein and hepatic arterial embolization have both been utilized in patients requiring extended hepatic resection to achieve a negative tumor margin (50,51). In these approaches, preoperative embolization of the hepatic lobe containing the extensive cholangiocarcinoma leads to atrophy of the lobe to be resected and hypertrophy of the contralateral lobe. Portal vein or hepatic arterial embolization is then followed 2 to 3 weeks later by extended hepatic resection. This approach may lower the morbidity and mortality from hepatic failure following extensive hepatic resection in jaundiced patients with hilar cholangiocarcinoma.
Page 737
Figure 4 Continued.
Another resectional option for patients with hilar and intrahepatic cholangiocarcinoma that has been received with some enthusiasm in the past is total hepatectomy and liver transplantation. However, the results of liver transplantation for perihilar cholangiocarcinoma have been disappointing, with early and widespread recurrence precluding tumorfree survival in most patients (2,53). Survival rates of less than 10% have been reported among patients undergoing liver transplantation for perihilar cholangiocarcinoma. Recently, liver transplantation has been advocated for patients with otherwise unresectable intrahepatic cholangiocarcinoma. Liver transplantation should be restricted to patients with single tumors confined to the liver and without lymph node metastases (48). Patients with resectable distal cholangiocarcinoma are managed, much like patients with other periampullary malignancies, with pancreatoduodenectomy. Over 90% of these tumors
Page 738
Figure 4 Continued.
will be resectable, and over 85% of these patients will be suitable for the pyloruspreserving modification (1). Gastrointestinal continuity is restored with an endtoend pancreatojejunostomy, an endtoside hepaticojejunostomy, and an endtoside duodenojejunostomy. The hepaticojejunostomy is usually stented with a T tube to decompress the system in the event of a biliary or pancreatic leak. VIII— Palliative Therapy Palliative therapy in patients with perihilar and distal cholangiocarcinoma is directed at relieving obstructive jaundice and pruritus, preventing recurrent cholangitis, and avoiding hepatic failure secondary to unrelieved biliary obstruction. In addition, palliative therapy in patients with distal cholangiocarcinoma is also aimed at preventing or relieving gastric outlet obstruction. Palliation can be achieved either nonoperatively with percutaneous or endoscopic techniques or by using an operative approach.
Page 739
A— Nonoperative Palliation Patients with unequivocal evidence of unresectable cholangiocarcinoma at initial evaluation are palliated nonoperatively. Tumor extension into the secondary biliary radicles of both right and left hepatic lobes, extensive bilobar disease or multiple tumors, encasement or occlusion of the main portal vein, main hepatic artery, or superior mesenteric vein or artery, or the presence of distant metastases excludes patients from curative resection. In addition, patients who refuse surgery or are in poor general medical condition are also not operative candidates. Nonoperative palliation can be achieved both endoscopically and percutaneously. A significant proportion of the functioning hepatic parenchyma should be decompressed, and this philosophy may require two or three percutaneously or endoscopically placed catheters (54,55). In a recent large retrospective study, bilateral endoscopic or percutaneous drainage was associated with significantly improved overall survival when compared to unilateral drainage (225 versus 80 days, p < 0.0001) (55). Bilateral percutaneous catheters should be placed unless one hepatic lobe has significant atrophy demonstrated by CT or MRI scan or proximal extension of tumor is extensive with isolation of multiple subsegments in one hepatic lobe. Percutaneous biliary drainage has several advantages over endoscopic management in patients with perihilar cholangiocarcinoma. Stent placement is more reliably achieved percutaneously than via endoscopic means (2,44,56). In addition, occluded percutaneous stents are easily changed over a guidewire on an outpatient basis, whereas replacement of an endoscopic endoprostheses requires an additional invasive procedure. In contrast, endoscopic palliation is the preferred approach in the small percentage of patients with unresectable distal cholangiocarcinoma. Placement of an endoprosthesis in experienced centers has been technically successful in approximately 95% of patients with advanced periampullary cancer, with relief of jaundice and pruritus in 80%. More recently, percutaneous or endoscopically placed metallic stents have been used to palliate patients with malignant biliary obstruction (57–59). Metallic stents remain patent longer than plastic stents and require fewer subsequent manipulations in patients with distal malignant biliary obstruction (60). Selfexpanding metallic stents have also been effective in patients with unresectable perihilar tumors with median stent patency rates of up to one year (58). Photodynamic therapy has also been used to palliate jaundice in patients with malignant hilar obstruction. Significant decreases in serum bilirubin and improvement in performance scores were achieved in a group of nine patients with unresectable hilar cholangiocarcinoma and a poor response to endoscopic catheter drainage (61). Photodynamic therapy was associated with a mean survival of 14 months and no procedurerelated mortality. The future role of photodynamic therapy in palliating malignant biliary obstruction or as an adjuvant in resected patients with positive microscopic margins remains to be established. B— Operative Palliation In goodrisk patients without preoperative evidence of unresectable cholangiocarcinoma, operative exploration is undertaken in an attempt to resect the primary tumor. At Johns Hopkins, approximately 45% of patients with perihilar cholangiocarcinoma have been found at exploration to have intraperitoneal or liver metastases (15%) or extensive tumor involvement of the porta hepatis (30%), precluding resection (1). In comparison, only 10% of patients with distal cholangiocarcinoma will have unresectable lesions at operative exploration. Patients with peritoneal carcinomatosis undergo minimal operative intervention. Cholecystectomy is performed to prevent the subsequent development of acute cholecystitis from cystic duct obstruction related to the percutaneous catheter (62). Postoperatively, the preoperatively placed transhepatic catheters are replaced with larger, softer transhepatic stents (44). In patients with locally advanced unresectable perihilar tumors, several operative approaches are available for palliation. These include a RouxenY choledochojejunostomy with intraoperative placement of silastic biliary catheters or a segment III cholangiojejunostomy.
Page 740
The operative procedure for placing transhepatic silastic catheters begins with obtaining a tissue diagnosis of cholangiocarcinoma. A biopsy is taken of the main tumor mass either directly or with the aid of a choledochoscope to confirm the diagnosis of malignancy. The tissue diagnosis is required prior to initiating postoperative radiation or chemotherapy (44). The gallbladder is then mobilized and the distal common bile duct divided (Fig. 5). This step improves the exposure of the hepatic hilum and removes the risk of postoperative catheterrelated acute cholecystitis. The distal common bile duct is then oversewn. The common hepatic duct is then divided just distal to the perihilar tumor. Next, the Ring catheters are cut off as they exit the abdominal cavity and the ends are brought into the operative field. The biliary tree is then dilated using sequentially larger coudé catheters passed over a guidewire. Finally, 16Fr silastic biliary catheters are placed into both the right and left hepatic ducts. A hepaticojejunostomy is then performed to a RouxenY limb of jejunum. The advantages of this
Figure 5 a. Diagram illustrating palliative intubation of Bismuth type IV unresectable perihilar cholangiocarcinoma with preoperatively placed transhepatic stents. b. The common bile duct is divided distal to the tumor and a cholecystectomy is performed. c. A RouxenY choledochojejunostomy is constructed over Silastic transhepatic stents distal to the tumor. (From Ref. 94.)
Page 741
Figure 5 Continued.
Page 742
approach over nonoperative palliation include (a) removal of the gallbladder; (b) placement of larger, softer silastic stents with a lower risk of hemobilia and improved patient comfort; and (c) positioning of the stent into a defunctionalized RouxenY jejunal limb. Although difficult to document, larger stents that drain into a defunctionalized limb of jejunum should reduce the incidence of subsequent cholangitis. The second approach to operative palliation of the unresectable patient with hilar cholangiocarcinoma is the segment III cholangiojejunostomy (44). This approach is particularly well suited for patients with an undilatable hilar cancer and extensive disease on the right side. It is most effective in relieving jaundice when an adequate communication exists between the left and right ductal systems. This procedure should not be used when the left lobe of the liver is atrophic, signifying limited functioning parenchyma on this side. The segment III hepatic duct is approached by following the falciform ligament into the recess of the left lobe in the umbilical fissure. A sidetoside biliary enteric anastomosis is performed between the segment III hepatic duct and a RouxenY limb of jejunum. Most proponents of this technique do not use transanastomotic stents. In patients with extensive leftsided cholangiocarcinoma, a right intrahepatic cholangiojunostomy to the segment V duct may be performed. This approach is considerably more difficult than the segment III cholangiojejunostomy. No anatomic landmarks exist to assist in identifying the segment V duct, and considerable parenchymal dissection may be necessary prior to isolating a sufficient segment for a biliary enteric anastomosis. In patients with locally unresectable or metastatic distal cholangiocarcinoma, operative palliation is directed at relieving both biliary and gastric outlet obstruction. Symptomatic gastroduodenal obstruction occurs prior to death in approximately 30% of patients with periampullary malignancies. Gastrojejunostomy at the time of initial presentation prevents this complication without increasing operative morbidity or mortality. Biliaryenteric continuity is restored most often with a hepaticojejunostomy or a choledochojejunostomy. A chemical splanchnicectomy should also be included to treat or prevent pain (2). IX— Survival Procedurerelated morbidity and mortality, quality of survival, and longterm survival are highly dependent on the stage of disease at presentation and on whether the patient is treated by a palliative procedure or complete tumor resection. A— Surgical Resection Thirtyfour patients with intrahepatic cholangiocarcinoma managed with surgical resection were recently reported by Casavilla et al. (48). Multiple tumors were present in 44% of these patients, and 41% had involvement of both hepatic lobes. Histologically negative surgical margins were obtained in 71% of patients. Operative morality was 6%. Overall patient survival was 60% at 1 year, 37% at 3 years, and 31% at 5 years after resection. Positive surgical margins, multiple tumors, and metastatic disease in regional lymph nodes were all associated with lower overall survival. Similar results have been reported from several other centers with 5year survival rates of 16 to 32% in patients with resected intrahepatic cholangiocarcinoma (63,65). Chu et al. also identified hilar lymph node metastases as a significant predictor of lower overall survival (64). A total of 109 patients with perihilar cholangiocarcinoma were managed with resection at The Johns Hopkins Hospital between 1973 and 1995 (1). Following resection, 36 patients had gross tumor remaining and 81 patients had a positive microscopic surgical margin; 94 of these patients underwent local hilar excision while 15 were managed with a hepatic lobectomy
Page 743
in addition to resection of the extrahepatic biliary tree. Four operative deaths (3.6%) occurred among these 109 resected patients including one death (6.6%) in the 15 patients managed with hepatic lobectomy. Actuarial survival among the 109 patients undergoing resection was 68% at 1 year, 30% at 3 years, and 11% after 5 years of followup. Survival was significantly prolonged in patients without residual microscopic tumor at the surgical margin. In the 28 patients with a negative margin, the actuarial survival was 68% at 1 year, 56% at 3 years, and 19% after 5 years. Of these patients 53% also received a combination of externalbeam radiotherapy and/or a boost of internal irradiation utilizing iridium 192. In recent years, the trend around the world has been to perform more hepatic resections for perihilar cholangiocarcinoma. In a collective review of 389 patients with hilar cholangiocarcinoma resected between 1980 and 1989, the differences between outcomes in patients undergoing local hilar resection and major liver resection for cholangiocarcinoma were compared (66). The 201 patients undergoing a local hilar resection had an operative mortality rate of 8% and a mean survival of 21 months; the percentage of patients alive at 5 years was 7%. In contrast, in 188 patients undergoing major liver resection, the operative mortality rate was 15%, the mean survival was 24 months, and the percentage of patients alive at 5 years was 17%. An additional 398 patients managed between 1990 and 1995 were recently reported (44). In 118 patients undergoing local hilar resection, the operative mortality rate remains about 8%. In 280 patients managed with combined hilar and hepatic resection, the operative mortality had improved to 8%. The mean survival rate in the locally resected patients ranged from 19 to 36 months. Similarly, the mean survival in patients undergoing combined hilar and hepatic resection ranged from 16 to 32 months. Reduction in operative mortality has been due to
Figure 6 Actuarial survival in 136 resected patients with perihilar cholangiocarcinoma according to status of resection margins. Surgical resection margin free of tumor (solid line, n = 106), with microscopic residual tumor (dotted line, n = 27), or with gross residual tumor (dashed line, n = 3). Hospital mortality was excluded. Residual tumor stage was an independent prognostic indicator in multivariate analysis. (From Ref. 68.)
Page 744
Figure 7 Prognosis according to type of resection in 136 patients with perihilar cholangiocarcinoma. Isolated resection of bile duct including bifurcation (solid line, n = 31), resection of bile duct and anatomic liver resection (dotted line, n = 71), and resection of the bile duct bifurcation combined with hepatic and vascular resections (dashed line, n = 34). Hospital mortality was excluded. Actuarial survival was not influenced by the operative procedure performed. (From Ref. 68.)
improvements in postoperative care and operative technique with the recognition of the sensitivity of the jaundiced liver to hepatic arterial ischemia. Several groups have demonstrated low operative mortality and prolonged longterm survival following combined hilar and hepatic resection (44,49,51). Nagino et al. have recently updated their series of 124 patients managed with biliary and hepatic resection for perihilar cholangiocarcinoma (51). This aggressive approach included caudate lobectomy in all patients, partial resection of the portal vein in 41 patients, and hepaticopancreatoduodenectomy in 16 patients in order to achieve negative histological margins. Operative mortality was 9.7%. At 3 and 5 years, overall survival was 43 and 26% in the 97 patients with a marginnegative resection. Sugiura et al. reviewed 83 patients with hilar carcinoma managed at several Japanese institutions with an operative mortality of 8% and a 5year survival of 20% (49). These series demonstrate that with proper preoperative preparation, major hepatic resection can be performed safely in patients with perihilar cholangiocarcinoma and results in an increase in the percentage of patients undergoing marginnegative resections and an improvement in survival. Several factors may be important in determining longterm prognosis following curative resection for hilar cholangiocarcinoma. Achieving negative histological margins is important in determining overall longterm survival (49,67,68). Bengmark et al. initially reported several 10year survivors following hepatic resection with negative histological margins for hilar cholangiocarcinoma (67). In two large series of patients managed with attempted curative resection for perihilar cholangiocarcinoma, both Klempnauer et al. and Sugiura et al. have reported a 33% overall 5year survival in patients with negative histological margins, whereas no patient with microscopic cancer at the surgical margins survived 5 years (Fig. 6) (49,68). Among
Page 745
patients with negative histological margins, the addition of a caudate lobectomy significantly prolonged 5year survival (46 versus 12%) (49). In addition, the presence of regional lymph node metastases also adversely affected survival. The extent of resection (hilar versus hilar and hepatic resection) did not significantly affect survival in this study; however, hospital mortality was not included in the survival analysis (Fig. 7) (68). Seventythree patients with distal cholangiocarcinoma undergoing pancreatoduodenectomy have been reported from The Johns Hopkins Hospital (1). The 1, 3, and 5year survival rates were 70, 31, and 28%, respectively. Actuarial 10year survival in patients with resected distal cholangiocarcinoma was 21%. Factors associated with prolonged survival in these patients include tumor differentiation and lymph node status. Negative nodes increased median survival from 17 to 27 months, and 5 year overall survival was significantly longer in the 63% of patients without lymph node metastases than in the 37% of patients with nodal metastases (38 versus 9%, Fig. 8) (1,69). Poorly differentiated tumors also had a lower median survival (10 versus 22 months). B— Palliative Therapy Almost 50% of patients with perihilar tumors explored with the intent to resect will be unresectable. The majority of these patients will undergo a biliaryenteric bypass for palliation. A number of studies have compared the results of operative palliation with nonoperative biliary drainage in patients with unresectable perihilar cholangiocarcinoma. Between 1973 and 1989, a total of 65 patients with unresectable hilar cholangiocarcinoma underwent either nonoperative percutaneous stenting or operative palliation at The Johns Hopkins Hospital (54). Of these, 21 patients were managed with percutaneous biliary stents and 44 underwent laparotomy, with placement of largebore Silastic transhepatic stents in 33. No significant differences with respect to age, mean preoperative laboratory data, and extent of tumor were observed between treatment groups. The procedurerelated morbidity was similar between patients undergoing nonoperative palliation and those undergoing laparotomy. Hospital mortality was 14% in the nonoperatively palliated group and 7% in those managed operatively. Survival at 1, 3, and 6 months
Figure 8 Influence of lymph node status on survival of patients with resected distal cholangiocarcinoma. Survival was significantly (p < 0.01) longer in patients with negative lymph nodes than in patients with regional lymph node metastases. (From Ref. 1.)
Page 746
after treatment was significantly greater in patients undergoing operative palliation when compared to patients managed nonoperatively. Mean survival was 8 months in the patients managed with operative palliation and 5 months in those managed with percutaneous stenting (p < 0.05). Quality of survival measured by the number of readmissions per month of survival, hospital days per month of survival, and episodes of cholangitis was better in the patients undergoing operative palliation. In addition, patients managed nonoperatively required more frequent stent changes than patients managed operatively. On the other hand, eight patients (15%) managed operatively required late operations. These procedures were performed most frequently for small bowel or duodenal obstruction. None of the patients managed nonoperatively required laparotomy during the followup period. Several additional studies have compared the results of operative and nonoperative palliative therapy in hilar cholangiocarcinoma (70–73). In a French report, the operative mortality and longterm survival were similar between patients managed with operatively placed biliary stents and patients managed with palliative biliary enteric anastomosis (70). Several retrospective analyses have also compared nonoperative versus operative palliation. Guthrie et al. demonstrated more effective palliation with a segment III cholangiojejunostomy when compared with endoscopic or percutaneous stenting (71). The operatively managed patients had a lower incidence of cholangitis and jaundice than the patients managed with nonoperative stenting. Lai et al. demonstrated a lower procedurerelated mortality in patients managed operatively than in patients palliated with endoscopic stenting (72). Overall survival and quality of survival assessed by frequency of cholangitis attacks, episodes of jaundice, and days in the hospital were similar between the two groups. Patients managed with endoscopic stents had a greater incidence of catheter related problems than patients managed with cholangioenteric bypass. In contrast, Washburn et al. have demonstrated better survival in patients palliated nonoperatively than in surgically palliated patients (73). All of these retrospective analyses have been limited, however, by biases in selecting patients for each treatment group. Patients with unequivocal evidence of unresectable disease on imaging studies are palliated with endoscopically or percutaneously placed biliary stents. Metallic stents have better patency rates than standard polyethylene stents. Median stent patency ranges from 3 to 12 months for endoscopically placed metallic wall stents in the management of inoperative malignant biliary obstruction (57,74,75). In a randomized trial of endoscopically placed polyethylene and metallic stents, the metallic stents were associated with shorter overall hospital stay and greater complicationfree survival (75). Metallic wall stents also provided a cost savings in patients surviving over 6 months. No survival differences were observed among the two types of stents. Several randomized, prospective trials have compared operative versus nonoperative palliation in patients with periampullary cancer. In general, these trials demonstrated lower procedurerelated morbidity and mortality for the nonoperatively managed patients. However, the procedurerelated mortality in the operatively managed patients ranged from 15 to 24%, which is much higher than has been reported from tertiary referral centers (76). In addition, a higher rate of recurrent jaundice or gastric outlet obstruction occurred in the nonoperatively managed patients. Survival was similar among the two treatment groups in each study (mean survival 12 to 22 weeks). Lower operative mortality has been reported from several centers in patients undergoing operative palliation of unresectable periampullary tumors. Between 1987 and 1991, a total of 118 patients with unresectable periampullary tumors were explored at The Johns Hopkins Hospital (76). Of these, 70% were over age 60. The most common operative procedures were gastrojejunostomy and hepaticojejunostomy, performed in 67% of patients. Perioperative mortality was 2.5%, and gastric outlet obstruction or recurrent jaundice developed prior to death in only 4 and 2.5% of patients, respectively. Median survival for patients with unresectable intrahepatic cholangiocarcinoma ranges from 2 to 3 months. Even with aggressive multimodal adjuvant therapy, median survival for patients with unresectable intrahepatic cholangiocarcinoma has been only 5 to 7 months (64,65).
Page 747
X— Adjuvant Therapy Most of the trials of chemotherapy for biliary tract cancers have included cholangiocarcinomas from all sites as well as gallbladder cancers (7). The majority of regimens have used 5FU alone or in combination with other agents. The use of 5FU alone produces partial responses in 8% of patients and median survival of 26 weeks in patients with cholangiocarcinoma (77). Combination chemotherapy using 5FU together with cisplatin, carboplatin, or interferon alpha2b has produced modest improvements in survival and response rates (Table 4) (78–85). The improved response rates and survival seen with gemcitabine in pancreatic cancer have not yet been duplicated in biliary tract malignancies (83). Unfortunately, none of the above regimens has been proven to enhance survival in patients with cholangiocarcinoma. Radiation therapy has been evaluated in patients with cholangiocarcinoma using a variety of methods including externalbeam radiotherapy, intraoperative radiotherapy, internal radiotherapy, radioimmunotherapy, and charged particle radiation (2,7). Externalbeam radiotherapy has been the most commonly used modality and is typically administered to a total dose of 45 to 60 Gy (7,86,87). Internal radiotherapy is normally delivered through either percutaneous or endoscopically placed biliary stents using iridium 192 as the radiation source (2). Total radiation doses may vary from 20 to 60 Gy up to 1 cm from the source. Radioimmunotherapy has also been performed with iodine131 antiCEA (88). Multiple retrospective analyses have suggested that radiation therapy may provide some benefit in patients with perihilar cholangiocarcinoma administered through one of these techniques. However, in many of these analyses, betterrisk patients receive the radiation therapy and, not surprisingly, have a more favorable survival. A recent prospective analysis from The Johns Hopkins Hospital has evaluated 84 patients with perihilar cholangiocarcinoma (86); 34 were excluded because of metastatic disease, poor Karnofsky performance status, prior cancer or radiotherapy, or hospital mortality. The remaining 50 patients were considered eligible for postoperative radiotherapy. Of these, 23 received Table 4 Chemotherapy for Cholangiocarcinoma Author
Year
Falkson et al. (77)
1984
No. of patients 12
Median survival
Chemotherapy Oral 5FU
Response rate
6 Months
8% a
Isacoff et al. (78)
1993
7
5FU infusion, leucovorin, mitomycin C
17 Months
43%
Rougier et al. (79)
1995
19
5FU infusion + cisplatin
10 Months
32%
Ellis et al. (80)
1995
20
5FU infusion, epirubicin, cisplatin
b
40%
Jones et al. (81)
1996
15
Paclitaxel
DiLauro et al. (82)
1997
15
5FU infusion, epirubicin, cisplatin
Mezger et al. (83)
1997
11
Gemcitabine
Patt et al. (84)
1997
35
5FU infusion + interferon 2b
12 Months
34%
Sanzaltamira et al. (85)
1998
14
5FU infusion, leucovorin + carboplatin
5 Months
21%
Median survival not yet reached. Duration of response (survival not reported). c NR, not reported. a
b
10 Months NRc
9.5 Months NR
0% 33% 0%
Page 748
Figure 9 Actuarial KaplanMeier survival for patients with perihilar cholangiocarcinoma who did or did not receive postoperative radiation therapy. Survival was similar among resected patients who received postoperative radiation and patients who did not. (From Ref. 86.)
radiation while 27 did not. Radiation dose ranged from 45 to 63 Gy and consisted of external beam plus iridium 192 seeds. None of the patients received adjuvant chemotherapy. Patients undergoing curative resection survived significantly longer than patients undergoing operative palliation. Among resected patients, radiation had no effect on mean (24 versus 24 months), median, or actuarial survival (Fig. 9). Similarly, among palliated patients, radiation had no effect on mean (10 versus 13 months), median, or actuarial survival. Multivariate analysis identified resection as the only positive predictive factor for prolonged survival, while the presence of diabetes was the only negative prognostic factor. Radiation therapy had no effect on survival. Table 5 Chemoradiation for Cholangiocarcinoma Author
Year
No. of patients
Minsky et al. (89)
1991
12
5FU infusion + mitomycin 65 Gy EBRT bolus
17 Months
Koyama et al. (90)
1993
20
Mitomycin bolusb
Co60 IORT + 32–40 GyEBRT
30 Months
Whittington et al. (91)
1995
9
5FU infusion
59 Gy EBRT
12 Months
Kraybill et al. (92)
1994
58 38
Various regimensc
<40 Gy EBRT 40 Gy EBRT
8 Months 16 Months
Robertson et al. (93)
1997
11
Hepatic artery floxuridine 48–66 Gy EBRT infusion
Chemotherapy
Radiation
Key: EBRT, external beam radiation therapy; IORT, intraoperative radiation therapy. a Includes resected and unresected patients. b Absorbed to charcoal particles and injected intraoperatively. c In 31 of 96 patients. Source: Adapted from Ref. 2.
Median survivala
16 Months
Page 749
Combinations of chemotherapy with radiation have been attempted for many localized tumors. The benefit of combining radiation with 5FU or cisplatin with radiation has been demonstrated with several tumor types but not for biliary tract cancers (7). Chemoradiation has been applied to patients with cholangiocarcinoma at several centers (Table 5) (89–93). In general, these regimens are well tolerated; however, the number of patients has been small, both resected and unresected patients have been treated, and no control patients have been included. XI— Summary Despite overall advances in the ability to diagnose and treat patients with cholangiocarcinoma, the prognosis for patients with this malignancy remains poor. Further improvements in survival of patients with cholangiocarcinoma will come with the early diagnosis of these lesions. New molecular techniques should improve our ability to screen highrisk patients such as those with primary sclerosing cholangitis, hepatolithiasis, choledochal cysts, and ulcerative colitis. Improvements in imaging will continue, and spiral CT, duplex ultrasonography, and MRI will improve our ability to noninvasively stage patients with cholangiocarcinoma. Complete surgical resection remains the only curative treatment for malignancies of the biliary tract. Aggressive surgical approaches are likely to continue, and the challenge remains in being able to perform these procedures safely in jaundiced and sometimes septic patients. For those patients with unresectable lesions, the optimal form of palliation, whether operative or nonoperative, remains to be defined. Finally, multicenter, prospective, randomized trials of chemoradiation must be performed to further delineate an effective adjuvant therapy and hopefully improve the overall prognosis of patients with cholangiocarcinoma. References 1. Nakeeb A, Pitt HA, Sohn TA, Coleman J, Abrams RA, Piantadosi S, Hruban RH, Lillemoe KD, Yeo CJ, Cameron JL. Cholangiocarcinoma: a spectrum of intrahepatic, perihilar, and distal tumors. Ann Surg 1996; 224:463–475. 2. Pitt AH, Dooley WC, Yeo CJ, Cameron JL. Malignancies of the biliary tree. Curr Probl Surg 1995; 32:1–100. 3. Nagorney DM, Donohue JH, Farnell MB, Schleck CD, Ilstrup DM. Outcomes after curative resections of cholangiocarcinoma. Arch Surg 1993; 128:871–877. 4. Altemeier WA, Gall EA, Zinninger MM. Sclerosing carcinoma of the major intrahepatic bile ducts. Arch Surg 1957; 75:450–455. 5. Klatskin G. Adenocarcinoma of the hepatic duct at its bifurcation within the porta hepatis: an unusual tumor with distinctive clinical and pathologic features. Am J Med 1965; 38:241–247. 6. Landis MPH, Murray T, Bolden S, Wingo PA. Cancer statistics, 1998. CA Cancer J Clin 1998; 48:6–29. 7. Pitt HA, Grochow LB, Abrams RA. Cancer of the biliary tree. In: DeVita VT, Hellman S, Rosenberg SA eds. Cancer: Principles and Practice of Oncology. Philadelphia: Lippincott, 1997, pp 1114–1128. 8. Ito Y, Kojiro M, Nakashima T, Mori T. Pathomorphologic characteristics of 102 cases of Thorotrastrelated hepatocellular carcinoma, cholangiocarcinoma and hepatic angiosarcoma. Cancer 1988; 62:1153–1158. 9. Yamamoto M, Takasaki K, Nakano M, Saito A. Minute nodular intrahepatic cholangiocarcinoma. Cancer 1998; 82:2146–2149.
Page 750
10. Ahrendt SA, Pitt HA, Nakeeb A, Klein AS, Lillemoe KD, Kalloo AN, Cameron JL. Diagnosis and management of cholangiocarcinoma in primary sclerosing cholangitis. J Gastrointest Surg 1999; 3:357–368. 11. Lipsett PA, Pitt HA, Colombani PM, Boitnott JK, Cameron JL. Choledochal cyst disease: a changing pattern of presentation. Ann Surg 1994; 220:644–652. 12. SheenChen SM, Chou FF, Eng HL. Intrahepatic cholangiocarcinoma in hepatolithiasis: a frequently overlooked disease. J Surg Oncol 1991; 47:131–135. 13. AbuElmagd KM, Malinchoc M, Dickson ER, Fung JJ, Murtaugh PA, Langworthy AL, Demetris AL, Krum RAF, Van Thiel DH, Starzl TE. Efficacy of hepatic transplantation in patients with primary sclerosing cholangitis. Surg Gynecol Obstet 1993; 177:335–344. 14. Farges O, Malassagne B, Sebagh M, Bismuth H. Primary sclerosing cholangitis: liver transplantation or biliary surgery. Surgery 1995; 117:146–155. 15. Ahrendt SA, Pitt HA, Kalloo AN, Venbrux AC, Klein AS, Herlong HF, Coleman J, Lillemoe KD, Cameron JL. Primary sclerosing cholangitis: resect, dilate, or transplant? Ann Surg 1998; 227:412–423. 16. Chapman RW, Arborgh BA, Rhodes JM, Summerfield TA, Dick R, Scheuer PJ, Sherlock S. Primary sclerosing cholangitis: a review of its clinical features, cholangiography and hepatic histology. Gut 1980; 21:870–877. 17. Rosen CB, Nagorney DM, Wiesner RH, Coffey RJ, LaRusso NF. Cholangiocarcinoma complicating primary sclerosing cholangitis. Ann Surg 1991; 231:21–25. 18. Cameron JL, Gayler BW, Sanfey H, Milligan F, Kaufman J, Maddrey WC, Herlong HC. Sclerosing cholangitis: anatomical distribution of obstructive lesions. Ann Surg 1984; 200:54–60. 19. Todani T, Tabuchi K, Watanabe Y, Kobayshi T. Carcinoma arising in the wall of congenital bile duct cysts. Cancer 1979; 44:1134–1139. 20. Tanaka K, Nishimura A, Yamada K, Ishibe R, Ishizaki N, Yoshimine M, Hamada N, Taira A. Cancer of the gallbladder associated with anomalous junction of the pancreatobiliary duct system without bile duct dilatation. Br J Surg 1993; 80:622–624. 21. Chijiiwa K, Ichimiya H, Kuroki S, Koga A, Nakayama F. Late development of cholangiocarcinoma after the treatment of hepatolithiasis. Surg Gynecol Obstet 1993; 177:279–282. 22. Sakamoto E, Nimura Y, Hayakawa N, Kamiya J, Kondo S, Nagino M, Kanai M, Miyachi M, Uesaka K. The pattern of infiltration at the proximal border of hilar bile duct carcinoma: a histologic analysis of 62 resected cases. Ann Surg 1998; 277:405–411. 23. Ohashi K, Nakajima Y, Kanehiro H, Tsutsumi M, Taki J, Aomatsu Y, Yoshimura A, Ko S, Kin T, Yagura K, Kanishi Y, Nakano H. Kiras mutations and p53 protein expression in intrahepatic cholangiocarcinomas: relation to gross tumor morphology. Gastroenterology 1995; 109:1612–1617. 24. Diamantis I, Karamitopoulou E, Perentes E, Zimmerman A. p53 protein immunoreactivity in extrahepatic bile duct and gallbladder cancer: correlation with tumor grade and survival. Hepatology 1995; 22:774–779. 25. Teh M, Wee A, Raju GC. An immunohistochemical study of p53 protein in gallbladder and extrahepatic bile duct/ampullary carcinoma. Cancer 1994; 74:1542– 1545. 26. Ohashi K, Tstsumi M, Nakajima Y, Nakano H, Konishi Y. Kiras point mutations and proliferation activity in biliary tract carcinomas. Br J Cancer 1996; 74:930–935. 27. Tomono H, Nimura Y, Aono K, Nakashima I, Iwamoto T, Nakashima N. Point mutations of the cKiras gene in carcinoma and atypical epithelium associated with congenital biliary dilatation. Am J Gastroenterol 1996; 91:1211–1214. 28. Matsubara T, Sakurai Y, Sasayama Y, Hori H, Ochiai M, Funabiki T, Matsumoto K, Hirono I. Kras point mutations in cancerous and noncancerous biliary epithelium in patients with pancreatobiliary malfunction. Cancer 1996; 77:1752–1757. 29. Hruban RH, Sturm PD, Slebos RJ, Wilentz RE, Musler AR, Yeo CJ, Sohn TA, Van Velthuysen MZ, Offerhaus GJ. Can Kras codon 12 mutations be used to distinguish
Page 751
benign bile duct proliferations from metastases to the liver? A molecular analysis of 101 liver lesions from 93 patients. Am J Pathol 1997; 151:943–949. 30. Yoshida S, Todoroki T, Ichikawa Y, Hanai S, Suzuki H, Hori M, Fukao K, Miwa M, Uchida K. Mutations of the p16Ink4/CDKN2 and p15Ink4B/MTS2 genes in biliary tract cancers. Cancer Res 1995; 55:2756–2760. 31. Hahn SA, Bartsch D, Schroers A, Galehdari H, Becker M, Ramaswamy A, SchwarteWaldhoff I, Masheck I, Schmiegel W. Mutations of the DPC4/Smad4 gene in biliary tract carcinoma. Cancer Res 1998; 58:1124–1126. 32. Ramage JK, Donaghy A, Farrant JM. Serum tumor markers for the diagnosis of cholangiocarcinoma in primary sclerosing cholangitis. Gastroenterology 1995; 108:865–869. 33. Nakeeb A, Lipsett PA, Lillemoe KD, FoxTalbot K, Coleman J, Cameron JL, Pitt MA. Biliary CEA levels are a marker for cholangiocarcinoma. Am J Surg 1996; 171:147–153. 34. Hann LE, Greatrex KV, Bach AM, Fong Y, Blumgart LH. Cholangiocarcinoma at the hepatic hilus: sonographic findings. Am J Roentgenol 1997; 168:985–989. 35. Han JK, Choi BI, Kim TK, Kim SW, Han MC, Yeon KM. Hilar cholangiocarcinoma: thinsection spiral CT findings with cholangiocarcinoma correlation. Radiographics 1997; 17:1475–1485. 36. Nakeeb A, Pitt HA. The role of preoperative biliary decompression in obstructive jaundice. Hepatogastroenterology 1995; 42:332–339. 37. Lomanto D, Pavone P, Laghi A, Panebianco V, Mazzocchi P, Fiocca F, Lezoche E, Passeriello R, Speranza V. Magnetic resonance cholangiopancreatography in the diagnosis of biliopancreatic disease. Am J Surg 1997; 174:33–38. 38. Lee M, Lee H, Kim MH, Kang E, Kim Y, Lee S, Kim P, Ha HK, Auh YH. Extrahepatic biliary diseases: 3D MR cholangiopancreatography compared with endoscopic retrograde cholangiopancreatography. Radiology 1997; 202:663–669. 39. Guthrie JA, Ward J, Robinson PJ. Hilar cholangiocarcinomas T2weighted spinecho and gadoliniumenhanced FLASH MR imaging. Radiology 1996; 201:347– 351. 40. Dooley WC, Pitt HA, Venbrux AD, Robertson L, Cameron JL. Is angiography useful in patients with perihilar cholangiocarcinoma (abstr). In: Proceedings of the International Hepatobiliary Pancreatic Association, 1992, p 32A. 41. Desa LA, Akosa AB, Lazzara S, Domizio P, Krausz T, Benjamin IS. Cytodiagnosis in the management of extrahepatic biliary stricture. Gut 1991; 32:1188– 1191. 42. American Joint Committee on Cancer. Gallbladder and extrahepatic bile ducts. In: Beahrs OH, Henson DE, Hutter RVP, Kennedy BJ. Manual for the Staging of Cancer. Philadelphia: Lippincott, 1993, pp 111–120. 43. Bismuth H, Nakache R, Diamond T. Management strategies in resection for hilar cholangiocarcinoma. Ann Surg 1992; 215:31–38. 44. Ahrendt SA, Cameron JL, Pitt HA. Current management of patients with perihilar cholangiocarcinoma. In: Cameron JL, ed. Advances in Surgery. Vol 30. St. Louis: Mosby, 1996, pp 427–452. 45. Pitt HA, Cameron JL, Postier RG, Gadacz TR. Factors influencing mortality in biliary tract surgery. Am J Surg 1981; 141:66–72. 46. Little JM. A prospective evaluation of computerized estimates of risk in the management of obstructive jaundice. Surgery 1987; 102:473–476. 47. Su CH, Tsay SH, Wu CC, Shyr YM, King KL, Lee CH, Lui WY, Liu TJ, P'eng FK. Factors influencing postoperative morbidity, mortality, and survival after resection for hilar cholangiocarcinoma. Ann Surg 1996; 223:384–394. 48. Casavilla FA, Marsh JW, Iwatsuki S, Todo S, Lee RG, Madariaga JR, Pinna A, Dvorchik KI, Fung JJ, Starzl TE. Hepatic resection and transplantation for periheral cholangiocarcinoma. J Am Coll Surg 1997; 195:429–436. 49. Sugiura Y, Nakamura S, Iida S, Hosoda Y, Ikeuchi S, Mori S, Sugiuka A, Tsuzuki T. Extensive resection of the bile ducts combined with liver resection for cancer of the main hepatic duct junction: a cooperative study of the Keio Bile Duct Cancer Study Group. Surgery 1994; 115:445–451.
Page 752
50. Kawasaki S, Makuuchi M, Miyagawa S, Kakazu T. Radical operation after portal embolization for tumor of hilar bile duct. J Am Coll Surg 1994; 178:480–486. 51. Nagino M, Nimura Y, Kamiya J, Kanai M, Uesaka K, Hayakawa N, Yamamoto H, Kondo S, Nishio H. Segmental liver resections for hilar cholangiocarcinoma. Hepatogastroenterology 1998; 45:7–13. 52. Vogl TJ, Balzer JO, Dette K, Hintze R, Pegios W, Maurer J, Keck H, Neuhaus P, Felix R. Initially unresectable hilar cholangiocarcinoma hepatic regeneration after transarterial embolization. Radiology 1998; 208:217–222. 53. Ringe B, Wittekind C, Bechstein WO, Bunzendahl H, Pichlmayr R. The role of liver transplantation in hepatobiliary malignancy: a retrospective analysis of 95 patients with particular regard to tumor stage and recurrence. Ann Surg 1989; 209:88–95. 54. Nordback IH, Pitt HA, Coleman JA, Venbrux AC, Dooley WC, Yeu NN, Cameron JL. Unresectable hilar cholangiocarcinoma: percutaneous versus operative palliation. Surgery 1994; 115:597–603. 55. Chang WH, Kortan P, Haber GB. Outcome in patients with bifurcation tumors who undergo unilateral versus bilateral hepatic duct drainage. Gastrointest Endosc 1998; 47:354–362. 56. Lameris JS, Stoker J, Dees J, Nix JA, Van Blankenstein M, Jeekel J. Nonsurgical palliative treatment of patients with malignant biliary obstruction: the place of endoscopic and percutaneous drainage. Clin Radiol 1987; 38:603–608. 57. Schima W, Prokesch R, Osterreicher C, Thurnher S, Fugger R, Schoff R, Havelec L, Lammer J. Biliary Wallstent endoprosthesis in malignant hilar obstruction: longterm results with regard to the type of obstruction. Clin Radiol 1997; 52:213–219. 58. Peters RA, Williams SG, Lombard M, Karani J, Westaby D. The management of highgrade hilar structures by endoscopic insertion of selfexpanding metal endoprosthesis. Endoscopy 1997; 29:10–16. 59. O'Brien S, Hatfield AR, Craig PI, Williams SP. A three year follow up of selfexpanding metal stents in the endoscopic palliation of longterm survivors with malignant biliary obstruction. Gut 1995; 36:618–621. 60. Becker CD, Glattie A, Mailbach R, Baer HU. Percutaneous palliation of malignant obstructive jaundice with wall stent endoprosthesis: followup and reintervention in patients with hilar and nonhilar obstruction. J Cardiovasc Int Radiol 1993; 4:597–604. 61. Ortner MAEJ, Liebetruth J, Schreiber S, Hanft M, Wruck U, Fusco V, Muller JM, Hortnagl H, Lochs H. Photodynamic therapy of nonresectable cholangiocarcinoma. Gastroenterology 1998; 114:536–542. 62. Lillemoe KD, Pitt HA, Kaufmann SL, Cameron JL. Acute cholecystitis occurring as a complication of percutaneous transhepatic drainage. Surg Gynecol Obstet 1989; 168:348–352. 63. Jan YY, Jeng LB, Hwang TL, Wang CS, Chen MT, Chen TJ. Factors influencing survival after hepatectomy for peripheral cholangiocarcinoma. Hepatogastroenterology 1996; 43:614–619. 64. Chu KM, Lai EC, AlHadeedi S, Arcilla CE, Lo C, Liu C, Fan ST, Wong J. Intrahepatic cholangiocarcinoma. World J Surg 1997; 21:301–306. 65. Berdah SV, Delpero JR, Garcia S, Hardwigsen J, Le Treut YP. A Western surgical experience of peripheral cholangiocarcinoma. Br J Surg 1996; 83:1517– 1521. 66. Boerma EJ. Research into the results of resection of hilar bile duct cancer. Surgery 1990; 108:572–580. 67. Bengmark S, Ekberg H, Evander A, KlofverStahl B, Tranberg KG. Major liver resection for hilar cholangiocarcinoma. Ann Surg 1988; 207:120–128. 68. Klempnauer J, Ridder GJ, von Wasielewski R, Werner M, Meimann A, Pichlmayr R. Resectional surgery of hilar cholangiocarcinoma: a multivariate analysis of prognostic factors. J Clin Oncol 1997; 15:947–954. 69. Yeo CJ, Sohn TA, Cameron JL, Hruban RH, Lillemoe KD, Pitt HA. Periampullary adenocarcinoma: analysis of 5 year survivors. Ann Surg 1998; 227:821–831.
Page 753
70. Reding R, Buard JL, Lebeau G, Launois B. Surgical management of 552 carcinomas of the extrahepatic bile ducts: results of the French Surgical Association survey. Ann Surg 1991; 213:236–241. 71. Guthrie CM, Haddock G, de Beaux AC, Garden OJ, Carter DC. Changing trends in the management of extrahepatic cholangiocarcinoma. Br J Surg 1993; 80:1434–1439. 72. Lai EC, Chu KM, Lo CY, Fan ST, Lo CM, Wong J. Choice of palliation for malignant hilar obstruction. Am J Surg 1992; 163:208–212. 73. Washburn WK, Lewis DW, Jenkins RL. Aggressive surgical resection for cholangiocarcinoma. Arch Surg 1995; 130:270–276. 74. Ede RJ, Williams SJ, Hatfield ARW, McIntyre S, Mair G. Endoscopic management of inoperable cholangiocarcinoma using iridium192. Br J Surg 1989; 76:867–871. 75. Prat F, Chapat O, Ducot B, Ponchon T, Pelletier G, Fritsch J, Choury AD, Buffet C. A randomized trial of endoscopic drainage methods for inoperable malignant strictures of the common bile duct. Gastrointest Endosc 1998; 47:1–7. 76. Lillemoe KD, Sauter PK, Pitt HA, Yeo CJ, Cameron JL. Current status of surgical palliation of periampullary carcinoma. Surg Gynecol Obstet 1993; 176:1–10. 77. Falkson G, MacIntyre JM, Moertel CG. Eastern Cooperative Oncology Group experience with chemotherapy for inoperable gallbladder and bile duct cancer. Cancer 1984; 54:965–969. 78. Isacoff WH, Botnick L, Tompkins R, Jacobs AD, Reber H, Taylor O. Treatment of patients with advanced tumors of the bile ducts with continuous infusion 5 fluorouracil in conjunction with calcium leucovorin, mitomycin C, and dipyramidole. Proc Am Soc Clin Oncol 1993; 12:225. 79. Rougier P, Fandi A, Ducreux M, Fandi L, Fabri MC, Zarba J, Kac J, Armand JP. Demonstrated efficiency of 5fluorouracil continuous infusion and cisplatin in patients with advanced biliary tract carcinoma (abstr). Proc Annu Meet Am Soc Clin Oncol 1995; 14:A498. 80. Ellis PA, Norman A, Hill A, O'Brien ME, Nicolson M, Hickish T, Cunningham D. Epirubicin, cisplatin, and infusional 5fluorouracil in hepatobiliary tumors. Eur J Cancer 1995; 31:1594–1598. 81. Jones DV, Lozano R, Hoque A. Phase II study of paclitaxel therapy for unresectable biliary tree carcinomas. J Clin Oncol 1996; 14:2306–2310. 82. DiLauro L, Carpano S, Capomolla E, Conti F, Rinaldi M, Lopez M, Vici P. Cisplatin, epirubicin, and fluorouracil for advanced biliary tract carcinoma (abstr). Proc Am Soc Clin Oncol 1997; 16:287a. 83. Patt YZ, Jones DV, Hoque A, Lozano R, Markowitz A, Raijman I, Lynch P, Charnsangavej C. Phase II trial of intravenous flourouracil and subcutaneous interferon alfab for biliary tract cancer. J Clin Oncol 1996; 14:2311–2315. 84. Sanzaltamira PM, Ferrante K, Jenkins RL, Lewis WD, Huberman MS, Stuart KE. A phase II trial of 5fluorouracil, leucovorin, and carboplatin in patients with unresectable biliary tree carcinoma. Cancer 1998; 82:2321–2325. 85. Mezger J, Sauerbruch T, Ko Y, Wolter H, Funk C. Phase II trial of gemcitabine in biliary tract cancers (abstr). Proc Am Meet Am Soc Clin Oncol 1997; 221:788–798. 86. Pitt HA, Nakeeb A, Abrams RA, Coleman J, Piantadosi S, Yeo CJ, Lillemoe KD, Cameron JL. Perihilar cholangiocarcinoma: postoperative radiotherapy does not improve survival. Ann Surg 1995; 221:788–798. 87. Verbeek PC, van Leeuwen DJ, van der Heyde MN, Gonzalesz D. Does additive radiotherapy after hilar resection improve survival of cholangiocarcinoma: an analysis in sixtyfour patients. Ann Chir 1991; 45:350–354. 88. Stillwagon GB, Order SE, Haulk T, Herpst J, Ettinger DS, Fishman EK, Klein JL, Leichner PK. Variable low dose irradiation (131IantiCEA) and integrated low dose chemotherapy in the treatment of nonresectable primary intrahepatic cholangiocarcinoma. Int J Radiat 1991; 21:1601–1605.
Page 754
89. Minsky BD, Kemeny N, Armstrong JG, Reichmann B, Botet J. Extrahepatic biliary system cancer: an update of a combined modality approach. Am J Clin Oncol 1991; 14:433–439. 90. Koyama K, Tanaka J, Sato Y, Seki H, Kato Y, Umezawa A. Experience in twenty patients with carcinoma of hilar bile duct treated by resection, targeting chemotherapy and intercavitary irradiation. Surg Gynecol Obstet 1993; 176:239–245. 91. Kraybill WG, Lee H, Picus J, Ramachandran G, Lopez MJ, Kucik N, Myerson RJ. Multidisciplinary treatment of biliary tract cancers. J Surg Oncol 1994; 55:239–245. 92. Whittington R, Neuberg D, Teste WJ, Benson AB, Haller DG. Protracted intravenous fluorouracil infusion with radiation therapy in the management of localized pancreaticobiliary carcinoma: a phase I Eastern Cooperative Oncology Group Trial. J Clin Oncol 1995; 13:227–234. 93. Robertson JM, McGinn CJ, Walker S, Marx MV, Kessler ML, Ensminger WD, Lawrence TS. A phase I trial of hepatic arterial bromodeoxyuridine and conformal radiation therapy for patients with primary hepatobiliary cancers or colorectal liver metastases. Int J Radiat Oncol 1997; 39:1087–1092. 94. Cameron JC. Atlas of Surgery. Vol 1. Philadelphia: Decker, 1990.
Page 755
34— Ampullary Tumors Keith D. Lillemoe The Johns Hopkins Medical Institutions, Baltimore, Maryland I— Introduction One century ago, William Stewart Halsted described the first successful surgical resection of an adenocarcinoma of the ampulla of Vater (1). He performed a local resection of the lesion with reimplantation of the bile and pancreatic ducts. Although the surgery was successful, the patient expired 6 months later. At autopsy ''it was found that the carcinoma had recurred in the head of the pancreas and duodenum." Although significant advances have been made since the days of Halsted, controversy continues today regarding the indications for local resection of tumors of the ampulla as opposed to more aggressive resection via pancreaticoduodenectomy. The purpose of this chapter is to review the epidemiology and pathogenesis, diagnosis, staging, and management of both benign and malignant ampullary tumors. II— Pathology The ampulla of Vater is a complex anatomic site representing the interface between the duodenum, pancreas, and pancreaticobiliary ductal system. Although small, representing an area of approximately 1 cm in diameter, it is the area of the small bowel with the highest incidence of neoplastic transformation and malignancy. This high incidence of malignancy likely reflects the local production of carcinogens through the combined interaction of the components of bile, duodenal contents, and pancreatic exocrine secretions. Although adenomas and adenocarcinoma constitute the predominant cell type, other benign and malignant tumors as well as metastatic and locally invasive tumors arise in the ampulla. Benign tumors include leiomyoma (gastrointestinal stromal tumors), lipomas, and carcinoid tumors. Metastatic tumors occurring from primary sites of melanoma, lymphoma, and renal cell carcinoma have also been reported. Finally, the ampulla can also be locally invaded by other primary periampullary malignancies, most commonly by tumors of the duodenum, pancreas, and bile duct. Adenomas of the ampulla of Vater usually appear as a protruding ampulla with a fine granular surface. Microscopically, the tumor is composed of papillary and/or tubular proliferation of atypical, tall columnar epithelial cells. The adenoma may grow intraluminally in the common channel of the bile duct/pancreatic duct and may extend onto the duodenal mucosa. Grossly, ampullary carcinomas are divided into protruding (intramural and exposed) and ulcerating types. The protruding tumor represents an earlier form of the carcinoma, while the
Page 756
ulcerated rumor is usually a more advanced stage. Microscopically, the majority of ampullary cancers are welldifferentiated adenocarcinomas, often showing a superficial papillary proliferation. The papillary growth may extend into the common channel, distal common bile duct, and/or pancreatic duct. The superficial portion of the tumor usually consists of a betterdifferentiated or lowgrade component, thus frequently leading to difficulty in differentiating an adenoma from a well differentiated adenocarcinoma. Ampullary carcinoma may invade, by direct extension, the adjacent duodenum, pancreas, and common bile duct. These neoplasms tend to metastasize to lymph nodes in the posterior and anterior pancreatic head regions and hepatoduodenal area. Lymph node metastases are present in about half of the patients with ampullary carcinoma and occur at a higher rate in patients with an exposed, protruding, or ulcerated tumor. III— Epidemiology and Pathogenesis Adenocarcinoma of the ampulla of Vater accounts for less than 10% of all cases of periampullary carcinoma, which includes cancers of the head of the pancreas, distal common bile duct, periampullary duodenum, and ampulla. However, if one considers only the subgroup of surgically resectable periampullary malignancies, ampullary carcinomas can be the final pathological diagnosis in up to 25% of patients who undergo resection. The incidence of adenocarcinoma of the ampulla is 2.9 per million population, whereas it is 8 in 1.6 per million population for gallbladder and bile duct carcinoma, respectively (2). The incidence of adenomatous lesions ranges between 0.04 and 0.12% in autopsy series. Ampullary adenocarcinoma arises from the mucosal cells of the ampulla through the process of malignant transformation. Benign ampullary adenomas undergo dysplastic degeneration with subsequent progression to adenocarcinoma in a similar fashion to the adenoma–carcinoma sequence, which has been well documented in colon cancer (3). The evidence supporting this theory has been nicely defined for ampullary carcinomas, including studies in which adenomas of the ampulla were followed with observation of a transition from a benign to a malignant histology (4). The rate of this malignant transformation is unknown but may go high as 25%. Similarly, several groups have demonstrated that 80 to 90% of invasive carcinomas of the ampulla of Vater have surrounding benign adenomatous changes, suggesting an origin from a site of mucosal adenoma or adenomatous change (5). Familial adenomatous polyposis (FAP), a dominantly inherited syndrome associated with multiple adenomas of the colon and the inevitable development of a colorectal carcinoma, has also been clearly associated with ampullary adenomas and carcinoma. Patients with FAP have a markedly increased incidence of adenomas of the ampulla, approaching 50 to 86% (6). Patients with FAP tend to develop adenomas in their second to fifth decades of life and thus are much younger than those patients with periampullary adenomas without FAP. It does appear, however, that these patients have a much lower incidence of malignant transformation in longterm followup as compared to those with colon adenomatous disease. Nevertheless, the risk of duodenal and periampullary cancer is over 100 times that of the normal population, supporting the role for routine screening and surveillance in FAP patients (7). It is currently recommended that patients with FAP undergo initial endoscopic screening with a wideviewing duodenoscope between the ages of 20 and 25, with repeat examinations every 3 years. Most recently, studies of its molecular genetics have also supported the adenoma—carcinoma concept. Immunoreactivity for the p53 tumor suppressor gene has been shown in the majority of neoplasms of the ampulla of Vater (8). The incidence of kras, the widely studied oncogene present in 80 to 90% of patients with pancreatic adenocarcinoma, has been found in only 13 to 40% of patients with ampullary adenocarcinoma (9,10). This frequency is more like that of bile duct carcinoma than that of pancreatic cancer.
Page 757
IV— Diagnosis and Staging A— Clinical Presentation The average age of diagnosis for ampullary adenomas is in the mid50s. The age range of patients at the time of diagnosis of carcinoma of the ampulla is 20 to 91 years, with a peak in the seventh decade of life. There does not appear to be any gender predilection. Jaundice is the most common presenting symptom and is seen in approximately 80% of patients with both adenomas and adenocarcinoma at the time of diagnosis. Nonspecific gastrointestinal symptoms such as dyspepsia, malaise, and anorexia can also be seen. Often these symptoms may predate the development of jaundice by several months. Jaundice is generally progressive, but it may be intermittent in a small percentage of patients. Weight loss occurs in 75% of patients, while abdominal pain occurs in half. Hematemesis or melena are rare, but occult gastrointestinal bleeding from neoplastic ulceration can be seen in up to onethird of all patients. The triad of intermittent, painless jaundice, anemia, and a palpable, large gallbladder has been considered relatively specific for ampullary carcinoma; however, this triad is seen in less than 10% of patients. Finally, rare patients with ampullary carcinoma can present with pancreatitis due to obstruction of the pancreatic duct. Typical findings on physical examination include scleral or cutaneous icterus, hepatomegaly and/or distended palpable gallbladder, and occasionally positive stool tests for occult blood. B— Laboratory Tests The majority of patients with ampullary adenomas and adenocarcinoma will have abnormalities of serum liver function tests. The earliest and often the only abnormality is an increase in alkaline phosphatase. As more biliary obstruction occurs, jaundice and hyperbilirubinemia occur, as well as increases in serum aminotransaminases. No tumor markers associated with ampullary neoplasms have been identified. C— Radiological Imaging The early diagnosis of periampullary carcinoma requires an appropriate level of clinical suspicion and aggressiveness in pursuing the diagnosis. The prompt evaluation of patients with jaundice offers the opportunity for an early diagnosis. Any patient presenting with jaundice should undergo diagnostic imaging with either ultrasonography or computed tomography (CT). Both tests will confirm the obstructive nature of the jaundice by demonstrating a dilated intra and extrahepatic biliary tree and therefore focuses the next step in evaluation. Conventional ultrasound is often useful for initial evaluation of patients presenting with abdominal pain or suspected of obstructive jaundice, as dilated intra and extrahepatic bile ducts are readily seen. Other important findings that can be routinely imaged with ultrasound include gallstones, ascites, and liver metastasis. However, a major limitation to ultrasound is the occurrence of a technically inadequate examination in 15 to 20% of patients. In contrast, the advantages include the ease and availability of the examination, the lack of radiation exposure, and its relatively inexpensive nature. CT, however, is probably the most costeffective test. It can detect the presence of a periampullary mass of at least 2 cm in size and also provides important information about the level of biliary obstruction with respect to the pancreatic parenchyma if no mass is seen (Fig. 1). Typically patients with biliary obstruction due to ampullary tumors will show dilatation of the bile duct into the pancreatic segment down to the ampulla. Intravenous and oral contrastenhanced spiral CT is currently the optimal technique for evaluation of the periampullary area. Scans obtained through the rapid intravenous injection of an iodinated contrast agent result in an increase in pancreatic parenchymal attenuation as well as excellent contrast enhancement of the major peripancreatic blood vessels. This technique not only results
Page 758
Figure 1 Computed tomography (CT) scans of a patient with obstructive jaundice due to ampullary carcinoma. a. Scan demonstrates a 3cm ampullary mass. b. Scan at a higher level demonstrates bile duct dilatation within the pancreatic parenchyma, indicating distal ductal obstruction. (From Ref. 42.) c. CT scan of a patient with an ampullary carcinoma. Note the dilated bile duct and pancreatic duct with no mass. (From Ref. 31.)
Page 759
Figure 1 Continued.
in clear delineation of the tumor but also demonstrates the involvement of adjacent major visceral vessels, suggesting unresectability. Finally, CT can detect liver metastases, ascites, and often evidence of peritoneal metastasis. Upper gastrointestinal (GI) barium studies or hypotonic duodenography can be used to evaluate the duodenum for mucosal abnormalities. These radiographic studies, however, have been essentially replaced by endoscopic evaluation. In the past, mesenteric angiography has been used to evaluate for major vessel invasion due to periampullary malignancy. The incidence of major vascular involvement with ampullary carcinoma, however, is very rare. Furthermore, with recent spiral CT availability, mesenteric angiography is seldom indicated. The most recent additions to the noninvasive imaging techniques for ampullary carcinoma include magnetic resonance imaging. The combination of current techniques can both demonstrate an ampullary mass and also, using magnetic resonance cholangiopancreatography (MRCP), provide noninvasive images of the biliary tree comparable to those obtained with invasive cholangiography. D— Endoscopy/Cholangiography Once biliary obstruction has been confirmed by imaging studies, traditionally the next step in the evaluation of the jaundiced patient has been cholangiography. Endoscopic retrograde cholangiography is the optimal procedure for patients suspected of having an ampullary neoplasm. The endoscopic component of this exam will define the extent, size, and gross appearance of the lesion and allow for simultaneous performance of endoscopic biopsy and cytologic brushings (Fig. 2). The endoscopic appearance of an ampullary tumor may be similar for benign and malignant tumors; furthermore, endoscopic biopsies of a periampullary malignancy may be inaccurate, yielding falsenegative results, largely due to sampling error in 15 to 25% of patients (11,12). The demonstration of malignancy on biopsy specimen is definitive, but a diagnosis of benign adenoma does not rule out the presence of an adenocarcinoma elsewhere in the adenoma. Therefore, regardless of whether the biopsy shows a malignant or benign lesion, complete resection (either operative or endoscopic) will be warranted, both to relieve biliary obstruction and to detect a potentially malignant lesion.
Page 760
Figure 2 a. Endoscopic appearance of a benign villous adenoma. (From Ref. 42.) b. Endoscopic appearance of an ulcerated ampullary adenocarcinoma.
Cholangiography in patients with an ampullary neoplasm will demonstrate a combination of proximal dilatation of the biliary tree and obstructing, irregular filling defect of the very distal common bile duct (Fig. 3). The choice of either endoscopic retrograde pancreatography (ERCP) or the percutaneous transhepatic route (PTC) depends on the apparent level of obstruction, the gut anatomy, the presence or absence of coagulopathy and local expertise. The advantages of visualizing and performing an endoscopic biopsy, however, make the endoscopic route preferable in patients with suspected ampullary lesions. After cholangiography, a biliary stent can be placed to relieve biliary obstruction in jaundiced patients. Stenting is not indicated
Page 761
Figure 3 Endoscopic retrograde pancreatography demonstrating an ampullary carcinoma obstructing the distal bile duct. (From Ref. 42.)
routinely; however, in selected patients with advanced malnutrition, sepsis, or correctable metabolic conditions, or while awaiting further workup or operation, preoperative biliary drainage can be useful. Recent reports, however, suggest that the use of biliary stents may be associated with an increase in perioperative morbidity and mortality associated with surgical resection (13). Endoscopic ultrasound (EUS) has been reported to be a reliable and accurate technique in assisting in the diagnosis and staging of ampullary carcinoma (Fig. 4). Real timeEUS enables one to evaluate and integrate in the same examination mucosal, vascular, ductal, and parenchymal abnormalities. To obtain similar information will require endoscopy for mucosa, visceral angiography for arterial and venous structures, ERCP for ducts, and CT and/or US for parenchyma and lymph nodes. In one prospective study of 14 ampullary tumors, 13 (93%) were imaged correctly by EUS, while successful detection of ampullary tumors occurred in only 7% and 29% of patients by ultrasound and CT scan, respectively (14). However, this imaging modality is extremely dependent upon the experience of the endoscopic ultrasonographer both in performing the procedure and in interpreting the images. EUS has also provided a method of selecting patients for local resection of ampullary tumors. It is essential to acknowledge, however, that EUS cannot replace histological evaluation. Although EUS cannot differentiate a T1 carcinoma (limited to the mucosa) from an adenoma, T3 and T4 tumors are easily differentiated from an adenoma or early carcinoma by EUS. In a series by Mukai and colleagues, EUS accurately defined depth of wall penetration in 78% of ampullary carcinomas (15). Similarly, Quirk and colleagues have found the sensitivity and specificity of EUS for differentiating local from advanced ampullary tumors were both 83% (16). Underestimating the depth of tumor penetration seldom occurs, while overestimation is more common and is often due to edema of the submucosa from associated pancreatitis, which occurs in up to onethird of T1 lesions. Finally, the most important criteria for determining malignancy of an ampullary neoplasm is confirmation by histological diagnosis. Although multiple endoscopic biopsies can detect
Page 762
Figure 4 Endoscopic ultrasonography of an ampullary tumor, represented by hypoechoic area on the right. An endoprosthesis can be seen running through the center of the tumor (black arrows). The tumor infiltrates beyond the muscularis propria into the pancreas (white arrows). (From Ref. 42.)
malignancy in most patients with ampullary carcinoma, up to 25% of such biopsies will be negative (11,12). A recent series from the Massachusetts General Hospital reported the sensitivity of endoscopic biopsy for detecting malignancy of only 42%, with a specificity of 70%. The positive predictive value of endoscopic biopsy for ampullary neoplasms was 50%, with a negative predictive value of 73% (17). The use of cytological brushings may also be helpful in the determination of malignancy in some tumors; however, it may still be associated with the same difficulties in distinguishing severe dysplasia from invasive carcinoma. The accuracy of the biopsy or brushings for diagnosis of carcinoma can be related to the gross morphological characteristics of the tumor: 50% for the intramural or intraampullary type, 64% for the protruding type, and 88% for the ulcerating type (18). The problem of inaccurate diagnosis can also be extended to frozensection analysis of resected specimens, which can fail to detect malignancy in 14% of patients (19). It must be concluded, therefore, that without complete excision and detailed histological evaluation, malignancy cannot be completely excluded. V— Management A— Staging The primary staging system currently used for ampullary carcinoma is the TMN system (Table 1). Ampullary carcinoma is classified according to the extent of tumor invasion, tumor size, presence of lymph node metastasis, and presence of distant metastasis. Because ampullary cancers are both more often resectable and when resected more often curable than pancreatic cancer, most surgeons feel that extensive preoperative staging is unnecessary. Spiral CT with bolus intravenous contrast will detect liver metastases of 1 cm in diameter or greater, major vessel invasion, and ascites. Although other staging modalities such as angiography and
Page 763 Table 1 TNM Staging of Ampullary Carcinoma Tumor T1
Tumor limited to ampulla of Vater
T2
Tumor invades duodenal wall
T3
Tumor invades 2 cm into pancreas
T4
Tumor invades >2 cm into pancreas and/or adjacent organs
Nodes N0
No regional lymph node metastases
N1
Regional lymph node metastases
Metastases M0
No distant metastases
M1
Distant metastases
Stages
T
N
M
Stage I
T1
N0
M0
Stage II
T2–T3
N0
M0
Stage III
T1–T3
N1
M0
Stage IV
T4
any T
M0
any T
any T
M1
laparoscopy have been advocated for pancreatic cancer, these invasive procedures would appear neither costeffective nor appropriate with respect to potential risks for patients with ampullary carcinoma. Therefore, patients who are good operative risks without evidence of distant metastasis should undergo surgical exploration. When preoperative evaluation reveals distant metastases or when the patient is not an operative candidate because of serious medical problems, nonoperative palliation with a biliary endoprosthesis should be performed and palliative chemotherapy considered. B— Local Excision In 1899, Halsted reported the first successful local excision of ampullary carcinoma, reimplanting the pancreatic and bile ducts into the duodenal wall (1). Currently, local resection of ampullary tumors has been advocated for patients with benign adenomas, rare patients with ampullary neuroendocrine tumors, and highly selected patients with ampullary carcinoma. The treatment options for local management of neoplasms of the ampulla are endoscopic snare excision or ablation or surgical ampullectomy. Small tubular adenomas of the papilla with a maximum diameter of less than 1 cm without severe dysplasia can be adequately treated by endoscopic means using snare excision, usually in conjunction with endoscopic sphincterotomy. Endoscopic excision of an adenoma, however, incurs the risk of incomplete resection and local recurrence in up to 20% of patients. This observation has been confirmed by Binmoeller and colleagues, who observed six recurrences after endoscopic resection after a median followup of 37 months (20). Operative resection of the ampulla is indicated in patients with large adenomas (Fig. 5). Ampullectomy is performed through either a midline or a subcostal incision. As with all resections, careful exploration to rule out metastatic disease or other evidence of resectability is imperative. A generous Kocher maneuver allows for complete inspection of the duodenum or pancreatic head. If the tumor is resectable, a longitudinal duodenotomy is made that is centered over the ampulla (passage of a biliary Fogarty catheter through the cystic duct into the common
Page 764
Figure 5 Technique of local resection of an ampullary neoplasm. a. A duodenotomy and wide transmural resection of the periampullary mass including a portion of the duodenal wall and the periampullary portions of the pancreatic and common bile ducts. b. Anastomosis of the common bile duct and pancreatic duct. c. Mucosatomucosa anastomosis of the common bile duct/pancreatic duct to duodenal mucosa. (From Ref. 21.)
Page 765
Figure 5 Continued.
duct and out into the duodenum may aid in the placement of the duodenotomy). One then excises the tumor completely, taking a full thickness of the duodenal wall and including segments of the bile and pancreatic ducts. If necessary, the two ducts are sutured together to form a common septum, and the posterior wall of the duodenum is approximated to the joined pancreatic and common duct openings. Following local resection, it is recommended that patients undergo annual upper GI endoscopy to detect evidence of local recurrence or the development of new lesions. Early postoperative morbidity after local resection of the ampulla is considerably less than after pancreaticoduodenectomy. Hospital mortality after ampullectomy in recently reported series has been below 2% (17,21–24), with the length of hospital stay generally in the range of 10 to 12 days. The longterm followup after ampullectomy for benign adenomas would suggest that the incidence of local recurrence is low, likely less than 20%. In most cases, followup endoscopic evaluation can allow endoscopic reexcision or laser ablation. In a review of the recent literature, Beger and colleagues identified 62 patients reported in the literature undergoing ampullectomy for benign neoplasms. All of these patients were free of disease at followup ranging from 1 to 156 months (25). In contrast, Branum and colleagues reported that 5 of 19 patients with benign disease resected by ampullectomy had recurrence at a mean of 35 months (range 8 to 72 months). Three of the five patients were treated with laserdiathermy ablation while one underwent local reexcision and one patient underwent a Whipple procedure. Of note, two of these patients also had familial polyposis syndromes. More concerning, however, is a recent report from the Mayo Clinic (24) suggesting that these patients may be at risk for the development of adenocarcinoma at the site of recurrence. In that series of 50 patients with benign villous tumors managed by local excision, 17 recurred, with actuarial rates of recurrence of 32% at 5 years and 43% at 10 years. Four of the recurrences (24%) were adenocarcinoma, with one of the four patients unresectable at the time of recurrence. The recurrence rate in this series was influenced by the presence of a known polyposis syndrome but not by tumor size. These results have led these authors to strongly support annual followup with GI endoscopy surveillance and biopsy following local excision for all patients and to strongly consider removal of all mucosa at risk (duodenum) by pancreaticoduodenectomy.
Page 766
In addition to local excision of benign lesions, some groups have advocated local ampullary excision for patients with T1 cancers. The limitations of this technique are related both to inadequate local resection as well as the uncertainty about the degree of extension of the cancer into the wall of the duodenum and pancreatic head. In addition, up to 10% of patients with a T1 lesion may have positive lymph nodes in the pancreaticoduodenectomy specimen (26). Review of recent series examining the longterm outcome following local resection for adenocarcinoma has demonstrated that the 5year survival is approximately 40% (22,26,27). These results would appear to be less favorable than recent survival results following pancreaticoduodenectomy for ampullary carcinoma with overall survival rates approaching 50% at 5 years. In patients in whom a local resection of an adenoma containing a small focus of carcinoma in situ or a T1NOMO cancer of the ampulla is being considered, the decision may be altered by operative findings. If intraoperative findings show a cancer more advanced than a T1 or a highgrade tumor on frozen section review of the ampullectomy specimen, the procedure should be converted to a pancreaticoduodenectomy. Similarly, if any lymphatic metastasis or lymphatic infiltration is detected, extension to a pancreaticoduodenectomy is also indicated. Unfortunately, sometimes the initial intraoperative frozen section review does not fully stage the disease. This creates a difficult problem when the pathologist finds a more advanced lesion on permanent section than was suspected by intraoperative frozen section. In such cases, it is generally recommended that in the early postoperative period after completion of a transduodenal ampullectomy, the patient be returned to the operating room for a pancreaticoduodenectomy to provide the best chance of cure. Because of these difficult uncertainties, many surgeons advocate a pancreaticoduodenectomy for all patients with any evidence of carcinomatous invasion in a periampullary adenoma regardless of the stage. C— Pancreaticoduodenectomy A report by Whipple and colleagues in 1935 described the first twostage pancreaticoduodenal resection in three patients with ampullary carcinoma (28). Since that initial report, pancreaticoduodenectomy has undergone a number of technical modifications. For years, acceptance of this procedure in the treatment of periampullary malignancy was limited by the high complication rate and operative mortality approaching 25%. Recent advancements in surgical technique, anesthesia, and pre and postoperative care have resulted in a decrease in morbidity rate (although still in the range of 40%) and, more importantly, a decrease in the perioperative mortality rate to less than 5% in most experienced centers (29–32). Therefore, pancreaticoduodenectomy is the curative procedure of choice for almost all patients with ampullary carcinoma. Either classic pancreaticoduodenectomy, which includes an antrectomy, or the pyloruspreserving modification are suitable alternatives in patients with carcinoma of the ampulla. In general, local lymph node resection appears appropriate, with no indication for extended lymphadenectomy to include the N2 lymph nodes. Regardless of the type of pancreaticoduodenectomy performed, the incidence of postoperative complications remains high and ranges from 25 to 50% (29–32). Delayed gastric emptying and anastomotic leaks at the pancreaticojejunostomy are the most common significant postoperative complications. Despite the high rate of complications, postoperative death occurs in less than 5% of patients, with several large series reporting consecutive series of patients with no mortalities (29–32). In 1997 the group at The Johns Hopkins Hospital reported the largest single institutional experience in the management of adenocarcinoma of the ampulla of Vater (33). A total of 120 patients with adenocarcinoma were managed over a 28year period. Resection was performed in 106 patients (88%), and 105 of these (99%) underwent either a pancreaticoduodenectomy (n = 103) or total pancreatectomy (n = 2). The resection rate at our institution increased from 62% in the 1970s to 82% in the 1980s to 96% in the 1990s. Overall mortality following resection was 3.8%, with no mortality in the 45 consecutive patients resected in the past 5
Page 767
years. One or more complications occurred in 49 resected patients, for an overall morbidity rate of 47%. The most common complication was pancreatic fistula, which occurred in 23 patients (25%). This incidence of pancreatic fistula would appear to be higher than that seen at our institution in other series (30,32,34). This likely represents the increased risk of a pancreatic anastomotic leak associated with the ''soft" gland frequently found in neoplasms of the duodenum and ampulla not associated with obstructive pancreatitis. D— Chemotherapy and Radiation Therapy Currently there are no data supporting the use of postoperative chemotherapy or radiation therapy in an adjuvant setting. Combined modality therapy using 5 fluorouracil—based chemotherapy with externalbeam radiation improves survival in patients with resected adenocarcinoma in the head of the pancreas (35). It is tempting to extrapolate these results to ampullary carcinoma, although no large series provides good supportive data. Limited data suggest that postoperative adjuvant combination chemotherapy alone (using fluorouracil, doxorubicin, and mitomycin C) delays tumor recurrence in patients with ampullary carcinoma, but more data are needed (36). E— Palliation In patients with ampullary carcinoma who, based on preoperative staging studies such as abdominal CT scan, reveal no evidence of unresectability, the resection is possible in more than 85% (33). In a minority of patients, surgical exploration reveals unanticipated metastatic tumor outside of the resection specimen or vascular involvement that precludes curative resection. In this situation, operative palliation is appropriate. Decompression of the biliary tree is performed as a biliaryenteric anastomosis. We prefer to perform a RouxenY hepaticojejunostomy, using a defunctionalized jejunal limb and performing a cholecystectomy. A gastrojejunostomy is performed in patients with a compromised duodenal lumen or who have symptoms of gastric outlet obstruction. A recent series of 256 patients with periampullary carcinoma at The Johns Hopkins Hospital reported at 3.1% overall mortality, 22% perioperative morbidity, and a 10.3day mean postoperative length of stay (37). Survival in patients with unresectable ampullary carcinoma is similar to that of other patients with unresectable periampullary cancers, with median survivals from 5 to 9 months (33,38). In patients determined preoperatively to have unresectable disease, surgical intervention for palliation is generally not indicated. The placement of a biliary endoprosthesis through the ampullary region to provide biliary decompression is the standard approach for the palliation of jaundice. Using either a plastic endoprosthesis or newer metallic expandable stents, this method allows internal drainage of the bile into the duodenum and avoids the placement of an external apparatus. VI— Survival The overall 5year survival rates for patients with ampullary cancer undergoing radical resection range from 25 to 55% (32,33,38–41). These results are significantly better than what has generally been reported for pancreatic carcinoma (Fig. 6). In the recent series of patients of 120 patients with carcinoma of the ampulla of Vater reported from Hopkins, the median and actuarial 5year survivals were 46 months and 38% respectively (33). Tumor size did not affect survival, but lymph node statues, tumor differentiation and perioperative blood transfusions were significant factors in longterm survival (Fig. 7). Adjuvant therapy had no discernible influence on survival, but the number of patients treated was small. Similar findings were recently reported from the Memorial SloanKettering Cancer Center (38). A total of 101 patients underwent resection of an ampullary carcinoma—99 by pancre
Page 768
Figure 6 The tumorspecific actual 5year survival curves for 242 patients treated by pancreaticoduodenectomy for periampullary adenocarcinoma. (From Ref. 39.)
Figure 7 a. Influence of resection on Outcome for ampullary carcinoma. b. Influence of blood transfusion on outcome for ampullary carcinoma. c. Influence of lymph node status on outcome for ampullary carcinoma. d. Influence of tumor differentiation on outcome for ampullary carcinoma. (From Ref. 33.)
Page 769
aticoduodenectomy and 2 by local excision (one of whom later underwent pancreaticoduodenectomy for recurrence). The median survival in that series was 58.8 months, with a 46% survival at 5 years. Factors that were significantly correlated with improved survival in resected tumors by univariate analysis included negative margins, good or moderate tumor differentiation, and negative nodes. When margins, differentiation, and nodal status were examined as covariates by Cox regression, only nodal status and margins were independently correlated with survival. Factors not correlated with survival included perineural invasion, tumor size, transfusion, T stage, and vascular invasion. Like the experience at Johns Hopkins, ampullary tumors have the second highest survival of the periampullary carcinomas, with better survival observed only for duodenal carcinoma. The pattern of failure following pancreaticoduodenectomy for ampullary carcinoma was addressed at The Massachusetts General Hospital in a retrospective review of 29 patients undergoing pancreaticoduodenectomy (41). The 5year survival rate was 55%. Patients were classified into low and highrisk groups based on pathological features suggesting a risk of local recurrence (tumor invasion of the pancreas, poorly differentiated histological findings, involved lymph nodes, or resected margins). The patients in the highrisk group had a 50% incidence of local control at 5 years, with an overall survival of 38%. In contrast, patients with a more favorable pathology had 100% local control and an 80% survival. In order the try to obtain better local control, postoperative radiation therapy was given to 12 of the 17 highrisk patients after pancreaticoduodenectomy. There was a trend toward better local control (83%) but no improvement in survival. Distant metastases were the dominant factor in predetermining the outcome of this group. VII— Conclusion Ampullary tumors, although not the most common, can be among the most rewarding of periampullary malignancy to manage. Patients with ampullary tumors present clinically with jaundice and pruritus in a similar fashion to those with other periampullary cancers. The key to diagnosis in the jaundiced patient is the CT image demonstrating an obstructive pattern and endoscopy to identify and potentially biopsy the ampullary mass. Although a positive endoscopic biopsy can provide pathological verification of the diagnosis in a large percentage of patients with ampullary carcinoma, tissue diagnosis from endoscopic biopsy may be inconclusive or inaccurate. Endoscopic ultrasound can provide information regarding the depth of tumor penetration, suggesting the benign versus malignant nature of the process. However, complete histological examination of the resected specimen is essential for obtaining the final diagnosis. In almost all patients with ampullary carcinoma, pancreaticoduodenectomy is the operation of choice. Local excision may be performed in carefully selected patients with benign ampullary neoplasms or particularly highrisk patients with lowgrade ampullary malignancies. The overall 5year survival rate in patients with resected ampullary adenocarcinoma is approximately 50% and is largely determined by the status of resected lymph nodes and margins at resection. At present there is no evidence that adjuvant therapy is beneficial in this disease. In those patients found to be unresectable at the time of presentation, either operative or endoscopic palliation is indicated due to the overall short expected life span. References 1. Halsted WS. Contributions to the surgery of the bile passages, especially of the common bile duct. Boston Med J 1899; 141:645–654. 2. Anderson JB, Cooper MJ, Williamson RCN. Adenocarcinoma of the extrahepatic biliary tree. Ann R Coll Surg Eng 1985; 67:139–143.
Page 770
3. Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell 1990; 61: 759–767. 4. Cattell RB, Braasch J, Kahn F. Polypoid epithelial tumors of the bile ducts. N Engl J Med 1962; 266:57–61. 5. Baczakok B, Büchler M, Beger HG, Kirkpatrick CJ, Haferkamp O. Morphogenesis and possible precursor lesions of invasive carcinoma of the papilla of Vater: epithelial dysplasia and adenoma. Hum Pathol 1985; 16:305–310. 6. Yao T, Iida M, Ohsato K, Watanabe H, Omae T. Duodenal lesions in familial polyposis of the colon. Gastroenterology 1977; 73:1086–1092. 7. Offerhaus GJA, Giardiello FM, Krush AJ, Booker SV, Tersmette AC, Kelly NC, Hamilton SR. The risk of upper gastrointestinal cancer in familial adenomatous polyposis. Gastroenterology 1992; 102:1080–1982. 8. The M, Wee A, Raju GC. An immunohistochemical study of p53 protein in gallbladder and extrahepatic bile duct/ampullary carcinomas. Cancer 1994; 74:1542– 1545. 9. Motojima K, Tsunoda T, Kanematsu T, Nagata Y, Urano T, Shiku H. Distinguishing pancreatic carcinoma from other periampullary carcinomas by analysis of mutations in the Kirstenras oncogene. Ann Surg 1991; 214:657–662. 10. Chung CH, Wilentz; RE, Polak MM, Ramsoekh TB, Noorduyn LA, Gouma DJ, Huibregste K, Offerhaus GJ, Slebos RJ. Clinical Significance of Kras oncogene activation in ampullary neoplasms. J Clin Pathol 1996; 49:460–464. 11. Komorowski RA, Beggs BK, Geenan JE, Veuu RP. Assessment of ampulla of Vater pathology: an endoscopic approach. Am J Surg Pathol 1991; 15:1188– 1196. 12. Ryan DP, Shapiro RH, Warshaw AI. Villous tumors of the duodenum. Ann Surg 1986; 203:301–306. 13. Povoski SP, Karpeh MS, Conlon KC, Blumgart LH, Brennan MF. Association of preoperative biliary drainage with postoperative outcome following pancreaticoduodenectomy. Ann Surg 1999; 230:131–142. 14. Rosch T, Braig C, Gain T, Feuerbach S, Siewert JR, Schusdziarva V, Classen M. Staging of pancreatic and ampullary carcinoma by endoscopic ultrasonography: comparison with conventional sonography, computed tomography, and angiography. Gastroenterology 1992; 102:188–199. 15. Mukai H, Nakajima M, Yasuela K, Mizuno S, Kawai K. Evaluation of endoscopic ultrasonography in the preoperative staging of carcinoma of the ampulla of Vater and common bile duct. Gastrointest Endosc 1992; 38:676–683. 16. Quirk DM, Rattner DW, Castillo, FC, Warshaw AL. The use of endoscopic ultrasonography to reduce the cost of treating ampullary tumors. Gastrointest Endosc 1997; 46:334–337. 17. Rattner DW, Fernandezdel Castillo C, Brugge WR, Warshaw AL. Defining the criteria for local resection of ampullary neoplasms. Arch Surg 1996; 131:366– 371. 18. Yamaguchi K, Enjoji M, Kitamura K. Endoscopic biopsy has limited accuracy in diagnosis of ampullary tumors. Gastrointest Endosc 1990; 36:588–592. 19. Sharp KW, Brandes JL. Local resection of tumors of the ampulla of Vater. Am Surg 1990; 58:214–217. 20. Binmoeller KF, Boaventura S, Ramsperger K, Soehendra N. Endoscopic snare excision of benign adenomas of the papilla of Vater. Gastrointest Endosc 1993; 39:127–131. 21. Asburn HJ, Rossi RL, Munson JL. Local resection for ampullary tumors: is there a place for it? Arch Surg 1993; 128:515–520. 22. Branum GD, Pappas TN, Meyers WC. The management of tumors of the ampulla of Vater by local resection. Ann Surg 1996; 224:621–627. 23. Bjork KJ, Davis CJ, Nagorney DM, Mucha P. Duodenal villous tumors. Arch Surg 1990; 125:961–965. 24. Farnell MB, Sakorafas GH, Sarr MG, Rowland CM, Tsiotos GG, Farley DR, Nargorney DM. Villous tumors of the duodenum: reappraisal of local versus extended resection. J Gastrointest Surg 2000; 4:13–21.
Page 771
25. Beger HG, Treitschke F, Poch P, Schoenberg MH. Adenoma of the ampulla of Vater: operative treatment and results. In: Beger HG, Warshaw AL, Buchler MW, CarrLocke DL, Russell C, Sarr MG, eds. The Pancreas. Oxford, England: Blackwell, 1998, pp 1324–1327. 26. Tarazi RY, Hermann RF, Voyt DP. Results of surgical management of periampullary tumors: a 35year experience. Surgery 1986; 100:716–723. 27. Klein P, Reingruber B, Kart LS, Dvorak O, Hohenberger W. Is local excision of pT1 ampullary carcinomas justified? Eur J Surg Oncol 1996; 22:366–371. 28. Whipple AO, Parsons WB, Mullins CR. Treatment of carcinoma of the ampulla of Vater. Ann Surg 1935; 102:763–779. 29. Trede M, Schwall G, Saeger HD. Survival after pancreaticoduodenectomy: 118 consecutive resections without an operative mortality. Ann Surg 1990; 211:447–458. 30. Cameron JL, Pitt HA, Yeo CJ, Lillemoe KD, Kaufman HS, Coleman J. One hundred and fortyfive consecutive pancreaticoduodenectomies without mortality. Ann Surg 1993; 217:430–438. 31. Fernandezdel Castillo C, Rattner DW, Warshaw AL. Standards for pancreatic resection in the 1990's. Arch Surg 1995; 130:295–300. 32. Yeo CJ, Cameron JL, Sohn TA, Lillemoe KD, Pitt HA, Talamini MA, Hruban RH, Ord SE, Sauter PK, Coleman J, Zahurak ML, Grochow LB, Abrams RA. Six hundred fifty consecutive pancreaticoduodenectomies in the 1990's. Ann Surg 1997; 226:248–260. 33. Talamini MA, Moesinger RC, Pitt HA, Sohn TA, Hruban RH, Lillemoe KD, Yeo CJ, Cameron JL. Adenocarcinoma of the ampulla of Vater: a 28 year experience. Ann Surg 1997; 225:590–600. 34. Yeo CJ, Cameron JL, Maher MM, Sauter PK, Zaharak ML, Talamini ML, Lillemoe KD, Pitt HA: A prospective randomized trial of pancreaticogastrostomy versus pancreaticojejunostomy after pancreaticoduodenectomy. Ann Surg 1995; 222:580–592. 35. Kalser MH, Ellenberg SS. Pancreatic Cancer. Adjuvant combined radiation and chemotherapy following curative resection. Arch Surg 1985; 120:880–903. 36. Bakkevold KE, Arnesjo B, Dahl O, Kambestad B. Adjuvant combined chemotherapy (AMF) following radical resection of carcinoma of the pancreas and papilla of Vater—results of a controlled prospective, randomized multicentre trial. Eur J Cancer 1993; 29A: 698–703. 37. Sohn TA, Lillemoe KD, Cameron JL, Huang JJ, Pitt HA, Yeo CJ. Surgical palliation of unresectable periampullary adenocarcinoma in the 1990's. J Am Coll Surg 1999; 188:658–669. 38. Howe JR, Klimstra DS, Moccia RD, Conlon KC, Brennan MF. Factors predictive of survival in ampullary carcinoma. Ann Surg 1998; 228:87–94. 39. Yeo CJ, Sohn TA, Cameron JL, Hruban RH, Lillemoe KD, Pitt HA. Periampullary adenocarcinoma: Analysis of 5year survivors. Ann Surg 1998; 227:821– 831. 40. Allema JH, Reinders ME, vanGulik TM, vanLeeuwen DJ, Verbeek PCM, deWit LT, Gouma DJ. Results of pancreaticoduodenectomy for ampullary carcinoma and analysis of prognostic factors for survival. Surgery 1995; 117:247–253. 41. Willett CG, Warshaw AL, Convery K, Compton CC. Patterns of failure after pancreaticoduodenectomy for ampullary carcinoma. Surg Gynecol Obstet 1993; 176:33–38. 42. Martin SA, Lillemoe KD. Periampullary tumors: clinical presentation and diagnostic strategy. In: Beger HG, Warshaw AL, Buchler MW, Carr Locke DL, Neptolemos JP, Russell C, Sarr MG, eds. The Pancreas. Oxford, England: Blackwell, 1998.
Page 773
35— Infections of the Bile Ducts Andrew P. Keaveny Boston University School of Medicine, Boston, Massachusetts I— Introduction The bile ducts may become colonized with bacterial, fungal, protozoal, and viral agents, resulting in a variety of clinical syndromes encompassed by the term cholangitis. The presentation varies from acute overwhelming sepsis to chronic infection complicated by biliary tract and liver parenchymal damage. Multiple factors determine the presentation and consequences of an infection. These include the patient's underlying condition and immune status as well as the infecting agent(s). Specific organisms are endemic to certain geographical regions and infect patients who originate from these areas. Biliary tract infections result in significant worldwide morbidity and mortality. This chapter discusses the most common clinical presentation of biliary infections, acute bacterial cholangitis. Fungal infections, recurrent pyogenic cholangitis (RPC), parasitic infestations of the biliary tract, and the cholangiopathy associated with the acquired immunodeficiency syndrome (AIDS cholangiopathy) are also reviewed. II— Acute Bacterial Cholangitis A— Introduction Acute bacterial cholangitis develops when bacterial infection complicates any form of obstruction within the biliary tract. In 1877, Charcot described the clinical triad of rightupperquadrant (RUQ) pain, fever, and jaundice (1). Reynolds and Dargan later added mental confusion and hypotension to Charcot's triad in describing Reynolds' pentad and the syndrome of acute obstructive cholangitis (2). Prompt evaluation and treatment of a patient with bacterial cholangitis is critical. The administration of appropriate antimicrobial therapy and biliary tract decompression remain the essential principles of management (3). Nonsurgical methods of biliary drainage have revolutionized the approach to cholangitis, reducing the mortality of this potentially devastating illness to less than 10% (4–6). B— Pathogenesis Bacterial cholangitis arises when bile becomes infected with bacteria in a partially or completely obstructed biliary tree. The source of biliary tract bacteria, or bactibilia, has received considerable attention over the years. Several routes have been proposed, including spread via the lymphatic system or through secretion from the liver or infected gallbladder (7,8). However,
Page 774
most studies support either direct extension of bacteria from the duodenum or hematogenous spread through the portal venous system (9,10). Bactibilia per se does not indicate cholangitis, as it has been found in 15% of patients undergoing elective cholecystectomy and in patients with common bile duct (CBD) stones, or who have undergone endoscopic sphincterotomy or surgical sphincteroplasty (11–14). Normally, the sphincter of Oddi acts to prevent the entry of bacteria into the CBD from the duodenum. Continuous bile flow, with secretion of mucus and immunoglobulin from the cholangiocytes, also acts to ensure bile duct sterility (14,15). The predominant immunoglobulin, secretory IgA, along with mucins probably act as antiadherence factors. However, duodenalbiliary reflux is facilitated when the sphincter of Oddi is disrupted after sphincterotomy, biliary stent placement, or biliaryenteric bypass surgery (16). Microbiological evidence supporting the role of ascending infection from the duodenum includes a recent study finding that 63% of patients undergoing elective biliary surgery for calculous disease had similar strains of bacteria isolated from the CBD, gallbladder, and duodenum (17). Suzuki et al. demonstrated that 61% of organisms cultured from intrahepatic bile were identical to those recovered from duodenal fluid in 30 patients with benign biliary tract disease (18). Bactibilia occurs in 80 to 90% of patients with choledocholithiasis and benign biliary strictures compared with 30 to 50% of patients with malignant obstruction (3,19). Therefore, bacterial movement from the duodenum to the CBD appears more likely to occur in the presence of partial rather than complete biliary obstruction (11). Stones usually cause intermittent obstruction, whereas malignant obstruction is slower to develop and will eventually become complete. Ampullary tumors appear to be the exception, with bactibilia reported in 65% of cases, presumably secondary to intermittent obstruction (18). However, it is not fully understood why different etiologies of biliary obstruction result in differences in the prevalence of bactibilia, and the duodenal origin of bacteria has been questioned (3,20). Presumably partial obstruction must, in general, precede complete CBD obstruction, thus providing sufficient time for ascending colonization to occur. Furthermore, early studies in healthy volunteers found that coliforms, which are the most commonly isolated organisms in cholangitis, colonized the proximal small bowel infrequently (21,22). However, changes with age and other factors such as hypochlorhydria may alter the bacterial population in the duodenum and jejunum (18,23–25). An alternative mechanism for bactibilia based on animal studies is bacterial translocation from the portal venous system (26,27). In the guinea pig model, the portal circulation was the most important route by which injected bacteria were delivered to the biliary tract compared with either the systemic or lymphatic systems (26). In addition, bile duct obstruction in that model caused further increases in biliary bacterial concentration after portal bacteremia was achieved through splenic injection. Sung et al. demonstrated that bacteria infused into the splenic vein were isolated earlier and at higher levels in bile of cats with ligated CBDs as compared with that of cats with unobstructed CBDs (27). Ong found that 40% of patients presenting with an acute episode of cholangitis had bacteria isolated from portal venous blood (28). However, this may be a result of bacterial reflux from bile into the portal vein under high intrabiliary pressure (27). Indeed, it is unclear whether the portal system in humans can harbor bacteria. Orloff et al. did not find any significant bacterial isolates after culturing the portal blood of 101 patients undergoing abdominal surgery for a variety of intraabdominal diseases (29). Approximately 25% of their patients received preoperative antibiotics; to counteract this, antibiotic inhibitors were added to all culture media. The effectiveness of the reticuloendothelial system (RES) would have to be impaired for successful bacterial colonization of the biliary tract to occur through portal translocation (10). Sung et al. showed that in cats with unobstructed biliary systems, the liver's RES was overwhelmed by the splenic vein infusion of 105 bacteria, resulting in bactibilia (27). Cats with chronic biliary obstruction but not those with acute obstruction had bactibilia after a lowdose infusion of 103 bacteria. The authors concluded that chronic obstruction impaired the liver's capacity to trap bacteria, making the biliary tract more vulnerable to bacterial invasion. Other experimental work in rats has suggested that the phagocytic activity of Kupffer cells is also
Page 775
depressed during biliary obstruction (30–33). Drivas et al. previously demonstrated that some patients with cholestasis had abnormal RES clearance for labeled albumin (34). Accordingly, these alterations would seem more likely to occur in complete biliary obstruction, increasing the likelihood of bactibilia. However, bactibilia is less common in cases of malignant obstruction compared with those of choledocholithiasis. Therefore, portal translocation of bacteria and impaired RES function do not fully explain the origin of bactibilia. Another problem with this theory is the disproportionately low rate of anaerobic biliary infections compared to their number in the gut. It has been suggested that bile salt inhibition of anaerobic growth may be responsible for this difference (7). Other mechanisms of bactibilia—such as bacterial translocation in the lymphatics or hepatic arteries and hepatic or gallbladder secretion of bacteria—appear unlikely or uncommon. The direction of lymphatic flow around the CBD appears to be away from the biliary tree (26). In the guinea pig model, bacteria did not reach the gallbladder or liver in high titers after duodenal lymphatic injection even after CBD ligation (26). Hepatic arterial bacteremia usually results in the development hepatic abscesses rather than cholangitis (7). Chronically infected gallbladders were suggested to be the source of bactibilia, as gallbladder bile was found to be frequently infected in chronic cholecystitis (35,36). One suggested means of entry from the bloodstream was through ulcerations in the gallbladder wall (36). However, like hepatic secretion of bacteria, a gallbladder source must be unusual, as patients with a completely obstructed CBD rarely develop cholangitis spontaneously. Furthermore, bacterial infection of the gallbladder remains unexplained. Bactibilia does not invariably result in the syndrome of cholangitis. Stasis within the biliary tract is essential for bacterial multiplication and increased biliary pressure is responsible for bacteremia (37). Indeed, high intrabiliary pressures (above 25 cmH2O) have been associated with increased mortality in patients with cholangitis (38). With increasing pressure, bacteria can reflux into the lymphatics and bloodstream as well as produce intrahepatic damage (39,40). Csendes et al. found that the mean biliary pressure in patients with choledocholithiasis and cholangitis was 26 cmH2O, compared with 10 cmH2O in control subjects (41). The mean biliary pressure was only 13 cmH2O in patients with uncomplicated choledocholithiasis. Canine studies have shown that organisms are not detected in blood or lymph when the biliary pressure is below 20 cmH2O but appear rapidly when the pressure reaches 25 cmH2O (40). Intrabiliary pressure, rather than a high bile acid concentration, appears to be the significant factor in biliary permeability (42). In CBD obstruction, tight junctions between hepatocytes become disrupted, providing a route for bacteria to penetrate into the systemic circulation (43–45). However, studies using transmission electron microscopy showed that the predominant route of bacterial passage into the bloodstream appeared to be through the hepatocytes themselves (37). Bacteria appeared to be entering pits on the surfaces of hepatocytes in rats with CBD obstruction and many cells were filled with large vacuoles. As a result of these changes, bacteria and endotoxin can reach the systemic circulation. In addition, hepatocyte function may be impaired when the ductal pressure is elevated. The maximal limiting secretory capacity of hepatocytes is approximately 30 cmH2O (41). If intrabiliary pressures exceed this level, the resulting reflux of excretory products from hepatocytes into the systemic circulation adds to the systemic toxicity of bacteria (46). The endoscopically placed CBD stent is uniquely predisposed to the development of cholangitis. The normal protective mechanisms preventing bactibilia are bypassed and stents are prone to become obstructed. The earliest event in stent occlusion may be adsorption of proteins, after which bacteria and other material can adhere to the endoprosthesis (47). Yu et al. have shown that biliary proteins appear to be adsorbed on the stent's surface within a short time after insertion (48). The stents become colonized with bacteria, which grow as glycocalyxenclosed microcolonies, forming an adherent biofilm (9,49,50). Bile immunoglobulins may facilitate bacterial adhesion on the stent surface (51). Based on in vitro experiments, Leung et al. have suggested that there is a synergistic effect between grampositive and gramnegative bacteria in adherence and biofilm formation (52). Bacterial enzymes deconjugate bilirubin and
Page 776
Figure 1 Rightupperquadrant ultrasound showing sludge (open arrows) within a biliary stent (closed arrows).
lecithin, precipitating calcium bilirubinate and calcium salts of fatty acids, which contribute to the progressive accumulation of bile sediment and sludge (Fig. 1) (53). The biofilm protects against antibacterial agents and host clearance mechanisms and ultimately causes stent occlusion. C— Etiology Obstruction of the biliary tract or disruption of the normal CBD is essential for the development of cholangitis. The principal causes of obstruction leading to cholangitis are listed in Table 1. The majority of patients with acute cholangitis in western countries have CBD calculi. Choledocholithiasis occurs in approximately 10 to 15% of patients undergoing cholecystectomy, and around 10% of these patients develop cholangitis (54,55). Choledochal sludge has also been implicated in the development of acute cholangitis (56). A recent retrospective review found that 8 of 164 patients (5%) with malignant biliary obstruction developed cholangitis (57). Of the 12 patients with ampullary carcinomas in this review, 5 developed cholangitis, compared with only 3 of the 152 patients with other pancreaticobiliary malignancies. The overall incidence of cholangitis secondary to either benign or malignant strictures has risen with time (7). Lipsett et al. found that these strictures accounted for 30% of their cases before 1974, compared with 69% between 1986 and 1989 (7). This may be a result of referral bias, but it also reflects the increased diagnostic and therapeutic interventions available for pancreatic and biliary tract malignancies, especially endoscopic retrograde cholangiopancreatography (ERCP) and endoprosthesis insertion. Endoscopic or radiological instrumentation of the biliary tract may result in cholangitis. Bacteria are introduced into the system and the injection of contrast material elevates the intrabiliary pressure, increasing the risk of bacteremia. The overall rate of cholangitis postERCP is around 1%. A recent prospective study of 2347 patients showed it was associated with combined percutaneousendoscopic procedures, stent placement for malignant strictures,
Page 777 Table 1 Causes of Biliary Obstruction and Cholangitis Choledocholithiasis and biliary sludge Biliary strictures Benign Primary sclerosing cholangitis Secondary to Iatrogenic injury Ischemia Pancreatitis AIDS cholangiopathy Malignant Biliary tract malignancies Pancreatic carcinoma Metastases Infections Ascaris Clonorchis sinensis Opisthorchis viverrini Fasciola hepatica Echinococcus granulosa and multilocularis Extrinsic compression Hilar lymphadenopathy Periampullary diverticulum Postoperative syndromes "Sump" syndrome postcholedochoenterostomy Choledochojejunostomy Stenosis of the papilla of Vater Foreign bodies Biliary tract instrumentation Congenital abnormalities of the common bile duct Choledochal cyst Choledochocele Congenital hepatic fibrosis Caroli's disease
and failed biliary access or inadequate drainage (58). Plastic biliary stents with a diameter of 10 Fr tend to clog on average 4 to 6 months after placement, predisposing the patient to cholangitis (59–61). This occurs in around 40 to 50% of patients with stents, as many die earlier from progression of the underlying malignant disease (61–63). Selfexpanding metal stents have better patency rates than their plastic counterparts, but they can still become occluded by tumor ingrowth (64–66). Postoperative cholangitis can occur after any form of biliary reconstruction. This is more likely to occur if a stricture develops at the biliaryenteric anastomosis. Patients with choledochoduodenostomies who have delayed emptying of the biliary tree are more likely to have positive bile cultures than those with normal emptying (67). The "sump syndrome" postcholedochoenterostomy arises when a sump, or pit, develops in the distal, nonfunctioning limb of the CBD (68). Bile and debris accumulate, resulting in obstruction of the surgical anastamosis and predisposing the patient to the development of cholangitis. Sphincter of Oddi dysfunction may also contribute to the pathogenesis of this syndrome. Other factors, such as the
Page 778
presence of intrahepatic stones or strictures, should also be considered in postoperative cholangitis. D— Pathology In acute bacterial cholangitis, the hepatic histological changes include bile ductal tortuosity and proliferation, interstitial edema, and a prominent neutrophilic infiltrate (69). These findings are not pathognomonic for obstruction and may be seen in systemic infection or secondary to a toxin or drug reaction. Therefore, a diagnosis of cholangitis should be based on clinical rather than pathological grounds. E— Clinical Features There is a wide spectrum of clinical presentations in acute cholangitis, ranging from a mild selflimiting episode to obtundation and hypotension, as described in Reynolds' pentad (2). Most patients are between 50 and 70 years of age and, unlike the case with acute cholecystitis, there is no female preponderance (5,7,10). Patients may give a history of previous surgery or instrumentation of the biliary tract. The classic presentation of Charcot's triad occurs in 20 to 70% of patients (5,14,70). Fever is seen in 65 to 90% of cases. Overall, jaundice and abdominal pain appear to be less common presentations now than in the past. In one series, jaundice was seen in 67% of patients after 1983, compared with 79 and 90% of patients before 1974 and between 1976 and 1978, respectively (7). Similarly, abdominal pain was seen in less than half of the more recent cases, compared with over 75% of patients before 1978. The change in presenting features may reflect the increased number of patients with malignant diseases that develop obstruction of endoprostheses. In patients with choledocholithiasis, over 80% will have jaundice and abdominal pain at presentation (5). While chills have been reported in 65 to 87% of patients at presentation, dark urine, pruritus, and pale bowel movements are uncommon (71). Severe cholangitis with septic shock still occurs in at least 5% of patients and appears more likely to occur in elderly patients (7). In a retrospective review of 61 patients, whose mean age was 76, shock was reported in 23% of cases (71). Lai et al. found that 60% of their patients with cholangitis complicating choledocholithiasis had septicemic shock, the majority within 24 h of admission (5). Patients with complete biliary obstruction are especially at risk of developing septicemia and hypotension after an unsuccessful attempt at drainage (72). Finally, cholangitis complicating gallstones can present nonspecifically in the elderly as mental and physical deterioration, without any of the classic features such as significant abdominal pain (73). Physical findings are often unimpressive in acute cholangitis. Jaundice and RUQ tenderness may be present in up to twothirds of patients, but peritoneal signs are uncommon and suggest an alternative diagnosis such as acute cholecystitis (see below) (7,10). F— Investigations 1— Laboratory Investigations In acute bacterial cholangitis, a leukocytosis is common, with over 70% of patients having values above 10,000/mm3 (74). The mean white blood cell count in one study was 13,600/mm3, and there was no significant difference in the count between benign and malignant causes of obstruction (75). The differential count may show increased numbers of immature forms (46). Occasionally, leukopenia is present in patients with overwhelming sepsis. Liver function tests (LFTs) are usually abnormal in acute cholangitis. Although hyperbilirubinemia occurs in the majority of patients, the actual concentration is less than 2 mg/dL in 20% of cases (5). Alkaline phosphatase is increased from two to five times normal in at least 75% of patients (74,76). Cases of malignant obstruction have more marked elevations in
Page 779
bilirubin and alkaline phosphatase. Thompson et al. reported mean alkaline phosphatase values of 531 and 278 IU in patients with malignant and benign disease, respectively (75). The transaminases rarely exceed 270 U/mL, with higher values suggesting underlying benign disease (5,75). An elevated serum amylase is seen in approximately onethird of patients and, as with the transaminases, higher values suggest benign disease (74). While serum CA 199 levels were found to be elevated in patients with acute cholangitis compared with other causes of benign biliary diseases, its usefulness in clinical practice is uncertain (77). A biliary tract infection score has been proposed as an aid in assessing patients with cholangitis and their response to therapy. It includes the presence and extent of local symptoms, fever, and elevations in seven hematological and biochemical tests (78). However, prospective studies will be required to determine the value of such a scoring system, in addition to the usual clinical assessment. 2— Radiological Investigations Several radiological studies are available that provide valuable information concerning the site and etiology of biliary obstruction. Complications of cholangitis can also be identified. Plain abdominal xrays are of little value. Occasionally calcified gallstones and air within the biliary tract or portal system may be seen (74). The most useful initial investigation in patients with RUQ pain is ultrasonography, as it can diagnose cholelithiasis in over 95% of cases (79). Ultrasound can usually differentiate extrahepatic obstruction from intrahepatic cholestasis when biliary dilatation is present (80). It has a sensitivity of around 75 and 50% for stone detection in dilated and nondilated bile ducts, respectively. In cholangitis, ultrasound may demonstrate thickening of the CBD wall, with the appearance of a hypoechoic stripe lying internal to the echogenic line of the normal duct (81). However, gas in the duodenum may obscure visualization of the distal CBD (79). Computed tomography (CT) evaluates the liver parenchyma, extrahepatic structures, retroperitoneum, abscesses, and fluid collections more readily than ultrasound. Accordingly, it can provide a more comprehensive evaluation of the upper abdomen in patients with suspected cholangitis. CT has a similar sensitivity to ultrasound for the detection of choledocholithiasis (79). Isotope scintigraphy by hepatobiliary dimethyl iminodiacetic acid (HIDA) is generally not useful in the investigation of acute bacterial cholangitis. The anatomic definition is inferior to that of other imaging modalities in establishing the site and cause of obstruction (82). However, it may be helpful in differentiating acute cholecystitis from cholangitis and can detect CBD obstruction before ductal dilatation develops (83). Magnetic resonance cholangiopancreatography (MRCP) allows for noninvasive imaging of the common bile and pancreatic ducts. It can detect dilated bile ducts, strictures, and biliary calculi (84,85). A recent study of 110 patients found that MRCP had a sensitivity and specificity of over 90% for the detection of CBD stones when compared with cholangiography or surgery (86). However, in another study, its sensitivity for bile duct stones was only 51.7%, frequently missing stones smaller than 6 mm in size. Experience with MRCP is clearly evolving and its precise role in cholangitis has yet to be defined. It may have a role in evaluating nontoxic patients when the diagnosis is uncertain and direct cholangiography is not readily available or is unsuccessful (87). Endoscopic ultrasound (EUS) can provide detailed imaging of the biliary tree without interference by bowel, air, or fat (88). It is superior to both CT and ultrasound and appears to be particularly useful for the detection of stones less than 1 cm in size in normaldiameter ducts (89). While ERCP is the test of choice in evaluating highrisk patients, as therapeutic intervention is often necessary, EUS may be useful in the assessment of nonacute patients for the presence of choledocholithiasis (88,90). The ''gold standard" for evaluating the etiology of cholangitis remains a direct cholangiogram. Oral and intravenous cholangiography are not useful, as the likelihood of sufficient contrast being excreted into the biliary tract in cholangitis is limited (91). The two available
Page 780
methods for visualizing the biliary tract directly are ERCP and percutaneous transhepatic cholangiography (PTC) (54,92). Both investigations can determine the site and cause of obstruction and define the anatomy. In addition, biliary decompression can be performed by either technique. In most centers, ERCP is the preferred method of investigation unless the obstruction is likely to be at or above the porta hepatitis or previous gastric or biliary bypass surgery precludes the retrograde approach. G— Differential Diagnosis While the differential diagnosis for acute cholangitis is extensive, patients presenting with Charcot's triad rarely pose a major diagnostic dilemma. Table 2 lists the conditions which form the differential diagnoses. Patients with acute cholecystitis usually have constant RUQ pain, fever, and signs in the RUQ including tenderness and possible guarding or rigidity secondary to a localized peritonitis. A gallbladder phlegmon may be palpable (91). Elevated levels of bilirubin and LFTs occur in less than onethird of patients with acute cholecystitis (55). Markedly abnormal values suggests the presence of choledocholithiasis (55,93). Other possible explanations for hyperbilirubinemia in cholecystitis include Mirizzi's syndrome (bile duct obstruction by the inflamed gallbladder); increased permeability of the gallbladder epithelium, allowing systemic absorption of bile; or sepsisrelated cholestasis (93,94). A marked leukocytosis is more commonly seen in septicemia associated with cholangitis. Other intra, retro, or extraabdominal processes can usually be differentiated from acute cholangitis without major difficulty on clinical grounds or after laboratory and radiological investigations have been performed. The entity of sepsisrelated cholestasis can sometimes pose management difficulties. It occurs in septic patients where the history and physical examination may be limited. The usual pattern of biochemical abnormality is marked conjugated hyperbilirubinemia with minimal elevations in the other LFTs. Imaging of the biliary tract reveals no evidence of intra or extrahepatic obstruction. Sepsisrelated cholestasis is probably multifactorial in origin. Affected patients are often receiving parenteral alimentation and medications Table 2 The Differential Diagnoses of Acute Cholangitis Gallbladder conditions Acute cholecystitis Perforated gallbladder Mirizzi's syndrome Torsion of the gallbladder Intraabdominal conditions Acute hepatitis Hepatic abscess Acute appendicitis Perforated duodenal ulcer Retroperitoneal conditions Acute pancreatitis Rightsided pyelonephritis Extraabdominal conditions Sepsisrelated cholestasis Rightlowerlobe pneumonia Pulmonary infarct Herpes zoster infection
Page 781
that can cause cholestasis. In addition, endotoxins and cytokines may play a significant role in hepatocellular inflammatory events, leading to downregulation of bile salt transporters and bile salt leakage through damaged biliary epithelium (94). The underlying sepsis should be treated and invasive procedures such as ERCP should be avoided. H— Microbiology of Acute Cholangitis The majority of bacteria associated with cholangitis are enteric in origin. Gramnegative bacteria are consistently the most commonly isolated organisms in bile cultures (Table 3). Escherichia coli and Klebsiella species were previously the dominant biliary tract organisms, but now other organisms are frequently being isolated (7). The changing microbiological profile over the past 30 years has been attributed to increased instrumentation within the biliary tract and the use of antibiotics (7,14,95). Bacteria such as Enterobacter, Pseudomonas species, and Streptococcus species are more often found in bile cultures from patients with malignant biliary tract strictures, immunosuppressed individuals, patients with diabetes, or those who have undergone previous instrumentation of the biliary tract or recent treatment with antibiotics (46,73). Bile cultures are positive in 80 to 100% of patients with cholangitis secondary to gallstones and in up to 50% of patients with complete CBD obstruction (4,5,7,70,96). Polymicrobial bile cultures are found in 20 to 88% of cases (4,5,17,70,97). The number of bacterial colonies per milliliter in bile was higher in patients with cholangitis complicating choledocholithiasis than in those who had CBD stones and no evidence of infection (98). Anaerobes, such as Bacteroides and Clostridium species, and facultative bacteria are usually isolated in conjunction with aerobic bacteria and not as the sole isolate in culture (99). Anaerobic bactibilia is most commonly seen in patients after biliary surgery and CBD manipulation (99). The variable culture rates for anaerobes (from 3 to 65%) may relate to differences in culture technique of bile specimens (3,7,18,97,99–103). Mixed infections with aerobic and anaerobic bac Table 3 Organisms Isolated from Bile and Blood Cultures of Patients with Acute Bacterial Cholangitis
Reported bacterial isolates from bile cultures (%)
Reported bacterial isolates from blood cultures (%)
Gramnegative organisms Escherichia coli
34–71
17–63
Klebsiella species
14–54
11–18
9–34
3–9
Pseudomonas species Citrobacter species
21
Enterobacter species
5–34
5
Proteus species
6–13
3–6
Serratia species
5
Aeromonas species
5
Grampositive organisms Streptococcus species
38
Enterococcus species Bacteroides species
12
10–34 15
Staphylococcus species
5
Clostridium species
5
Sources: Data from Refs. 4, 54, 70, 75, and 96.
4–9
3–12
Page 782
teria are associated with a more serious clinical condition and a high incidence of postoperative infectious complications (99,102). Blood cultures are positive in 21 to 63% of patients with acute cholangitis (4,5,54,70,96). E. coli and Klebsiella species have been the most frequently isolated organisms in blood (Table 3). The discrepancy between positive blood and bile cultures may be due to the intermittent release of bacteria into the systemic circulation. Streptococcus and Enterococcus species are infrequently recovered from blood cultures in comparison with their relatively high prevalence in bile cultures. Multiple organisms are isolated in around 25% of patients, with a similar number having the same organism isolated from both bile and blood cultures (5,70). Anaerobic bacteremia is very uncommon in acute cholangitis. Biliary endoprostheses usually become colonized with multiple bacteria. In one study, the intraluminal sections of 25 biliary stents were cultured (104). The mean number of isolates recovered from obstructed and nonobstructed stents were 3.5 and 2, respectively. In total, 81 microorganisms were isolated, 54 Enterobacteriaceae, 5 Pseudomonas aeruginosa, 19 Enterococcus species, and 3 Candida albicans species. Stenotrophomonas maltophilia is a multiresistant, gramnegative bacillus that is increasingly recognized as a cause of nosocomial infections. Five cases of cholangitis caused by S. maltophilia have been reported (105,106). All patients had hepatobiliary malignancy and previously underwent biliary tract instrumentation. Invasive Salmonella species, such as S. dublin and S. cholerasuis, are thought to enter the biliary tract as a result of transient portalvenous bacteremia that occurs during primary intestinal disease (107). Repeatedly positive bile cultures for S. typhi were documented in a patient with choledocholithiasis; stone removal resulted in clearance of the infection (108). It was suggested that the surface biofilm on CBD stones might play a role in maintaining the carrier state for Salmonella infection. I— Management 1— Overview The mainstays of treatment are adequate resuscitation, appropriate antibiotic therapy, and the reestablishment of biliary drainage (3). Initially, the patient with cholangitis should fast and receive adequate hydration with intravenous fluids. He or she should be monitored closely with special attention being paid to the urinary output. Essential laboratory investigations include a complete blood count, renal and liver function tests, as well as a prothrombin time or International Normalized Ratio (INR). Antibiotic therapy should be instituted once blood cultures have been drawn. Vitamin K administration is recommended if the INR is elevated. Fresh frozen plasma will be necessary for rapid correction of a coagulopathy when urgent intervention is required. Antibiotic therapy should be modified once blood culture results are known. The majority of patients will stabilize with this approach. Some 10 to 15% of patients will require urgent biliary decompression because of their condition on presentation, if they fail to stabilize, or if they deteriorate despite these initial measures (70,109). The remainder will require elective biliary drainage. It appears that urgent intervention is more likely in older patients, in those who have had previous biliary manipulation, and in the setting of RPC (6,71). 2— Antibiotic Therapy The principal goals of antibiotic treatment are to control sepsis and local inflammation secondary to cholangitis (3). Broadspectrum coverage is necessary initially, which can then be adjusted when culture results become available. The usual duration of treatment is 7 to 10 days. Factors that may affect antibiotic efficacy include the agent's spectrum of activity; its pharmacokinetic properties, including extent of biliary excretion; and bactericidal activity in bile (110).
Page 783
Ideally, the antibiotic selected in acute cholangitis should achieve adequate biliary and tissue concentrations to be effective against the most commonly isolated organisms in the biliary tract (110,111). As a result, there has been considerable interest in studying the biliary excretion of antimicrobial agents. The biliary levels of quinolones are approximately 2 to 10 times the simultaneous serum levels for all quinolones (111,112). Mezlocillin and cefoperazone also have a high degree of biliary penetration (113). Other antibiotics, such as netilmicin and imipenem, are secreted into the bile to a lesser degree. However, the biliary excretion of many antibiotics is impaired in patients with an obstructed CBD. Cefoperazone, imipenem, ceftazidime, and netilimicin were not detected in the bile of patients immediately after drainage of obstructed systems, while ciprofloxacin was present at 20% of the mean peak serum level (113). This may be due to high tissue penetration of the quinolones across the gallbladder wall independent of hepatic excretion (113,114). Mezlocillin also seems to be less affected by biliary obstruction compared with ampicillin or gentamicin (115). Passive and active mechanisms of antibiotic excretion into the biliary tree have been described. Passive excretion of ceftazidime and active excretion of cefoperazone were found in patients without evidence of biliary obstruction (116). In patients with obstruction, both forms of excretion were impaired, the passive mechanism recovering quickly (within 60 min) after decompression. Active excretion of cefoperazone did not recover for at least 24 h. Therefore it appears that once the obstruction is relieved, biliary excretion of antibiotics gradually recovers but levels remain lower than in bile of freely draining systems (113). The different excretory mechanisms in hepatocytes and bile duct epithelium may be affected to a variable extent by biliary obstruction and recover at different rates after decompression (72). Since the biliary excretion of most antibiotics does not occur at high intrabiliary pressures, the antibacterial spectrum of an antibiotic is a more important consideration in the treatment of cholangitis than its biliary levels (7). Effective antibiotics are active against the commonly isolated organisms and achieve adequate blood and tissue levels around the biliary tract (7,117–119). Multiple regimens have been suggested as empiric therapy, and some of the current options are listed in Table 4 (120). Initially all regimens should be effective against E. coli and Klebsiella species, but the value of routine empiric treatment for Enterococcus, Pseu Table 4 Currently Available Antibiotics for the Treatment of Acute Bacterial Cholangitis First Line Agents Fourquinolone (Ciprofloxacin, Ofloxacin, Levofloxacin) ± Metronidazolea Thirdgeneration cephalosporin ± metronidazolea With antiPseudomonas activity: ceftazidime cefoperazone Without antiPseudomonas activity: Ceftriaxone, ceftizoxime and cefotaxime Ampicillin + aminoglycoside (gentamicin, tobramycin, amikacin) ± metronidazole Ureidopenicillins (mezlocillin, piperacillin) + aminoglycoside Ampicillin/sulbactam + aminoglycoside Piperacillin/tazobactam Ticarcillin/clavulanate Alternative agentsb Imipenem or meropenem Aztreonam ± metronidazolea a
Additional coverage with ampicillin, or vancomycin in patients allergic to penicillin, is necessary to cover for Enterococcus species. b Data are limited on efficacy of these agents in cholangitis. Empiric broadspectrum antibiotic coverage should be considered in elderly patients, patients with severe cholangitis, and those at risk from cholangitis secondary to Enterococcus, Pseudomonas, and anaerobic species (after biliary tract instrumentation or surgery, recent antibiotic exposure, patients with malignant obstruction, or immunosuppressed individuals).
Page 784
domonas, and anaerobic species is uncertain in view of their relatively low rates of bacteremia (Table 3). However, patients at risk for cholangitis secondary to these organisms include those who have undergone biliary tract instrumentation or have recently been treated with antibiotics, patients with malignant obstruction, and immunosuppressed individuals. Administration of metronidazole should be considered in any patient who has undergone previous biliary surgery to cover for potential anaerobic infection. Factors that influence antibiotic selection include the patient's age, clinical condition, and allergy history as well as the presence of comorbidities, such as renal dysfunction. Knowledge of local antibiotic sensitivity patterns is also important. Traditionally, a commonly prescribed empiric regimen consisted of a penicillin such as ampicillin and an aminoglycoside (gentamicin, tobramycin, or amikacin). Concerns have been raised about the ototoxicity and nephrotoxicity of the aminglycosides, particularly in elderly patients receiving these agents for more than 7 days (121,122). Aminoglycosides should be used cautiously if at all in patients with evidence of hearing loss or vestibular dysfunction. The risk of developing nephrotoxicity varies widely, depending on many factors including the definition of nephrotoxicity used, monitoring methods, and patient selection. One study reported impairment of renal function in 27% of patients treated with gentamicin for cholangitis (115). Risk factors for aminoglycosiderelated nephrotoxicity include morbid obesity, baseline renal insufficiency or unstable renal function, and prolonged therapy. Critically ill patients with intravascular volume depletion or hemodynamic instability, patients with diabetes and endorgan damage, and those concurrently taking vancomycin, non steroidal antiinflammatory agents, cyclosporine, or amphotericin B are also at increased risk. In these situations, we recommend that an alternative antibiotic be used. The ureidopenicillins, mezlocillin and piperacillin, are popular agents in the treatment of cholangitis. They have a broad spectrum of activity against gramnegative organisms, enterococci, Pseudomonas, and anaerobes (72). Mezlocillin was more effective and less toxic than a combination of ampicillin and gentamicin in a randomized trial of 46 patients with cholangitis (115). In another randomized study of 96 patients, there was no significant difference in clinical cure rates between monotherapy with piperacillin versus ampicillin and tobramycin (69 versus 70%, respectively) (75). However, there have been reports of resistance of E. coli and Klebsiella species to ureidopenicillins (123). In our institution, current sensitivity rates of piperacillin to E. coli and Klebsiella are 60 to 74% and 71 to 86%, respectively. Some authors suggest prescribing an aminoglycoside with a ureidopenicillin initially, to broaden gramnegative coverage, until blood culture results become available (3,120). An alternative approach is to use a penicillin combined with a betalactamase inhibitor—such as sulbactam, clavulanic acid, or tazobactam sodium—which may overcome some gramnegative resistance mechanisms (124,125). Ampicillinsulbactam has a much more limited spectrum of activity against gramnegative bacteria than either ticarcillin/clavulanate or piperacillin/tazobactam, the only other currently marketed parenteral betalactam combination products. Therefore, the addition of an aminoglycoside should be considered in patients with severe cholangitis when ampicillinsulbactam is being administered. Ticarcillin/clavulanate and piperacillin/tazobactam have broad activity, covering most biliary pathogens, and may be used as monotherapy in cholangitis (120). In our institution, 85% of Enterococcus isolates are susceptible to ampicillin; thus administration of any of these penicillins should provide adequate empiric coverage for Enterococcus species. The cephalosporins are valuable antibiotics in the treatment of intraabdominal sepsis, including acute cholangitis. Secondgeneration agents (for example, cefuroxime) have been used, usually in combination with an aminoglycoside. However, such regimens have not been fully evaluated (3). The thirdgeneration agents, ceftazidime and cefoperazone, have broad coverage against gramnegative bacteria, including Pseudomonas species (120). The other thirdgeneration cephalosporins— ceftriaxone, ceftizoxime, and cefotaxime—are also useful agents in cholangitis but do not have activity against P. aeruginosa. In one study, initial treatment of severe biliary tract infections with cefoperazone was more effective than a combination of ampicillin and tobramycin (126). A second study found that cefoperazone had a cure rate
Page 785
of only 56% in patients with cholangitis, compared with 85% for the combination of ampicillin and tobramycin (127). Overall, 13% of patients receiving cefoperazone had an increased prothrombin time. Cefoperazone may have contributed to the disturbance in coagulation by inhibiting vitamin K—dependent synthesis of clotting factors. However, more patients in the cefoperazone group had cholangitis complicating malignant biliary obstruction, which is associated with a poorer outcome and more antibiotic related toxicity, than did patients with underlying benign biliary disease (75). Features that make the thirdgeneration cephalosporins attractive in treatment compared to penicillins and aminoglycosides include ease of dosage, the lack of significant nephrotoxicity or ototoxicity, and a lower sodium load per treatment than the penicillins. In our institution, over 90% of E. coli, P. aeruginosa, and Klebsiella isolates are sensitive to ceftazidime. Despite their broadspectrum coverage, the thirdgeneration cephalosporins should be administered with metronidazole in patients with evidence of severe cholangitis in order to cover for an anaerobic infection (120). Depending upon the cephalosporin being used, additional antipseudomonal coverage may be required if Pseudomonas is isolated in bile or blood cultures (Table 3). There is concern that continuing cephalosporin usage may select out resistant enterococci from the patient's endogenous flora (128). The quinolones are extremely useful antibiotics in the management of cholangitis because of their pharmacokinetics and spectrum of activity. Agents such as ciprofloxacin normally achieve higher concentrations in bile than in serum or plasma. Furthermore, ciprofloxacin continues to be actively excreted into an obstructed biliary tree at levels 10 times the minimum inhibitory concentration for gramnegative bacteria (113,114,129). The newer fluoroquinolone levofloxacin is absorbed within 3 h of ingestion and is 90 to 100% bioavailable from the oral route (112,130). Furthermore, it has a better dosing profile than ciprofloxacin because of its high bioavailability and long halflife. However, there are no data on the biliary excretion of levofloxacin in the presence of obstruction. The quinolones as a class are generally safe antibiotics and have activity against most common biliary tract bacteria. In our institution, over 90% of E. coli and Klebsiella isolates are sensitive to ciprofloxacin and levofloxacin; 75 to 85% and 68% of P. aeruginosa isolates are sensitive to ciprofloxacin and levofloxacin, respectively. However, depending upon the patient, there is a potential for drug interactions when the quinolones are used, especially ciprofloxacin. These include an elevation in theophylline levels and an enhancement of warfarin's anticoagulant effect. The oral absorption of quinolones is reduced if concurrently administered with iron, zinc, calcium, or antacids. Clinical trials using the quinolones have confirmed their value in the treatment of cholangitis. In one study, 100 patients were randomized to receive either intravenous ciprofloxacin or combination therapy of ceftazidime, ampicillin, and metronidazole. There was no significant difference in clinical response rates, recurrence of fever, duration of hospital stay, or need for emergency drainage between the groups (131). Another recent study compared ofloxacin with ceftriaxone in patients with acute biliary infections (cholecystitis and cholangitis) (132). The rates of clinical cure were similar in both groups, 89 and 85%, respectively. Further studies are required to define the role of the newer quinolones, either as mono or combination therapy, in the treatment of acute cholangitis. Based on all the factors discussed above, it is acceptable to use the quinolones as firstline agents in acute cholangitis. However, empiric monotherapy with one of the quinolones may provide insufficient coverage against anaerobic bacteria (130). Accordingly, when a mixed infection is more likely—as in the elderly, in patients with a prior biliaryenteric anastamosis, or in cases of severe sepsis—coadministration of ampicillin and metronidazole should be considered along with ciprofloxacin, ofloxacin, or levofloxacin. Imipenem has a wide spectrum of activity against gramnegative and grampositive organisms, as well as against anaerobes (120). It has been suggested as an alternative agent for patients with cholangitis (3,113). However, specific studies using imipenem in cholangitis are not yet available. Multiple factors in relation to specific agents need to be considered when empiric treatment is begun. In addition, the condition of the patient and his or her potential for cholangitis
Page 786
secondary to bacteria other than E. coli and Klebsiella species are critical factors in determining which antibiotic(s) should be administered. Patients with mild cholangitis may require only a short intravenous course of monotherapy with a ureidopenicillin, quinolone, or thirdgeneration cephalosporin, quickly changing to the oral equivalent once biliary drainage has been established. Patients with severe cholangitis and/or those with risk factors for Pseudomonas, Enterococcus, or anaerobic infections initially require broadspectrum coverage. Treatment options in these situations include a ureidopenicillin with an aminoglycoside, a penicillin with a betalactamase inhibitor, a thirdgeneration cephalosporin (which has Pseudomonas activity) with metronidazole and ampicillin, or a quinolone, such as levofloxacin, with metronidazole and ampicillin. In patients who are allergic to penicillin, vancomycin provides coverage against Enterococcus species. After biliary drainage, intravenous therapy should be continued until the patients's condition has remained stable for at least 48 h. Finally, treatment should be adjusted when blood and bile cultures become available, thus reducing the patient's exposure to unnecessary antibiotics. 3— Biliary Drainage Decompression of the biliary tract is essential in the management of acute cholangitis. Persistent obstruction, even with clinical improvement on antibiotic therapy, predisposes the patient to recurrent bouts of cholangitis and hepatic abscess formation. If obstruction is prolonged, secondary biliary cirrhosis may develop (133). Drainage can be accomplished by endoscopic, percutaneous, or surgical techniques. The timing of intervention is determined by the patient's overall condition. Between 70 and 85% of patients can have elective decompression once antibiotic therapy has been commenced (134). However, patients with evidence of severe systemic septic complications or those who deteriorate while receiving antibiotics must undergo urgent biliary drainage. The choice of procedure depends on several factors, including the site of the obstruction and local expertise. ERCP is now generally accepted as the firstline therapy for biliary decompression in the majority of patients. Surgery or percutaneous drainage is usually reserved for situations where the endoscopic route is not appropriate or is unsuccessful. Endoscopic Drainage Endoscopic drainage of the biliary tree can be successfully performed in the acute or elective setting. The procedure of choice for choledocholithiasis is endoscopic sphincterotomy (ES) and stone extraction (5). Alternatively, in severely ill patients, a biliary stent or nasobiliary drain may be placed as a temporary measure to decompress the obstructed system (91). Definitive therapy can then be performed electively, when the patient's condition has stabilized. The patient's clinical condition improves rapidly with successful drainage. Endotoxemia is effectively treated as endoscopic therapy lowers bile and serum endotoxin values (135). A detailed discussion of endoscopic technique in cholangitis is beyond the scope of this chapter. However, several points are worth mentioning. Minimal sedation should be administered to patients who are ill. Indeed, there are reports of ERCP being performed without sedation in patients who are hypotensive (136). The procedure may be completed in the intensive care unit using a Carm for fluoroscopy (137). In centers where this is not available, after cannulation, bile should be aspirated to confirm catheter placement in the CBD, before any therapeutic procedure is performed (134). If possible, deep cannulation should always be achieved and infected bile aspirated back prior to any contrast injection. The bile can then be sent for microbiological studies. Large bolus injections of contrast into the CBD and repeated injections should be avoided to prevent cholangiovenous reflux and worsening of the ongoing sepsis (138). A full cholangiogram is not essential in patients who are critically ill (6). The procedure should be performed as expeditiously as possible, with the principal aim of achieving adequate biliary drainage. This is confirmed endoscopically and radiographically by free drainage of bile and contrast, respectively. Several trials have shown that in the acute setting, endoscopic therapy is safer than surgical intervention for cholangitis secondary to choledocholithiasis. A retrospective, nonran
Page 787
domized series of 82 patients found endoscopic treatment to be superior to both medical and surgical interventions (54). The 30day mortality rates for medical, surgical, and endoscopic therapy were 36.4, 21.4, and 4.7%, respectively. The difference in outcomes is particularly impressive, as patients who underwent ES were older and had a greater number of medical comorbidities than those who had surgical drainage. All CBD stones were extracted in 70% of patients at the initial ERCP. Overall, 90% of patients responded clinically to endoscopic intervention. There has been one prospective, randomized, controlled trial comparing endoscopic with surgical decompression for severe acute cholangitis secondary to choledocholithiasis (5). Eightytwo patients were randomized to either surgical decompression by choledocholithotomy or endoscopic papillotomy and nasobiliary drainage with a 7Fr catheter, later followed by definitive therapy. Patients treated endoscopically had fewer complications (34 versus 66%, respectively) and a lower hospital mortality than those who underwent initial surgery (10 versus 32%, respectively). Ideally, ES and stone extraction should be completed at the initial intervention in stable patients (139). However, ES prolongs the procedure and is associated with a complication rate of up to 10%. Complications include hemorrhage, perforation, pancreatitis, and empyema of the gallbladder (58,140,141). Therefore, ES should be avoided in unstable patients, who would not tolerate any additional morbidity (6). The alternative methods to ES for biliary decompression are placement of either a nasobiliary catheter or internal biliary stent. These should be considered in any patient when drainage is incomplete or ES cannot be performed, for whatever reason. A 7Fr nasobiliary catheter can be placed without sphincterotomy, providing simple, safe, and effective decompression (96,140,142). Bile may be sampled for bacteriology; subsequently, chemical dissolution therapy can be administered through the catheter (136). However, definitive therapy is usually undertaken when the patient improves, reducing the need for prolonged nasobiliary access. Cholecystography via the nasobiliary drain should be avoided because of an increased risk of retrograde infection (140). The main disadvantages of a nasobiliary catheter are patient discomfort and catheter displacement by the patient or by nursing staff unfamiliar with its maintenance. Largebore (10Fr) biliary endoprostheses can also be placed without a sphincterotomy (6,143). Stents are thought to be effective by preventing stone impaction at the papilla rather than providing a channel for bile flow (109,144). They cannot be displaced at the bedside and cost less than nasobiliary catheters (136). A recent study of 60 patients with acute suppurative cholangitis comparing nasobiliary catheter drainage with endoscopic biliary stent placement confirmed that both are equally efficacious in providing adequate decompression (145). Failure to improve rapidly after endoscopic drainage should alert the clinician to the possibility of an undrained hepatic segment (especially if there is obstruction at the hilum), the presence of a hepatic abscess, or coexisting acute cholecystitis (14). Symptoms may also persist if stones remain in the CBD. After ES has been performed in patients with cholangitis secondary to choledocholithiasis, elective cholecystectomy should be considered. However, in the elderly or patients at high surgical risk, definitive surgery is not essential unless the patient has recurrent symptoms or complications (54). Percutaneous Transhepatic Biliary Drainage The use of percutaneous transhepatic biliary drainage (PTBD) to decompress an obstructed biliary system was first described in 1974 by Molnar and Stockum (146). PTBD can successfully drain patients with suppurative cholangitis in over 80% of cases, depending upon the operator's expertise (92,147–149). It is usually a temporary measure, stabilizing toxic patients prior to definitive therapy (149). However, the tract created at PTBD may be used for interventions such as biliary stent placement or stone retrieval especially in patients with contraindications to surgery (147,149). PTBD may be employed without sedation, thus avoiding the risks of the hemodynamic and pulmonary complications that can occur at endoscopy. Complications of PTBD include sepsis, bleeding, biliary peritonitis secondary to a biliary leak, and fistulous communications between the biliary and vascular trees (147,149–152). Var
Page 788
ious series have the complication rate ranging from 7 to 30% (147,149,151,153). Major acute periprocedural complications (septicemia and bleeding) and death occur in 5 to 10% of patients, usually in those with severe cholangitis (25,147,149). It appears that the underlying disease causing obstruction is more important than the presence of cholangitis in determining the outcome of percutaneous drainage (151). Patients undergoing PTBD for palliation of malignant biliary obstruction have higher rates of morbidity and mortality than those with benign biliary disease. As in ERCP, highpressure contrast injections should be avoided during the procedure to prevent reflux of endotoxin into the systemic circulation (46,149). Complications may arise after initial decompression, while the drainage catheter is left in situ. It may become occluded or dislodged, resulting in the recurrence of cholangitis or development of a hepatic abscess (147,150,154). Surgical Drainage Surgical drainage was the original means of biliary decompression for acute cholangitis, first described by Rogers in 1903 (155). The patient's clinical status and underlying condition determine the nature and extent of the surgical intervention. In emergencies, decompression may be achieved by choledochotomy, removal of any obvious stones, gentle irrigation of the duct, and placement of a T tube into the CBD (46). Further therapy should be deferred until the patient is more stable. However, internal drainage with the creation of a choledochoduodenostomy may be more effective in reducing biliary pressure (54,156). If the patient is stable, a cholecystectomy should be performed along with the CBD exploration (5). Intraoperative choledochoscopy is a valuable means of assessing the CBD for stones and it reduces the incidence of retained stones after exploration (157,158). It can be safely performed in patients with acute cholangitis provided that highpressure irrigation is avoided (157,158). As choledochoscopy may prolong the operation time, its use in patients who are critically ill is questionable. Biliary drainage through a cholecystostomy alone is rarely adequate and is associated with a high mortality rate (70,159). While surgical decompression is effective in most patients, morbidity and mortality rates are substantial. Morbidity includes postoperative sepsis (bronchopneumonia, wound infection, and abscesses), wound dehiscence, and renal impairment; it occurs in 17 to 67% of patients (5,54,71,96,160,161). The reported mortality rate for surgical management ranges from 6 to 33% (5,54,70,71,96,160,161). In reality, some patients may be excluded from surgical intervention because of their critical condition, further biasing the survival rates. In the only randomized study directly comparing endoscopic with surgical intervention in severe acute cholangitis, the hospital mortality rate was significantly higher in those treated surgically (see above) (5). The outcome appears to be related to the severity of cholangitis, concomitant medical problems, underlying biliary disease, and level of preoperative hyperbilirubinemia (160–163). Recurrent cholangitis in the absence of extrabiliary obstruction may be due to residual bacteria in an abnormal intrahepatic biliary system that is not draining adequately (164). Elective and semielective surgical management of patients who undergo acute endoscopic or percutaneous biliary decompression will vary depending on the cause of cholangitis. Biliary bypass or resection and bypass may be required. J— Complications Acute renal failure and development of hepatic abscesses are the most common complications of acute cholangitis (7,109). Patients with jaundice are especially prone to the development of renal failure, manifest as acute tubular necrosis, which is multifactorial in origin. In acute biliary obstruction, there is an initial diuresis followed by a significant reduction in intra and extravascular volumes, resulting in a decrease in real blood flow (46,165,166). Impaired cardiovascular responsiveness in the presence of jaundice and ongoing sepsis contributes to the development of systemic hypotension and hypoperfusion of the kidneys (167–169). Endotoxemia per se may play an important role in renal vasoconstruction, while endotoxemia in obstructive jaundice may result in disruption of renal vascular autoregulation (170,171). As a
Page 789
result of these changes, it is apparent that prompt correction of hypovolemia is essential in the management of patients with acute cholangitis (7). In addition, potentially nephrotoxic antibiotics should be avoided or closely monitored, if administered. The oral administration of lactulose or sodium deoxycholate to patients with jaundice may provide additional protection against renal injury by reducing systemic endotoxemia (172–174). The development of intrahepatic abscesses contributes to the mortality of patients with cholangitis (7). Ascending infection leads to neutrophil accumulation around the terminal bile ductules and, if biliary drainage is inadequate, multiple abscesses may subsequently develop. Biliary peritonitis has been reported as a rare complication of cholangitis after rupture of a bile ductule located close to the liver surface (175). K— Prognosis Acute bacterial cholangitis is a potentially devastating illness; but, with appropriate management, including the use of effective nonsurgical methods of biliary decompression, mortality now is approximately 5 to 10% (5,7,176). Table 5 lists the factors identified in three series that were associated with increased morbidity and mortality, either at diagnosis or after intervention. Other investigators have found that a low serum albumin and elevated serum urea and creatinine were associated with increased mortality in patients with cholangitis secondary to choledocholithiasis (54). Patients with underlying malignancies or cirrhosis and/or those who develop complications related to cholangitis have the highest mortality rates. In one prospective study, 59% of patients with biliary tract malignancies were cured of cholangitis, compared with 83% who had benign disease (75). Prognosis worsens in severe cholangitis when biliary decompression is delayed, emphasizing the need for rapid assessment and institution of therapy (91). L— Bacteria, Antibiotics, and the Biliary Tract: Special Situations 1— Antibiotic Prophylaxis for Biliary Surgery The goals of prophylactic therapy are to prevent cholangitis and the complications that may arise after instrumentation or surgery, such as wound infections and abscesses. Cox et al. found that the bile culture results from 451 patients correlated with specific biliary tract abnormalities and postoperative complications (177). The microorganisms responsible for postoperative cholangitis and septicemia were usually cultured from the biliary tract at the time of operation, and antibiotics reduced the incidence of complications after surgery. Patients with sterile bile cultures have a lower rate of postoperative infections (178). Accordingly, efforts have been made to identify those patients who would benefit most from prophylactic antibiotic therapy Table 5 Factors Identified with Increased Morbidity and Mortality in Patients with Acute Bacterial Cholangitis Gigot et al. (4)
Thompson et al. (75)
Lai et al. (96)
Acute renal failure
Malignancy
Concomitant medical illnesses
Liver abscess/cirrhosis
Bacteremia
Acidosis, pH < 7.4
High malignant stricture
Bilirubin 17.4 mmol/L
Total bilirubin > 90 mmol/L
a
2 organisms in bile
PostPTHC
Platelet count < 150 × 109 K/U
Female gender
Panresistant organism isolated
Serum albumin < 30 g/L
Advanced age
( 3 factors: mortality of 55% 2 factors: mortality of 6%)
a
PTHC, Percutaneous transhepatic cholangiogram.
Page 790
prior to surgery. Keighley et al. recommended antibiotic prophylaxis for elderly patients (age over 70 years), patients who were jaundiced, and those who had a recent history of rigors, CBD obstruction, or choledocholithiasis (12). Other indications for prophylaxis were patients requiring an emergency operation, if a patient was within 4 weeks of an emergency admission, or if he or she had had previous biliary tract surgery. The presence of one or more of these ''highrisk factors" correlated strongly with bactibilia. More recently, other investigators have found an incidence of bactibilia in at least 36% of patients undergoing biliary surgery with similar risk factors (179,180). In a metanalysis of 42 randomized, controlled trials of antibiotic prophylaxis in biliary surgery, there was an overall difference in infection rates of 9% in favor of antibiotics (181). The protective effect was up to 25% in highrisk patients who underwent wound inspection after discharge from hospital. Highrisk patients were defined as those who had acute cholecystitis within 4 weeks of surgery, emergency cholecystectomy, or CBD stone exploration. Other risk factors included jaundice at the time of surgery, age above 60 years, previous biliary tract surgery, morbid obesity, diabetes mellitus, concomitant alimentary procedures, or nonvisualization of the gallbladder on oral cholecystography. However, bactibilia rates of 11 to 54% have been reported in lowrisk groups of patients (180,182,183). As a result, some have recommended antibiotic prophylaxis for all patients undergoing biliary surgery (183). A variety of antimicrobial agents have been used for prophylaxis. Keighley et al. demonstrated that plasma and tissue levels of antibiotic appear to be more important than biliary excretion for effective prophylaxis (118). Patients who received gentamicin, which is poorly concentrated in bile, had lower postoperative infection rates than those who received rifamide, an agent that is predominately excreted via the biliary tract. In a metanalysis, Meijer et al. found there was no significant difference in efficacy between first, second, or thirdgeneration cephalosporins or between single or multidose regimens in postoperative wound infection rates (181). Minor or major wound infection rates were no different in a trial involving 1004 patients comparing a single preoperative dose of cefuroxime versus a threedose regimen, consisting of one dose preoperatively and two doses postoperatively (184). Cefazolin and ceftizoxime were equally effective in preventing infections in 150 patients undergoing elective biliary tract surgery (185). Kujath found that intravenous ciprofloxacin was as effective as ceftriaxone in prophylaxis for highrisk patients (186). In another study, there was no significant difference between amoxicillinclavulanic acid and cefamandole in preventing wound infections after elective biliary surgery (187). Many agents may therefore be used for prophylaxis. Factors such as local antibiotic susceptibility profiles and cost influence agent selection. We recommend using a single dose of a secondgeneration cephalosporin, such as cefuroxime or cefotetan, in highrisk patients because of efficacy and ease of administration. Timing of antibiotic administration is another important factor in surgical prophylaxis. Prophylactic therapy should be given at least 30 min prior to the procedure, so that adequate levels in the blood, gallbladder bed, liver, peritoneal cavity, and the wound will be achieved during the intervention (46). 2— Antibiotic Prophylaxis for Endoscopic and Percutaneous Biliary Drainage Several placebocontrolled studies have examined the question of antibiotic prophylaxis prior to ERCP. A variety of agents were used, including ciprofloxacin, cefotaxime, tetracycline, and piperacillin (188–193). Routine antibiotic prophylaxis was not necessary for all patients (194,195). However, patients who presented with biliary obstruction did benefit from prophylactic therapy. Current guidelines recommend antibiotic prophylaxis prior to ERCP for all patients with known or suspected biliary obstruction (196). It is accepted that incomplete biliary drainage is the single most important risk factor for cholangitis after ERCP (197). Undoubtedly, endoscopic expertise reduces the likelihood of inadequate drainage after ERCP. However, certain patients are at particular risk, including those who have proximal biliary strictures, hilar malignancies, or primary sclerosing cholangitis (PSC) (198). In these situations, cholangitis may recur or a liver abscess may develop in an
Page 791
undrained lobe that has been injected with contrast (3,14). Therefore, prophylactic antibiotics have a role in patients where it is suspected that drainage is unlikely to be complete after intervention. Antibiotics started prior to or at the time of the ERCP should be continued until adequate drainage is achieved. The antibiotics available for prophylaxis are the same as those employed for the treatment of acute cholangitis (Table 4). Factors that influence the actual agent selected include local bacterial susceptibility patterns, potential toxicities, and cost as well as a patient's history of drug allergy. Aminoglycosides may be given, as their usage is usually limited to a single dose. Intravenous thirdgeneration cephalosporins or the quinolones are other alternatives. Oral ciprofloxacin has been used successfully and compared favorably with intravenous cephalosporins with respect to cost and convenience (190). The newer fluoroquinolones (ofloxacin and levofloxacin) have even better bioavailability than ciprofloxacin (130). All oral antibiotics should be administered 1 h before the procedure for maximum efficacy. Antibiotic prophylaxis prior to PTBD is accepted as standard practice (152). 3— Antibiotics, T Tubes, and Postoperative Choledochoscopy In a study of 164 patients who underwent postoperative Ttube cholangiography, 62 received cephalothin intravenously prior to the test, followed by oral cephalexin for 3 days, while 71 patients received no antibiotics (199). There was no significant difference in the rates of complications between either group, suggesting that routine antibiotic prophylaxis is not necessary prior to Ttube cholangiography. However, antibiotics should be considered in patients with positive bile cultures who would tolerate bacteremia poorly, such as elderly patients and those with significant comorbidities, or in patients who were febrile immediately prior to the procedure (200). Chen and Jan studied 100 patients prospectively to determine the frequency of bacteremia after postoperative choledochoscopy (201). Of these, 15 patients were found to have bacteremia within 30 min of the procedure, 6 of whom developed cholangitis within 24 h. Only aerobic bacteria were isolated on culture, Enterococcus, E. coli, and Klebsiella species being the most common isolates. All patients recovered with antibiotic therapy. Therefore, in postoperative choledochoscopy, it seems reasonable to individualize prophylactic therapy, taking into account the patient's underlying condition and the procedure to be performed. 4— Recurrent Cholangitis and Maintenance Antibiotic Therapy Patients at risk for recurrent cholangitis include those with a biliaryenteric bypass, sphincteroplasty, or a biliary stent in situ. Others at risk include those with a diffusely abnormal biliary tree, such as PSC (3). Maintenance antibiotic therapy has been suggested as a means to prevent or decrease the frequency of recurrent septic episodes (202). Biliary excretion of antibiotics may be more important in this setting than in the treatment of acute cholangitis. Effective therapy may reduce the bacterial concentration in the biliary tract below a certain critical level or prevent septicemia if organisms reach the systemic circulation. Other important features of maintenance therapy include availability in oral form at an appropriate cost and effectiveness against the common biliary pathogens. One suggested agent is trimethoprimsulfamethoxazole, which has been used extensively in the prevention of recurrent urinary tract infections (202). Alternatives include the fluoroquinolones, which are much more expensive but have excellent biliary excretion; amoxicillinclavulanic acid, which has activity against E. coli but may have limited coverage against Klebsiella species; and the cephalosporins, such as cefixime (203). A selection of antibiotics may also be taken on a rotating schedule in an effort to prevent the emergence of resistance to any one particular agent. The duration of maintenance therapy is unknown, but it may have to continue for as long as the patient's biliary tract remains abnormal.
Page 792
5— Biliary Stents Stent patency after placement is influenced by stent design and composition. Factors that may contribute to stent clogging include stent diameter and shape, the presence of side holes, and the roughness of the stent's inner surface (60). Stents with diameters smaller than 10 Fr are more likely to become blocked. Biomechanical approaches to prevent stent clogging have centered on altering the materials used in the manufacture of the prosthetic devices to reduce microbial attachment (204). These include the synthesis of hydrophilic polymers, alteration in the number of stent side holes, or the incorporation of compounds with antimicrobial properties (205– 209). However, none have been shown to be better than the standard polyethylene stent in the clinical setting (9,210). Selfexpanding metal stents have better stent patency rates than their plastic counterparts, although tumor occlusion remains a problem (211,212). Prophylactic administration of antibiotics and bile salts has also been suggested as a measure to prevent stent blockage (213,214). However, the results from clinical studies involving small numbers of patients have been inconsistent. Furthermore, there is a high dropout rate because of progression of the underlying malignancy, which necessitated stent placement (215). Barrioz et al. compared norfloxacin and ursodeoxycholic acid (UDCA) with conservative treatment in 20 patients who had a polyethylene stent placed for malignant biliary obstruction (61). Patients receiving the drug combination had longer stent patency rates and a prolonged median survival (67 versus 18 weeks in the conservatively treated group). Ghosh and Palmer found no difference in stent occlusion rates when a regimen consisting of antibiotics (rotating between ampicillin, metronidazole, and ciprofloxacin) and UDCA were compared with no treatment (62). Similarly, Luman et al. did not demonstrate any prolongation in stent patency or patient survival in a prospective study comparing ciprofloxacin and Rowachol, a choleretic agent, with no active treatment (63). The large number of bacterial species with variable antibiotic sensitivities isolated from biliary stents may render singleagent therapy for either prophylaxis or therapy of acute cholangitis ineffective. In one study, 43% of the Enterobacteriaceae isolated from the biliary stents were resistant to piperacillin (104). However, further clinical trials will be necessary to determine whether prophylactic antibiotics provide any additional benefit to patients who have stems placed for malignant biliary obstruction. III— Fungal Infections of the Biliary Tract The commonest fungal infection of the biliary tract is Candida albicans. This organism is more likely to be isolated in bile cultures of patients with malignant than benign disease (4,216). In one study, Candida was found in the bile of 5% of patients with benign biliary obstruction, compared with 28% of those with malignant obstruction (75). Another study found biliary Candida in 61% of 90 cancer patients who underwent therapeutic biliary procedures (217). Candida frequently colonizes sites other than the biliary tree in patients with positive bile cultures, but candidemia is rare (216). Since Candida is usually isolated in bile with a mixed bacterial flora, broadspectrum antibiotics should be administered along with antifungal therapies (216). No studies have been performed comparing fluconazole with amphotericin B in the treatment of biliary candidiasis. IV— Recurrent Pyogenic Cholangitis A — Introduction The presence of hepatolithiasis and recurrent bouts of cholangitis characterize recurrent pyogenic cholangitis (RPC), also known as Oriental cholangiohepatitis. In primary hepatolithiasis, stones form within the intrahepatic ducts, whereas in the secondary form, retrograde migration of stones occurs from the extrahepatic biliary system (218,219). RPC is endemic to East Asia,
Page 793
with the highest prevalence being found in Taiwan (219–221). Recurrent fever, jaundice, and abdominal pain are the usual presenting features of RPC. Patients with this condition are predisposed to the development of hepatic parenchymal destruction, biliary cirrhosis, and cholangiocarcinoma. Management requires a multidisciplinary approach involving radiological, endoscopic, and surgical interventions when appropriate. B— Epidemiology The incidence of hepatolithiasis in East Asia is between 3 and 54%, compared with less than 2% in western countries (222,223). As a result, the clinical syndrome of RPC is an important cause of morbidity in countries where hepatolithiasis is common. However, RPC is increasingly being recognized in the West as a result of increased emigration from Asia (224,225). In Kashmir, RPC constitutes 13% of all biliary diseases (226). Over the last 20 years, there has been a gradual increase in gallstone prevalence among the Asian countries, with a concomitant decline in the incidence of hepatolithiasis, especially in Japan and Taiwan (14,221,227). This has been attributed to improved environmental conditions and changes in dietary habits, with increased westernization. Males and females are affected by hepatolithiasis with a similar frequency. The mean age at presentation is now usually in the sixth and seventh decades (227). C— Pathogenesis There are three different types of stones in primary hepatolithiasis—calcium bilirubinate (also known as brown pigment), cholesterol, and mixed stones (219). The vast majority of patients with RPC have calcium bilirubinate stones, which consist of up to 60% calcium bilirubinate and contain less than 20% cholesterol. They are brown to orange in color, have a soft consistency, and are usually multiple in number. Microscopic examination reveals a glycoprotein matrix composed of calcium salts of bilirubin, fatty acid, and cholesterol. Microcolonies of bacteria may be mixed with the crystals. Intrahepatic cholesterol stones are mainly composed of cholesterol, while mixed stones contain varying amounts of cholesterol and calcium bilirubinate. Primary cholesterol hepatolithiasis appears to be secondary to altered regulation of cholesterol and bile acid synthesis and is not associated with bactibilia (228). The formation of brown pigment stones results from a complex combination of bile infected with enteric bacteria, stasis within the biliary tract, and diminished host defenses due to malnutrition (229,230). Enteric bacteria like E. coli have been found to colonize the biliary tract in up to 95% of cases (231). These bacteria can elaborate enzymes—such as ß Dglucuronidase, which hydrolyzes watersoluble bilirubin, resulting in the formation of waterinsoluble unconjugated bilirubin and the precipitation of calcium bilirubinate (229,232). Lecithin is broken down by phospholipase, precipitating calcium palmitate. Calcium bilirubinate and palmitate can be detected in the stones by scanning electron microscopy with xray microanalysis (233). Malnutrition and a lowprotein diet may be important in stone pathogenesis as they result in a deficiency of biliary inhibitors of ß Dglucuronidase (14). Biliary tract infestation with the parasites Ascaris lumbricoides and Clonorchis sinensis has been implicated in the development of hepatolithiasis (234,235). Much of the evidence supporting a causative role for Ascaris in RPC pathogenesis comes from Kashmir, where Ascaris is endemic. One study found that 5% of patients with Ascaris infection of the biliary tract developed RPC over a 2year followup and 10% of patients with RPC had evidence of ascariasis (236). The parasite may invade the papilla, causing papillitis, or an edematous papilla seen on endoscopy (226,237). Khuroo et al. described papillitis in 84 of 227 patients with RPC who underwent endoscopy (226). Motor abnormalities of the sphincter of Oddi may be induced by this process, resulting in impaired biliary drainage (237). Khuroo et al. found that 8 of 15 patients with RPC had motor abnormalities of the sphincter, although only 6 of the 15 patients had papillitis (237). Furthermore, the Ascaris worms may transport bacteria into the biliary tract during their migration to the liver (69). They can cause obstruction once in the CBD, and
Page 794
dead worm fragments may act as the nidus for the formation of biliary calculi (69,238,239). Clonorchis sinensis, has been documented radiologically in patients with RPC in countries where this parasite is endemic (240). Clonorchis infection results in hyperplasia and desquamation of biliary epithelium, leading to stricture formation (69). However, the precise role of parasitic infestation in the etiology of hepatolithiasis is unclear. Intrahepatic stones are far more prevalent than Clonorchis infestation in Taiwan and remain common in Japan, despite a decline in the incidence of Ascaris infestation in the last 40 years (223,241). In western countries, hepatolithiasis usually develops in the setting of stasis as a result of benign or malignant stricture formation, choledochal cysts, or Caroli's disease. Benign strictures develop in sclerosing cholangitis or as a complication of surgery, ischemia, or chronic pancreatitis or after chemoembolization for hepatocellular carcinoma (220,242). D— Pathology In RPC, the intrahepatic changes consist of strictured and dilated ducts, with stones forming within the abnormal ducts. The left hepatic duct system is more commonly affected than the right (219). Repeated bouts of inflammation results in periportal fibrosis, bile duct proliferation, granuloma formation, and neutrophil infiltration. The sequelae of recurrent cholangitis include parenchymal damage with abscess formation, liver atrophy, and progression to secondary biliary cirrhosis (243). The development of cholangiocarcinoma is associated with RPC and can arise in any portion of the biliary tree (244). The initial histological changes are those of chronic infection in the bile ducts with desquamation of biliary epithelium, followed by hyperplasia and adenomatous proliferation. Atypical epithelial hyperplasia and dysplasia are precancerous findings (219,245). The actual incidence of cholangiocarcinoma in patients with hepatolithiasis ranges from 2 to 10%, with some cases only being detected at autopsy (246–248). E— Clinical Features Patients with RPC usually present with some or all of Charcot's triad. The development of Reynolds' pentad is associated with a poor outcome (219). Patients may complain of pruritus secondary to cholestasis. Features suggestive of malignant transformation include anemia, weight loss, and intractable pain (249,250). F— Investigations Biochemical abnormalities during an acute attack are similar to those seen in acute bacterial cholangiris. Bile cultures are usually positive and polymicrobial infections are common (113,251,252). In recent studies, E. coli, P. aeruginosa, and Enterococcus were the most frequently isolated biliary organisms (251,252). Anaerobes may be isolated in up to 12% of patients with positive bile cultures (251). On admission, blood cultures have been reported to be positive in 21 to 47% of patients (113,252). Multiple organisms are isolated in less than 10% of blood cultures. Ascaris worms may be visualized invading the ampullary orifice at endoscopy and Clonorchis eggs have been demonstrated in the bile of patients with RPC (236,253). Radiological investigations accurately define the anatomy of the biliary tree and hepatic parenchyma, providing critical information prior to any intervention (254). Ultrasound is recommended as the primary noninvasive examination because it can identify abnormal ducts and stones in a large proportion of patients (254). Abdominal CT (Fig. 2) better assesses parenchymal damage and complications of RPC, such as hepatic abscesses. CT is particularly helpful in patients with recurrent disease after previous biliary surgery and to localize obstructed ducts, which cannot be visualized by direct cholangiography. Although MRI is not as readily available as CT, it is more sensitive in the diagnosis of small ductal stones and acutely inflamed ducts
Page 795
Figure 2 Abdominal CT scan demonstrating multiple intrahepatic calculi in the right lobe of the liver.
or parenchyma (254). It is likely that the combination of MRI of the liver and MRCP will be increasingly used for the noninvasive evaluation of the liver, portal system, and biliary tree (255,256). While technetiumlabeled HIDA scans have been used to assess the hepatic function in RPC, they rarely provide additional information to CT or MRI (219). Contrast cholangiography, with either ERCP or PTC, remains the gold standard in defining the extent of ductal disease prior to biliary intervention. In addition, suspicious strictures may be biopsied and brushed for cytology (254). ERCP is less invasive than PTC but may not visualize obstructed proximal segments unless an occlusion cholangiogram is performed. This maneuver may elevate intrahepatic pressure and further aggravate the systemic manifestations of cholangitis. Therefore, small amounts of contrast should be injected during cholangiography to avoid overdistention of the biliary tree (226). PTC is important for both diagnosis and treatment of patients with RPC. It may demonstrate intrahepatic disease more satisfactorily than ERCP, and the presence of a biliaryenteric anastamosis often precludes the retrograde approach. In addition, cholangioscopy can be performed through an appropriately sized percutaneous tract. Accurate preoperative diagnosis of a cholangiocarcinoma associated with hepatolithiasis remains challenging, despite the use of CT and cholangiography (244,257– 259). The majority of cases of cholangiocarcinoma were previously diagnosed postoperatively, but now most cases can be diagnosed before or at laparotomy (247). The role of imaging techniques such as liver MRI and MRCP in evaluating patients with RPC for suspected cholangiocarcinoma has yet to be determined. G— Treatment 1— Overview The initial goals of therapy are to control infection, localize the abnormality, and determine the best approach for further treatment (219). The administration of broad spectrum antibiotics, intravenous fluids, and analgesics is essential. Around 30% of patients presenting with acute
Page 796
cholangitis secondary to hepatolithiasis will require emergency intervention because of septicemia, persistent fever, or worsening peritonitis (227). In a series of 88 patients, the two factors with independent significance in predicting the need for emergency intervention were a tachycardia of more than 100 beats per minute within 24 h of presentation and an admission platelet count less than 150 × 109/L (252). Both these findings reflect systemic sepsis secondary to the underlying cholangitis. The aims of intervention are to establish adequate biliary drainage, remove all stones, and prevent residual stasis. Providing a means to access the biliary tract that will permit further therapy if required is also important (219,220). The extent of any intervention will depend upon the clinical condition of the patient. Ideally, the biliary system is decompressed, stones are removed, and strictures are corrected in one procedure (252). In addition, residual or recurrent stones should be able to pass spontaneously into the intestinal tract. However, in a critically ill patient, once biliary decompression is performed, definitive therapy should be deferred until the patient is more stable. Therefore initial intervention may be with ERCP or percutaneous transhepatic biliary drainage (PTBD) (252). It is essential that the liver and biliary tree be fully evaluated radiologically prior to embarking upon definitive surgery. 2— Endoscopic and Percutaneous Therapy Extrahepatic stones are amenable to endoscopic removal in over 90% of cases, with a complication rate of less than 10% (219,260). After successful ES and stone extraction, patients may obtain a prolonged symptomfree interval, with significant resolution of abnormalities on repeat cholangiography (226). No longterm complications were reported in 21 patients who had complete clearance of intrahepatic stones postES (261). However, stones located above strictures pose significant endoscopic challenges. Stricture dilatation is necessary prior to stone extraction, and the stones themselves may be impacted or not accessible for removal by basket or other endoscopic devices (219,227). Furthermore, ES without adequate treatment of intrahepatic strictures and stones may increase the risk of subsequent cholangitis (219,261). This may be related to preexisting papillitis at the time of ES (226). PTBD has an important role in the treatment of RPC. Stones may be removed by basket or forceps or fragmented by mechanical crushing or lithotripsy, using electrohydraulic shockwave or laser techniques. The percutaneous approach is particularly useful in patients who are poor surgical candidates or who refuse surgery and when the disease is not amenable to endoscopic therapy because of location or previous biliary surgery (262,263). The success of percutaneous stone removal is over 80%, but it is influenced by multiple factors, including intrahepatic duct angulation, the site and consistency of stones, and the extent and location and of strictures (262–264). In addition, percutaneous drainage is rendered more difficult and hazardous in the presence of cirrhosis. Cholangioscopy using a transhepatic tract created by sequential catheter dilation allows targeted removal of intrahepatic stones with lithotripsy (265,266). Multiple procedures are frequently required to dilate the percutaneous tract sufficiently so the choledochoscope can be passed into the intrahepatic ducts (263). Complications include hemobilia and sepsis, and the rate of stone recurrence after percutaneous therapy may be considerable. Yeh et al. used percutaneous cholangioscopy with Dormia basket: extraction of stones or electrohydraulic shock wave lithotripsy as primary therapy in 165 patients (263). They reported that 21% of patients had at least one episode of cholangitis during or just after percutaneous intervention and two patients died during treatment. The overall recurrence rate for intrahepatic stones in their patients was 33%, after a mean followup of 58 months. Jan and Chen followed 48 patients for up to 10 years after successful percutaneous transhepatic cholangioscopic lithotripsy for hepatolithiasis (262). The cumulative recurrent stone rate was 28.5 and 40% at 5 and 7 years, respectively. No stones were retained in patients without strictures. Secondary biliary cirrhosis developed in 1 patient who had successful percutaneous treatment and in 3 of 7 for whom the procedure failed. Cholangiocarcinoma developed in 2 patients, both within 3 years of initial
Page 797
therapy. Yang et al. used PTBD to enable cholangioscopy and electrohydraulic lithotripsy in 19 patients (267). Complete stone removal was possible in 18 of 19 patients (95%). The mean number of cholangioscopic sessions was 2.7. Minor complications occurred in 5 patients, while 1 patient required transarterial embolization to control severe bleeding. Alternative methods for treating hepatolithiasis include extracorporeal shockwave lithotripsy (ESWL), dissolution therapy, and laser lithotripsy. Stone fragmentation can be achieved by ESWL, but fragment removal is frequently incomplete (268). Better results may be achieved when ESWL is combined with ERCP if stones are not associated with difficult ductal strictures (269). ESWL requires that a cholangiogram be performed to facilitate targeting, which usually results in the patient undergoing two or more ERCPs as well as the lithotripsy sessions (270). Stone dissolution therapy does not appear to have a significant role in treating brown pigment stones (271). The rhodamine6G pulsed dye laser, which has been used in the treatment of giant and impacted CBD stones, can also be used to treat hepatolithiasis (272,273). This unit contains a special stone recognition system, which minimizes bile duct exposure to laser energy when the stone is not in contact with the probe (274). Conventional fluoroscopy at ERCP will provide sufficient targeting, as only 5 to 8% of the total pulse energy is delivered unless the returning reflected light indicates a hard target (270). Intraductal stones can be fragmented with the laser system by either the percutaneous or endoscopic approach, with subsequent fragment extraction by balloon or basket (219,273,275,276). Nonsurgical approaches to the management of biliary strictures have been investigated, as persistent intrahepatic strictures predispose patients to recurrent stone formation. In one series of 57 patients, postoperative balloon dilatation of biliary strictures through percutaneous or Ttube tracts increased the immediate complete stone clearance rate from 0 to 95% (277). The cumulative probability of restricturing was 8% at 3 years. Complications of stricture dilatation included septicemia and hemobilia, each occurring in 10.5% of patients. However, dilatation can be limited by the presence of acute and/or multiple angulated strictures (278). Biliary stents may be employed after stricture dilatation to facilitate stone removal or to prevent stricture recurrence (279). Jeng et al. described 20 patients with predominately longsegment (1.5 cm or greater) rightsided strictures who underwent plastic stent placement for at least 6 months (280). Ten patients with similar strictures refused stenting. The cumulative probability of restricturing was 10% in the stented group compared with 80% in those patients who were not stented. While plastic stents are useful in treating hepatolithiasis as an adjunct to other therapies, the role of expandable metal stents in RPC appears to be limited. These stents can be inserted percutaneously, endoscopically, or along Ttube tracts, but their longterm patency rates are low. Yoon et al. reported patency rates of 75 and 46% at 12 and 36 months, respectively, in a group of 23 patients who underwent metallic stent placement (281). Recurrent stones or sludge and epithelial hyperplasia resulted in stent obstruction. The overall nonsurgical treatment—related mortality for RPC has been estimated between 0.5 and 2% (219). Percutaneous stone removal is unsuccessful in approximately 20% of cases, and intrahepatic stones recur in around 30%, usually in patients with persistent strictures (262,263). Sharply angulated strictures associated with rightsided hepatolithiasis are particularly difficult to manage (282). Furthermore, nonsurgical interventions do not provide definitive therapy for complications of hepatolithiasis, such as cholangiocarcinoma. As a result of these problems, a combination of surgical and nonsurgical approaches is essential for successful management of RPC. 3— Surgical Therapy Intrahepatic stones and strictures may be managed surgically in a variety of ways. In addition, flexible choledochoscopy and intraoperative ultrasonography are useful imaging tools that visualize stones and surrounding structures, facilitating appropriate therapy. For access, a choledochotomy or hepaticodochotomy with or without extension into the right or left hepatic ducts may be employed (219). As the CBD may be obscured by adhesions, peridochal venous plexus,
Page 798
or left lobe hypertrophy, biliary access can be achieved through the left duct, left lateral inferior duct, or right inferior duct (227). Biliary drainage is possible through techniques such as sphincterotomy and surgical sphincteroplasty or the formation of a biliaryenteric anastamosis. Routine use of flexible choledochoscopy and electrohydraulic lithotripsy for large or impacted stones has been recommended (227). Combined surgical and radiological techniques in the management of RPC results in stone clearance rates approaching 90% (242,283). With careful preoperative evaluation, complete removal of stones and possible formation of a biliodigestive anastamosis, stone recurrence rates can be reduced below 16% (227,283). Prophylactic antibiotics appear to be beneficial in patients undergoing surgery for hepatolithiasis. Lee et al. found that appropriate perioperative antibiotic therapy against bacteria cultured from bile was associated with a postoperative complication rate of 17%, compared with 85% in patients where antibiotics were ineffective (251). Adequate perioperative antibiotic therapy must cover gramnegative bacilli, especially Pseudomonas and E. coli, Enterococcus species, and anaerobic bacteria. Hepatic resection is indicated in hepatolithiasis when the involved liver segment is destroyed by recurrent infection that has resulted in atrophy or hepatic abscess formation (219,284). Left lobectomy or left lateral segmentectomy can be considered for isolated leftsided disease (220). Right lobectomy is rarely performed because of its high complication rate (219). Combining cholangioscopy with resection can increase the stone clearance rate as the recovering biliary tract is evaluated more completely (285,286). Operative morbidity for hepatic resection ranges from 12 to 32% and operative mortality is around 2% (284,285). The most common complications include wound infection, subphrenic abscess, and biliary fistula formation (284). These may be related to the presence of infected bile and liver abscesses at the time of surgery. The stone recurrence rate after hepatic resection is less than 20%, as a result of the contralateral lobe being affected by RPC (250,284–287). However, several studies have demonstrated a lower rate of stone recurrence in patients who have undergone resection compared with those who do not undergo hepatectomy (250,287). Furthermore, Jeng et al. showed that patients who underwent partial left hepatectomy had a reduced postoperative complication rate and length of hospitalization compared with those who did not have a resection, even when the disease affected both lobes of the liver (286). Finally, a retrospective review of 614 patients with hepatolithiasis found that patients who underwent hepatic resection were more likely to be symptomfree and to have a lower incidence of secondary biliary cirrhosis and cholangiocarcinoma than those who did not have resection (250). Surgical approaches to managing recurrent disease have been developed that allow for repeated biliary instrumentation but minimize the need for reexploration of affected patients. The formation of a hepaticocutaneous jejunostomy provides permanent access to the biliary tree (227,288,289). Stone extraction and stricture dilatation can be performed through the stoma under light sedation (288). After therapy is completed, the stoma can be closed and buried subcutaneously, but it may be reopened if required (290). Problems associated with this stoma include leakage of bile and mucus, resulting in cutaneous excoriation when it is open. Parastomal hernias develop in approximately 15% of patients with a hepaticocutaneous jejunostomy after stomal closure (291). An alternative approach is the formation of hepaticojejunal biliary access loop attached to the subparietal anterior abdominal wall (292). The subparietal access loop appears to provide reliable longterm biliary access and is not associated with hernia development (293). Hepatic resection is the only treatment available for patients with cholangiocarcinoma that can provide any prospect of prolonged survival (294,295). Resection is less likely to be performed if the tumor is located in the right lobe (296). In one series of 20 patients with cholangiocarcinoma complicating hepatolithiasis, overall operative morbidity and mortality rates after hepatic resection were 36 and 7%, respectively (296). The same group from Hong Kong reported a median survival of 12.2 months in 39 patients who underwent resection (294). An important adverse factor in outcome is the presence of a pyogenic hepatic abscess. In 11 patients with cholangiocarcinoma complicating hepatolithiasis who presented with a liver abscess, mortality within 30 days of treatment was 55% (258).
Page 799
Another lesion that may cause diagnostic and therapeutic difficulties in patients with RPC is hepatocellular carcinoma, the leading cause of cancerrelated death in Southeast Asian men. This tumor may develop in patients with coexisting hepatolithiasis; its presence may be masked by features of infection (297). Hepatic resection for either hepatocellular carcinoma or cholangiocarcinoma is limited by the presence of biliary infection and lobar atrophy, reducing the likelihood of longterm survival. V— Parasitic Infestations of the Biliary Tract A— Ascaris Lumbricoides 1— Introduction Ascaris lumbricoides is the most prevalent helminthic infestation in the world, with over 1.4 billion people being infected (236). Women are more commonly affected than men (298,299). Ascaris infestation is predominately seen in tropical and subtropical countries, where it is maintained by favorable environmental conditions. Up to 60% of people living in South Africa, Southeast Asia, India, and South America may be infected by Ascaris (219). It is most prevalent in crowded rural areas with poor sanitation, especially where fresh human feces is used as fertilizer (236). In the United States, it is the third most common helminthic infection, after hookworm and Trichuris (236). Frequent travel to endemic countries and increased emigration from the Far East have increased the likelihood of encountering Ascaris infection in the West (299). In endemic areas, ascariasis is as common as gallstones in causing biliary and pancreatic diseases (236). A prospective study from India found Ascaris in 40 of 109 patients (37%) presenting with biliary and pancreatic diseases, compared with gallstones in 38 patients (35%) (238). 2— Pathogenesis A. lumbricoides requires moist, shady soil for embryonation of the fertilized eggs (236). Humans become infected after ingesting eggs that are present in contaminated food, water, and soil. The larvae hatch in the jejunum and penetrate into the lymphatics and portal circulation of the cecum, migrating to the thoracic duct and liver, respectively (69). Larvae can move freely once in the hepatic sinusoids. Some may pass to the lungs via the hepatic veins or the thoracic duct. From there they can penetrate the alveoli and ascend to the hypopharynx; they may subsequently be coughed up and swallowed into the gut. The adult forms migrate into locations such as the biliary and pancreatic ducts, leading to obstruction. Previous biliary tract surgery or endoscopic sphincterotomy predisposes patients in endemic areas to disease by facilitating passage of the worms into the CBD (300). In one large series of 300 patients with pancreaticbiliary acariasis, 80 and 77% had a history of cholecystectomy and sphincterotomy, respectively (299). A higher rate of Ascarisrelated complications has been reported during the period of Ramadan, when Muslims observe a daylong fast (299). 3— Clinical Features Clinical symptoms depend upon the size and location of the worm. Ascarides in the duodenum can enter the ampulla and advance along the biliary, hepatic, cystic, and pancreatic ducts, resulting in biliary colic, acute cholangitis, acalculous cholecystitis, and pancreatitis, respectively (299). Abdominal pain is the commonest presenting symptom, occurring in practically all patients (299). In addition, some patients may give a history of worm emesis, either before or at the time of the acute presentation. Ascending cholangitis was diagnosed in 48 of 300 patients (16%) with pancreaticbiliary ascariasis (299). The development of liver abscess with subsequent rupture has also been reported (236,301). Ascaris infection has been associated with the development of RPC (see above).
Page 800
4— Diagnosis The finding of adult worms, larvae, or eggs makes the diagnosis of Ascaris infestation. It is most readily established by examination of the stool for eggs. The thick smear technique of Kato and Miura and simple sedimentation may increase the field of egg detection (236). Ultrasound of the biliary tract is usually the first radiological investigation performed in patients with RUQ pain, and it can detect worms within the biliary tract in over 85% of cases (302). In addition, repeated examinations can be performed to monitor worm movement. Various ultrasonographic appearances have been described, including multiple parallel echogenic strips or long echogenic structures (302). The liver may be lifted off the main portal vein, resulting in obliteration of the CBD lumen (303). However, as ultrasound cannot diagnose duodenal ascariasis, approximately 50% of cases of pancreaticbiliary ascariasis will be missed (298,299). Ascaris worms may be identified in the intestinal tract by contrast studies or at upper endoscopy. ERCP is the investigation of choice if it can be carried out soon after the onset of pain (238). Several days after the acute episode has resolved, the worms may have migrated from the biliary tract and reside in the duodenum. In biliary ascariasis, the ampulla usually appears injured, with a dilated and/or lacerated orifice. Sandouk et al. found that 278 of 300 patients (93%)had ampullary damage and/or worms protruding from the orifice (299). Most patients have a dilated CBD on cholangiography and single or multiple filling defects may be noted (299). 5— Treatment and Prevention In acute cholangitis secondary to Ascaris, patients should receive adequate hydration, analgesia, and antispasmodics. Specific treatment in Ascarisrelated intestinal and biliary obstruction consists of orally administered piperazine, which causes an immediate flaccid paralysis of the worms (236,304). These are then expelled distally by peristalsis. If performed promptly after symptom onset, endoscopic extraction is possible once the worms have been identified (299). While ERCP may not be readily available in some areas when patients present acutely, it should be performed in those patients who are not responding to intensive medical treatment or when worms are still present in the ducts at 3 weeks (305). Worm extraction by Dormia basket can usually be achieved, even in patients who have not had a sphincterotomy, because of Ascarisrelated damage to the ampulla (236). Complete clearance of worms from the biliary tract can be achieved in over 95% of patients at ERCP (299). Surgical extraction is necessary only in the occasional patient where medical and endoscopic treatments have been unsuccessful (299). The administration of antihelminthic agents (albendazole and piperazine) into the biliary tract has been described (306,307). However, intraductal antihelminthic treatment may impede migration of live worms out of the affected ducts into the duodenum (236). Endoscopic sphincterotomy itself may increase the risk of worm reinvasion and recurrence of cholangitis (226). Oral antihelminthic therapy is required to eradicate residual gut infection (236). Mebendazole, albendazole, or pyrantal pamoate are the agents of first choice. Symptomatic recurrence was reported in 62 of 300 patients (21%) after initial extraction, necessitating a second therapeutic ERCP (299). Health education and improved sanitation, combined with mass chemotherapy to populations at risk, should control ascariasis in endemic areas. Boiling kills Ascaris eggs within minutes, but they are resistant to the standard methods of chemical water purification (236). B— Liver Fluke Cholangitis The liver flukes that usually cause disease in humans are Opisthorchis viverrini, Clonorchis sinensis, and Fasciola hepatica (69). These are trematodes with a life cycle comprising sexual phases in freshwater snails (the intermediate host) and mammals (the definitive host). Eggs passed from the definitive host are excreted in feces, hatch in freshwater into miricidia, and penetrate the intermediate host. The miricidia develop into cercariae, which penetrate freshwater
Page 801
fish and are infective when the fish is eaten raw (Opisthorchis viverrini and Clonorchis sinensis), or they may encyst on plants such as watercress and infect humans when ingested (Fasciola). 1— Clonorchis Sinensis Introduction. Clonorchis sinensis is found primarily in Asia, eastern Europe, and Siberia (69). While many people may be infested with this trematode, only subjects with a heavy worm burden will be symptomatic. Clonorchis infection does not develop de novo in the United States, as there is no intermediate snail host. However, since Clonorchis has the ability to survive for several decades within the biliary system, it may be found in emigrants from endemic areas. As a result of this long life span, 26% of Asian immigrants had evidence of active liver fluke infection when stool specimens were screened (308). Pathogenesis. After contaminated raw fish has been ingested, the metacercariae excyst in the duodenum. The worms migrate up the biliary tree, lodging in small bile ducts (309). The biliary tract is thought to be injured by mechanisms including mechanical irritation and fluke secretions (69). Ductal inflammation, epithelial hyperplasia, and progressive portal fibrosis result in obstruction, recurrent cholangitis, and cirrhosis. In endemic areas, a significant association has been found between cholangiocarcinoma and prior Clonorchis infection (310). The hyperplastic bile duct epithelium becomes dysplastic and finally undergoes malignant transformation to cholangiocarcinoma. Clinical Features and Diagnosis. Clonorchis infection can produce either an acute or chronic illness. Acute clonorchiasis presents as a virallike syndrome of abdominal pain, diarrhea, hepatomegaly, jaundice, and eosinophilia. Features of chronic infection include abdominal pain, anorexia, fever, and hepatomegaly (301). The symptoms and signs of biliary obstruction may be evident, and eventually those of cirrhosis or cholangiocarcinoma may develop. The diagnosis of clonorchiasis is confirmed by the presence of an adult worm in duodenal aspirates or bile or by ova in stool samples (219,244). Abdominal ultrasound and CT may demonstrate thickened, dilated intrahepatic bile ducts, but the worms are usually too small to be identified by either of these techniques. They may appear as filamentous, wavy, and/or elliptical filling defects on cholangiography (311). In addition, bile duct findings include a dilated extrahepatic system with diffuse intrahepatic tapering, the presence of a solitary cyst, multiple cystic dilatations of the intrahepatic ducts, or any combination of these changes (301). Treatment and Prevention. Praziquantel is the treatment of choice for clonorchiasis (219,312). Biliary drainage is necessary for patients presenting with acute cholangitis. The biliary tract abnormalities persist after treatment despite eradication of the infection (313). Reinfection is prevented by the thorough washing and cooking of freshwater fish. 2— Opisthorchis Viverrini Opisthorchis viverrini infestation has similar features to Clonorchis and infection with this trematode is rare in the United States (314). It is associated with cholangitis and an increased risk of cholangiocarcinoma in populations where Opisthorchis infection is endemic. A significant correlation was found between parasite specific antibodies and hepatobiliary abnormalities (315). As a result, it has been suggested that an immunopathological mechanism may be responsible for the development of Opisthorchisrelated biliary disease (315). Furthermore, serum IgG antibody levels may have a role in screening atrisk populations for this trematode. Many individuals who are infected with O. viverrini have nonspecific gastrointestinal complaints (316). Portable ultrasonography is a useful noninvasive technique for the assessment of highrisk communities (316). Praziquantel is the treatment of choice for Opisthorchis infestation (312). Clinical, biochemical, and radiographic improvements have been documented with this agent.
Page 802
3— Fasciola Hepatica Humans become accidental hosts of Fasciola hepatica after eating watercress in areas where sheep and cattle are maintained. An acute clinical syndrome with RUQ pain, fever, and eosinophilia occurs during larval migration in the liver (69). This trematode can reside in the biliary tree and cases of biliary fascioliasis causing pain and jaundice have been reported when there is heavy infestation (317). Imaging demonstrates multiple conglomerated filling defects in the CBD (318). The worms may be seen at ERCP and endoscopic extraction can be performed successfully (317). Bithionol is the recommended treatment for F. hepatica infestation (312). C— Echinococcal Cholangitis 1— Echinococcus Granulosus Echinococcus granulosus is a worldwide problem and is associated with poor sanitary conditions, especially where humans, dogs, and cattle live in close proximity (69). The adult form develops in the definitive host, usually a dog: it is passed in feces and ingested by intermediate hosts (humans or livestock). The embryo is released into the duodenum and passes to the liver and lung and subsequently other sites. Embryos develop into twolayered cysts, which contain fluid, protoscolices, and occasional daughter cysts. Echinococcal cysts usually involve the biliary tract when they rupture into a duct, producing obstruction, pain, and cholangitis (319). Other complications include cyst compression of bile ducts and portal veins or erosion into adjacent cavities or viscera. Ultrasound is helpful in establishing the diagnosis of cholangitis from a ruptured cyst (319,320). ERCP may demonstrate a dilated CBD with hydatid membranes or daughter cysts (321). Definitive therapy requires that all cysts and fluid be removed surgically. The associated cholangitis is treated with antibiotics and biliary drainage (69,320). Endoscopic sphincterotomy can be performed pre or postoperatively to clear the CBD of hydatid material and relieve the obstruction (321). Cystbiliary communication after surgical or percutaneous drainage can usually be confirmed and treated endoscopically (320). The cyst contents may be retrieved by basket, and sphincterotomy usually results in closure of the biliary fistula (320,322). Praziquantel should be administered if cyst spillage into the peritoneal cavity occurs during drainage. Albendazole is the scolicidal agent of choice (312,320). 2— Echinococcus Multilocularis The definitive host of Echinococcus multilocularis is the fox. Eggs are passed in the feces, contaminating grass and wild fruits, which are then eaten by humans (323). The eggs spread from the duodenum to sites including the liver. Affected patients usually remain asymptomatic for many years, until the hepatic lesions, consisting of cysts and fibrous tissue, invade the intrahepatic bile ducts and blood vessels. These ducts and vessels are compressed and destroyed by the intense granulomatous host reaction (301). Patients typically complain of pain. Jaundice and irregular hepatomegaly are found on examination. Complications of E. multilocularis infection include bacterial cholangitis, hepatic abscess formation, and secondary biliary cirrhosis. Diagnosis may be made using serology and imaging studies. A diagnostic technique employing the reverse transcription polymerase chain reaction for the detection of echinococcalspecific messenger RNA from biopsy specimens has recently been described (324). Further definition of the extent of disease can be made with percutaneous cholangiography and angiography. The treatment options for E. multilocularis infection are partial hepatectomy with restoration of biliary drainage or liver transplantation (69,301). VI— AIDS Cholangiopathy A— Epidemiology AIDS cholangiopathy describes the biliary tract abnormalities occurring in patients with AIDS. The characteristic changes involve sclerosing cholangitis and/or papillary stenosis (325,326).
Page 803
AIDS cholangiopathy is most likely a result of chronic inflammation by one or more opportunistic infections (OI). The actual incidence of this disorder is unknown, as definitive diagnosis requires cholangiography. Biliary tract disease usually occurs in patients who have established AIDS and CD4 counts less than 50/mm3 (327,328). However, AIDS cholangiopathy has been reported as the first presentation of HIV infection (329–331). With the introduction of highly active antiretroviral therapy (HAART), the occurrence and natural history of OI in AIDS have been dramatically altered. This has led to a decline in the morbidity and mortality associated with AIDS (332–334). AIDS cholangiopathy may be similarily affected by HAART in the future. B— Pathogenesis The stricturing along the biliary tract is thought to be due to inflammation secondary to OI. While AIDS cholangiopathy was originally associated with infection by either CMV or Cryptosporidium, many other pathogens—including Mycobacterium avium complex (MAC), microsporidia, Herpes simplex virus, Pseudomonas aeruginosa, Candida, and Cyclospora cayetanenis—have since been implicated in the development this condition (10,325,329,330,335–342). No pathogen may be isolated in up to 50% of cases (329,343). The HIV virus itself does not appear to directly infect the biliary epithelium. Some retrospective series have suggested that male homosexuality is a major risk factor for the development of AIDS cholangiopathy. This association may be related to selection bias or the higher rate of gut pathogen carriage occurring in homosexuals (327,330). C— Clinical Features Patients usually present with RUQ pain, cholestatic LFT elevations, and morphological abnormalities of the biliary tree. Patients with papillary stenosis may have more severe pain and develop clinical cholangitis compared with those who only have intrahepatic disease. Other less specific symptoms of AIDS cholangiopathy include fever, nausea, and vomiting (344). Diarrhea may occur because some OI, such as microsporidia, can involve the small bowel as well. Cholangitis may be associated with cholecystitis in patients with AIDS (345). D— Investigations The canalicular enzymes ALP and GGT are commonly elevated 10 to 20 times above normal. Jaundice is rare, as obstruction is usually incomplete (327,346–348). Ultrasound may reveal dilated and thickened intra and extrahepatic ducts as well as a hyperechoic nodule in the distal CBD, which correlates with papillary stenosis seen at ERCP (349). Definitive diagnosis requires direct imaging of the biliary tree, as the symptoms and biochemical abnormalities can be indistinguishable from that of hepatic MAC infection. Cholangiography is performed in patients demonstrating biliary dilatation on RUQ ultrasound or in patients without biliary dilatation where a liver biopsy has failed to identify any mycobacterial infection (329,344,350). Abnormalities at ERCP include papillary stenosis, a narrowing of the terminal CBD with proximal dilatation and extrahepatic and/or intrahepatic bile duct stricturing (Fig. 3a) (327,329). The ducts may have the classic ''beaded" mucosal pattern of sclerosing cholangitis (343). Papillary stenosis may also involve the pancreas, resulting in juxtaampullary pancreatic duct strictures (330,343). The commonest finding is papillary stenosis in association with intrahepatic sclerosing cholangitis. Although MRCP may also demonstrate the classic biliary findings of AIDS cholangiopathy, no study has directly compared MRCP with ERCP in this condition. ERCP offers the additional advantages of allowing for tissue sampling and sphincterotomy. Duodenal, papillary, and biliary tract biopsies sent for both histological and microbiological analysis may identify specific organisms (Fig. 3b) (330,344). Duodenal aspiration of
Page 804
Figure 3 a. ERCP of a patient with AIDS cholangiopathy showing a dilated common bile duct (closed arrow) with beading and stricturing of the intrahepatic ducts (open arrows). b. Common bile duct biopsy from the same patient showing numerous Cryptosporidium organisms along the luminal surface of the biliary epithelium. (From Ref. 359.)
Page 805
stimulated bile has also been studied as a means of identifying HIVrelated infections in the upper GI and biliary tracts (351). Stool examination may reveal organisms such as microsporidia (338). Specific pathogen identification often requires the use of special stains on light microscopy or even electron microscopy. The diagnostic accuracy of bile culture is poor (352). E— Treatment Endoscopic sphincterotomy relieves the pain associated with papillary stenosis in 45 to 86% of patients (327,344,348,353). Responses may be sustained for the remainder of the patient's course. Serum biochemical values do not generally improve after sphincterotomy and the radiographic changes may progress (327,344,348,352,353). We do not recommend sphincterotomy in patients without papillary stenosis as it does not improve symptoms and may increase the risk of bacterial cholangitis in patients with more proximal disease. If a dominant CBD stricture is present, endoscopic balloon dilatation and/or stenting may be considered. Collazos et al. recently reported successful celiac plexus nerve block in three patients with pain unresponsive to conventional analgesia (354). There is no specific treatment for patients with diffuse sclerosing cholangitis. While numerous pathogens have been associated with AIDS cholangiopathy, the disease only rarely responds to treatment of specific viruses or organisms (355). Ganciclovir and foscarnet have been shown to eliminate CMV from the duodenum without producing an improvement in the radiographic images (327,331). Clearly effective treatment for cryptosporidia and microsporidia are lacking (339,340). There are reports of patients with Cyclospora infection of the biliary tract responding to trimethoprimsulfamethoxazole (337). One patient who received paromomycin and letrazuril for cryptosporidial cholangitis responded clinically and radiologically (355). There also have been reports of patients responding symptomatically and biochemically to ursodeoxycholic acid (330,356). In patients presenting with features suggesting cholecystitis in addition to suspected AIDS cholangiopathy, the decision to perform a cholecystectomy should be based on clinical grounds (345,357). Cholecystectomy can provide rapid symptom relief in such patients, although the features of AIDS cholangiopathy may continue to progress after surgery. No information is currently available concerning the use of HAART as treatment for AIDS cholangiopathy. It is not known if the symptoms or the biochemical and/or radiographic changes will improve on HAART as the CD4 count recovers. F— Prognosis In the preHAART era, the prognosis for AIDS cholangiopathy was poor, with a reported 1year survival of 41% in one series (344). The most important prognostic indicator was the patients' degree of immunosuppression, as the usual cause of death was from progression of AIDS itself (328,331). There is clear evidence demonstrating a significant reduction in mortality and prolonged AIDSfree survival on HAART (334,358). Intensive antiretroviral therapy may yet prove to be the best means of preventing and treating AIDS cholangiopathy. Acknowledgments Elizabeth Buonpane, Pharm. D., for reviewing the manuscript, the Microbiological Laboratory at Boston Medical Center for providing microbiological data, Helen Fenlon, M.B., for assistance with the radiological images, and T.V. Keaveny, M.B., M.Ch., F.A.C.S., and Gurmeet Sandhu for their advice and support during the chapter's preparation.
Page 806
References 1. Charcot JM. Lecons sur les maladies du foi, des voiles billare et des reins. Paris: Faculte de Medicine de Paris 1877, pp 194–195. 2. Reynolds BM, Dargan EL. Acute obstructive cholangitis. Ann Surg 1959; 150:299–303. 3. van den Hazel SJ, Speelman P, Tytgat GN, Dankert J, van Leeuwen DJ. Role of antibiotics in the treatment and prevention of acute and recurrent cholangitis. Clin Infect Dis 1994; 19:279–286. 4. Gigot JF, Leese T, Dereme T, Coutinho J, Castaing D, Bismuth H. Acute cholangitis: multivariate analysis of risk factors. Ann Surg 1989; 209:435–438. 5. Lai EC, Mok FP, Tan ES, Lo CM, Fan ST, You KT, Wong J. Endoscopic biliary drainage for severe acute cholangitis. N Engl J Med 1992; 326:1582–1586. 6. Lee JG. Role of endoscopic therapy in cholangitis. Am J Gastroenterol 1998; 93:2016–2018. 7. Lipsett PA, Pitt HA. Acute cholangitis. Surg Clin North Am 1990; 70:1297–1312. 8. Pitt HA. Does cholangiovenous reflux cause cholangitis? HPB, Surg 1990; 2:220–223. 9. Sung JJ. Bacterial biofilm and clogging of biliary stents. J Ind Microbiol 1995; 15:152–155. 10. Hanau LH, Steigbigel NH. Cholangitis: pathogenesis, diagnosis, and treatment. Curr Clin Top Infect Dis 1995; 15:153–178. 11. Flemma RJ, Flint LM, Osterhout S, Shingleton WW. Bacteriologic studies of biliary tract infection. Ann Surg 1967; 166:563–572. 12. Keighley MR, Flinn R, AlexanderWilliams J. Multivariate analysis of clinical and operative findings associated with biliary sepsis. Br J Surg 1976; 63:528–531. 13. Csendes A, Burdiles P, Maluenda F, Diaz JC, Csendes P, Mitru N. Simultaneous bacteriologic assessment of bile from gallbladder and common bile duct in control subjects and patients with gallstones and common duct stones. Arch Surg 1996; 131:389–394. 14. Lee DW, Chung SC. Biliary infection. Baillieres Clin Gastroenterol 1997; 11:707–724. 15. Sung JY, Costerton JW, Shaffer EA. Defense system in the biliary tract against bacterial infection. Dig Dis Sci 1992; 37:689–696. 16. Sand J, Airo I, Hiltunen KM, Mattila J, Nordback I. Changes in biliary bacteria after endoscopic cholangiography and sphincterotomy. Am Surg 1992; 58:324– 328. 17. Bapat RD, Supe AN, Patwardhan A, Kocher HM, Parab S, Sathe MJ. Biliary sepsis: an ascending infection. Indian J Gastroenterol 1996; 15:126–128. 18. Suzuki Y, Kobayashi A, Ohto M, Tsuchiya Y, Saisho H, Kimura K, Ono T, Okuda K. Bacteriological study of transhepatically aspirated bile: relation to cholangiographic findings in 295 patients. Dig Dis Sci 1984; 29:109–115. 19. Ohdan H, Oshiro H, Yamamoto Y, Tanaka I, Inagaki K, Sumimoto K, Hinoi T. Bacteriological investigation of bile in patients with cholelithiasis. Surg Today 1993; 23:390–395. 20. O'Connor MJ, Schwartz ML, McQuarrie DG, Sumner HW. Cholangitis due to malignant obstruction of biliary outflow. Ann Surg 1981; 193:341–345. 21. Kalser MH, Cohen R, Arteaga I, Yawn E, Mayoral L, Hoffert WR, Frazier D. Normal viral and bacterial flora of the human small and large intestine. N Engl J Med 1966; 274:500–505, 558–563. 22. Plant AG, Gorbach SL, Nahab L, Weinstein L, Spanknebel G, Levitan R. Studies of intestinal microflora: III. The microbial flora of human small intestinal mucosa and fluids. Gastroenterology 1967; 53:868–873. 23. Edlund YA, Mollstedt BO, Ouchterlony O. Bacteriological investigation of the biliary system and liver in biliary tract disease correlated to clinical data and microstructure of the gallbladder and liver. Acta Chir Scand 1958; 116:461–476. 24. Cetta F. The route of infection in patients with bactibilia. World J Surg 1983; 7:562. 25. Kinoshita H, Hirohashi K, Igawa S, Nagata E, Sakai K. Cholangitis. World J Surg 1984; 8:963–969.
Page 807
26. Dineen P. The importance of the route of infection in experimental biliary tract obstruction. Surg Gynecol Obstet 1964; 119:1001–1008. 27. Sung JY, Shaffer EA, Olson ME, Leung JWC, Lam K, Costerton JW. Bacterial invasion of the biliary system by way of the portalvenous system. Hepatology 1991; 14:313–317. 28. Ong GB. A study of recurrent pyogenic cholangitis. Arch Surg 1962; 84:199–225. 29. Orloff MJ, Peskin GW, Ellis HL. A bacteriologic study of human portal blood: implications regarding hepatic ischemia in man. Ann Surg 1958; 148:738–746. 30. Katz S, Grosfeld JL, Plager DA, Ross D, Rosenthal RS, Hull M, Weber TR. Impaired bacterial clearance and trapping in obstructive jaundice. Ann Surg 1984; 199:14–20. 31. Tanaka N, Christensen P, Ryden S, KlofverStahl B, Bengmark S. Decreased clearance of Escherichia coli from the bile in rats with obstructive jaundice. Eur Surg Res 1985; 17:281–285. 32. Tanaka N, Christensen P, Ryden S, KlofverStahl B, Bengmark S. Biliary obstruction and susceptibility to biliary sepsis in rats. Res Exp Med 1985; 185:115– 119. 33. Ding JW, Andersson R, Soltesz V, Willen R, Bengmark S. Obstructive jaundice impairs reticuloendothelial function and promotes bacterial translocation in the rat. J Surg Res 1994; 57:238–245. 34. Drivas G, James O, Wardle N. Study of reticuloendothelial phagocytic capacity in patients with cholestasis. BMJ 1976; 1:1568–1569. 35. Scott AJ, Kahn GA. Origin of bacteria in bileduct bile. Lancet 1967; 2:790–792. 36. Justesen T, Lykkegaard Nielsen M. Anaerobic infection of the liver and biliary tract in experimental common duct occlusion. Scand J Infect Dis 1979; 19 (suppl):35–41. 37. Raper SE, Barker ME, Jones AL, Way LW. Anatomic correlates of bacterial cholangiovenous reflux. Surgery 1989; 105:352–359. 38. Lygidakis NJ, Brummelkamp WH. The significance of intrabiliary pressure in acute cholangitis. Surg Gynecol Obstet 1985; 161:465–469. 39. Williams RD, Fish JC, Williams DD. The significance of biliary pressure. Arch Surg 1967; 95:374–379. 40. Huang T, Bass JA, Williams RD: The significance of biliary pressure in cholangitis. Arch Surg 1969; 98:629–632. 41. Csendes A, Sepulveda A, Burdiles P, Braghetto I, Bastias J, Schutte H, Diaz JC, Yarmuch J, Maluenda F. Common bile duct pressure in patients with common bile duct stones with or without acute suppurative cholangitis. Arch Surg 1988; 123:697–699. 42. Toyota N, Miyai K, Hardison WG. Effect of biliary pressure versus high bile acid flux on the permeability of hepatocellular tight junction. Lab Invest 1984; 50:536–542. 43. Boyer JL. Tight junctions in normal and cholestatic liver: does the paracellular pathway have functional significance? Hepatology 1983; 3:614–617. 44. Metz J, Bressler D. Reformation of gap and tight junctions in regenerating liver after cholestasis. Cell Tissue Res 1979; 199:257–270. 45. Devos R, Desmet VJ. Morphological changes of the junctional complex of the hepatocytes in rat liver after bile duct ligation. J Exp Pathol 1978; 59:220–227. 46. Rege R. Adverse effects of biliary obstruction: implications for treatment of patients with obstructive jaundice. AJR 1995; 164:287–293. 47. Groen A, Out T, Huibregtse K, Delzenne B, Hoek F, Tytgat GN. Characterization of the content of occluded biliary endoprostheses. Endoscopy 1987; 19:57– 59. 48. Yu JL, Andersson R, Ljungh A. Protein adsorption and bacterial adhesion to biliary stent materials. J Surg Res 1996; 62:69–73. 49. Leung J, Ling T, Kung J, VallanceOwen J. The role of bacteria in the blockage of biliary stents. Gastrointest Endosc 1988; 34:19–22. 50. Speer AG, Cotton PB, Rode J, Seddon AM, Neal CR, Holton J, Costerton JW. Biliary stent blockage with bacterial biofilm: a light and electron microscopy study. Ann Intern Med 1988; 108:546–553.
Page 808
51. Chan FK, Suen M, Li JY, Sung JJ. Bile immunoglobulins and blockage of biliary endoprosthesis: an immunohistochemical study. Biomed Pharmacother 1998; 52:403–407. 52. Leung JW, Liu YL, Desta T, Libby E, Inciardi JF, Lam K. Is there a synergistic effect between mixed bacterial infection in biofilm formation on biliary stents? Gastrointest Endosc 1998; 48:250–257. 53. Sung JY, Leung JW, Shaffer EA, Lam K, Costerton JW. Bacterial biofilm, brown pigment stone and blockage of biliary stents. J Gastroenterol Hepatol 1993; 8:28–34. 54. Leese T, Neoptolemos JP, Baker AR, CarrLocke DL. Management of acute cholangitis and the impact of endoscopic sphincterotomy. Br J Surg 1986; 73:988–992. 55. Jarvinen H. Abnormal liver function tests in acute cholecystitis; the predicting of common duct stones. Ann Clin Res 1978; 10:323–327. 56. Grier JF, Cohen SW, Grafton WD, Gholson CF. Acute suppurative cholangitis associated with choledochal sludge. Am J Gastroenterol 1994; 89:617–619. 57. Nomura T, Shirai Y, Hatakeyama K. Cholangitis in malignant biliary obstruction. Br J Surg 1998; 85:407. 58. Freeman M, Nelson D, Sherman S, Haber G, Herman M, Dorsher P, Moore J, Fennerty M, Ryan M, Shaw M, Lande J, Pheley A. Complications of endoscopic biliary sphincterotomy. N Engl J Med 1996; 335:909–918. 59. Dowidar N, Moesgaard F, Matzen P. Clogging and other complications of endoscopic biliary endoprostheses. Scand J Gastroenterol 1991; 26:1132–1136. 60. Gilbert D, DiMarino A, Jensen D, Katon R, Kimmey M, Laine LA, MacFadyen B, MichaletzOnody P, Zuckerman G. Status evaluation: biliary stents. Gastrointest Endosc 1992; 38:750–752. 61. Barrioz T, Ingrand P, Besson I, de Ledinghen V, Silvain C, Beauchant M. Randomised trial of prevention of biliary stent occlusion by ursodeoxycholic acid plus norfloxacin. Lancet 1994; 344:581–582. 62. Ghosh S, Palmer KR. Prevention of biliary stent occlusion using cyclical antibiotics and ursodeoxycholic acid. Gut 1994; 35:1757–1759. 63. Luman W, Ghosh S, Palmer K. A combination of ciprofloxacin and Rowachol does not prevent biliary stent occlusion. Gastrointest Endosc 1999; 49:316–321. 64. Davids P, Groen AK, Rauws EA, Tytgat GN, Huibregtse K. Randomized trial of selfexpanding metal stents versus polyethylene stents for distal malignant biliary obstruction. Lancet 1992; 340:1488–1492. 65. Knyrim K, Wagner HJ, Pausch J, Vakil N. A prospective, randomized, controlled trial of metal stents for malignant obstruction of the common bile duct. Endoscopy 1993; 25:207–212. 66. Biliary stents. Gastrointest Endosc 1994; 40:798. 67. Lygidakis NJ. Incidence of bile infection in patients with choledocholithiasis. Am J Gastroenterol 1982; 77:12–17. 68. Siegel JH. Duodenoscopic sphincterotomy in the treatment of the "sump syndrome." Dig Dis Sci 1982; 26:922–928. 69. Carpenter H. Bacterial and parasitic cholangitis. Mayo Clin Proc 1998; 73:473–478. 70. Boey JH, Way LW. Acute cholangitis. Ann Surg 1980; 191:264–270. 71. Himal HS, Lindsay T. Ascending cholangitis: surgery versus endoscopic or percutaneous drainage. Surgery 1990; 108:629–633. 72. Gumaste VV, Leung JW. Antibiotics and cholangitis. Gastroenterology 1995; 109:323–325. 73. Cobden I, Lendrum R, Venables CW, James OF. Gallstones presenting as mental and physical debility in the elderly. Lancet 1984; 1:1062–1064. 74. Saharia PC, Cameron JL. Clinical management of acute cholangitis. Surg Gynecol Obstet 1976; 142:369–372. 75. Thompson JE, Jr., Pitt HA, Doty JE, Coleman J, Irving C. Broad spectrum penicillin as an adequate therapy for acute cholangitis. Surg Gynecol Obstet 1990; 171:275–282.
Page 809
76. Saik RP, Greenburg AG, Farris JM, Peskin GW. Spectrum of cholangitis. Am J Surg 1975; 130:143–150. 77. Albert MB, Steinberg WM, Henry JP. Elevated serum levels of tumor marker CA 199 in acute cholangitis. Dig Dis Sci 1988; 33:1223–1225. 78. Krastev Z, Vladimirov B, Mateva L, Alexiev A. Quantitative assessment of severity of biliary tract infection. Hepatogastroenterology 1996; 43:792–795. 79. Saini S. Imaging of the hepatobiliary tract. N Engl J Med 1997; 336:1889–1894. 80. Taylor KJ, Rosenfield AT, Spiro HM. Diagnostic accuracy of gray scale ultrasonography for the jaundiced patient. Arch Intern Med 1979; 139:60–63. 81. Gaines P, Markham N, Leung J, Metreweli C. The thick common bile duct in pyogenic cholangitis. Clin Radiol 1991; 44:175–177. 82. Taylor TV, Sumerling MD, Carter DC, McLoughlin GP, Miller AM. An evaluation of 99Tcmlabelled HIDA in hepatobiliary scanning. Br J Surg 1980; 67:325– 328. 83. Kaplun L, Weissmann HS, Rosenblatt RR, Freeman LM. The early diagnosis of common bile duct obstruction using cholescintigraphy. JAMA 1985; 254:2431– 2434. 84. Chan YL, Chan AC, Lam WW, Lee DW, Chung SS, Sung JJ, Cheung HS, Lai AK, Metreweli C. Choledocholithiasis: comparison of MR cholangiography and endoscopic retrograde cholangiography. Radiology 1996; 200: 85–89. 85. Hintze RE, Adler A, Veltzke W, AbouRebyeh H, Hammerstingl R, Vogl T, Felix R. Clinical significance of magnetic resonance cholangiopancreatography (MRCP) compared to endoscopic retrograde cholangiopancreatography (ERCP). Endoscopy 1997; 29: 182–187. 86. Reinhold C, Taourel P, Bret PM, Cortas GA, Mehta SN, Barkun SN, Wang L, Tafazoli F. Choledocholithiasis: evaluation of MR cholangiography for diagnosis. Radiology 1998; 209:435–442. 87. Adamek HE, Weitz M, Breer H, Jakobs R, Schilling D, Reimann JF. Value of magnetic resonance cholangiopancreatography (MRCP) after unsuccessful endoscopicretrograde cholangiopancreatography (ERCP). Endoscopy 1997; 29:741–744. 88. Brugge WR. Endoscopic ultrasonography: the current status. Gastroenterology 1998; 115:1577–1583. 89. Amouyal P, Amouyal G, Levy P, Tuzet S, Palazzo L, Vilgrain V, Gayet B, Belghiti J, Fekete F, Bernades P. Diagnosis of choledocholithiasis by endoscopic ultrasonography. Gastroenterology 1994; 106:1062–1067. 90. Burtin P, Palazzo L, Canard JM, Person B, Oberti F, Boyer J. Diagnostic strategies for extrahepatic cholestasis of indefinite origin: endoscopic ultrasonography or retrograde cholangiography? Results of a prospective study. Endoscopy 1997; 29:349–355. 91. Williams JG, Neoptolemos JP. Cholangitis. In: Taylor MB, ed. Gastrointestinal Emergencies, 2nd ed. Baltimore: Williams & Wilkins, 1997, pp 275–288. 92. Kadir S, Baassiri A, Barth KH, Kaufman SL, Cameron JL, White RI Jr. Percutaneous biliary drainage in the management of biliary sepsis. AJR 1982; 138:25– 29. 93. Dumont AE. Significance of hyperbilirubinemia in acute cholecystitis. Surg Gynecol Obstet 1976; 142:855–857. 94. Crawford JM, Boyer JL. Clinicopathological conferences: inflammationinduced cholestasis. Hepatology 1998; 28:253–260. 95. Thompson J, Bennion RS, Pitt HA. An analysis of infectious failures in acute cholangitis. HPB Surg 1994; 8:139–144. 96. Lai EC, Paterson IA, Tam PC, Choi TK, Fan ST, Wong J. Severe acute cholangitis: the role of emergency nasobiliary drainage. Surgery 1990; 107:268–272. 97. Brook I. Aerobic and anaerobic microbiology of biliary tract disease. J Clin Microbiol 1989; 27:2373–2375. 98. Csendes A, Mitru N, Maluenda F, Diaz JC, Burdiles P, Csendes P, Pinones E. Counts of bacteria and pyocites of choledochal bile in controls and in patients with gallstones or common bile duct stones with or without acute cholangitis. Hepatogastroenterology 1996; 43:800–806.
Page 810
99. Bourgault AM, England DM, Rosenblatt JE, Forgacs P, Bieger RC. Clinical characteristics of anaerobic bactibilia. Arch Intern Med 1979; 139:1346–1349. 100. Nielsen ML, Justesen T. Anaerobic and aerobic bacteriological studies in biliary tract disease. Scand J Gastroenterol 1976; 11:437–446. 101. Shimada K, Urayama K, Noro T, Inamatsu T. Biliary tract infection with anaerobes and the presence of free bile acids in bile. Rev Infect Dis 1984; 6(suppl 1):S147–S151. 102. Jackaman FR, Hilson GR, Smith L. Bile bacteria in patients with benign bile duct stricture. Br J Surg 1980; 67:329–332. 103. Malden ES, Picus D, Dunagan WC. Anaerobic culture yield in interventional radiologic drainage procedures. J Vasc Intervent Radiol 1995; 6:933–937. 104. Molinari G, Pugliese V, Schito GC, Guzman CA. Bacteria involved in the blockage of biliary stents and their susceptibility to antibacterial agents. Eur J Clin Microbiol Infect Dis 1996; 15:88–92. 105. Papadakis KA, Vartivarian SE, Vassilaki ME, Anaissie EJ. Stenotrophomonas maltophilia: an unusual cause of biliary sepsis. Clin Infect Dis 1995; 21:1032– 1034. 106. Zuravleff JJ, Yu VL. Infections caused by Pseudomonas maltophilia with emphasis on bacteremia: case reports and a review of the literature. Rev Infect Dis 1982; 4:1236–1246. 107. Acheson DWK, Keusch GT. Intestinal infections with Salmonella and Yersinia species. In: LaMont JT, ed. Gastrointestinal Infections: Diagnosis and Management. New York: Marcel Dekker, 1997, pp 149–189. 108. Lai CW, Chan RC, Cheng AF, Sung JY, W. LJ. Common bile duct stones: a cause of chronic salmonellosis. Am J Gastroenterol 1992; 87:1198–1199. 109. Kadakia SC. Biliary tract emergencies: acute cholecystitis, acute cholangitis and acute pancreatitis. Med Clin North Am 1993; 77:1015–1036. 110. Westphal JF, Brogard JM. Biliary tract infections: a guide to drug treatment. Drugs 1999; 57:81–91. 111. Westphal JF, Blickle JF, Brogard JM. Management of biliary tract infections: potential role of the quinolones. J Antimicrob Chemother 1991; 28:486–490. 112. Neuman M. Clinical pharmacokinetics of the newer antibacterial 4quinolones. Clin Pharmacokinet 1988; 14:96–121. 113. Leung JWC, Ling TKW, Chan RCY, Cheung SW, Lai CW, Sung JJY, Chung SCS, Cheng AFB. Antibiotics, biliary sepsis, and bile duct stones. Gastrointest Endosc 1994; 40:716–721. 114. van den Hazel SJ, de Vries XH, Speelman P, Dankert J, Tytgat GN, Huibregtse K, van Leeuwen DJ. Biliary excretion of ciprofloxacin and piperacillin in the obstructed biliary tract. Antimicrob Agents Chemother 1996; 40:2658–2660. 115. Gerecht WB, Henry NK, Hoffman WW, Muller SM, LaRusso NF, Rosenblatt JE, Wilson WR. Prospective randomized comparison of mezlocillin therapy alone combined with ampicillin and gentamicin therapy for patients with cholangitis. Arch Intern Med 1989; 149:1279–1284. 116. Leung JW, Chan RC, Cheung SW, Sung JY, Chung SC, French GL. The effect of obstruction on the biliary excretion of cefoperazone and ceftazidime. J Antimicrob Chemother 1990; 25:399–406. 117. Keighley MR, Drysdale RB, Quoraishi AH, Burdon DW, AlexanderWilliams J. Antibiotic treatment of biliary sepsis. Surg Clin North Am 1975; 55:1379– 1390. 118. Keighley MR, Drysdale RB, Quoraishi AH, Burdon DW, AlexanderWilliams J. Antibiotics in biliary disease: the relative importance of antibiotic concentrations in the bile and serum. Gut 1976; 17:495–500. 119. Maluenda F, Csendes A, Burdiles P, Diaz J. Bacteriological study of choledochal bile in patients with common bile duct stones, with or without acute suppurative cholangitis. Hepatogastroenterology 1989; 36:132–135. 120. Anonymous. The choice of antibacterial drugs. Med Lett 1998; 40:33–42.
Page 811
121. Paterson DL, Robson JM, Wagener MM. Risk factors for toxicity in elderly patients given aminoglycosides once daily. J Gen Intern Med 1998; 13:735–739. 122. Pitt HA, Postier RG, Cameron JL. Consequences of preoperative cholangitis and its treatment on the outcome of operation for choledocholithiasis. Surgery 1983; 94:447–452. 123. Chamberland S, L'Ecuyer J, Lessard C, Bernier M, Provencher P, Bergeron MG. Antibiotic susceptibility profiles of 941 gramnegative bacteria isolated from septicemic patients throughout Canada: The Canada Study Group. Clin Infect Dis 1992; 15:615–628. 124. Livermore DM. Betalactamasemediated resistance and opportunities for its control. J Antimicrob Chemother 1998; 41(suppl D):25–41. 125. Okereke C, Dudley MN. Betalactam/betalactamase inhibitor combinations: pharmacodynamic considerations and possible role in the management of bacterial infections in the neutropenic host. J Antimicrobial Chemother 1998; 41(suppl D):43–49. 126. Bergeron MG, Mendelson J, Harding GK, Mandell L, Fong IW, Rachlis A, Chan R, Biron S, Feld R, Segal NB. Cefoperazone compared with ampicillin plus tobramycin for severe biliary tract infections. Antimicrob Agents Chemother 1988; 32:1231–1236. 127. Muller EL, Pitt HA, Thompson JE, Jr., Doty JE, Mann LL, Manchester B. Antibiotics in infections of the biliary tract. Surg Gynecol Obstet 1987; 165:285–292. 128. Graninger W, Ragette R. Nosocomial bacteremia due to Enterococcus faecalis without endocarditis. Clin Infect Dis 1992; 15:49–57. 129. Ball CS, Manson JM, Reid F, Tweedle DE. The pharmacokinetics of the biliary excretion of ciprofloxacin. HPB Surg 1989; 1:319–326; discussion 326–327. 130. Hooper DC. Expanding uses of fluoroquinolones: opportunities and challenges. Ann Inter Med 1998; 129:908–910. 131. Sung JJ, Lyon DJ, Suen R, Chung SC, Co AL, Cheng AF, Leung JW, Li AK. Intravenous ciprofloxacin as treatment for patients with acute suppurative cholangitis: a randomized, controlled clinical trial. J Antimicrob Chemother 1995; 35:855–864. 132. Karachalios GN, Nasiopoulou DD, Bourlinou PK, Reppa A. Treatment of acute biliary tract infections with ofloxacin: a randomized, controlled clinical trial. Int J Clin Pharmacol Ther 1996; 34(12):555–557. 133. Scobie BA, Summerskill WHJ. Hepatic cirrhosis secondary to obstruction of the biliary system. Am J Dig Dis 1965; 10:135–146. 134. Siegel JH, Rodriguez R, Cohen SA, Kasmin FE, Cooperman AM. Endoscopic management of cholangitis: critical review of an alternative technique and report of a large series. Am J Gastroenterol 1994; 89:1142–1146. 135. Lau JY, Chung SC, Leung JW, Ling TK, Yung MY, Li AK. Endoscopic drainage aborts endotoxaemia in acute cholangitis. Br J Surg 1996; 83:181–184. 136. Misra SP, Dwivedi M. Biliary endoprosthesis as an alternative to endoscopic nasobiliary drainage in patients with acute cholangitis. Endoscopy 1996; 28:746– 749. 137. Ramirez FC, McIntosh AS, Dennert B, Harlan JR. Emergency endoscopic retrograde cholangiopancreatography in critically ill patients. Gastrointest Endosc 1998; 47:368–371. 138. Yoshimoto H, Ikeda S, Tanaka M, Matsumoto S. Relationship of biliary pressure to cholangiovenous reflux during endoscopic retrograde balloon catheter cholangiography. Dig Dis Sci 1989; 34:16–20. 139. Cohen SA. Biliary tract emergencies: endoscopic and medical management. Crit Care Clin 1995; 11:273–294. 140. Sugiyama M, Atomi Y. The benefits of endoscopic nasobiliary drainage without sphincterotomy for acute cholangitis. Am J Gastroenterol 1998; 93:2065–2068. 141. Cotton PB, Lehman G, Vennes J, Greenen JE, Russell RC, Meyers WC, Liguory C, Nickl N. Endoscopic sphincterotomy complications and their management: an attempt at consensus. Gastrointest Endosc 1991; 37:383–393. 142. Chawla YK, Sharma BC, Dilawari JB. Endoscopic nasobiliary drainage in acute suppurative cholangitis. Indian J Gastroenterol 1994; 13:83–85.
Page 812
143. Kiil J, Kruse A, Rokkjaer M. Large bile duct stones treated by endoscopic biliary drainage. Surgery 1989; 105:51–56. 144. Chopra KB, Peters RA, O'Toole PA, Williams SG, Gimson AE, Lombard MG, Westaby D. Randomised study of endoscopic biliary endoprosthesis versus duct clearance for bile duct stones in highrisk patients. Lancet 1996; 348:791–793. 145. Chan A, Lam Y, Lee D, Law B, Ng E, Lau J, Chung S. Biliary decompression by nasobiliary catheter versus biliary stent in acute suppurative cholangitis: a randomized trial (abstr). Digestion 1998; 59(suppl 3):40. 146. Molnar W, Stockum AE. Relief of obstructive jaundice through percutaneous transhepatic catheter—a new therapeutic method. Am J Roentgenol Radium Ther Nucl Med 1974; 122:356–367. 147. Mueller PR, van Sonnenberg E, Ferrucci JT Jr. Percutaneous biliary drainage: technical and catheterrelated problems in 200 procedures. AJR 1982; 138:17– 23. 148. Gould RJ, Vogelzang RL, Neiman HL, Pearl GJ, Poticha SM. Percutaneous biliary drainage as an initial therapy in sepsis of the biliary tract. Surg Gynecol Obstet 1985; 160:523–527. 149. Pessa ME, Hawkins IF, Vogel SB. The treatment of acute cholangitis: percutaneous transhepatic biliary drainage before definitive therapy. Ann Surg 1987; 205:389–392. 150. Szabo S, Mendelson MH, Mitty HA, Bruckner HW, Hirschman SZ. Infections associated with transhepatic biliary drainage devices. Am J Med 1987; 82:921– 926. 151. Lois JF, Gomes AS, Grace PA, Deutsch LS, Pitt HA. Risks of percutaneous transhepatic drainage in patients with cholangitis. AJR 1987; 148:367–371. 152. Cohan RH, Illescas FF, Saeed M, Perlmutt LM, Braun SD, Newman GE, Dunnick NR. Infectious complications of percutaneous biliary drainage. Invest Radiol 1986; 21:705–709. 153. Joseph PK, Bizer LS, Sprayregen SS, Gliedman ML. Percutaneous transhepatic biliary drainage: results and complications in 81 patients. JAMA 1986; 255:2763–2767. 154. McPherson GA, Benjamin IS, Habib NA, Bowley NB, Blumgart LH. Percutaneous transhepatic drainage in obstructive jaundice: advantages and problems. Br J Surg 1982; 69:261–264. 155. Rogers L. Biliary abscesses of the liver with operation. BMJ 1903; 2:706. 156. Baker AR, Neoptolemos JP, Leese T, Fossard DP. Choledochoduodenostomy, transduodenal sphincteroplasty and sphincterotomy for calculi of the common bile duct. Surg Gynecol Obstet 1987; 164:245–251. 157. Lau WY, Chong KK, Fan ST. Chu KW, Yip WC, Poon GP, Wong KK. A scientific evaluation of operative choledochoscopy in acute cholangitis. Ann Surg 1987; 206:142–147. 158. Lau WY, Chu KW, Yuen WK, Poon GP, Hwang JS, Lai AK. Operative choledochoscopy in patients with acute cholangitis: a prospective, randomized study. Br J Surg 1991; 78: 1226–1229. 159. Gagic N, Frey CF. The results of cholecystostomy for the treatment of acute cholecystitis. Surg Gynecol Obstet 1975; 140:255–257. 160. Lai EC, Tam PC, Paterson IA, Ng MM, Fan ST, Choi TK, Wong J. Emergency surgery for severe acute cholangitis: the highrisk patients. Ann Surg 1990; 211:55–59. 161. Chijiiwa K, Kozaki N, Naito T, Kameoka N, Tanaka M. Treatment of choice for choledocholithiasis in patients with acute obstructive cholangitis and liver cirrhosis. Am J Surg 1995; 170:356–360. 162. Thompson JN, Edwards WH, Winearls CG, Blenkharn JI, Benjamin IS, Blumgart LH. Renal impairment following biliary tract surgery. Br J Surg 1987; 74:843–847. 163. Csendes A, Diaz JC, Burdiles P, Maluenda F, Morales E. Risk factors and classification of acute suppurative cholangitis. Br J Surg 1992; 79:655–658. 164. Goldman LD, Steer L, Silen W. Recurrent cholangitis after biliary surgery. Am J Surg 1983; 145:450–454.
Page 813
165. Levy M, Finestone H. Renal response to four hours of biliary obstruction in the dog. Am J Physiol 1983; 244:F516–F525. 166. Gillet DJ. The effect of obstructive jaundice on the blood volume in rats. J Surg Res 1971; 11:447–449. 167. Binah O, Bomzon A, Blendis LM, Mordohovich D, Better OS. Obstructive jaundice blunts myocardial contractile response to isoprenaline in the dog: a clue to susceptibility of jaundiced patients to shock? Clin Sci 1985; 69:647–653. 168. Green J, Beyar R, Sideman S, Mordechovitz D, Better OS. The ''jaundiced heart": a possible explanation for postoperative shock in obstructive jaundice. Surgery 1986; 100: 14–20. 169. Bomzon A, Rosenberg M, Gali D, Binah O, Mordechovitz D, Better OS, Greig PD, Blendis LM. Systemic hypotension and decreased pressor response in dogs with chronic bile duct ligation. Hepatology 1986; 6:595–600. 170. Mitaka C, Hirata Y, Yokoyama K, Nagura T, Tsunoda Y, Amaha K. Improvement of renal dysfunction in dogs with endotoxemia by a nonselective endothelin receptor antagonist. Crit Care Med 1999; 27:146–153. 171. Inan M, Sayek I, Tel BC, SahinErdemli I. Role of endotoxin and nitric oxide in the pathogenesis of renal failure in obstructive jaundice. Br J Surg 1997; 84:943–947. 172. Cahill CJ. Prevention of postoperative renal failure in patients with obstructive jaundice—the role of bile salts. Br J Surg 1983; 70:590–595. 173. Cahill CJ, Pain JA, Bailey ME. Bile salts, endotoxin and renal dysfunction in obstructive jaundice. Surg Gynecol Obstet 1987; 165:519–522. 174. Pain JA, Cahill CJ, Gilbert JM, Johnson CD, Trapnell JE, Bailey ME. Prevention of postoperative renal dysfunction in patients with obstructive jaundice: a multicentre study of bile salts and lactulose. Br J Surg 1991; 78:467–469. 175. Eyres KS, Williams L, Ali HH, MacDonald RC. Spontaneous rupture of liver due to cholangitis. Br J Clin Pract 1990; 44:740–742. 176. Leung JW, Chung SC, Sung JJ, Banez VP, Li AK. Urgent endoscopic drainage for acute suppurative cholangitis. Lancet 1989; 1:1307–1309. 177. Cox JL, Helfrich LR, Pass HI, Osterhaut S, Shingleton WW. The relationship between biliary tract infections and postoperative complications. Surg Gynecol Obstet 1978; 146: 233–236. 178. Edwards GF, Lindsay G, Taylor EW. A bacteriological assessment of ampicillin with sulbactam as antibiotic prophylaxis in patients undergoing biliary tract operations: The West of Scotland Surgical Infection Study Group. J Hosp Infect 1990; 16:249–255. 179. KasholmTengve B. Selective antibiotic prophylaxis in biliary tract operations. Surg Gynecol Obstet 1991; 173:25–28. 180. Landau O, Kott I, Deutsch AA, Stelman E, Reiss R. Multifactorial analysis of septic bile and septic complications in biliary surgery. World J Surg 1992; 16:962–964; discussion 964–965. 181. Meijer WS, Schmitz PI, Jeekel J. Metaanalysis of randomized controlled clinical trials of antibiotic prophylaxis in biliary tract surgery. Br J Surg 1990; 77:283– 290. 182. Wells GR, Taylor EW, Lindsay G, Morton L. Relationship between bile colonization, highrisk factors and postoperative sepsis in patients undergoing biliary tract operations while receiving a prophylactic antibiotic: West of Scotland Surgical Infection Study Group. Br J Surg 1989; 76:374–377. 183. Aloj G, Bianco C, Covelli I, Blenkharn JI, Blumgart LH, Benjamin IS. Antibiotic prophylaxis for biliary tract surgery: selection of patient and agent. Int Surg 1991; 76:131–134. 184. Meijer WS, Schmitz PI. Prophylactic use of cefuroxime in biliary tract surgery: randomized controlled trial of single versus multiple dose in highrisk patients: Galant Trial Study Group. Br J Surg 1993; 80:917–921. 185. Jewesson PJ, Stiver G, Wai A, Frighetto L, Nickoloff D, Smith J, Schwartz L, Sleigh K, Danforth D, Scudamore C, Chow A. Doubleblind comparison of cefazolin and cef
Page 814
tizoxime for prophylaxis against infections following elective biliary tract surgery. Antimicrob Agents Chemother 1996; 40:70–74. 186. Kujath P. Antibiotic prophylaxis in biliary tract surgery. Ciprofloxacin versus ceftriaxone. Am J Med 1989; 87(suppl 5A):255S–257S. 187. Krige JE, Isaacs S, Stapleton GN, McNally J. Prospective, randomized study comparing amoxycillinclavulanic acid and cefamandole for the prevention of wound infection in highrisk patients undergoing elective biliary surgery. J Hosp Infect 1992; 22(suppl A): 33–41. 188. Brandes JW, Scheffer B, LorenzMeyer H, Korst HA, Littmann KP. ERCP: complications and prophylaxis a controlled study. Endoscopy 1981; 13:27–30. 189. Sauter G, Grabein B, Huber G, Mannes GA, Ruckdeschel G, Sauerbruch T. Antibiotic prophylaxis of infectious complications with endoscopic retrograde cholangiopancreatography: a randomized controlled study. Endoscopy 1990; 22:164–167. 190. Alveyn CG, Robertson DA, Wright R, Lowes JA, Tillotson G. Prevention of sepsis following endoscopic retrograde cholangiopancreatography. J Hosp Infect 1991; 19(suppl C):65–70. 191. Niederau C, Pohlmann U, Lubke H, Thomas L. Prophylactic antibiotic treatment in therapeutic or complicated diagnostic ERCP: results of a randomized controlled clinical study. Gastrointest Endosc 1994; 40:533–537. 192. Byl B, Deviere J, Strulens MJ, Roucloux I, De Coninck A, Thys JP, Cremer M. Antibiotic prophylaxis for infectious complications afer therapeutic endoscopic retrograde cholangiopancreatography: a randomized, doubleblind, placebocontrolled study. Clin Infect Dis 1995; 20:1236–1240. 193. van den Hazel SJ, Speelman P, Dankert J, Huibregtse K, Tytgat GN, van Leeuwen DJ. Piperacillin to prevent cholangitis after endoscopic retrograde cholangiopancreatography: a randomized, controlled trial. Ann Intern Med 1996; 125:442–447. 194. Byl B, Deviere J. Antibiotic prophylaxis before endoscopic retrograde cholangiopancreatography. Ann Intern Med 1997; 126:1001. 195. Meyer GW. Antibiotic prophylaxis for gastrointestinal procedures: who needs it? Gastrointest Endosc 1994; 40:645–646. 196. Dajani AS. Prevention of bacterial endocarditis: recommendations by the American Heart Association. JAMA 1997; 277:1794–1801. 197. Motte S, Deviere J, Dumonceau JM, Serruys E, Thys JP, Cremer M. Risk factors for septicemia following endoscopic biliary stenting. Gastroenterology 1991; 101:1374–1381. 198. Nomura T, Shirai Y, Hatakeyama K. Cholangitis after endoscopic biliary drainage for hilar lesions. Hepatogastroenterology 1997; 44:1267–1270. 199. SheenChen SM, Cheng YF, Chou FF, Lee TY. Postoperative Ttube cholangiography: is routine antibiotic prophylaxis necessary? A prospective, controlled study. Arch Surg 1995; 130:20–23. 200. Pitt HA, Postier RG, Cameron JL. Postoperative Ttube cholangiography: is antibiotic coverage necessary? Ann Surg 1980; 191:30–34. 201. Chen MF, Jan YY. Bacteremia following postoperative choledochofiberoscopy: a prospective study. Hepatogastroenterology 1996; 43:586–589. 202. van den Hazel SJ, Speelman P, Tytgat GN, van Leeuwen DJ. Successful treatment of recurrent cholangitis with antibiotic maintenance therapy. Eur J Clin Microbiol Infect Dis 1994; 13:662–665. 203. Westphal JF, Jehl F, Schloegel M, Monteil H, Brogard JM. Biliary excretion of cefixime: assessment in patients provided with Ttube drainage. Antimicrob Agents Chemother 1993; 37:1488–1491. 204. Hoffman BJ, Cunningham JT, Marsh WH, O'Brien JJ, Watson J. An in vitro comparison of biofilm formation on various biliary stent materials. Gastrointest Endosc 1994; 40: 581–583.
Page 815
205. Coene PP, Groen AK, Cheng J, Out MM, Tytgat GN, Huibregtse K. Clogging of biliary endoprostheses: a new perspective. Gut 1990; 31:913–917. 206. Jansen B, Goodman LP, Ruiten D. Bacterial adherence to hydrophilic polymercoated polyurethane stents. Gastrointest Endosc 1993; 39:670–673. 207. McAllister EW, Carey LC, Brady PG, Heller R, Kovacs SG. The role of polymeric surface smoothness of biliary stents in bacterial adherence, biofilm deposition, and stent occlusion. Gastrointest Endosc 1993; 39:422–425. 208. Seitz U, Vadeyar H, Soehendra N. Prolonged patency with a newdesign Teflon biliary prosthesis. Endoscopy 1994; 26:478–482. 209. Rees EN, Tebbs SE, Elliott TS. Role of antimicrobialimpregnated polymer and Teflon in the prevention of biliary stent blockage. J Hosp Infect 1998; 39:323– 329. 210. van Berkel AM, Boland C, Redekop WK, Bergman JJ, Groen AK, Tytgat GN, Huibregtse K. A prospective randomized trial of Teflon versus polyethylene stents for distal malignant biliary obstruction. Endoscopy 1998; 30:681–686. 211. Davids PH, Rauws EA, Coene PP, Tytgat GN, Huibregtse K. Endoscopic stenting for postoperative biliary strictures. Gastrointest Endosc 1992; 38:12–18. 212. Knyrim K, Wagner HJ, Pausch J, Vakil N. A prospective, randomized, controlled trial of metal stents for malignant obstruction of the common bile duct. Endoscopy 1993; 25:207–212. 213. Tsang TK, Pollack J, Chodash HB. Inhibition of biliary endoprostheses occlusion by ampicillinsulbactam in an in vitro model. J Lab Clin Med 1997; 130:643– 648. 214. Sung JY, Shaffer EA, Lam K, Rususka I, Costerton JW. Hydrophobic bile salt inhibits bacterial adhesion on biliary stent material. Dig Dis Sci 1994; 39:999– 1006. 215. Smit JM, Out MM, Groen AK, Huibregtse K, Jansen PL, van Marle J, Tytgat GN. A placebocontrolled study on the efficacy of aspirin and doxycycline in preventing clogging of biliary endoprostheses. Gastrointest Endosc 1989; 35:485–489. 216. Diebel LN, Raafat AM, Dulchavsky SA, Brown WJ. Gallbladder and biliary tract candidiasis. Surgery 1996; 120:760–764; discussion 764–765. 217. Khardori N, Wong E, Carrasco CH, Wallace S, Patt Y, Bodey GP. Infections associated with biliary drainage procedures in patients with cancer. Rev Infect Dis 1991; 13:587–591. 218. Saunders KD, Cates JA, Roslyn JJ. Pathogenesis of gallstones. Surg Clin North Am 1990; 70:1197–1216. 219. Leung JW, Yu AS. Hepatolithiasis and biliary parasites. Baillieres Clin Gastroenterol 1997; 11:681–706. 220. Neuhaus H. Management of intrahepatic stones. Gastrointest Endosc 1995; 42:94–96. 221. Su CH, Lui WY, P'eng FK. Relative preponderance of gallstone diseases in Taiwan: nationwide cooperative study. Dig Dis Sci 1992; 37:764–768. 222. Nakayama F, Soloway RD, Nakama T, Miyazaki K, Ichimiya H, Sheen PC, Ker CG, Ong GB, Choi TK, Boey J, et al. Hepatolithiasis in East Asia. Retrospective study. Dig Dis Sci 1986; 31:21–26. 223. Cheung KL, Lai EC. The management of intrahepatic stones. Adv Surg 1996; 29:111–129. 224. Sperling RM, Koch J, Sandhu JS, Cello JP. Recurrent pyogenic cholangitis in Asian immigrants to the United States: natural history and role of therapeutic ERCP. Dig Dis Sci 1997; 42:865–871. 225. Harris HW, Kumwenda ZL, SheenChen SM, Shah A, Schecter WP. Recurrent pyogenic cholangitis. Am J Surg 1998; 176:34–37. 226. Khuroo MS, Dar MY, Yattoo GN, Khan BA, Boda MI, Zargar SA, Javid G, Allai MS. Serial cholangiographic appearances in recurrent pyogenic cholangitis. Gastrointest Endosc 1993; 39:674–679. 227. Fan ST, Choi TK, Lo CM, Mok FPT, Lai ECS, Wong J. Treatment of hepatolithiasis: improvement of result by a systematic approach. Surgery 1991; 109:474–480.
Page 816
228. Shoda J, Tanaka N, Matsuzaki Y, Yamamori S, Osuga T. Primary dual defect of cholesterol and bile acid metabolism in liver of patients with intrahepatic calculi. Gastroenterology 1995; 108:1534–1546. 229. Kaufman HS, Magnuson TH, Lillemoe KD, Frasca P, Pitt HA. The role of bacteria in gallbladder and common duct stone formation. Ann Surg 1989; 209:584– 591. 230. Wen CC, Lee HC. Intrahepatic stones: a clinical study. Ann Surg 1972; 175:166–177. 231. Tabata M, Nakayama F. Bacteriology of hepatolithiasis. Prog Clin Biol Res 1984; 152: 163–174. 232. Tabata M, Nakayama F. Bacteria and gallstones: etiological significance. Dig Dis Sci 1981; 26:218–224. 233. Kaufman HS, Magnuson TH, Pitt HA, Frasca P, Lillemoe KD. The distribution of calcium salt precipitates in the core, periphery and shell of cholesterol, black pigment and brown pigment gallstones. Hepatology 1994; 19:1124–1132. 234. Maki T. Pathogenesis of calcium bilirubinate gallstone: role of E. coli, betaglucuronidase and coagulation by inorganic ions, polyelectrolytes and agitation. Ann Surg 1966; 164:90–100. 235. Seel DJ, Park YK. Oriental infestational cholangitis. Am J Surg 1983; 146:366–370. 236. Khuroo MS. Ascariasis. Gastroenterol Clin North Am 1996; 25:553–577. 237. Khuroo MS, Zargar SA, Yattoo GN, Allai MS, Khan BA, Dar MY, Boda MI, Javid G. Oddi's sphincter motor activity in patients with recurrent pyogenic cholangitis. Hepatology 1993; 17:53–58. 238. Khuroo MS, Zargar SA. Biliary ascariasis: a common cause of biliary and pancreatic disease in an endemic area. Gastroenterology 1985; 1985:418–423. 239. Tech TB. A study of gallstone and included worms in recurrent pyogenic cholangitis. J Pathol Bacteriol 1963; 86:123–129. 240. Sullivan WG, Koep LJ. Common bile duct obstruction and cholangiohepatitis in clonorchiasis. JAMA 1980; 243:2060–2061. 241. Maki T. Cholelithiasis in the Japanese. Arch Surg 1961; 82:599–612. 242. Pitt HA, Venbrux AC, Coleman J, Prescott CA, Johnson MS, Osterman FA, Jr, Cameron JL. Intrahepatic stones: the transhepatic team approach. Ann Surg 1994; 219:527–535; discussion 535–537. 243. Fan ST, Wong J. Complications of hepatolithiasis. J Gastroenterol Hepatol 1992; 7:324–327. 244. Kim KH, Kim CD, Lee HS, Lee SJ, Jeen JT, Chun HJ, Song CW, Lee SW, Um SH, Choi JH, Ryu HS, Hyun JH. Biliary papillary hyperplasia with clonorchiasis resembling cholangiocarcinoma. Am J Gastroenterol 1999; 94:514–517. 245. Ona FV, Dytoc JN. Clonorchisassociated cholangiocarcinoma: a report of two cases with unusual manifestations. Gastroenterology 1991; 101:831–839. 246. SheenChen SM, Chou FF, Eng HL. Intrahepatic cholangiocarcinoma in hepatolithiasis: a frequently overlooked disease. J Surg Oncol 1991; 47:131–135. 247. Chen MF, Jan YY, Wang CS, Hwang TL, Jeng LB, Chen SC, Chen TJ. A reappraisal of cholangiocarcinoma in patients with hepatolithiasis. Cancer 1993; 71:2461–2465. 248. Ohta T, Nakanuma Y, Terada T. Pathology of hepatolithiasis: cholangitis and cholangiocarcinoma. In: Okuda K, Nakayama F, Wong J, eds. Intrahepatic Calculi. New York: Liss, 1984, pp 91–113. 249. Chen MF, Jan YY, Wang CS, Jeng LB, Hwang TL, Chen SC. Intrahepatic stones associated with cholangiocarcinoma. Am J Gastroenterol 1989; 84:391–395. 250. Jan YY, Chen MF, Wang CS, Jeng LB, Hwang TL. Chen SC. Surgical treatment of hepatolithiasis: longterm results. Surgery 1996; 120:509–514. 251. Lee WJ, Chang KJ, Lee CS, Chen KM. Surgery in cholangitis: bacteriology and choice of antibiotic. Hepatogastroenterology 1992; 39:347–349. 252. Fan ST, Lai EC, Mok FP, Choi TK, Wong J. Acute cholangitis secondary to hepatolithiasis. Arch Surg 1991; 126:1027–1031.
Page 817
253. Wong WT, TeohChan CH, Huang CT, Cheng FC, Ong GB. The bacteriology of recurrent pyogenic cholangitis and associated diseases. J Hyg (Lond) 1981; 87:407–412. 254. Chan FL, Chan JK, Leong LL. Modern imaging in the evaluation of hepatolithiasis. Hepatogastroenterology 1997; 44:358–369. 255. Kubo S, Hamba H, Hirohashi K, Kinoshita H, Lee KC, Yamazaki O, Nishio H, Yamada R. Magnetic resonance cholangiography in hepatolithiasis. Am J Gastroenterol 1997; 92:629–632. 256. Soong TC, Lee RC, Cheng HC, Chiang JH, Tseng HS, Lin CW, Teng MM. Dynamic MR imaging of hepatolithiasis. Abdom Imaging 1998; 23:515–519. 257. Chen MF, Jan YY, Wang CS, Jeng LB, Hwang TL. Clinical experience in 20 hepatic resections for peripheral cholangiocarcinoma. Cancer 1989; 64:2226– 2232. 258. Jan YY, Yeh TS, Chen MF. Cholangiocarcinoma presenting as pyogenic liver abscess: is its outcome influenced by concomitant hepatolithiasis? Am J Gastroenterol 1998; 93: 253–255. 259. Lauffer JM, Baer HU, Schajor M, Halter F, Buchler MW. Choledocholithiasis at the hepatic confluence mimicking a hilar cholangiocarcinoma. Hepatogastroenterology 1998; 45:2339–2343. 260. Lam SK. A study of endoscopic sphincterotomy in recurrent pyogenic cholangitis. Br J Surg 1984; 71:262–266. 261. Tanaka M, Ikeda S, Ogawa Y, Yokohata K, Matsumoto S, Chijiiwa K. Divergent effects of endoscopic sphincterotomy on the longterm outcome of hepatolithiasis. Gastrointest Endosc 1996; 43:33–37. 262. Jan YY, Chen MF. Percutaneous transhepatic cholangioscopic lithotomy for heptolithiasis: longterm results. Gastrointest Endosc 1995; 42:1–5. 263. Yeh YH, Huang MH, Yang JC, Mo LR, Lin J, Yueh SK. Percutaneous transhepatic cholangioscopy and lithotripsy in the treatment of intrahepatic stones: a study with 5year followup. Gastrointest Endosc 1995; 42:13–18. 264. Hwang MH, Tsai CC, Mo LR, Yang CT, Yeh YH, Yau MP, Yueh SK. Percutaneous choledochoscopic biliary tract stone removal: experience in 645 consecutive patients. Eur J Radiol 1993; 17:184–190. 265. Chen MF, Jan YY. Percutaneous transhepatic cholangioscopic lithotripsy. Br J Surg 1990; 77:530–532. 266. Lo CM, Fan ST. Percutaneous transhepatic choledochoscopic electrohydraulic lithotripsy for common bile duct stones: experience in four highrisk patients. Am J Gastroenterol 1991; 86:840–842. 267. Yang US, Kim GH, Ok CM, Lee SJ, Huh J, Song CS, Song GA, Cho M. Percutaneous transhepatic electrohydraulic lithotripsy for stones in biliary tracts. Korean J Gastroenterol 1998; 31:92–99. 268. Choi BI, Han JK, Park YH, Yoon YB, Han MC, Kim CW. Retained intrahepatic stones: treatment with piezoelectric lithotripsy combined with stone extraction. Radiology 1991; 178:105–108. 269. Binmoeller KF, Bruckner M, Thonke F, Soehendra N. Treatment of difficult bile duct stones using mechanical, electrohydraulic and extracorporeal shock wave lithotripsy. Endoscopy 1993; 25:201–206. 270. Wilkinson ML. Does laser lithotripsy hit the target? Gut 1998; 43:740–741. 271. Ros E, Navarro S, Bru C, Gilabert R, Bianchi L, Bruguera M. Ursodeoxycholic acid treatment of primary hepatolithiasis in Caroli's syndrome. Lancet 1993; 342:404–406. 272. Hochberger J, Bayer J, May A, Muhldorfer S, Maiss J, Hahn EG, Ell C. Laser lithotripsy of difficult bile duct stones: results in 60 patients using a rhodamine 6G dye laser with optical stone tissue detection system. Gut 1998; 43:823–829. 273. Brambs HJ, Duda SH, Rieber A, Scheurlen M, Claussen CD. Treatment of bile duct stones: value of laser lithotripsy delivered via percutaneous endoscopy. Eur Radiol 1996; 6:734–740.
Page 818
274. Ell C, Hochberger J, May A, Fleig WE, Bauer R, Mendez L, Hahn EG. Laser lithotripsy of difficult bile duct stones by means of a rhodamine6G laser and an integrated automatic stonetissue detection system. Gastrointest Endosc 1993; 39:755–762. 275. Neuhaus H, Hoffmann W, Zillinger C, Classen M. Laser lithotripsy of difficult bile duct stones under direct visual control. Gut 1993; 34:415–421. 276. Jakobs R, Maier M, Kohler B, Riemann JF. Peroral laser lithotripsy of difficult intrahepatic and extrahepatic bile duct stones: laser effectiveness using an automatic stonetissue discrimination system. Am J Gastroenterol 1996; 91:468–473. 277. Jeng KS, Yang FS, Ohta I, Chiang HJ. Dilatation of intrahepatic biliary strictures in patients with hepatolithiasis. World J Surg 1990; 14:587–592; discussion 592–593. 278. Cheng YF, Chen TY, Ko SF, Huang CC, Huang TL, Weng HH, Lee TY, SheenChen SM. Treatment of postoperative residual hepatolithiasis after progressive stenting of associated bile duct strictures through the Ttube tract. Cardiovasc Intervent Radiol 1995; 18:77–81. 279. SheenChen SM, Cheng YF, Chen FC, Chou FF, Lee TY. Ductal dilatation and stenting for residual hepatolithiasis: a promising treatment strategy. Gut 1998; 42:708–710. 280. Jeng KS, Yang FS, Chiang HJ, Ohta I. Bile duct stents in the management of hepatolithiasis with longsegment intrahepatic biliary strictures. Br J Surg 1992; 79:663–666. 281. Yoon HK, Sung KB, Song HY, Kang SG, Kim MH, Lee SG, Lee SK, Auh YH. Benign biliary strictures associated with recurrent pyogenic cholangitis: treatment with expandable metallic stents. AJR 1997; 169:1523–1527. 282. Jeng KS, Ohta I, Yang FS, Liu TP, Shih SC, Chang WS, Wan HY, Huang SH. Coexisting sharp ductal angulation with intrahepatic biliary strictures in right hepatolithiasis. Arch Surg 1994; 129:1097–1102. 283. Chijiiwa K, Yamashita H, Yoshida J, Kuroki S, Tanaka M. Current management and longterm prognosis of hepatolithiasis. Arch Surg 1995; 130:194–197. 284. Fan ST, Lai ECS, Wong J. Hepatic resection for hepatolithiasis. Arch Surg 1993; 128: 1070–1074. 285. Chijiiwa K, Kameoka N, Komura M, Yamasaki T, Noshiro H, Nakano K. Hepatic resection for hepatolithiasis and longterm results. J Am Coll Surg 1995; 180:43–48. 286. Jeng KS, Ohta I, Yang FS. Reappraisal of the systematic management of complicated hepatolithiasis with bilateral intrahepatic biliary strictures. Arch Surg 1996; 131:141–147. 287. Chen MF, Jan YY, Wang CS, Hwang TL, Jeng LB, Chen SC, Chao TC. Role of hepatic resection in surgery for bilateral intrahepatic stones. Br J Surg 1997; 84:1229–1232. 288. Saing H, Chan KL, Mya GH, Cheng W, Fan ST, Chan FL. Cutaneous stoma in the roux limb of hepaticojejunostomy (hepaticocutaneous jejunostomy): useful access for intrahepatic stone extraction. J Pediatr Surg 1996; 31:247–250. 289. Stain SC, Incarbone R, Guthrie CR, Ralls PW, RiveraLara S, Parekh D, Yellin AE. Surgical treatment of recurrent pyogenic cholangitis. Arch Surg 1995; 130:527–532. 290. Gott PE, Tieva MH, Barcia PJ, Laberge JM. Biliary access procedure in the management of oriental cholangiohepatitis. Am Surg 1996; 62:930–934. 291. Fan ST, Mok F, Zheng SS, Lai EC, Lo CM, Wong J. Appraisal of hepaticocutaneous jejunostomy in the management of hepatolithiasis. Am J Surg 1993; 165:332–335. 292. Beckingham IJ, Krige EJ, Beningfield SJ, Bornman PC, Terblanche J. Subparietal hepaticojejunal access loop for the longterm management of intrahepatic stones. Br J Surg 1998; 85:1360–1363. 293. Beningfield S, Beckingham I, Terblanche J. Subparietal hepaticojejunal access loop for intrahepatic stones. Hepatology 1998; 28(4, pt 2):243A. 294. Chu KM, Lai EC, AlHadeedi S, Arcilla CEJ, Lo CM, Liu CL, Fan ST, Wong J. Intrahepatic cholangiocarcinoma. World J Surg 1997; 21:301–305. 295. Su CH, Shyr YM, Lui WY, P'Eng FK. Hepatolithiasis associated with cholangiocarcinoma. Br J Surg 1997; 84:969–973.
Page 819
296. Chu KM, Lo CM, Liu CL, Fan ST. Malignancy associated with hepatolithiasis. Hepatogastroenterology 1997; 44:352–357. 297. Yeh TS, Chen TC, Hsieh LL, Jan YY, Jeng LB, Hwang TL, Chen MF. Hepatocellular carcinoma complicated with coexisting hepatolithiasis: pitfalls in diagnosis and management. Dig Dis Sci 1998; 43:2483–2488. 298. Khuroo MS, Zargar SA, Mahajan R. Hepatobiliary and pancreatic ascariasis in India. Lancet 1990; 335:1503–1506. 299. Sandouk F, Haffar S, Zada MM, Graham DY, Anand BS. Pancreaticbiliary ascariasis: experience of 300 cases. Am J Gastroenterol 1997; 92:2264–2267. 300. Khuroo MS, Mahajan R, Zargar SA. Javid G, Sapru S. Prevalence of biliary tract disease in India: a sonographic study in adult population in Kashmir. Gut 1989; 30:201–205. 301. Goldman IS, Brandborg LL. Parasitic diseases of the liver. In: Zakim D, Boyer TD, eds. Hepatology: A Textbook of Liver Disease. Philadelphia: Saunders, 1996, pp 1206–1231. 302. Khuroo MS, Zargar SA, Mahajan R, Bhat RL, Javid G. Sonographic appearances in biliary ascariasis. Gastroenterology 1987; 93:267–272. 303. Schulman A. Ultrasound appearances of intra and extrahepatic biliary ascariasis. Abdom Imaging 1998; 23:60–66. 304. Martin RJ. Electrophysiological effects of piperazine and diethylcarbamazine on Ascaris suum somatic muscle. Br J Pharmacol 1982; 77:255–265. 305. Khuroo MS, Zargar SA, Yattoo GN, Javid G, Dar MY, Boda MI, Khan BA. Worm extraction and biliary drainage in hepatobiliary and pancreatic ascariasis. Gastrointest Endosc 1993; 39:680–685. 306. Kamath PS, Joseph DC, Chandran R, Rao SR, Prakash ML, D'Cruz AJ. Biliary ascariasis: ultrasonography, endoscopic retrograde cholangiopancreatography, and biliary drainage. Gastroenterology 1986; 91:730–732. 307. van den Bogaerde JB, Jordaan M. Intraductal administration of albendazole for biliary ascariasis. Am J Gastroenterol 1997; 92:1531–1533. 308. Schwartz DA. Cholangiocarcinoma associated with liver fluke infection: a preventable source of morbidity in Asian immigrants. Am J Gastroenterol 1986; 81:76–79. 309. Sun T. Pathology and immunology of Clonorchis sinensis infection of the liver. Ann Clin Lab Sci 1984; 14:208–215. 310. Chou ST, Chan CW. Mucinproducing cholangiocarcinoma: an autopsy study in Hong Kong. Pathology 1976; 8:321–328. 311. Leung JW, Sung JY, Chung SC, Metreweli C. Hepatic clonorchiasis—a study by endoscopic retrograde cholangiopancreatography. Gastrointest Endosc 1989; 35:226–231. 312. Anonymous. Drugs for parasitic infections. Med Lett 1993; 35:111–122. 313. Leung JWC, Sung JY, Banez VP, Agas FV, Ling TKW, Chung SCS. Endoscopic cholangiopancreatography in hepatic clonorchiasis—a followup study. Gastrointest Endosc 1990; 36:360–363. 314. Roslyn JJ. Biliary infections. Curr Opin Gastroenterol 1993; 8:756–760. 315. HaswellElkins MR, Sithithaworn P, Mairiang E, Elkins DB, Wongratanacheewin S, Kaewkes S, Mairiang P. Immune responsiveness and parasitespecific antibody levels in human hepatobiliary disease associated with Opisthorchis viverrini infection. Clin Exp Immunol 1991; 84:213–218. 316. Pungpak S, Viravan C, Radomyos B, Chalermrut K, Yemput C, Plooksawasdi W, Ho M, Harinasuta T, Bunnag D. Opisthorchis viverrini infection in Thailand: studies on the morbidity of the infection and resolution following praziquantel treatment. Am J Trop Med Hyg 1997; 56:311–314. 317. elNewihi HM, Waked IA, Mihas AA. Biliary complications of Fasciola hepatica: the role of endoscopic retrograde cholangiography in management. J Clin Gastroenterol 1995; 21:309–311. 318. Han JK, Choi BI, Cho JM, Chung KB, Han MC, Kim CW. Radiological findings of human fascioliasis. Abdom Imaging 1993; 18:261–264.
Page 820
319. Ovnat A, Peiser J, Avinoah E, B arki Y, Charuzi I. Acute cholangitis caused by ruptured hydatid cyst. Surgery 1984; 95:497–500. 320. Khuroo MS, Wani NA, Javid G, Khan BA, Yatoo GN, Shah AH, Jeelani SG. Percutaneous drainage compared with surgery for hepatic hydatid cysts. N Engl J Med 1997; 337:881–887. 321. Ponchon T, Bory R, Chavaillon A. Endoscopic retrograde cholangiography and sphincterotomy for complicated hepatic hydatid cyst. Endoscopy 1987; 19:174–177. 322. Iscan M, Duren M. Endoscopic sphincterotomy in the management of postoperative complications of hepatic hydatid disease. Endoscopy 1991; 23:282–283. 323. Miguet JP, BressonHadni S. Alveolar echinococcus of the liver. J Hepatol 1989; 8:373–379. 324. Kern P, Frosch P, Helbig M, Wechsler JG, Usadel S, Beckh K, Kunz R, Lucius R, Frosch M. Diagnosis of Echinococcus multilocularis infection by reverse transcription polymerase chain reaction. Gastroenterology 1995; 109:596–600. 325. Margulis SJ, Honig CL, Soave R, Govoni AF, Mouradian JA, Jacobson IM. Biliary tract obstruction in the acquired immunodeficiency syndrome. [Published erratum appears in Ann Intern Med 1986 Oct;105:634.] Ann Intern Med 1986; 105:207–210. 326. Schneiderman DJ, Cello JP, Laing FC. Papillary stenosis and sclerosing cholangitis in the acquired immunodeficiency syndrome. Ann Intern Med 1987; 106:546–549. 327. Benhamou Y, Caumes E, Gerosa Y, Cadranel JF, Dohin E, Katlama C, Amouyal P, Canard JM, Azar N, Hoang C, et al. AIDSrelated cholangiopathy: critical analysis of a prospective series of 26 patients. Dig Dis Sci 1993; 38:1113–1118. 328. Vakil NB, Schwartz SM, Buggy BP, Brummitt CF, Kherellah M, Letzer DM, Gilson IH, Jones PG. Biliary cryptosporidiosis in HIVinfected people after the waterborne outbreak of cryptosporidiosis in Milwaukee. N Engl J Med 1996; 334:19–23. 329. Cello JP. Human immunodeficiency virusassociated biliary tract disease. Semin Liver Dis 1992; 12:213–218. 330. Bouche H, Housset C, Dumont JL, Carnot F, Menu Y, Aveline B, Belghiti J, Boboc B, Erlinger S, Berthelot P, et al. AIDSrelated cholangitis: diagnostic features and course in 15 patients. J Hepatol 1993; 17:34–39. 331. Forbes A, Blanshard C, Gazzard B. Natural history of AIDS related sclerosing cholangitis: a study of 20 cases. Gut 1993; 34:116–121. 332. Sepkowitz KA. Effect of HAART on natural history of AIDSrelated opportunistic disorders. Lancet 1998; 351:228–230. 333. Jacobson MA, French M. Altered natural history of AIDSrelated opportunistic infections in the era of potent combination antiretroviral therapy. AIDS 1998; 12(suppl A): S157–163. 334. Palella FJ, Jr., Delaney KM, Moorman AC, Loveless MO, Fuhrer J, Satten GA, Aschman DJ, Holmberg SD. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection: HIV Outpatient Study Investigators. N Engl J Med 1998; 338:853–860. 335. Pitlik SD, Fainstein V, Garza D, Guarda L, Bolivar R, Rios A, Hopfer RL, Mansell PA. Human cryptosporidiosis: spectrum of disease: report of six cases and review of the literature. Arch Intern Med 1983; 143:2269–2275. 336. Pol S, Romana C, Richard S, Carnot F, Dumont JL, Bouche H, Pialoux G, Stern M, Pays JF, Berthelot P. Enterocytozoon bieneusi infection in acquired immunodeficiency syndromerelated sclerosing cholangitis. Gastroenterology 1992; 102:1778–1781. 337. SifuentesOsornio J, PorrasCortes G, Bendall RP, MoralesVillarreal F, ReyesTeran G, RuizPalacios GM. Cyclospora cayetanensis infection in patients with and without AIDS: biliary disease as another clinical manifestation. Clin Infect Dis 1995; 21:1092–1097. 338. Gunnarsson G, Hurlbut D, DeGirolami PC, Federman M, Wanke C. Multiorgan microsporidiosis: report of five cases and review. Clin Infect Dis 1995; 21:37– 44. 339. Pol S, Romana CA, Richard S, Amouyal P, DesportesLivage I, Carnot F, Pays JF,
Page 821
Berthelot P. Microsporidia infection in patients with the human immunodeficiency virus and unexplained cholangitis. N Engl J Med 1993; 328:95–99. 340. Willson R, Harrington R, Stewart B, Fritsche T. Human immunodeficiency virus 1—associated necrotizing cholangitis caused by infection with Septata intestinalis. Gastroenterology 1995; 108:247–251. 341. Bryan CS. AIDS: where do we stand? J S C Med Assoc 1995; 91:389–390. 342. von Wichmann MA, Castiella A, RodriguezArrondo F, Iribarren JA, Arrizabalaga J, Lopez P. Pseudomonas aeruginosa cholangitis in a HIV patient. Am J Gastroenterol 1998; 93:483–484. 343. Farman J, Brunetti J, Baer JW, Freiman H, Comer GM, Scholz FJ, Koehler RE, Laffey K, Green P, Clemett AR. AIDSrelated cholangiopancreatographic changes. Abdom Imaging 1994; 19:417–422. 344. Ducreux M, Buffet C, Lamy P, Beaugerie L, Fritsch J, Choury A, Liguory C, Longuet P, Gendre JP, Vachon F, et al. Diagnosis and prognosis of AIDSrelated cholangitis. AIDS 1995; 9:875–880. 345. Wind P, Chevallier JM, Jones D, Frileux P, Cugnenc PH. Cholecystectomy for cholecystitis in patients with acquired immune deficiency syndrome. Am J Surg 1994; 168: 244–246. 346. Schneiderman DJ. Hepatobiliary abnormalities of AIDS. Gastroenterol Clin North Am 1988; 17:615–630. 347. Cello JP. Acquired immunodeficiency syndrome cholangiopathy: spectrum of disease. Am J Med 1989; 86:539–546. 348. Cello JP, Chan MF. Longterm followup of endoscopic retrograde cholangiopancreatography sphincterotomy for patients with acquired immune deficiency syndrome papillary stenosis. Am J Med 1995; 99:600–603. 349. De Silva R, Boudghene F, Lecomte I, Delage Y, Grange JD, Bigot JM. Sonography in AIDSrelated cholangitis: prevalence and cause of an echogenic nodule in the distal end of the common bile duct. AJR 1990; 175:449–453. 350. Miller FH, Gore RM, Nemecek AA, Fitzgerald SW. Pancreatobiliary manifestations of AIDS. AJR 1996; 166:1269–1274. 351. Franzen C, Salzberger B, Fatkenheuer G, Ziegenhagen D, Cornely O, Diehl V, Schrappe M. Stimulated duodenal/bile juice aspiration for diagnosis of enteric pathogens in HIVinfected patients. J Clin Gastroenterol 1995; 21:33–37. 352. Urbain D, Jeanmart J, Lemone M, Kiromera A, Muls V, Arendt V, Dewit S. Cholestasis in patients with the acquired immune deficiency syndrome: comparison between ultrasonographic and cholangiographic findings. Am J Gastroenterol 1991; 86:574–576. 353. Wilcox CM, Rabeneck L, Friedman S. AGA technical review: malnutrition and cachexia, chronic diarrhea, and hepatobiliary disease in patients with human immunodeficiency virus infection. Gastroenterology 1996; 111:1724–1752. 354. Collazos J, Mayo J, Martinez E, Callejo A, Blanco I. Celiac plexus block as treatment for refractory pain related to sclerosing cholangitis in AIDS patients. J Clin Gastroenterol 1996; 23:47–49. 355. Hamour AA, Bonnington A, Hawthorne B, Wilkins EG. Successful treatment of AIDSrelated cryptosporidial sclerosing cholangitis. AIDS 1993; 7:1449–1451. 356. Castiella A, Iribarren JA, Lopez P, Arrizabalaga J, Rodriguez F, von Wichmann MA, Arenas JI. Ursodeoxycholic acid in the treatment of AIDSassociated cholangiopathy. Am J Med 1997; 103:170–171. 357. Adolph MD, Bass SN, Lee SK, Blum JM, Schreiber H. Cytomegaloviral acalculous cholecystitis in acquired immunodeficiency syndrome patients. Am Surg 1993; 59:679–684. 358. Hogg RS, Heath KV, Yip B, Craib KJ, O'Shaughnessy MV, Schechter MT, Montaner JS. Improved survival among HIVinfected individuals following initiation of antiretroviral therapy. JAMA 1998; 279:450–454. 359. Keaveny AP, Karasik MS. Hepatobiliary and pancreatic infections in AIDS: Part II. AIDS Patient Care STDs 1998; 12:451–456.
Page 823
36— Bile Duct Injuries Noel N. Williams and Daniel Kreisel University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania I— Introduction The possibility of injury to bile ducts has existed for over a century, since the introduction of surgical removal of the gallbladder. While injuries to the extrahepatic biliary system have been a wellrecognized complication of traditional or open cholecystectomies, it was the introduction of minimally invasive techniques in the late 1980s that led to a resurgence of interest in this topic. The incidence of this complication has increased significantly with the widespread enthusiasm for laparoscopic removal of the gallbladder. Historically, bile duct injuries occur in approximately 0.1 to 0.2% of open procedures, whereas most studies report an incidence of approximately 0.5% during laparoscopic cholecystectomies. With over 500,000 laparoscopic cholecystectomies performed annually for calculous disease in the United States, over 2000 bile duct injuries are thought to occur every year (1). The associated potential for morbidity has enormous medical, legal, and economic consequences. In recent years there has been a surge in the literature on multidisciplinary approaches to management of these injuries. This chapter addresses the historical aspects of this problem and describes the different approaches utilized in correcting the injury. II— Historical Aspects Carl Langenbuch performed the first open cholecystectomy on July 15, 1882, in Berlin, Germany (2). Wilhelm Daniels, his 43yearold patient, had suffered from biliary colic for over 20 years. Daniels was admitted to the hospital 5 days prior to the operation, which was performed through a Tshaped incision in the right upper quadrant. The procedure was uncomplicated except for some venous bleeding from the liver bed, and the gallbladder was found to contain two cholesterol stones. The patient recovered uneventfully and was discharged from hospital approximately 6 weeks later. Some 20 years later, in 1905, William Mayo described different methods for the repair of bile duct injuries (3). He reported primary endtoend anastomosis of an inadvertently divided common bile duct utilizing catgut sutures as well as hepaticoduodonostomy for a stricture of the common bile duct that followed a cholecystectomy and choledochotomy. During the same year, Jenckel repaired a common bile duct stricture by suturing one end of a rubber tube to the duct and inserting the other end into the duodenum, following the Witzel method. This repair was complicated by the development of a duodenal fistula. However, through epithelialization, a nexus formed between the stump of the distal common bile duct and the duodenum
Page 824
and the patient was discharged home over 1 year after the repair. During the same time period, Hertzler utilized a portion of the gallbladder to repair a lacerated bile duct (3). The development of the T tube for bile duct drainage resulted from animal experiments by Sullivan in 1912. He demonstrated that insertion of rubber tubes into resected bile ducts facilitated successful reconstruction through epithelialization. In 1923, McArthur recognized that bile duct injuries were most commonly associated with ''former surgical invasion of the bile tracts for disease and resultant stones." He reported on the successful use of rubber tubes as transanastomotic stents, with the distal end of the tube being carried into the duodenum (4). In 1948, Cole and coworkers published a series of 49 patients who had suffered trauma to the extrahepatic biliary system (5). Of these injuries, 65% were sequelae of operative trauma. They reported favorable results with an anastomosis between hepatic ducts and a RouxenY arm of the jejunum, finding this method to be superior to an anastomosis between hepatic ducts and duodenum. Contrary to the recommendations of Cole et al., Lahey and Pyrtek believed that preservation of the sphincter of Oddi was important in order to avoid cholangitis (6). Therefore, they advocated an endtoend anastomosis between the hepatic duct and the distal common bile duct over a T tube in all cases where sufficient hepatic duct was preserved. Saypol and Kurian addressed the issue of recurrences of strictures after repair of bile duct injuries (7). They recognized that mucosatomucosa approximation with fine sutures was essential for favorable results. In order to reduce the incidence of postoperative strictures, they recommend leaving transanastomotic stents in place for up to 2 years. In 1975, Wexler and Smith reviewed their experience with jejunal mucosal grafts for repair of high bile duct strictures (8). The cornerstone of their technique is adequate mucosatomucosa apposition to decrease the incidence of restenosis. The biliary tree is identified by transecting scar tissue in the liver hilum. The duct opening is split and a curved forceps introduced into the biliary tree and passed through the anterior liver surface. A rubber tube is drawn through the liver out the hepatic duct opening. Subsequently, a RouxenY jejunostomy is constructed; a small seromuscular disk is then removed near the end of the loop and a protruding mucosal diverticulum is created. The transhepatic tube is passed into the mucosal diverticulum, and the jejunal mucosa is approximated to the bile duct by withdrawing the transhepatic tube. Finally, the jejunal wall is anchored to the liver capsule with a few interrupted sutures. The advantages of this technique were the relative ease of the procedure and the avoidance of placing sutures through bile duct mucosa, which could potentially compromise the blood supply. In the 1980s, endoscopic techniques that had initially been utilized for gynecological operations were adopted for general surgical procedures. In 1983, Semm reported his experience with endoscopic appendectomies (9). Laparoscopic cholecystectomy was first popularized by Dubois and colleagues in France; in 1980, they published their experience with 36 patients operated on for nonacute cholecystitis (10). Their results were favorable, with complications in only two patients. They recognized several advantages of this new technique, including shorter hospital stay, earlier return to work, better cosmetic results, and decreased incidence of wound dehiscence. The indications for laparoscopic cholecystectomies were rapidly expanded, and significant increases in the total number of gallbladder procedures were reported over the ensuing years (11). III— Classification of Injuries Iatrogenic injury to the biliary tree has been of interest to surgeons and nonsurgeons for many decades. Several studies reported a bile duct injury rate of approximately 0.2% with open cholecystectomies. The introduction of laparoscopic methods in the late 1980s and the attendant learning curve, renewed interest in this topic. The injury rate in laparoscopic cholecystectomies is in a range between 0.4 and 0.6%. The spectrum of injuries is wideranging from minor
Page 825
leakage of bile to transections and excisions of ductal structures, with subsequent development of debilitating strictures. The incidence of postoperative bile leaks has increased with laparoscopic cholecystectomy. Minor bile leaks are usually clinically insignificant. Using a radionuclide study, Gilsdorf demonstrated bile leakage in 44% of patients following open cholecystectomies (12). This study did not demonstrate increased morbidity from minor postoperative bile leaks. Van der Linden et al. found a correlation between postoperative bile leakage and the degree of denuation of the liver surface at the time of operation (13). On the other hand, major leaks can lead to biliary fistulas, bilomas, ascites, or peritonitis. Biliary drainage of more than 100 mL per day usually indicates injury of a major duct, such as the hepatic, common, or cystic ducts. Biliary drainage in excess of 500 mL per day suggests an obstruction of the biliary system. The term biloma was first used by Gould and Patel to describe a sonographically anechoic fluid collection in the subhepatic space following hepatic trauma (14). Bilomas are frequently loculated bile collections that are usually secondary to leakage from a major duct; as the leakage commonly persists, bilomas frequently expand in size. Rightupperquadrant discomfort associated with anorexia and possibly nausea and vomiting are the early signs indicating a significant leakage of bile. The diagnosis may be missed because of early hospital discharge on the day of operation or on the first postoperative day. Fever and chills as well as abdominal pain in the postoperative period indicate infection of a biloma or bile peritonitis. Free accumulation of bile into the peritoneal cavity can lead to either bile ascites or bile peritonitis. Bile peritonitis is due to either leakage of infected bile or to sudden release of large quantities of concentrated bile, which can result in chemical irritation. Bile ascites is secondary to slow leakage of noncontaminated bile into the peritoneal cavity, which occurs in the absence of obstructions of the biliary system. Progressive abdominal distention without fever is the hallmark of this condition. Lacerations of bile ducts seem to be less frequent than transections or excisions. This may be partially due to the fact that more complicated bile duct injuries are being transferred to tertiary care centers, while simple lacerations may be repaired at community hospitals and subsequently do not get reported in the literature. This would lead to an underestimation of the incidence of simple lacerations. Transections and excisions of ductal structures are more likely to lead to the development of biliary strictures after repair; this may be due to damage to the microcirculation of the ducts. Benign bile duct strictures can be divided into congenital, posttraumatic, postradiotherapy, postoperative, and postinflammatory types. Conditions predisposing to bile duct strictures include chronic pancreatitis, cholangitis, duodenal penetrating ulcers, and sclerosing cholangitis. However, the vast majority of benign biliary strictures occur following operations on the gallbladder and biliary tree. The location and the extent of benign bile duct strictures are critical factors in predicting prognosis and determining treatment. Bismuth classified strictures according to their location (15) (Fig. 1). This system relates the site of the stricture to its distance from the confluence of the right and left hepatic ducts. Grade I strictures are located in the low common hepatic duct or common bile duct and are more than 2 cm from the confluence; grade II strictures are less than 2 cm from the confluence while still leaving remnant hepatic duct; grade III strictures extend to the confluence and leave the distal right and left hepatic ducts intact; grade IV strictures are high strictures involving the right and left hepatic ducts; and grade V strictures involve an accessory right hepatic duct with or without involvement of the common bile duct. Frattaroli modified the Bismuth classification and described eight types of bile duct strictures to include those involving the papilla and intrahepatic segmental ducts as well as diffuse strictures (16). IV— Mechanisms of Injury and Strategies of Prevention Johnston recognized that three factors contributed to iatrogenic bile duct injuries during cholecystectomies: dangerous disease, dangerous anatomy, and dangerous surgery (17).
Page 826
Figure 1 Bismuth classification of bile duct strictures relating the site of the stricture to its distance from the confluence of the right and left hepatic ducts.
Moossa reviewed 81 patients who were referred to the University of California at San Diego between 1970 and 1988 for treatment of bile duct strictures that occurred following open cholecystectomy or common bile duct explorations (1). This study showed that low strictures were usually due to trauma to the lumen of the duct, as most of these injuries occurred following exploration of the common bile duct. Forceful manipulation can lead to damage to the mucosa, with creation of false passages. Compromising the blood supply to the duct can also predispose to the development of strictures. High strictures were the result of inadvertent transection or excision of ductal structures during dissections with inadequate exposure. An important observation was that more than 80% of all injuries occurred in obese patients, where small incisions led to inadequate exposure. The authors suggest that starting the dissection at the fundus rather than in the triangle of Calot lowers the incidence of injuries. This recommendation appears to be especially important in the setting of inflammatory changes. A remarkable finding of this series, which summarizes bile duct injuries in the prelaparoscopic era, is that most operations were performed by experienced surgeons. Asbun reviewed 21 patients who were referred to the Lahey Clinic Medical Center for the management of bile duct injuries following laparoscopic cholecystectomies (18). Fibrosis at the triangle of Calot, acute cholecystitis, obesity, local hemorrhage, and aberrant anatomy were identified as local risk factors. The most common problem is inadequate identification of the anatomy in Calot's triangle. This is reflected in failure to recognize the injury in the vast majority of patients at the time of the initial operation. The authors recommend obtaining lateral and inferior retraction of Hartmann's pouch in order to avoid alignment of the cystic duct and common bile duct, with the dissection beginning high on the neck of the gallbladder. Hunter's recommendations to minimize bile duct injuries during laparoscopic cholecystectomies were to use a 30degree telescope, exert firm cephalic traction on the fundus and lateral traction on the infundibulum for better exposure of the cystic duct, dissect the cystic duct at the infundibulum, and use routine fluoroscopic cholangiography (19). Figure 2 illustrates an example of misidentification of the cystic duct with clip placement on the common bile duct and subsequent transection.
Page 827
Figure 2 Laparoscopic bile duct injury with inadverent division of the common bile duct. The risk of this injury can be minimized by lateral traction of Hartmann's pouch and opening of Calot's triangle.
Thermal injury due to injudicious use of electrocautery for hemostasis or dissection can have devastating consequences by predisposing to severe scarring and stricture formation. Davidoff and colleagues reviewed videotapes of 12 patients with bile duct injuries (20). In three cases, overzealous use of electrocautery in the hilar area was thought to be the causative factor for stricture formation. Contrary to Moossa's observation in open cholecystectomies, a study by The Southern Surgeons Club in 1995 showed that the incidence of bile duct injuries in laparoscopic cholecystectomies correlated with the inexperience of the surgeon (1,21). They reported that 90% of injuries in a series of 8839 laparoscopic cholecystectomies occurred within the first 30 cases performed by an individual surgeon. A commonly cited prospective study of 1518 laparoscopic cholecystectomies by the same group in 1991 showed that the incidence of bile duct injuries in the first 13 patients operated on by each individual surgeon was 2.2%, as compared with 0.1% for subsequent patients (22). The subject of routine versus selective use of intraoperative cholangiography has been discussed controversially in the surgical literature. A report by Gregg discourages the use of intraoperative cholangiography in patients with no evidence of common bile duct obstruction (23). He believes that only patients with jaundice, moderately dilated common bile ducts, or evidence of pancreatitis should be subjected to cholangiography. On the other hand, Flowers and coworkers believe that routine intraoperative cholangiography during laparoscopic chole
Page 828
cystectomies helps delineate the exact anatomy and thus reduces the incidence of iatrogenic trauma to the bile ducts (24). They argue that routine use of intraoperative cholangiography helps to recognize injuries immediately and thus minimize the higher morbidity associated with delayed detection. In addition, its routine use helps surgeons master the technique. Hunter reported that routine cholangiograms altered the operative behavior in 9% of his first 100 laparoscopic cholecystectomies and helped to prevent injury to the common bile duct (19). Opponents of routine cholangiography claim that it may carry an increased risk of biliary tract injury and unnecessarily adds to operative time and cost. In 1993, Brodish and Fink conducted a survey of the Society of American Gastrointestinal Endoscopic Surgeons regarding the use of laparoscopic cholecystectomies, endoscopic retrograde cholangiopancreatography (ERCP), and intraoperative cholangiography (25). In all, 42% of surgeons who performed laparoscopic cholecystectomies recommended routine intraoperative cholangiography in patients with normal liver function tests. A multi institutional study conducted by Woods and colleagues at three tertiary hepatobiliary centers analyzed the role of intraoperative cholangiography in the recognition of a bile duct injury (26). Intraoperative cholangiography was completed in 40% of their patients who had sustained bile duct injuries. A total of 52% of patients with injuries who had intraoperative cholangiography were converted to an open procedure, while 37% of patients who did not have intraoperative cholangiography were converted. An important finding was that the bile duct injuries were missed on 16% of all cholangiograms. Possible explanations for "normal" cholangiograms were that the injury occurred after completion of the cholangiogram, misinterpretation by the surgeon, low sensitivity, or delayed stricture formation secondary to factors such as thermal injury or ischemia secondary to disruption of the ductal microcirculation. Based on their results, the authors suggested that the benefits of intraoperative cholangiography outweigh the risks. V— Surgical Management It is well recognized that injury to the biliary tract is a serious complication that is frequently associated with high morbidity or even mortality. Moossa et al. reported in his review of bile duct injuries following open cholecystectomies that 18.5% of patients eventually died and 13.5% suffered longterm morbidity as a consequence of their injury (1). Bile duct injuries frequently require multiple interventions in their management and are a source of frustration for patients as well as physicians. It appears obvious that surgeons performing the primary biliary operations should be very meticulous and careful to avoid these complications. Conversion to an open procedure in case of uncertainty is one of the key elements during laparascopic cholecystectomies. However, once an injury occurs, it is imperative to recognize it promptly. With the surge in laparoscopic gallbladder removal came multidisciplinary approaches to the management of early and delayed presentations of bile duct injuries. The local expertise of individual departments usually determines the therapeutic algorithm. While some injuries are recognized intraoperatively, the majority are not detected until the early postoperative period. It can even take months or years before the patient develops jaundice or cholangitis. In the series of bile duct strictures that developed after open cholecystectomies reported by Chapman et al., only 18% of all injuries were recognized intraoperatively (27). The goal of surgical intervention is to reestablish uninterrupted bile flow from the liver to the gastrointestinal tract. This can only be optimally achieved if a tensionfree anastomosis between scarfree tissues is created while maintaining maximum ductal length. There is general agreement that if an injury is recognized at the time of operation, the repair should be performed immediately. If the operating surgeon is uncomfortable with biliary reconstruction, transfer to a hepatobiliary center should be initiated expeditiously. Stewart and Way compared results of immediate bile duct repairs by the primary surgeons with those that were referred to tertiary care centers (28). While 94% of repairs performed by hepatobiliary surgeons were successful without the need for further intervention, the success rate was only
Page 829
11% in the hands of the primary surgeons. An important observation was that primary surgeons continued to treat patients after failed initial repair for an average of 236 days prior to transfer to a tertiary center, which contributed to significant morbidity. Complications of initial unsuccessful repair included leaks, stricture recurrences, cholangitis, and segmental hepatic necrosis. Mirza and colleagues also observed a significant period of delay of approximately 3 weeks—between observation of the first symptoms suggesting a biliary complication following laparoscopic cholecystectomy—before eventual referral to a hepatobilary center (29). In their series, 56% of the patients had undergone at least one attempt at surgical repair prior to referral. Strasberg and coworkers suggest that simple drainage of the right upper quadrant and referral to a hepatobiliary referral center is a safe strategy for injuries that are recognized intraoperatively at a primary center (30). A short delay in repair is usually not detrimental to the patient. Carroll's group reported that primary surgeons were successful with their surgical reconstructions in 27% of their patients while hepatobiliary surgeons had a success rate of 79% (31). Similarly, Blumgart and associates believe that the best results are obtained by initial adequate repair (32). Repeated attempts at anastomosis is associated with the development of complex strictures and ultimately cholestatic liver disease. The classic bile duct injury in the laparoscopic era involves inadverent transection and segmental excision of the common bile duct or the common hepatic duct. Segmental bile ducts are less commonly affected and received less attention in the literature. Anatomic variations of the biliary system predispose to damage to segmental or lobar ducts. These include low convergence of right and left hepatic ducts; insertion of accessory ducts into the common hepatic duct, cystic duct, or common bile duct; a cystic duct inserting into the right hepatic duct; as well as rare cholecystohepatic ducts. These subsegmental ducts are prone to injury because they traverse Calot's triangle. Swanson and Stewart demonstrated in animal experiments that ligation of segmental bile ducts ultimately leads to atrophy of hepatic tissue (33,34). Similarly, atrophy of hepatic tissue with compensatory hypertrophy of unaffected parenchyma has been documented in humans with segmental bile duct obstruction. Controversy exists regarding the management of segmental bile duct injuries. Attempts at surgical reconstruction of these small bile ducts is associated with a high complication rate including cholangitis. Hadjis and Blumgart believe that the decision regarding management of segmental bile duct injuries has to be made on an individual basis (35). It is often justified to ligate an inadvertently transected segmental duct rather than to undertake surgical reconstruction. Rutledge reported the use of a saphenous vein patch to repair a noncircumferential biliary defect during a left hepatectomy (36). He advocated stenting the patched area for approximately 2 to 4 months. Healing occurs by ingrowth of columnar biliary epithelium with the patch acting as a scaffold. Several methods of patching bile duct defects have been described, including duodenal or jejunal onlay patches, gallbladder patches, and patches using the remains of the cystic duct or prosthetic material. Browder's group reviewed their experience with surgical management of bile duct injuries that had occurred during open gallbladder procedures (37). The surgical approach was influenced by the type of injury as well as by the time of recognition. The authors observed that the only type of injury that was recognized intraoperatively was partial or complete transection of the bile duct. The most common location was the cystic duct—common duct junction. On the other hand, ligation of the common bile duct was usually recognized in the postoperative period. The majority of intraoperatively recognized injuries were repaired by primary reanastomosis; after an average followup period of 2 1/2 years, none of the patients had developed biliary strictures. These comparatively favorable results following primary reanastomosis could be due to the fact that the majority of these patients had partial transections where a portion of the duct had remained intact. Primary reanastomosis is usually not the surgical treatment of choice for injuries that are recognized in the postoperative period. Several reports from the 1980s documented a high incidence of stricture formation following such attempts at reconstruction. This is especially the case when a portion of the bile duct has been inadvertently excised, which would lead to significant tension at the anastomosis. In addition, there is more excessive scar formation with
Page 830
the passage of time. Lillemoe advocates an endtoend anastomosis with placement of a T tube through a separate choledochostomy either above or below the anastomosis only for lesions where the two ends of the bile duct can be approximated without tension (38). In these cases, the length of the excised bile duct segment usually does not exceed 1 cm. Important surgical considerations in creating a tensionfree anastomosis are mobilization of the duodenum and avoidance of skeletonization of the bile duct so as not to cause ischemic injuries by devascularization. Despite establishing a repair with "normal" anatomic status and drainage of bile through the sphincter of Oddi, primary reanastomosis with endtoend repair of the bile ducts has a high rate of stricture recurrence. Consequently many hepatobiliary centers do not use this method of reconstruction. Csendes and coworkers reported a 78% failure rate of endtoend repairs of complete and incomplete transections of common hepatic and common bile ducts at an average of 4 years' followup (39). Patients with incomplete transection had a worse prognosis than patients with complete transection of the common bile duct. The authors recommend limiting primary endtoend reconstructions to patients who meet the following criteria: (a) the injury is recognized immediately, (b) proximal and distal ducts are well debrided and irrigated, (c) the ductal diameter is greater than 4 mm, and (d) the distance between the proximal and distal duct is less than 2 cm (40). Ischemic damage to the duct during excessive dissection is thought to be the causative factor for the failure of primary endtoend reconstructions. It has been shown by Northover and Terblanche that the arterial blood supply to the extrahepatic biliary tree from the confluence of right and left hepatic ducts to the first portion of the duodenum is axial (41). The main arteries run adjacent to the bile duct at 3 and 9 o'clock. Some 60% of the blood supply comes from below, with the remainder arising from above. On the other hand, the bile ducts in the liver hilum have a richer arterial blood supply from adjacent arteries. The authors suggested that ischemic damage to the mucosa of the bile ducts leads to exposure of the ductal wall to bile, which causes an inflammatory reaction with subsequent fibrosis and stricturing. Similarly, choledochoduodenostomy, except for strictures of the retropancreatic portion, is not suitable for the repair of most biliary strictures because an adequate length of bile duct for creating a tensionfree anastomosis to the duodenum cannot be usually obtained. Further disadvantages of this procedure include possible reflux of ingested food particles into the biliary system and resultant cholangitis despite widely patent anastomoses. Cholangitis can occur secondary to stasis and bacterial overgrowth in the distal segment of the bile duct. In addition, an anastomotic leak with this reconstruction would lead to a duodenal fistula. Despite limiting this procedure to highly select populations including patients who had undergone Billroth II resections in the past, Frattaroli and coworkers obtained unsatisfactory results in 40% of their cases (16). The most commonly performed surgical procedure for bile duct injuries that are recognized in the postoperative period is a tensionfree biliaryenteric anastomosis. Based on the level of injury, restoration of biliaryenteric continuity can be achieved with a defunctionalized RouxenY jejunal loop by means of either hepaticojejunostomy, choledochojejunostomy, or intrahepatic cholangiojejunostomy. The main advantage of a Roux segment is reduction of biliary reflux of food particles and consequently a decreased incidence of cholangitis. Matthews and colleagues recognized anastomotic stenoses, intrahepatic strictures, intraluminal debris, improperly constructed enteric conduits, and miscellaneous conditions predisposing to abnormal intestinal flora and bacterial overgrowth in the biliary tree as risk factors for recurrent episodes of cholangitis after biliaryenteric bypasses (42). The proximal jejunum is divided, and the biliaryenteric anastomosis is performed between the bile duct and the distal. end of the divided jejunum. The anastomosis is usually fashioned by a single layer of interrupted absorbable sutures to obtain mucosatomucosa approximation. The length of the diverted jejunum. should be approximately 40 cm in length. Matthews and coworkers observed recurrent bouts of cholangitis in patients where the Roux segment was less than 25 cm in length. Recognizing the potential need for postoperative cannulation of the biliary system for dilatation of recurrent strictures, a modification of RouxenY biliaryenteric reconstructions was intro
Page 831
duced in the 1980s. Because a traditional RouxenY reconstruction makes the biliary system inaccessible to endoscopic methods, a method was developed where the afferent limb is placed subcutaneously and secured to the right anterior abdominal wall. The jejunal limb usually measures 60 to 70 cm for this modified reconstruction. A metal ring can be placed between the bowel and the abdominal wall to facilitate future fluoroscopic cannulation. Hutson et al. examined the efficacy of percutaneous dilatation of biliary strictures through the afferent limb of modified RouxenY choledochojejunostomies or hepaticojejunostomies (43). They found that subcutaneously placed limbs are more accessible than those that were placed subfascially. However, they recognized the risk of developing parajejunal hernias with subcutaneous limbs. Gastrointestinal bleeding and pain have also been reported as potential complications. The authors found that recurrent biliary strictures can be successfully managed by repeat percutaneous balloon dilatation through the afferent limb, thus obviating the need for repeated surgical procedures. In their series, several patients underwent revisions of prior traditional RouxenY biliary jejunal reconstructions with creation of subcutaneously placed afferent limbs. The majority of these patients did not undergo a revision of their biliaryenteric anastomosis. Reviewing an institutional experience with benign bile duct strictures between 1955 and 1990 Millis and coworkers observed a more liberal use of percutaneous balloon dilatation in the management of anastomotic strictures (44). They reported a success rate of 93% with fewer than three dilatations in their series with percutaneous transhepatic balloon dilatation. In patients who have intraabdominal abscesses or bile collections at the time of presentation, control of sepsis through drainage procedures with radiological guidance should be performed prior to definitive surgical reconstruction. As outlined by Blumgart, portal hypertension needs to be ruled out in the preoperative phase (32). These patients have an increased morbidity and mortality and should be considered for less invasive procedures, such as interventional radiological procedures. The value of transanastomotic stents is discussed controversially in the literature and practices vary widely among surgical teams. While some groups stent all reconstructions for prolonged periods of time, others use stents only for technically difficult anastomoses. Arguments in favor of stenting include decrease in fibrotic narrowing during the early phase of healing, ease of suture placement, and the ability to provide a conduit for bile flow. On the other hand, others believe that stents can irritate the ductal mucosa and thereby promote fibrosis at the anastomotic site—that they can facilitate the entry of bacteria into the biliary system and may require reexploration for clogging or dislodgment. Innes et al. reported their experience with biliaryenteric reconstructions without transanastomotic stents for iatrogenic bile duct injuries. The vast majority of their patients had failed previous attempts at surgical reconstruction. A total of 95% of their patients had excellent results after an average followup period of 3 years. Pellegrini's group believes that stents are not necessary when a wide anastomosis is performed and that their use should be limited to cases where mucosal apposition is questionable (45). Their practice is to remove the stents after a period of 6 to 8 weeks. Similarly, Genest et al. remove stents after approximately 6 weeks as they believe that prolonged stenting can lead to an inflammatory reaction (46). They reported a higher incidence of stricture recurrence with prolonged stent placement of greater than 3 months. The success rate of the reconstructive procedure is mainly defined by the rate of recurrence of strictures and by the incidence of cholangitis. Choledochojejunostomies have higher rates of complications when compared with hepaticojejunostomies (42,47,48). RouxenY hepaticojejunostomy is currently the most commonly performed surgical reconstructive procedure for benign biliary strictures. Terblanche and coworkers addressed the importance of the level of the biliaryenteric anastomosis and observed fewer recurrences of strictures when the anastomosis was performed at a higher level (47). It is their practice to perform high anastomoses even for Bismuth type I strictures; for low strictures, the bile duct is opened to the level of the hepatic confluence. The higher complication rate with low anastomoses was felt to be secondary to relative ischemia of the bile duct. As outlined above, the arterial blood supply to the bile ducts distal to the confluence of right and left hepatic ducts is tenuous. Accordingly,
Page 832
inadequate blood supply and stricture formation can be expected if the biliaryenteric anastomosis is being performed at the level of the damaged bile duct. The authors recommend the transection of the bile duct at a higher level through normal tissue and to check for adequate backflow of blood from the cut edges of the upper bile duct (47). Similarly, Saber and ElManialawi report a high incidence of restenosis in cases where the proximal, unhealthy scarred end of the injured bile duct was used for the anastomosis (48). Bismuth et al. reviewed their experience with RouxenY hepaticojejunostomies and choledochojejunostomies for benign, nonprogressive biliary lesions between 1962 and 1974 (49). Of their 123 patients, 23% underwent the procedure for biliary strictures that had occurred secondary to operative injury. Over the mean followup period of 5.5 years, 83% of their patients remained without symptoms related to biliary disease. The only anastomotic stenosis in this series occurred in a patient who had undergone the surgical reconstruction for operative transection of the bile duct in a purulent field on the 15th day following the initial injury. The bile duct tissue used for the anastomosis was of suboptimal quality. The authors observed that the use of fine monofilament suture material reduced the incidence of cholangitis. Roethlin and coworkers reported their experience with hepaticojejunostomies for biliary strictures that were due to iatrogenic injuries or chronic pancreatitis (50). Their followup period averaged 7.6 years. In their report, Blumgart and Kelley (51), recognize portal hypertension and cirrhosis as important factors leading to increased morbidity and mortality. Minimally invasive procedures should be given serious consideration in this patient population. A total of 33% of the patients treated by Blumgart and Kelley experienced complications in the early postoperative period, including wound infections, pancreatic fistulas, anastomotic leaks, or stenoses. The majority of these complications were observed in patients who had undergone prior operations in the hepatobiliary area. The longterm failure rate, defined by cholangitis and stricturing of the anastomoses, was 18%. Tocchi et al. published a series of 84 patients who underwent surgical reconstruction of benign bile duct strictures (52). Hepaticojejunostomies were performed for Bismuth III strictures, and Bismuth IV strictures were treated with intrahepatic cholangiojejunostomies. The average followup period was 9 years. Favorable outcome, which was defined by no biliary symptoms or only transitory complaints, was reported in 83% of these patients. This figure is comparable to the data from Roethlin's series (50). Restenoses were managed both through interventional radiological techniques as well as surgically. The authors favor hepaticojejunostomies over choledochojejunostomies, as they experienced a lower complication rate with the high biliary enteric anastomosis. Bauer and coworkers reviewed immediate and shortterm outcomes of surgical reconstructions for major bile duct injuries that were sustained during laparoscopic cholecystecomy (53). The majority of patients had sustained Bismuth class I or II injuries. RouxenY hepaticojejunostomies were performed in 94% of patients, with the remainder undergoing primary bile duct anastomosis over a stent for resection of a segment of the common bile duct. Contrary to Tocchi's practice, transanastomotic stents were utilized in 97% of the patients (52,53). At an average of 1year followup, 62% of the patients remained asymptomatic with normal liver function values; 28% experienced symptoms related to intermittent cholangitis or stricture; and 10% remained asymptomatic with abnormal liver function values. The outcome in patients with Bismuth class III and IV injuries was worse than in those with Bismuth class I and II injuries. At a mean followup period of 9.3 years, Frattaroli et al. obtained almost 80% favorable results with surgical reconstruction of benign biliary strictures (16). Patients with lowlevel or midlevel biliary injuries (Bismuth classes I and II) had the best results. The investigators consider RouxenY hepaticojejunostomy the procedure of choice for these injuries. For very high lesions with technically complicated hilar dissection, they advocate resection of hepatic segments to eradicate bile stasis by removing the stenotic bile duct and anastomosis between bile ducts of the remaining liver and the small bowel. Frattaroli's group utilizes transanastomotic stents on a select basis. While they do not feel that technically correct anastomoses need stenting, they place silicon stents in technically unsatisfactory anastomoses, particularly in the proximal bile ducts. The most important factors determining outcome include site of stricture, the number of previous biliary reconstructions, the type surgical procedure, and the surgeon's experience.
Page 833
In patients with very high strictures or when the right and left hepatic ducts converge within a deep hilum, it may be technically impossible to perform an anastomosis between the common hepatic duct and the small intestine. In 1956, Hepp and Couinaud first described an adequate drainage procedure by an anastomosis with the left hepatic duct (54). An alternate procedure that was also initially described in France is a drainage procedure involving the ducts draining the left liver lobe lateral to the ligamentum teres. As outlined by Blumgart and Kelley, ductal anomalies are usually related to the manner of confluence of right and left hepatic ducts and of the cystic duct with the common hepatic duct, while the anatomy of the left hepatic duct and its branches is usually consistent. The left hepatic duct always has an extrahepatic course. The left hepatic duct can be surgically exposed by lowering it from the undersurface of the quadrate lobe or by dissection at the base of the ligamentum teres. After incising the left hepatic duct longitudinally, a RouxenY loop of jejunum can be anastomosed in a sidetoside fashion. Blumgart and Kelley reported the successful performance of a biliaryenteric anastomosis involving the left hepatic ductal system in 36 of 40 patients with benign or malignant bile strictures (51). Another surgical approach for high strictures of the extrahepatic bile ducts has been described by Longmire and Sanford (55). This procedure is particularly valuable in cases of anatomic distortion of the liver hilum, which makes dissection in that area technically difficult. The dissection is performed to the left of the ligamentum teres and thereby avoids the liver hilum. One of the intrahepatic ducts of the left hepatic lobe is anastomosed to a loop of jejunum following partial resection of the left hepatic lobe. In their original communication, Longmire and Sanford described the successful treatment of a patient with an acquired stricture of the common bile duct who had failed two previous attempts at surgical reconstruction. The authors felt that in that case a bilaryenteric anastomosis could be performed more accurately with a selected intrahepatic duct than a short stump of the common hepatic duct, which was scarred from previous surgery. VI— Role of Interventional Radiology Radiological techniques are of importance in both the diagnosis and management of bile duct injuries. The surge in laparoscopic cholecystectomies in the last decade has also led to greater experience with interventional radiology management of postoperative complications. Dawson and Mueller defined several roles of interventional radiology techniques in the management of bile duct injuries (56). These included anatomic outline of the injury, percutaneous drainage of fluid collections, percutaneous transhepatic decompression of obstructed bile ducts, palliation and temporizing prior to definitive surgical repair as well as preoperative placement of stents as adjuncts in identifying landmarks during surgical dissection. In the late 1970s, Burhenne reported his experience with biliary stricture dilatation utilizing balloon catheters (57). Percutaneous techniques gained more widespread recognition during the 1980s as an alternative to surgical intervention in the management of strictures of the biliary tract. In 1986, Mueller et al. reported a series of 73 patients with biliary strictures that were managed with percutaneous balloon dilatation from 1976 to 1985 (58). The etiology of these strictures—which occurred in the era preceding the widespread use of laparoscopic cholecystectomy—was either anastomotic, iatrogenic, or secondary to sclerosing cholangitis. Fiftynine percent of the patients had undergone attempts at surgical repair of their strictures prior to balloon dilatation. The strictures in this series were mostly shorter than 3 cm in length. The author reported a 67% patency rate at 3 years, and 60% of their stricture recurrences were treated with repeated percutaneous balloon dilatation. In light of comparable success rates following surgical intervention, the author felt that the low morbidity of the percutaneous approach warrants its use as the first line of treatment of select patients. Mueller et al. considered stenting of the stricture to be controversial and felt that the decision in favor or against stenting should be made on an individual basis.
Page 834
In 1989, vanSonnenberg and coworkers reviewed the role of interventional radiology in the diagnosis and treatment of major complications following open cholecystectomies (59). They felt that close collaboration between surgeons and interventional radiologists was very fruitful and led to a significant decrease in mortality in this patient population. They underlined the importance of preoperative percutaneous biliary drainage in decompression of obstructed ducts and thus alleviation of symptoms of cholangitis and septic physiology. This guideline is in agreement with the algorithm outlined by Ress and colleagues, who advocate the use of percutaneous transhepatic cholangiography in patients with biliary dilatation following laparascopic cholecystectomies as evidenced by ultrasound (60). In select patients, they favor the placement of a transhepatic biliary drainage tube prior to surgical repair. Furthermore, vanSonnenberg et al. discuss the benefit of stents as anatomic landmarks during biliary reconstruction. In 1992, Trerotola and coworkers were among the first to review an early experience with interventional radiology procedures in the diagnosis and treatment of biliary tract complications following laparoscopic cholecystectomies (61). Major complications—such as ductal obstruction or transection—had occurred in 8 of 14 patients in their small series, with the remainder having minor complications, such as leaks. All patients with major complications underwent interventional procedures such as percutaneous biliary drainage or attempts at percutaneous balloon dilatation. In contrast to the vanSonnenberg series, only a small percentage of biliary strictures was amenable to percutaneous management. The authors suggested that primary strictures that developed following laparoscopic cholecystectomies are more resistant to percutaneous techniques than those that develop following open procedures. On the other hand, recurrent strictures following previous surgical repair can be successfully treated utilizing percutaneous balloon dilatation. Quinn and colleagues reviewed the role of interventional radiological techniques in the management of minor biliary tract complications following laparoscopic cholecystectomies (62). They examined nonsurgical treatment strategies of 20 postoperative bile leaks, the vast majority of which were from the cystic duct. Placement of a percutaneous drain under either computed tomography or ultrasound guidance and/or ERCP with stent placement were the mainstays of therapy. Cessation of bile output was the criterion to remove the drain. This strategy was successful in avoiding the need for surgery. This algorithm is contrary to that of Trerotola's group, who felt that nonoperative treatment of bile leaks was more timeconsuming than surgical intervention and that the biliary drainage catheter was unacceptable to certain patients. The mean duration of catheter drainage in the Quinn study was 11 days. In 1993, vanSonnenberg et al. reported their experience at the University of California at San Diego with coordinated radiological and surgical management of complications following laparoscopic cholecystectomies. The authors recognize the value of radiological techniques in diagnosis and relief of initial symptoms as well as recovery and characterization of offending microorganisms. In addition, they can serve as an intraoperative landmark, aid in drainage of fluid from ongoing leaks, and help avoid surgical intervention altogether in select patients. Like their series in open cholecystectomies, the authors attributed avoidance of surgical intervention to interventional radiological techniques in approximately onehalf of their patient population. Most of these patients had experienced bile leaks and were treated with percutaneous drainage of fluid collections. In addition, this series included one case in which a postoperative biliary stricture was successfully treated with percutaneous biliary drainage and balloon dilatation. Another patient, who had undergone three successful surgical interventions for the treatment of a high ductal transection, was successfully treated with percutaneous biliary drainage and metal stent placement. VII— Endoscopic Techniques Treatment of certain types of bile duct injuries by nonsurgical endoscopic techniques has been well described. It is obvious that both extent of injury and timing of diagnosis determine
Page 835
treatment modalities. Choices of specific strategies are strongly influenced by the expertise and experience of the departments of surgery, interventional radiology, and gastroenterology at the specific institution. Endoscopic approaches are particularly valuable in the diagnosis and management of bile leaks and biliary strictures. In the 1980s, endoscopic transpapillary drainage became the treatment of choice in patients with inoperable, malignant obstruction of the biliary tract. In a study by Stanley and coworkers, a comparison between endoscopic and percutaneous decompression of the biliary tract in patients with obstructive jaundice secondary to both malignant and benign conditions was addressed (63). Advantages of the endoscopic approach included decreased risk of bleeding and less discomfort for the patient. The authors reported cholangitis as a potential complication of endoscopic biliary decompression. In 1986, Sauerbruch and associates reported six cases where postoperative leaks from the biliary tract were diagnosed by means of endoscopic retrograde cholangiography (64). These patients were subsequently successfully managed with internal drainage either through a nasobiliary tube or an endoprosthesis. Placement of an endoprosthesis was more comfortable for the patient and avoided ongoing bile loss. Smith et al. reported similar results with placement of biliary stents for persistent biliary fistulas (65). Ponchon's group reported their experience with endoscopic treatment of persistent biliary fistulas (66). In their series of 24 patients, they included 9 patients who had sustained bile duct injuries during hepatic, biliary, or gastric surgery. The lesions were diagnosed with ERCP and the fistulas were treated by either sphincterotomy alone or by nasobiliary drainage and insertion of endoprostheses. Sphincterotomy is indicated to reduce the pressure in the biliary tract, thus facilitating closure of the fistula. A nasobiliary drain was used as a complementary method to reduce biliary pressures further. Like the Sauerbruch report, this one emphasizes the fact that nasobiliary drainage is uncomfortable for the patient and can lead to metabolic acidosis. The authors' practice was to leave endoprostheses in place for 4 months while the fistulas were healing and to obtain cholangiographic imaging prior to their removal. Ponchon et al. recognized that cystic duct leaks are most amenable to endoscopic treatment, while intrahepatic bile leaks are rather difficult to manage by endoscopic means alone. Similarly, Liguory and coworkers advocate percutaneous transhepatic placement of endoprostheses as an alternative to endoscopic techniques mainly with intrahepatic bile leaks (67). In 1991, Kozarek and Traverso were among the first to report the treatment of a patient who presented with a cystic duct leak 1 week following an uneventful laparoscopic cholecystectomy (68). The leak was presumed to have been due to a malpositioned or slipped clip. Following percutaneous drainage of a perihepatic biloma, a biliary stent was placed endoscopically. The patient recovered uneventfully and the stent was removed 4 weeks later. Utilizing an endoprosthesis, Goldin reported the successful treatment of a bile leak from a proximal branch of the right hepatic duct that had occurred following excision of a right hepatic echinococcal cyst (69). Without bridging the defect, the endoprosthesis was thought to act mainly by decompressing the biliary system rather than bypassing it, as is the case in leaks from the cystic duct. The fistula healed within 10 days. Foutch and coworkers summarized their experience with endoscopic management of 23 patients with postoperative bile leaks, 8 of which had occurred following laparoscopic cholecystectomies (70). Leaks occurred at the cystic duct and common duct as well as at an anomalous branch of the right hepatic duct. The patients were treated with a combination of sphincterotomy, endoprostheses, and nasobiliary drainage. Permanent closure of the leaks occurred in all 23 patients without the need for surgical intervention. Based on their experience, the authors suggest that sphincterotomy by itself may be sufficient to decompress bile ducts with distal obstruction. On the other hand, they advocate the use of endoprostheses alone without sphincterotomy in patients without distal obstruction. Extravasation of bile usually stopped within 3 to 5 days following endoscopic intervention. The mean duration before stent removal in the Foutch group's series was approximately six weeks. Nonoperative management has evolved as an alternative in select cases because surgical therapy of postoperative biliary strictures can be associated with significant morbidity and mortality and oftentimes with the need for repeat interventions. Several series report successful
Page 836
treatment of benign biliary strictures by means of balloon dilatation and through the use of endoprostheses. Berkelhammer and colleagues evaluated the efficacy of endoprosthetic stenting as treatment for benign postoperative bile duct strictures (71). Their patient population consisted of 29 patients who had developed bile duct strictures following a variety of surgical procedures, including open cholecystectomies, repairs of benign biliary strictures, removal of hepatic cysts, and pancreatic resections. The time of presentation was between 10 days and 27 years following surgery. Stricture sites were located between the common bile duct and the bifurcation of the intrahepatic ducts. Successful treatment was determined by radiological criteria, laboratory values, and absence of symptoms. The followup periods were between 2 and 42 months. The authors reported favorable results in 74% of the patients in whom placement of an endoprosthesis was successful after mean followup periods of less than 2 years. Of particular interest was the fact that they were able to successfully treat 75% of their patients who presented with recurrent bile duct strictures. They observed a better response in patients who presented early after their surgical injury as compared to those who present after long periods of time. A likely explanation was the higher pliability of scar tissue during the early stages of fibroblastic proliferation. The mean duration of endoprosthetic stenting in this series was approximately 13 weeks. The authors postulate that endoscopic stenting is especially valuable as a first therapeutic intervention in patients, who are at high operative risk, in recurrent strictures, and in patients who present early after the surgical injury. Similar success rates were reported by Walden and associates after followup periods of more than 6 years (72). Geenan's group reported their results with endoscopic balloon dilatation alone or in combination with placement of endoprostheses in 23 patients with benign bile duct strictures (73). The strictures were the result of surgical injury in more than 70% of their patients. Of note, all strictures were located at the junction of the cystic duct or in the common bile duct, and—in contrast to Berkelshammer's practice—Geenan felt that the placement of endoprostheses following balloon dilatation was unnecessary if the structure were easily dilated and showed effacement of the stricture zone. With a mean followup period of 4 years, the authors reported a good response in 78% of their patients, which was defined by complete absence of symptoms, normalization of liver enzymes, and total radiographic improvement of the radiographic features. In 1991 Davids and colleagues published a series of 70 patients with incomplete postoperative biliary strictures in whom they evaluated the efficacy of treatment with endoprosthetic stenting (74). Of these injuries, 78% had occurred at the time of a cholecystectomy, and stent placement was successful in 94% of the patients. The stents were exchanged every 3 months in an attempt to avoid cholangitis secondary to clogging. The Davids group's policy to use stents was based on unsatisfactory experiences with balloon dilatations alone, where prompt recurrences had been observed. This observation is contrary to that made by Geenan's group: 83% of their patients had favorable results after stent removal with a mean followup period of 42 months. The results from this study indicated that stenting beyond 1 year was of no proven benefit. It is noteworthy that the Davids group achieved favorable results in three patients with Bismuth type IV strictures. The same group compared the results of endoscopic and surgical treatment of benign biliary strictures that had developed after surgery for gallstones or trauma. After followup periods of approximately 4 years, the authors found that the surgically and endoscopically treated patients had comparable results. In both groups, 83% of patients had favorable outcomes and 17% of patients in both groups developed recurrent strictures. The interval between intervention and restricturing was significantly shorter in the endoscopically treated group. In the endoscopy group 20% of patients developed cholangitis, while none of the surgically treated patients experienced this complication. In the endoscopy group, 10% of patients failed nonoperative treatment and required RouxenY hepaticojejunostomies. In another study, Kozarek and coworkers reported results with endoscopic interventions in the management of bile duct strictures with and without concomitant leak that had occurred during laparoscopic or open cholecystectomy (75). Endoscopic treatment included stent placement as well as balloon dilatation. Endoscopic therapy alone or in combination with other modalities is particularly valuable if the injury is recognized early. The authors recognize
Page 837
that certain types of strictures are not amenable to endotherapy; stenotic hepaticojejunostomies with long RouxenY limbs and nearly complete transections are among the injuries that require surgical and percutaneous approaches. VIII— Recommendations The treatment of patients with injuries to their bile ducts needs to be viewed as a multidisciplinary challenge. Surgeons, gastroenterologists, and interventional radiologists need to be involved in the management of this complex problem to successfully reduce the morbidity associated with this injury. In addition to the extent of injury and the treatment options that have been discussed in a detailed fashion in the preceding paragraphs, the management of injuries at our institution is greatly influenced by the time of recognition. It is important for the surgeon to have a high index of suspicion at the time of the initial operation, since delay in diagnosis is a major cause of a complicated course. Lesions that are detected immediately at the time of the initial surgery should be repaired expeditiously. The reconstructive efforts should be guided by the key principle of providing the patient with the best possible intervention to minimize the incidence of future complications that would necessitate additional procedures. Since the majority of general surgeons who perform cholecystectomies in the community have not received training in the highly complex area of hepatobiliary reconstruction, we favor drainage of the right upper quadrant and expeditious referral to a tertiary center for definitive repair. We feel that the quality of the initial intervention is critical for the eventual outcome and therefore believe that it should be performed by experts. However, reality has taught us that, unfortunately, most injuries are not being recognized until the patients develop symptoms postoperatively. The clinical condition of the patient, the severity of the injury, and the time interval since the initial operation influence our treatment strategies. Noncircumferential injuries or injuries to minor ducts in stable patients can be amenable to nonoperative interventions with stenting and drainage. Although some centers prefer nonoperative intervention for common bile duct occlusions with balloon dilatation and stenting, we have had excellent results with operative reconstruction. It is crucial to precisely identify the anatomy of the biliary system preoperatively and to plan the repair accordingly. In addition, septic physiology needs to be addressed through drainage prior to undertaking the operation. In this subpopulation of patients, the surgeon may decide to postpone the definitive repair until the inflammatory process subsides. As long as the patient remains stable after drainage, this policy will ease the dissection. At our institution, we commonly use preoperatively placed transhepatic drains as intraoperative guides. It is our policy to leave stents postoperatively to aid with cholangiography; these stents are then removed during the sixth postoperative week. In patients who present late after their original operation, scar tissue often complicates the reconstruction. It is imperative to perform a tensionfree mucosatomucosa anastomosis between a healthy, unscarred bile duct and small intestine. The level of dissection depends on the level of the original injury as well as the extent of scarring. In conclusion, high levels of suspicion combined with expeditious referral to a center with expertise in hepatobiliary surgery, advanced endoscopy, and interventional radiology are critical to lower the morbidity in this unfortunate patient population. IX— Legal Aspects The rapid increase in adoption of laparascopic techniques for cholecystectomy has led to a rapid rise in litigation claims related to injuries of the biliary tract. A 20fold increase in medical malpractice litigation has been reported with laparoscopic cholecystectomies when compared to the open method. Besides the lower incidence of bile duct injuries with open cholecystectomies, this phenomenon may be partially due to the fact that minimally invasive procedures
Page 838
are often presented as simpler and less disabling alternatives to the open methods and the patient is left with the perception that the procedure is relatively riskfree. Several aspects of bile duct injuries make them particularly prone to medical malpractice litigations. They usually affect young people, who subsequently suffer physical as well as economic compromise. Loss in quality of life is an important aspect of malpractice litigations. Adverse financial consequences include the patient's lost wages during periods of hospitalization and recovery, lost wages from the spouse, day care expenses for children, and expenses for supplies and medication as outpatients. Savader's group conducted a study at a hepatobiliary referral center analyzing the financial consequences of treatment of laparoscopic cholecystectomy related bile duct injuries (76). Charges for hospital rooms, laboratory services, pharmacy, surgery, diagnostic and interventional radiology, as well as consultations were included. The average cost to complete a durable bile duct repair was approximately $51,000. The subgroup of patients with the highest costs were those who sustained their bile duct injuries at outside institutions and were referred to the tertiary center after attempted repair. The average costs for this population, which generally had more complicated and protracted hospital courses, was approximately $130,000. On the other hand, patients who sustained their injuries at the tertiary center and whose injuries were recognized intraoperatively and repaired immediately had an average cost of approximately $22,000. Interventional radiology procedures accounted for 44% of the total charges. The authors acknowledge that the total costs associated with a bile duct injury cannot be accurately determined, as restenoses can occur many years following the initial repair. Chronicity of injury—with patients requiring invasive procedures several years after the injury—and delay in diagnosis are important factors contributing to a high incidence of litigation. Because of increased public awareness and the aggressive behavior of medical malpractice lawyers, patients strongly believe that the injury is a direct consequence of negligence on the surgeon's part. The ratio between the number of adverse events and the number of resultant malpractice cases is referred to as the negligence ratio. Brennan and associates studied large populations in metropolitan areas and reported negligence ratios of 1:25 (77). This ratio appears to be significantly higher with bile duct injuries. Several large studies have reported that the incidence of bile duct injuries following laparascopic cholecystectomies was approximately 0.5%. Some 500,000 laparoscopic cholecystectomies are performed annually in the United States, which result in approximately 2500 injuries to the biliary system. Kern estimates that approximately 200 cases of litigation involving laparoscopic cholecystectomies are filed annually in the United States (78). Because of protracted time courses from injury to resolution of claim, it was not until recently that outcomes of malpractice litigations involving bile duct injuries from laparoscopic cholecystectomies were analyzed. Kern reported on 27 cases of bile duct injuries from 19 different states following laparoscopic cholecystectomies (79). The operations had been performed between 1989 and 1992. The median time from injury to resolution of litigation was 37 months; 13 cases were resolved by jury trials and 14 cases were resolved by outofcourt settlements. A single jury verdict was in favor of the plaintiff. The average cost of an outofcourt settlement was $506,588. When compared to outcomes of malpractice litigations following open cholecystectomies, there was a 25% increase in the proportion of outofcourt settlements and, remarkably, a 34% decrease in jury verdicts in favor of the plaintiff (80). Important arguments used during the litigations were the learning curve of the individual surgeon, with a higher rate of injuries during the first 13 cases (2.2 versus 0.5%), or negligent behavior if the injury occurred after the 14th case. Kern concluded that cases involving laparoscopic cholecystectomies are more defensible if a rapid diagnosis of complications is made and expert repair is performed expeditiously. Carroll reviewed 46 malpractice cases involving bile duct injuries sustained from 1990 to 1996 during laparoscopic cholecystectomy in the state of California (31). Contrary to Kern's observations, 74% of the injuries in this series occurred in operations performed by surgeons who had previously performed more than 10 laparoscopic cholecystectomies. Surgeons who had performed more than 50 laparoscopic cholecystectomies accounted for 33% of all bile duct injuries. Intraoperative cholangiographic results were misinterpreted in twothirds of the cases. At the time of their
Page 839
publication, litigation had been resolved in 30 of the 46 cases. In all, 70% of the cases were settled with an average payment of $221,000; in 17% of the cases, the plaintiffs prevailed with an average award of $214,000, and 13% of the cases ended with a jury verdict for the defense. As with most medical malpractice cases, the quality of the physicianpatient relationship is important in determining whether a complication will result in a litigation claim. It is particularly critical that the surgeon outline all potential complications, so that the patient will not have false expectations. References 1. Moossa AR, Mayer AD, Stabile B. Iatrogenic injury to the bile duct. Arch Surg 1990; 125:1028–1031. 2. Langenbuch CJA. Ein Fall von Extirpation der Gallenblase wegen chronischer Cholelithiasis. Berl Klin Worchenschr 1882; 19:725–728. 3. Mayo WJ. Some remarks on cases involving operative loss of continuity of the common bile duct. Ann Surg 1905; 42:90–96. 4. McArthur LL. Repair of the common bile duct. Ann Surg 1923; 78:129–138. 5. Cole WH, Reynolds JT, Ireneus C Jr. Strictures of the common duct. Ann Surg 1948; 128:332–347. 6. Lahey FH, Pyrtek LJ. Experience with the operative management of 280 strictures of the bile ducts, with description of a new method and complete followup study of end results in 229 of the cases. Surg Gynecol Obstet 1950; 91:25–56. 7. Saypol GM, Kurian G. A technique of repair of stricture of the bile duct. Surg Gynecol Obstet 1969; 129:1070–1076. 8. Wexler MJ, Smith R. Jejunal mucosal graft. Am J Surg 1975; 129:204–211. 9. Semm. K. Endoscopic appendectomy. Endoscopy 1983; 15(2):59–64. 10. Dubois F, Icard P, Berthelot G, Levard H. Coelioscopic cholecystectomy. Ann Surg 1990; 211:60–62. 11. Nenner RP, Imperato PJ, Rosenberg C. Increased cholecystectomy rates among Medicare patients after the introduction of laparoscopic cholecystectomy. J Commun Health 1994; 19:409–416. 12. Gilsdorf JR, Phillips M, McLeod MK, Harness JK, Hoversten GH, Woodbury D, Daley K. Radionuclide evaluation of bile leakage and the use of subhepatic drains after cholecystectomy. Am J Surg 1986; 151:259–262. 13. Van der Linden W, Kempi V, Gedda S. A radionuclide study on the effectiveness of drainage after elective cholecystectomy. Ann Surg 1981; 193:155–160. 14. Gould L, Patel A. Ultrasound detection of extrahepatic encapsulated bile: ''biloma." AJR 1970; 132:1014–1015. 15. Bismuth H. Postoperative strictures of the bile duct. In: Blumgart LH, ed. The Biliary Tract. V. Edinburgh: Churchill Livingstone, 1982, pp 209–218. 16. Frattaroli FM, Reggio D, Guadalaxara A, Illomei G, Pappalardo G. Benign biliary strictures: a review of 21 years of experience. J Am Coll Surg 1996; 183:506– 513. 17. Johnston GW. Iatrogenic bile duct stricture: an avoidable surgical hazard? Br J Surg 1986; 73:245–247. 18. Asbun HJ, Rossi RL, Lowell JA, Munson JL. Bile duct injury during laparoscopic cholecystectomy: mechanism of injury, prevention, and management. World J Surg 1993; 17:547–552. 19. Hunter JG. Avoidance of bile duct injury during laparoscopic cholecystectomy. Am J Surg 1991; 162:71–76. 20. Davidoff AM, Pappas TN, Murray EA, Hilleren DJ, Johnson RD, Baker ME, Newman GE, Cotton PB, Meyers WC. Mechanisms of major biliary injury during laparoscopic cholecystectomy. Ann Surg 1992; 215:196–202.
Page 840
21. The Southern Surgeons Club, Moore MJ, Bennett CL. The learning curve for laparoscopic cholecystectomy. Am J Surg 1995; 170:55–59. 22. The Southern Surgeons Club. A prospective analysis of 1518 laparoscopic cholecystectomies. N Engl J Med 1991; 324:1073–1078. 23. Gregg RO. The case for selective cholangiography. Am J Surg 1988; 155:540–544. 24. Flowers JL, Zucker KA, Graham SM, Imbembo AL, Bailly RW. Laparoscopic cholangiography. Ann Surg 1992; 215:209–216. 25. Brodish RJ, Fink AS. ERCP, cholangiography, and laparoscopic cholecystectomy. Surg Endosc 1993; 7:3–8. 26. Woods MS, Traverso LW, Kozarek RA, Tsao J, Rossi RL, Gough D. Donohue JH. Characteristics of biliary tract complications during laparoscopic cholecystectomy: a multiinstitutional study. Am J Surg 1994; 167:27–34. 27. Chapman WC, Halevy A, Blumgart LH, Benjamin IS. Postcholecystectomy bile duct strictures. Arch Surg 1995; 130:597–604. 28. Stewart L, Way LW. Bile duct injuries during laparoscopic cholecystectomy. Arch Surg 1995; 130:1123–1128. 29. Mirza DF, Narsimhan KL, Neto BH, Mayer AD, McMaster P, Buckels JAC. Bile duct injury following laparoscopic cholecystectomy: referral pattern and management. Br J Surg 1997; 84:786–790. 30. Strasberg SM, Hertl M, Soper NJ. An analysis of the problem of biliary injury during laparoscopic cholecystectomy. J Am Coll Surg 1995; 180:101–125. 31. Carroll BJ, Birth M, Phillips EH. Common bile duct injuries during laparoscopic cholecystectomy that result in litigation. Surg Endosc 1998; 12:310–313. 32. Blumgart LH, Kelley CJ, Benjamin IS. Benign bile duct stricture following cholecystectomy: critical factors in management. Br J Surg 1984; 71:836–843. 33. Swanson EA, Millians WS, Sotus PC. Liver cell regeneration and degeneration after lobar biliary obstruction in dogs. Am J Gastroenterol 1967; 47:280–286. 34. Stewart HL, Cantarow A, Morgan DR. Changes in the liver of the cat following ligation of single hepatic ducts. Arch Surg 1937; 23:641–652. 35. Hadjis NS, Blumgart LH. Injury to segmental bile ducts. Arch Surg 1988; 123:351–353. 36. Rutledge RH. Methods of repair of noncircumferential bile duct defects. Surgery 1983; 93:333–342. 37. Browder IW, Dowling JB, Koontz K, Litwin MS. Early management of operative injuries to the extrahepatic biliary tract. Ann Surg 1987; 205:649–658. 38. Lillemoe KD. Treatment of laparoscopic bile duct injuries. Curr Tech Gen Surg 1997; 6:1–8. 39. Csendes A, Diaz JC, Burdiles P, Maluenda F. Late results of immediate primary end to end repair in accidental section of the common bile duct. Surg Gynecol Obset 1989; 168:125–130. 40. AndrenSandberg A, Johansson S, Bengmark S. Accidental lesions of the common bile duct at cholecystectomy. Ann Surg 1985; 201:452–455. 41. Northover JMA, Terblanche J. A new look at the arterial supply of the bile duct in man and its surgical implications. Br J Surg 1979; 66:379–384. 42. Matthews JB, Baer HU, Schweizer WP, Gertsch P, Carrel T, Blumgart LH. Recurrent cholangitis with and without anastomotic stricture after biliaryenteric bypass. Arch Surg 1993; 128:269–272. 43. Hutson DG, Russell E, Yrizarry J, Levi JU, Livingstone AS, Guerra J, Reddy R, Jeffers L, Schiff ER, Scagnelli T, Mendez K. Percutaneous dilatation of biliary strictures through the afferent limb of a modified RouxenY choledochojejunostomy or hepaticojejunostomy. Am J Surg 1998; 175:108–113. 44. Millis JM, Tompkins RK, Zinner MJ, Longmire WP, Roslyn JJ. Management of bile duct injuries. Arch Surg 1992; 127:1077–1084. 45. Pellegrini CA, Thomas MJ, Way LW. Recurrent biliary stricture. Am J Surg 1984; 147:175–180.
Page 841
46. Genest JF, Nanos E, GrundfestBroniatowski S, Vogt D, Hermann RE. Benign biliary strictures: an analytic review (1970 to 1984). Surgery 1986; 99:409–413. 47. Terblanche J, Worthley CS, Spence RAJ, Krige JEJ. High or low hepaticojejunostomy for bile duct strictures? Surgery 1990; 108:828–834. 48. Saber K, ElManialawi M. Repair of bile duct injuries. World J Surg 1984; 8:82–89. 49. Bismuth H, Franco D, Corlette MB, Hepp J. Long term results of RouxenY hepaticojejunostomy. Surg Gynecol Obstet 1978; 148:161–167. 50. Roethlin MA, Loepfe M, Schlumpf R, Largiader F. Longterm results of hepaticojejunostomy for benign lesions of the bile ducts. Am J Surg 1998; 175:22–26. 51. Blumgart LH, Kelley CJ. Hepaticojejunostomy in benign and malignant high bile duct stricture: approaches to the left hepatic ducts. Br J Surg 1984; 71:257–261. 52. Tocchi A, Costa G, Lepre L, Liotta G, Mazzoni G, Sita A. The longterm outcome of hepaticojejunostomy in the treatment of benign bile duct strictures. Ann Surg 1996; 224:162–167. 53. Bauer TW, Morris JB, Lowenstein A, Wolferth C, Rosato FE, Rosato EF. The consequences of a major bile duct injury during laparoscopic cholecystectomy. J Gastrointest Surg 1998; 2:61–66. 54. Hepp J, Couinaud C. L'abord de l'utilisation du canal hepatique gauche dans les reparation de la voie biliare principale. Presse Med 1956; 64:947–948. 55. Longmire WP, Sanford MC. Intrahepatic cholangiojejunostomy with partial hepatectomy for biliary obstruction. Surgery 1948; 128:264–276. 56. Dawson SL, Mueller PR. Interventional radiology in the management of bile duct injuries. Surg Clin North Am 1994; 74(4):865–874. 57. Burhenne HJ. Dilatation of biliary tract strictures: a new roentgenologic technique. Radiol Clin 1975; 44:153–159. 58. Mueller PR, van Sonnenberg E, Ferrucci JT, Weyman PJ, Butch RJ, Malt RA, Burhenne HJ. Biliary stricture dilatation: multicenter review of clinical management in 73 patients. Radiology 1986; 160:17–22. 59. vanSonnenberg E, Casola G, Wittich GR, Christensen R, Varney RR, Neff CC, D'Agostino HB, Moossa AR. The role of interventional radiology for complications of cholecystectomy. Surgery 1990; 107:632–638. 60. Ress AM, Sarr MG, Nagorney DM, Farnell MB, Donohue JH, McIlrath DC. Spectrum and management of major complications of laparoscopic cholecystectomy. Am J Surg 1993; 165:655–662. 61. Trerotola SO, Savader SJ, Lund GB, Venbrux AC, Sostre S, Lillemoe KD, Cameron JL, Osterman FA. Biliary tract complications following laparoscopic cholecystectomy: imaging and intervention. Radiology 1992; 184:195–200. 62. Quinn SF, Sangster W, Standage B, Schuman E, Gross G. Biliary complications related to laparoscopic cholecystectomies: radiologic diagnosis and management. Surg Laparasc Endosc 1992; 2:279–286. 63. Stanley J, Gobien RP, Cunningham J, Andriole J. Biliary decompression: an institutional comparison of percutaneous and endoscopic methods. Radiology 1986; 158:195–197. 64. Sauerbruch T, Weinzierl M, Holl J, Pratschke E. Treatment of postoperative bile fistulas by internal endoscopic biliary drainage. Gastroenterology 1986; 90:1998–2002. 65. Smith AC, Schapiro RH, Kelsey B, Warshaw AL. Successful treatment of nonhealing biliarycutaneous fistulas with biliary stent. Gastroenterology 1986; 90:764– 769. 66. Ponchon T, Gallez JF, Valette PJ, Chavaillon A, Bory R. Endoscopic treatment of biliary tract fistulas. Gastrointest Endosc 1989; 35:490–498. 67. Liguory C, Vitale GC, Lefebre JF, Bonnel D, Cornud F. Endoscopic treatment of postoperative biliary fistulae. Surgery 1991; 110:779–784. 68. Kozarek RA, Traverso LW. Endoscopic stent placement for cystic duct leak after laparoscopic cholecystectomy. Gastrointest Endosc 1991; 37:71–73. 69. Goldin E, Katz E, Wengrower D, Kluger Y, Haskel L, Shiloni E, Libson E. Treatment of
Page 842
fistulas of the biliary tract by endoscopic insertion of endoprostheses. Surg Gynecol Obstet 1990; 170:418–423. 70. Foutch PG, Harlan JR, Hoefer M. Endoscopic therapy for patients with a postoperative biliary leak. Gastrointest Endosc 1993; 39:416–421. 71. Berkelhammer C, Kortan P, Haber GB. Endoscopic biliary prostheses as treatment for benign postoperative bile duct strictures. Gastrointest Endosc 1989; 35:95–101. 72. Walden D, Raijman I, Fuchs E, Kandel G, Marcon N, Kortan P, Haber G. Long term followup of endoscopic stenting for benign postoperative biliary strictures. Gastrointest Endosc 1993; 39:335. 73. Geenan DJ, Geenan JE, Hogan WJ, Schenck J, Venu RP, Johnson GK, Jackson A Jr. Endoscopic therapy for benign bile duct strictures. Gastrointest Endosc 1989; 35:367–371. 74. Davids PHP, Tanka AKF, Rauws EAJ, van Gulik TM, van Leeuwen DJ, de Wit LT, Verbeek PCM, Huibregtse K, van der Heyde N, Tytgat GNJ. Benign biliary strictures. Ann Surg 1993; 217:237–243. 75. Kozarek RA, Ball TJ, Patterson DJ, Brandabur JJ, Raltz S, Traverso W. Endoscopic treatment of biliary injury in the era of laparoscopic cholecystectomy. Gastrointest Endosc 1994; 40:10–16. 76. Savader SJ, Lillemoe KD, Prescott CA, Winick AB, Venbrux AC, Lund GB, Mitchell SE, Cameron JL, Osterman FA. Laparoscopic cholecystectomy related bile duct injuries. Ann Surg 1997; 225:268–273. 77. Brennan TA, Leape LL, Laird NM, Herbert L, Localio AR, Lawthers AG, Newhouse JP, Weiler PC, Hiatt HH. Incidence of adverse effects and negligence in hospitalized patients. N Engl J Med 1991; 324:370–376. 78. Kern KA. Medicolegal perspectives on laparoscopic bile duct injuries. Surg Clin North Am 1994; 74:979–984. 79. Kern KA. Malpractice litigation involving laparoscopic cholecystectomy: cost, cause, and consequences. Arch Surg 1997; 132:392–398. 80. Kern KA. Medicolegal analysis of bile duct injury during open cholecystectomy and abdominal surgery. Am J Surg 1994; 168:217–222.
Page 843
37— The Management of Benign and Malignant Biliary Strictures Laurence S. Bailen and Eric D. Libby New England Medical Center, Boston, Massachusetts I— Introduction The approach to management of biliary strictures continues to evolve. Once considered the exclusive domain of hepatobiliary surgeons, strictures can now be treated successfully using percutaneous, endoscopic, or laparoscopic techniques. Each approach has potential advantages and drawbacks. This chapter reviews the current treatment options for benign and malignant biliary strictures and suggests algorithms for the management of different clinical situations. However, optimal treatment of individual patients depends on specific patient characteristics as well as the local availability of skilled personnel within the surgical and nonsurgical disciplines. II— Presentation of Biliary Stricture Biliary strictures present most commonly with signs of cholestasis. Jaundice and pruritus are often the first symptoms; more subtle findings may include acholic stools or signs of malabsorption. Occasionally, patients present with acute cholangitis, but this is unusual in the absence of prior biliary interventions. Many patients are discovered incidentally, when screening serum biochemistry panels show abnormalities of liver function. Painless jaundice has been regarded as a sign of malignant biliary obstruction. Common bile duct (CBD) strictures may also be found incidentally in patients being evaluated for pain associated with chronic pancreatitis. Rarely, patients may present with secondary biliary cirrhosis due to prolonged or intermittent biliary obstruction. Secondary biliary cirrhosis may lead to portal hypertension and hepatic failure (1,2). Chronic biliary obstruction may also lead to malabsorption of fat soluble vitamins A, D, and K. III— Diagnosis of Biliary Strictures The diagnosis of biliary stricture relies primarily on cholangiography. Dilatation of bile ducts is often identified by conventional computed tomography (CT) or transabdominal ultrasound scans, but these tests are relatively insensitive and cannot be relied upon to exclude pathology. Even when ductal obstruction is accurately detected by these modalities, they perform poorly
Page 844
at defining the exact location of the lesion within the biliary tree and determining its cause or characteristics. Direct cholangiography remains the "gold standard." Radiological views are obtained by injection of iodinated contrast into the biliary tree and may be achieved by endoscopic, percutaneous, or surgical approaches. The fine resolution of direct cholangiography is superior to that possible with scanning techniques, and there is less potential for artifact. Furthermore, direct techniques afford the opportunity to perform therapeutics during the same procedure. The major disadvantage of direct cholangiography is the invasiveness of the procedures, with higher risks of complications. Magnetic resonance cholangiopancreatography (MRCP) has emerged as a noninvasive alternative to direct cholangiography (3). Reconstruction of data acquired via magnetic resonance provides biliary and pancreatographic images similar to those obtained via endoscopic retrograde cholangiopancreatography (ERCP). The sensitivity of MRCP appears superior to that of either CT or ultrasound, although it has yet to prove as reliable as direct cholangiography, particularly in patients with biliary strictures. MRCP is purely diagnostic; thus treatment of any lesion identified will require surgery or direct cholangiographic techniques. Nevertheless, this is a rapidly evolving technology and is likely to play an increasingly important role when diagnosis is the only goal (4,5). IV— Causes of Biliary Strictures The term stricture describes a narrowing of the bile duct caused by scar tissue or tumor. Strictures are considered to be irreversible unless direct mechanical dilation is performed. A stenosis can be any ductular narrowing, including one caused by inflammatory, infectious, or muscular processes or by extrinsic compression. Although many biliary narrowings are more accurately described as stenoses, the term stricture has been widely applied, and this chapter generally refers to these lesions as strictures. Malignant tumor growth is the most common cause of biliary stricture. Adenocarcinoma of the head of the pancreas is by far the most frequent etiology underlying malignant biliary obstruction (6–8). Other causes include ampullary neoplasm, cholangiocarcinoma, and tumor metastatic to the liver or biliary tract. Malignant biliary strictures typically present insidiously with painless jaundice. Painful presentations are not uncommon, however, particularly when lesions are advanced or lead to pancreatitis. There are numerous benign causes of biliary stricture. Chronic choledocholithiasis can lead to scarring of the distal common bile duct (CBD). Impaction of a gallstone in the cystic duct may lead to narrowing and obstruction of the CBD in Mirizzi's syndrome. Biliary infestations by nematodes such as Ascaris lumbricoides or liver flukes such as Clonorchis sinensis can cause chronic scarring, while viral or parasitic infections have been associated with biliary stricture formation in patients with the acquired immunodeficiency syndrome (AIDS). Primary sclerosing cholangitis (PSC), a cholestatic liver disease of unknown etiology, typically causes biliary strictures and inflammatory changes in the liver that eventually lead to cirrhosis. The biliary strictures in PSC patients are located anywhere in the biliary tract and cause a characteristic cholangiographic picture. Extrabiliary diseases may cause obstructive jaundice via extrinsic compression of the bile duct. In acute pancreatitis, patients often develop narrowing of the intrapancreatic portion of the CBD due to pancreatic edema; this stenosis is rarely symptomatic and usually resolves without specific treatment. In patients with chronic pancreatitis, biliary stricture in the same location is typically caused by pancreatic fibrosis and may lead to secondary biliary cirrhosis. Pancreatic pseudocysts develop in either the acute or chronic setting and can externally compress the biliary tree. One of the more common causes of benign stricture is surgical injury. Most postoperative biliary strictures occur after cholecystectomy. Biliary strictures may also complicate pancreatic,
Page 845
gastric, or hepatic resections, and they occur commonly after orthotopic liver transplantation (OLT). Postoperative strictures may develop as a result of technical errors, ischemia, or infections. V— Treatment of Biliary Strictures The primary objective in the management of biliary strictures is to establish unrestricted bile flow. In so doing, symptoms of jaundice, pruritus, pain, and malabsorption are alleviated and secondary biliary cirrhosis is prevented. In the setting of infection, cholangitis is best treated by providing adequate biliary drainage. An additional goal in the management of the patient with malignant stricture is to establish the diagnosis definitively and thus facilitate other antineoplastic therapy. A number of treatment options are available to accomplish these objectives; the general use of these techniques is reviewed below. The appropriate application of these treatments to specific clinical settings is discussed in more detail under the individual disease processes. A— Open Surgery Open surgery has long been the mainstay of biliary stricture management. Direct resection of the stricture with endtoend anastomosis of the bile duct would appear to be the simplest solution, but this approach is seldom followed today due to high rates of restenosis. Most often, a biliaryenteric anastomosis is created to bypass the stricture. Depending on the level of the biliary stricture, the surgeon may perform choledochoduodenostomy, choledochojejunostomy, or hepaticojejunostomy, usually creating a RouxenY limb for jejunal anastomosis. An advantage of open surgery is the ability to create a large anastomosis. This ensures generous biliary drainage while potentially decreasing the incidence of subsequent stricture formation. At open laparotomy, the surgeon may resect malignant or ischemic tissue and is afforded the best opportunity to sample tissue for an accurate pathological diagnosis. If there is concomitant gastric outlet obstruction, gastrojejunostomy or similar bypass can be performed during the same operation. The creation of a bilioenteric anastomosis results in altered anatomy, with potential disturbance of the normal physiology of digestion and absorption. The wide anastomosis also permits reflux of intestinal contents up into the biliary tree, with potential for bacterial colonization and cholangitis. Bypassed segments may develop problems related to stasis or poor drainage, leading to the "sump syndrome" or "stump syndrome" (9–11). The main disadvantage of open surgery is its inherent invasiveness, with a typically prolonged recovery. Patients remain confined to bed longer and require longer hospitalization following open bypass surgery than after treatment by less invasive approaches. For elderly or infirm patients, these factors alone may lead to increased complications and mortality. For those with malignant strictures and short life expectancy, this additional time in the hospital is particularly important. B— Percutaneous Transhepatic Therapy Percutaneous transhepatic cholangiography (PTC) is performed by interventional radiologists who access the biliary tree via a puncture of the skin and liver capsule. Once a catheter is securely placed within the bile duct, drainage is established through an external catheter or internally via a stent placed across the stricture. PTC is less invasive, requires less anesthesia, is generally associated with a shorter recovery period than surgery. PTC is particularly advantageous for treatment of proximal strictures in the intrahepatic ducts, for which either surgical or endoscopic approaches are technically challenging.
Page 846
There are some disadvantages to percutaneous drainage. Compared with endoscopic techniques, PTC is more likely to be complicated by bile leakage or bleeding, and it is associated with more patient discomfort (12). If the intrahepatic ducts are not dilated, puncture of the bile ducts is technically difficult, often leading to failure of the procedure. External biliary drainage may lead to fluid and electrolyte imbalances, and close monitoring is required. Furthermore, external drainage leads to bile salt loss that may affect nutrient and drug absorption. With percutaneous treatment, multiple procedures are usually required, increasing hospital length of stay and patient discomfort. The frequent need to maintain external catheters may significantly impair a patient's quality of life. C— Endoscopy Endoscopic treatment of biliary strictures is accomplished via ERCP. The biliary tree is visualized and strictures are identified by retrograde injection of contrast through the ampulla of Vater. Once identified, a stricture may be crossed with a hydrophilic guidewire. Treatment options may include rigid or balloon dilatation or insertion of a stent. Sphincterotomy is often performed to facilitate other procedures, such as stenting, but it has also been used as a primary therapy for ampullary tumors. The endoscopic approach is the least invasive method of establishing biliary drainage. Patients usually experience less discomfort, recover more quickly, and are able to leave the hospital sooner than with other therapeutic modalities. However, biliary access is not always achieved via an endoscopic approach, particularly among patients with altered gastric or duodenal anatomy due to tumor or prior surgery. Although ERCP is associated with a lower risk of complications than either a surgical or percutaneous approach, there is a higher risk of pancreatitis (13). Even when the common bile duct is successfully cannulated, endoscopic treatment of hilar or intrahepatic strictures may be difficult due to the relatively long distance from the papilla and the working channel of the endoscope. For proximal biliary strictures, ERCP is associated with a treatment failure rate higher than that of PTC (14,15). The most common complication of either endoscopic or percutaneous approaches is late stent occlusion. Retrospective reviews have found occlusion rates ranging from 10 to 30% (16–18). Much higher rates are reported from prospective randomized studies, with stent blockage occurring in 21 to 54% of patients, for an overall incidence of 42% (8,19–24) (Table 1). Among randomized trials, the average functional patency of a largediameter plastic stent is 5 Table 1 Reported Rates of Stent Occlusion from Prospective, Randomized Trials of 10 to 12Fr Straight Plastic Stents Occluded %
Median patency (months)
10
43
NSa
10
30
54
4.2
28
11.5
12
43
6.0
1994
67
10
35
52
1.8
1994
95
10
36
38
4.5
Barrioz (23)
1994
10
10–12
7
70
1.4
Ghosh (24)
1994
70
10–12
15
21
9.2
145
42
4.9
Stent size (Fr)
n
Study
Date
Shepherd (20)
1988
23
10
Davids (19)
1992
56
Knyrim (21)
1993
Sung (22) Smith (8)
Total a
NS, not stated. Source: From Ref. 16.
349
n
Page 847
months. While expandable metal stents remain open significantly longer, tissue ingrowth limits patency and prevents stent removal should occlusion occur. D— Laparoscopic Surgery Laparoscopic techniques for treatment of biliary strictures remain in an early stage of development. The most widely used procedure is cholecystojejunostomy, chosen for its ease of performance rather than for any superior efficacy. Gastrojejunostomy may also be performed laparoscopically to treat gastric outlet obstruction. More traditional bilioenteric anastomoses such as choledochojejunostomy are difficult to construct with current laparoscopic techniques (25). Although potentially less painful and requiring shorter hospitalization than is needed for open surgery, laparoscopic methods remain more invasive than endoscopic or percutaneous techniques, and current knowledge of effectiveness is based on very limited experience. VI— Management of Specific Diseases A— Malignant Biliary Strictures Adenocarcinoma of the head of the pancreas is the most common cause of malignant biliary obstruction. Pancreatic cancer affects 10 per 100,000 population and is the fifth leading cause of cancer death in the United States (6). Most patients are elderly and succumb to the disease within a year of presentation. Surgical cure is feasible for only a small percentage of patients, as the vast majority have advanced disease by time of diagnosis (26,27). Thus, palliative therapy is the best option for most. Other tumors causing obstructive jaundice include ampullary neoplasm, cholangiocarcinoma, gallbladder carcinoma, lymphoma, and metastasis from carcinoma of the colon, ovary, or breast. For all but the ampullary tumors, attempts at resection are unlikely to result in cure. 1— Diagnosis of Malignancy In most cases, the diagnosis of malignant biliary obstruction is apparent by history and crosssectional imaging. The finding of a mass lesion associated with upstream biliary ductal dilation on CT or ultrasound scan is usually sufficient to indicate malignancy. Nevertheless, when several treatment options are available, unequivocal diagnosis is desirable before proceeding to definitive therapy. Serological tumor markers such as CA199 can suggest malignancy, but these levels may be elevated by biliary obstruction or cholangitis in the absence of tumor. Tissue for histological evaluation may be obtained by CT or ultrasoundguided needle biopsy of the mass. Alternatively, strictures may be sampled at time of direct cholangiography (Fig. 1). Unfortunately, the sensitivity of cytological brushings, scrapings, and needle aspiration during cholangiography is suboptimal, generally between 40 and 70% (28–31). Even biopsies obtained at open surgery may fail to reveal the true identity of a malignant stricture. Thus, it may be impossible to rule out malignancy short of complete surgical excision of the lesion. For goodrisk surgical candidates in whom the diagnosis remains unknown, this last option should be considered. 2— Management of Distal Obstruction For patients with pancreatic cancer, ampullary carcinoma, or distal cholangiocarcinoma, initial evaluation should assess for resectability. Surgery is the only treatment that offers the potential for cure. Crosssectional imaging studies such as CT, ultrasonography, or magnetic resonance imaging (MRI) are used to assess for signs of metastasis or vascular invasion (32). Doppler ultrasonography should assess for vascular involvement if other imaging is negative. For most patients, decisions regarding a treatment plan can be made without staging laparotomy or other
Page 848
Figure 1 Cholangiographic appearance of malignant biliary stricture due to pancreatic adenocarcinoma.
surgery (33). Endoscopic ultrasound is a promising new modality for tumor staging (see Chap. 19). When surgical cure appears feasible in otherwise goodrisk candidates, an attempt at complete resection should be offered. Complete excision of a pancreatic or distal biliary malignancy requires a pancreaticoduodenectomy, or Whipple resection. This procedure entails partial antrectomy, cholecystectomy, and resection of the pancreatic head, distal CBD, and duodenum. Anastomoses are created between the jejunum and the bile duct, the tail of the pancreas, and the common hepatic duct. Although the operative mortality rates for Whipple resection have fallen markedly in the past decade, the procedure is associated with a relatively high incidence of postoperative complications (8,20). Among the larger group of patients without significant hope of curative resection, treatment goals are purely palliative. In most cases, relief of obstructive jaundice is the primary consideration. Duodenal obstruction may complicate pancreatobiliary malignancies, but this problem occurs in less than 20% of patients and should not figure into the choice of treatments for most patients. Early comparisons of different treatments for malignant obstruction found superior results for those treated surgically. However, there was a tendency to choose surgical treatment for healthier patients, while the frail and elderly patients were treated with less invasive methods (34). To overcome this inherent bias, several randomized trials have been conducted to compare operative and nonoperative management of surgical candidates (8,20,35–37) (Table 2). Stents were placed endoscopically in four of these studies (8,20,35,37) (Fig. 2) and percutaneously in the other (36). Each found comparable rates of successful biliary drainage, but in four of the five, 30day mortality was lower in the nonoperative group (8,20,36,37). Initial hospital length of stay was shorter in the nonoperative group in all studies. However, late complications were more common in the stented groups. These were primarily due to stent clogging, but duodenal obstruction by tumor developed in a smaller number of patients. There was no difference in longterm survival between groups. The only study addressing cost found significant savings with the endoscopic approach (38). A randomized comparison of percutaneous and endoscopic approaches to stenting found a higher success rate and lower rate of complications and mortality in the patients stented endoscopically (12). Endoscopic stenting is associated with less patient discomfort and shorter hospitalization and should be the preferred or initial approach in patients with distal biliary obstruction.
Page 849 Table 2 Management of Distal Malignant Strictures
N
Study Dowsett (37) Shepherd (20)
Complications (%)
Success (%) Surg
Endo
Surg
Endo
Surg
101
103
94
91
10
28
23
25
91
92
33
56
Endo
Duodenal obstruction (%)
Median survival (weeks)
Surg
Endo
6
1
21
26
9
4
22
8
Endo
Surg
25
25
96
88
36
20
0
0
12
14
Smith (8)
100
101
92
92
11
29
26
10
21
26
Total
249
254
93
91
15
30
14
5
20
23
Andersen (35)
Page 850
Figure 2 Endoscopic image of plastic biliary stent in duodenum.
Approximately 14% of patients presenting with malignant jaundice eventually develop duodenal obstruction due to tumor (34,39). Gastrojejunostomy remains the most established therapy; when this is undertaken, a biliary bypass is usually created during the same operation. Gastrojejunostomy is most often performed with the open approach, although laparoscopic techniques have been described (40). Recently, endoscopic duodenal stenting combined with biliary stenting has been reported as a successful alternative to surgical double bypass (41–43), but experience remains limited. There have been no randomized comparisons of endoscopic and surgical approaches for biliary and duodenal obstruction. Metal Stents. The major limitation to endoscopic or percutaneous stenting is late occlusion. Once a plastic stent is placed in the bile duct, an amorphous sludge begins to accumulate on its surfaces. Given sufficient time, the lumen becomes occluded, bile flow ceases, and the patient develops symptoms of recurrent biliary obstruction. Since the stent is a contaminated foreign body, clogging is often complicated by cholangitis and sepsis. Such complications can be managed with antibiotics and replacement of the stent, but this requires repeated hospitalization and endoscopic procedures that limit the patient's functional independence and the quality of life (see Chaps. 28 and 33). Expandable metal stents were developed to help overcome the problem of stent occlusion. These devices are assembled in a lowprofile delivery system that allows easy passage through an endoscope channel or via percutaneous puncture. Once placed across the stricture, the stent is deployed and allowed to expand three to four times its initial diameter (Figs. 3 and 4). Because they provide a much larger internal lumen, expandable stents remain patent significantly longer. Following deployment, a metal stent quickly becomes embedded in the bile duct wall and is then nearly impossible to remove. Randomized comparisons of plastic and metallic stents for malignant jaundice show significant advantages for the expandable stents (19,21,44–47). Stent occlusion occurs less often, and median patency is approximately twice that of plastic stents. Although expandable stents cost more than their plastic counterparts, their lower occlusion rate leads to an overall decrease in number of procedures and hospitalizations needed for stent exchange. Thus, except when used for patients with a life expectancy of under 3 months, these devices appear to be costeffective (48).
Page 851
Figure 3 Radiographic appearance of expandable metal stent in bile duct.
Trials of metallic biliary stents generally were restricted to endoscopic techniques, but similar benefits were seen for expandable stents placed percutaneously (46). There have been no direct comparisons of metallic stent placement versus surgical bypass. Given the reduction in late stent clogging afforded by these new devices, one would expect that endoscopic stenting with expandable metallic stents would prove superior to surgery. A prospective randomized trial has yet to be performed, however.
Figure 4 Endoscopic image of metal biliary stent in duodenum.
Page 852
In summary, nonoperative palliation of malignant jaundice is as effective as open surgical bypass but is associated with lower procedurerelated mortality and shorter hospital length of stay. There are as yet insufficient data to determine the role of laparoscopic bypass. For distal lesions, the endoscopic approach is preferable to percutaneous stenting, but either approach may be preferable to open surgical palliation. Expandable metallic stents have superior patency as compared with plastic stents and may reduce the incidence of late complications. When duodenal obstruction is present or appears imminent, surgical biliary bypass combined with gastrojejunostomy remains the most desirable approach. 3— Management of Hilar Malignant Biliary Obstruction Tumors of the hepatic duct bifurcation account for less than 20% of all malignant strictures. Hilar obstruction is most commonly caused by cholangiocarcinoma, hepatic metastases, hepatocellular carcinoma, lymphoma, or local extension of gallbladder or pancreatic cancer. Resection for cure is usually not feasible. Less than 10% of patients with malignant hilar obstruction are alive 5 years after diagnosis; most die within 1 year (49). Thus, less invasive methods of palliation are attractive in this patient population. The traditional surgical approach to palliation involves hepaticojejunostomy, frequently with placement of an internal or external drain to maintain potency of the anastomosis (50,51). The placement of a drain across the anastomosis is controversial and not practiced by all hepatobiliary surgeons. Proximal anastomoses are difficult to construct, and the complication rate is higher than for surgical bypass of more distal lesions (52). Stenting of malignant hilar strictures can be achieved by either endoscopic or percutaneous routes. It may be difficult to place a stent across a proximal biliary stricture by endoscopic techniques because of the distance of the lesion from the papilla. In some patients, it is not possible to gain access to both left and right hepatic ducts when approaching the strictures from the common duct. Selective entrance into one of these dilated intrahepatic ducts may be more reliably attained by separate puncture to either lobe of the liver via a percutaneous approach. Nonetheless, the endoscopic route is preferred as the initial approach, as it is considered less traumatic and is associated with less patient discomfort, fewer procedure sessions, and shorter hospitalizations. In cases where ERCP fails to result in drainage of the desired biliary system, the percutaneous approach is usually successful. For some situations, a percutaneousendoscopic ''rendezvous" procedure is most appropriate, combining the advantages of percutaneous intrahepatic duct access with decreased trauma from endoscopic stent placement (53). An important issue in treatment of hilar strictures is the question of whether both sides of the liver must be stented. When right and left hepatic ducts do not communicate, a single stent will not drain the entire liver. Drainage of one lobe is usually adequate to alleviate jaundice, but cholangitis may develop in the other lobe, particularly if it has been instrumented or visualized with contrast injection. Some series have reported longer survival and a lower incidence of septic complications among patients treated with bilateral stents (54). However, others have found excellent palliation of jaundice with a low incidence of cholangitis when only one stent was placed, with no overall difference in outcomes (55). Perhaps the key factor is whether both left and right hepatic ducts are visualized, and hence contaminated, by injection of contrast. In a recent retrospective review, Chang and colleagues (15) examined the effect of ductal opacification with different drainage strategies. One group of patients had a single lobe visualized and the same lobe drained, another had both lobes opacified and both lobes drained, and a third group had both lobes injected with contrast and only one lobe drained. Acute cholangitis occurred significantly more often in the last group. The best survival was seen for patients treated with bilateral drainage, and the worst survival was in those with contrast filling of both lobes but drainage achieved in only one. Metallic stents may be particularly useful for treatment of proximal malignant obstruction. The openmesh design permits bile flow through the sides of the stent, and side branches
Page 853
of the intrahepatic are not obstructed. Furthermore, a second stent can often be placed through the meshwork and directed to the opposite lobe when bilateral drainage is desired. Since metallic stents are not removable, they need not extend the entire distance from the hilum to the papilla; thus, a shorter stent can be used, which may decrease the chance of late blockage. Nonsurgical techniques have lower success rates for hilar lesions than for distal biliary obstruction. Complications are also more common and are most often infectious due to incomplete drainage. Unfortunately, surgical drainage of hilar tumors is similarly associated with high failure and complication rates and thus does not offer a greater advantage for these lesions than it does for distal biliary malignancies. Brachytherapy. Stents provide palliation of jaundice for the majority of patients with bile duct tumors, but they do not affect tumor growth. Local extension may cause malignant ascites, vascular invasion, or portal vein occlusion. Tumor may grow in through the interstices of metal stents or overgrow the proximal or distal margins of a stent, leading to recurrent biliary obstruction. Brachytherapy offers yet another option for the management of patients with malignant tumors of the biliary tract. The technique involves endoscopic, percutaneous, or surgical placement of radioactive sources within the biliary tree, in close proximity to the tumor or across the malignant stricture. Typically, the radioisotope "seeds" are left within in the biliary lumen 24 to 72 h and then are removed. Brachytherapy has shown some effectiveness in prolonging survival of patients with localized bile duct carcinomas, particularly when used in combination with other therapies, such as externalbeam radiation or chemotherapy (56,57). The treatment is usually well tolerated and has few complications. Brachytherapy should be considered for patients with unresectable malignant biliary tumors who have good performance status and are expected to have prolonged survival. 4— Treatment of Malignant Obstruction: Overview The management of patients with malignant biliary obstruction requires several decision points (Fig. 5). First, it must be determined whether the patient is an operative candidate based on general health status and the extent of the malignant disease. An elderly patient or one with multiple comorbidities, such as cirrhosis or active cardiac or pulmonary disease, is expected to have a high operative risk. A comprehensive assessment for significant medical problems should be undertaken preoperatively, even in those with potentially resectable disease. Those with high performance status and radiological studies suggesting resectability should be offered surgery for cure. For the majority of patients with advanced disease, the goal of management is optimal palliation. Surgery, endoscopy, and percutaneous techniques all have potential roles. The greater early morbidity of surgical palliation should be weighed against the higher incidence of late complications with noninvasive techniques. Relatively fit patients may benefit most from surgically performed biliary and gastric bypass, giving them the potential for prolonged survival without stent exchanges or fear of gastric outlet obstruction. Those with poor Karnofski performance status or extensive metastases are unlikely to live very long, even if they tolerate the insults of surgery. These patients are best palliated by the least invasive approaches. Many patients are neither poor operative candidates nor particularly young and vigorous. For these, all treatment options should be offered and explained, with patient preference the deciding factor. Local technical expertise may also influence the choice of procedures. However, if a treatment is unavailable or not performed well at the particular hospital, the physician should consider referral to treatment centers with more experience with these techniques. B— Benign Biliary Strictures 1— Postoperative Biliary Stricture The most common cause of benign biliary stricture is surgical injury. Postoperative stricture is an uncommon complication of biliary surgery, but it is not rare. Most postoperative biliary
Page 854
Figure 5 Algorithm for management of malignant biliary strictures.
strictures occur after cholecystectomy. Other surgeries associated with bile duct strictures are gastrectomy, hepatic resection, and liver transplantation. Published series of patients treated with open cholecystectomy report an incidence of postoperative biliary strictures of 0.2 to 0.5% (58,59). Laparoscopic cholecystectomy was introduced in 1988 and virtually replaced open cholecystectomy as the firstline surgical technique for management for gallstones. Early experience with the laparoscopic approach was associated with a high rate of bile duct injury. However, more recent audits of laparoscopic complications show much lower rates of bile duct injury (60–62). When performed by experienced surgeons, laparoscopic cholecystectomy carries some risk of postoperative stricture, but the risk is not elevated compared with the open technique. Pathogenesis Postoperative bile duct strictures may form as a result of several factors. Partial transection or clipping of the common duct will lead to narrowing of the involved bile duct. Extensive dissection and use of cautery around the bile duct may also lead to a compromised blood supply or "skeletonization" of the biliary tree, with resultant ischemia (63). Even relatively minor irritation of the bile duct from thermal injury or mechanical trauma may cause fibrosis and scarring because of an initial intense acute inflammatory response. Finally, bile
Page 855
leakage may initiate a local acute inflammatory response in tissues adjacent to the bile duct, which may lead to stricture formation. Presentation Patients with postoperative biliary strictures may be asymptomatic for long periods of time or may develop nausea, abdominal pain, jaundice, or cholangitis. Some present only with cholestatic elevations in liver chemistries. Symptoms of postoperative biliary strictures have been reported to occur up to 30 years after surgery (64– 69). Rarely, a stricture remains unsuspected until the patient manifests signs of secondary biliary cirrhosis. The diagnosis is generally apparent when ultrasound or CT scan shows dilated bile ducts upstream from the injury. There is usually no mass, although inflammation or a localized biloma may confuse the issue. MRCP may be useful for noninvasive diagnosis, but is less reliable than direct cholangiography. For most cases of suspected bile duct injury, direct contrast injection provides the definitive information necessary to establish the diagnosis and plan treatment. Evaluation and Management of Postoperative Biliary Strictures Operative Management Major bile duct injuries such as complete transections or large lacerations are usually recognized intraoperatively. These complications should be managed surgically. The surgical approach in these situations is addressed in Chap. 36. However, the goal of operative management is to establish bile flow to the intestine via a tension free anastomosis between healthy tissue in the form of an endtoend repair, a RouxenY bilioenteric connection (hepaticojejunostomy or choledochojejunostomy), or a choledochoduodenostomy. Successful management of postoperative strictures requires the involvement of the surgeon, radiologist, and endoscopist. The initial evaluation should include cholangiography. If a T tube is in place, this may suffice to perform contrast injection. In most other circumstances, ERCP offers the best initial evaluation. ERCP will not provide visualization of the entire biliary tree if there is complete occlusion or transection of the duct. In this setting, percutaneous transhepatic cholangiography (PTC) or magnetic resonance cholangiopancreatography (MRCP) may be employed to visualize the proximal biliary tree. Once the location and nature of the injury is identified, treatment can be planned accordingly. No large, randomized trials have evaluated the optimal management of postoperative biliary strictures. As operative injury is a relatively uncommon problem, much of the data come from retrospective reviews of experience from tertiary care medical centers. Thus, results may be affected by selection, referral, and reporting biases. The largest series reporting on surgical management for benign strictures are from the prelaparoscopic era. The specific type of procedure performed depends on the location of the stricture. Successful outcomes (no symptoms, jaundice, or cholangitis) have been reported in 72 to 95% of cases (67,69–75). Surgical morbidity occurs in 20 to 30% and includes complications common to all abdominal operations as well as some specific to biliary repair such as anastomotic leaks. Surgical mortality rates have decreased to the range of 5% or less. Surgically managed strictures recur in 12 to 34% of cases after mean followup of up to 10 years (64,67,68,71,73,75). Stricture recurrence has been managed by repeat operation, which may be required in 25 to 35% of patients (73). In addition to stricture recurrence, bacterial reflux from the intestine and cholangitis complicate bilioenteric anastomoses. Therefore, traditional operative management of biliary strictures after conventional surgery is marked by relatively high morbidity. The longterm results of management of strictures related to laparoscopic cholecystectomy is less well defined. Recent experience with surgical repair of post laparoscopic cholecystectomy bile duct injuries is encouraging (76). Onethird of injuries were noted during the initial operation and managed at that time with open surgical reconstruction of the biliary tree. Of the remaining patients who presented after laparoscopic cholecystectomy, surgical repair led to complete resolution of symptoms in the majority. Although these initial results are promising, the followup is relatively brief, and other nonsurgical approaches may be equally beneficial in many situations.
Page 856
Nonoperative Management Percutaneous Transhepatic Balloon Dilatation and Stent Placement. Percutaneous treatment of postoperative strictures is achieved by gaining transhepatic access to the affected duct upstream from the stricture, crossing it with a guidewire, and then serially dilating the stricture with balloons. Several balloon inflations are usually performed, which are generally tolerated under conscious sedation but occasionally require general anesthesia due to pain (77–79). The patient is typically left with an external catheter and the procedure is repeated several times over a 5 to 7day period. Most catheters are subsequently removed, although in some patients the externalized catheter is left in place for up to 6 months to provide access for further treatment (78). Success rates for percutaneous management of postoperative strictures vary widely, depending on the series. At followup 3 to 5 years of after dilatation, 64 to 85% of post operative strictures remained (71,74,76–78,80–84). While generally considered less invasive than surgical intervention, percutaneous treatment appears to be associated with a higher rate of hemobilia and septic complications. When strictures recur after dilation, longterm stenting may be necessary. As for stenting of malignant strictures, clogging remains the limiting factor and necessitates exchange in most patients. Routine exchange via the percutaneous route requires maintenance of external access catheters, which can be bothersome to patients. Self expanding metal stents have been placed percutaneously to manage postoperative biliary strictures in some patients in an effort to prolong patency (85). Ingrowth of hyperplastic biliary epithelium occurs commonly, leading to eventual stent occlusion and necessitating further intervention. Metal stents cannot be removed and thus should not be placed in benign strictures except in cases where the patient has failed other nonoperative treatments and is at prohibitively high surgical risk. Endoscopic Management. Endoscopic balloon dilatation followed by stent insertion is now a wellestablished treatment for postoperative biliary strictures. This technique avoids the complications associated with transhepatic punctures, such as hemobilia and bile leakage, and is associated with a lower incidence of sepsis. Patient discomfort is minimized with endoscopic instrumentation, and the metabolic derangements and inconvenience associated with external catheters and drainage are avoided. Most often, a sphincterotomy is performed to facilitate the placement of multiple stents in the bile duct simultaneously. This use of parallel stents after balloon dilation maintains a larger diameter than would be possible with single stents. The period of stenting is often 6 months to 1 year; thus several procedures for endoscopic stent exchange are typically required. Experience with endoscopic management of benign postoperative strictures is less extensive than that reported from the surgical literature (68,86–89). In the two largest series (68,89), strictures were balloondilated and then stented for periods of 6 to 12 months. Patients typically required stent exchange five times during this period. Three to five years after ultimate stent removal, 74 to 83% of patients remained symptomfree without any sign of recurrence. Complications and success rates were felt to be equivalent to those with surgical management. Endoscopic treatment of benign strictures with expandable metal stents has been reported (86). Despite the absence of tumor to grow in through the mesh, tissue ingrowth due to reactive biliary epithelial hyperplasia eventually occurred in all patients (Fig. 6). Because metal stents are irretrievable, they cannot be exchanged when occluded. Patency can usually be maintained by placing additional stents inside the original one, but the permanence of the metal stent contributes further complexity to the stricture. At present, there is little evidence to suggest that metal stents offer significant advantages over plastic stents for benign biliary disease, and their use is discouraged in all but exceptional circumstances. Management of Postoperative Biliary Strictures: A Summary The most extensive experience with treatment of postoperative biliary strictures comes from the surgical literature. Studies of patients treated with endoscopic or percutaneous methods involve smaller numbers with shorter followup (Table 3). Nevertheless, experience with nonoperative management is growing, and longer periods of surveillance have not yielded higher recurrence rates
Page 857
Figure 6 Cholangiogram demonstrating tissue ingrowth into metal stent placed for benign disease.
than those seen with surgery. Currently, there is no convincing evidence that surgical outcomes are superior to those achieved less invasively. A multidisciplinary approach to the management of postoperative biliary strictures is advisable (90). In general, complete transections of the bile duct or complete occlusion due to clip or suture require surgical repair, typically with hepaticojejunostomy. Those patients whose strictures may be traversed by guidewires are candidates for initial nonoperative therapy with balloon dilatation and stenting. If strictures recur after a reasonable attempt at nonoperative management, surgery can still be offered. 2— Bile Duct Strictures after Liver Transplantation The biliary tract has been considered the "Achilles heel" of orthotopic liver transplantation (OLT). Injury to the bile duct occurs in 11 to 35% of patients undergoing transplantation, and this problem accounts for up to onefourth of all operative complications in the first posttransplant year (91,92). Posttransplant strictures present in a similar fashion to those seen in nontransplant patients. Cholestatic elevation of the liver chemistries is usually the first manifestation. These changes may be accompanied by abdominal pain and signs of cholangitis. However, it may be difficult to distinguish cholestasis caused by strictures from that caused by hepatic allograft rejection, cytomegalovirus (CMV) infection, or recurrent viral hepatitis. Etiology At the time of liver transplantation, anastomosis of the common duct is usually accomplished with an endtoend choledochocholedochostomy. RouxenY choledochojejunostomy is performed for patients with primary sclerosing cholangitis or those with extensive prior biliary surgery. Anastomotic strictures occur at the site of choledochocholedochostomy or the choledochojejunostomy, while nonanastomotic strictures involve the donor hepatic ducts or the intrahepatic ducts (93,94). The etiology of postOLT biliary strictures is multifactorial. Possible causes include technical errors, ischemia, infection, chronic rejection, or prolonged cold ischemia time of the graft. Because the collateral blood supply to the bile duct is interrupted when the liver is harvested, the donor bile duct relies solely on perfusion from the hepatic artery and is highly vulnerable to ischemia due to hepatic artery thrombosis or stenosis (95–102). Prolonged cold ischemia of the donor organ prior to transplantation is associated with increased risk of graft loss and may contribute to injury to the donor duct (103). Other potential causes of postOLT biliary strictures
Page 858 Table 3 Treatment of Benign Postoperative Stricturesa
N
Study Surgery
Longterm Mean followup patency rate (months) (%)
Pitt (73)
66
86
60
Pelligrini (75)
60
78
102
Genest (71)
105
82
60
Innes (70)
22
95
72
Pitt (74)
25
88
57
122
76
86
35
83
50
194
79
111
629
81
84
Chapman (72) Davids (69) Frattoroli (67) Total surgery Percutaneous
Vogel (79)
13
85
24
Mueller (78)
28
76
36
Williams (81)
64
78
28
Moore (77)
18
83
33
Pitt (74)
20
55
59
25
64
28
168
74
33
Lillemoe (76) Total percutaneous Endoscopy
Foutch (87)
9
55
6
Davids (68)
66
83
42
Geenen (88)
25
88
48
Berkelhammer (89)
25
74
19
27
81
44
152
80
37
Dumonceau (86) Total endoscopy a
Excluding postliver transplant.
include infection with opportunistic organisms such as CMV (104), chronic rejection, ABO incompatibility, and sphincter of Oddi dysfunction (105). Evaluation The first step in evaluating abnormal liver chemistries in the transplant patient is often a liver biopsy. If cholestasis is apparent or the diagnosis is unclear, the next step is to image the bile duct directly. The approach to cholangiography depends on the presence or absence of an indwelling T tube and on the type of surgical bile duct anastomosis. If a T tube remains in place, a cholangiogram can be obtained by instilling radiopaque contrast through the tube. For patients without direct biliary access, endoscopic cholangiography remains the preferred approach, although MRCP may soon replace diagnostic ERCP due to its lower morbidity. It is technically difficult to perform ERCP in a patient with RouxenY choledochojejunostomy; thus, a percutaneous transhepatic approach to cholangiography may be required for patients with this anastomosis. Management Like treatments for other forms of postoperative stricture, those for postOLT strictures should reestablish adequate bile flow to relieve cholestasis as well as prevent cholangitis, choledocholithiasis, and secondary biliary cirrhosis. Traditionally, the surgical approach has been favored, with bypass of the stricture using hepatico or choledochojejunostomy
Page 859
(106–108). Liver transplant patients are often severely compromised in the early postoperative period, suffering from malnutrition, immunosuppression, and impaired wound healing. Thus, morbidity and mortality rates for reoperation are high (109), and nonoperative avenues of therapy are increasingly utilized. Endoscopic treatment methods are similar to those applied to other benign strictures. Dilation and stenting are performed during the initial ERCP. Some anastomotic lesions detected early in the postoperative period are merely inflammatory stenoses and will resolve once the edema subsides; these are best managed with brief periods of stenting. For patients with true fibrotic strictures, repeated dilation sessions and multiple stents are typically required. Largebore stents should be placed and exchanged electively every 2 to 4 months. Strictures that fail to improve following dilation should be held open with multiple parallel stents between dilation sessions (Figs. 7 and 8). Data concerning endoscopic management for postOLT strictures have been culled primarily from nonrandomized retrospective reviews, and followup has been limited. Nevertheless, it appears that more than 70% of patients treated endoscopically will have ultimate resolution of the stricture without any need for surgery or further interventions (110–113). Patients with intrahepatic (donor duct) lesions generally fare less well than those with anastomotic strictures (110). Percutaneous transhepatic therapy is appropriate for patients who lack endoscopic access to the biliary tree due to choledochojejunal anastomosis or other surgical limitations. The percutaneous route may also be preferred for those patients with proximal intrahepatic biliary strictures, particularly if these cannot be crossed via the endoscopic approach. Because of higher rates of complications, patient discomfort, and prolonged hospitalization associated with percutaneous therapy, endoscopic treatment should remain the initial method attempted in all others. PostOLT bile duct strictures are a relatively common problem, affecting both graft and patient survival. The management of these complications requires the cooperation of surgeons, endoscopists, and interventional radiologists. Given the relatively high rate of morbidity and mortality associated with surgical repair of these strictures, nonoperative management should
Figure 7 Posttransplant anastomotic biliary stricture.
Page 860
Figure 8 Posttransplant stricture following course of endoscopic biliary dilatation.
be attempted first. The endoscopic approach is preferred for most patients, with PTC employed for those with RouxenY choledochojejunostomies. Reoperation should be reserved for patients who fail to respond to less invasive treatments. The majority of patients will have resolution of the stricture with nonoperative means. 3— Common Bile Duct Stricture Due to Chronic Pancreatitis Incidence Chronic pancreatitis leads to progressive fibrosis and scarring of the pancreas. As the common bile duct (CBD) traverses the pancreas in its distal segment, chronic pancreatitis may lead to stricture of the bile duct secondarily. Pancreatic pseudocysts may also cause biliary obstruction due to extrinsic compression. The reported incidence of CBD stricturing due to chronic pancreatitis is 3 to 46% (114–118). Most patients with CBD strictures have pancreatitis as a result of alcohol abuse (118). Clinical Presentation The clinical manifestations of biliary strictures associated with chronic pancreatitis are varied. Many CBD strictures are clinically silent and are discovered incidentally at the time of ERCP performed for evaluation of pancreatitis. An isolated elevated alkaline phosphatase is observed in 63 to 100% of cases, and the serum bilirubin is intermittently elevated in 53 to 100% (116,119–121). However, these biochemical parameters are not reliable indicators of the severity of CBD obstruction, as partial obstruction may lead to significant blood test abnormalities (120,121). Patients may experience transient episodes of jaundice with dark urine and acholic stools. Others have abdominal pain with jaundice or signs of cholangitis such as fever. The natural history of the stricture may depend on whether it is due to chronic pancreatic scarring, compression by a pancreatic pseudocyst, or acute pancreatic edema in the setting of underlying chronic pancreatitis. Longterm complications of CBD strictures due to chronic pancreatitis include intermittent episodes of cholangitis or sepsis and secondary biliary cirrhosis (2,116,117,121,122). Cholangitis occurs in approximately 9% of patients and secondary biliary
Page 861
cirrhosis in approximately 7% (115). It is difficult to predict which patients will develop these complications, however. Biliary cirrhosis can develop within 1 year of diagnosis or up to 10 years later (1,123). Some authors advocate therapeutic intervention when the alkaline phosphatase is twice the upper limit of normal (119), but levels of alkaline phosphatase or bilirubin often do not correlate with the severity or extent of the stricture. Diagnosis A biliary stricture due to chronic pancreatitis often appears cholangiographically as a long, smooth, tapered narrowing in the intrapancreatic portion of the CBD (124) (Fig. 9). Occasionally, there is a shelflike appearance to the duct, making it difficult to differentiate from a malignant stricture. Although typically considered a finding of malignancy, a "doubleduct" stricture in both the CBD and pancreatic duct may be benign (125). Brush cytology has a poor sensitivity and does not reliably distinguish between a benign and a malignant stricture when negative. Positive malignant cytology is definitive, however; thus brushings should usually be obtained from the pancreatic and bile ducts at the time of ERCP. Management The management of biliary obstruction due to chronic pancreatitis remains controversial. Because of the possibility of cholangitis and secondary biliary cirrhosis, many authors advocate definitive management with surgical bypass whenever a stricture is encountered. The most common operations performed include choledochoduodenostomy, choledochojejunostomy, and RouxenY hepaticojejunostomy (2,114,117,126–128). Surgical morbidity and mortality have declined in the last decade. In the late 1970s and early 1980s surgical inhospital mortality averaged 8.3% (129). Operative mortality for biliary bypass procedures is now reported to be in the range of 0 to 5% (74,130,131). Morbidity remains high, however, averaging around 20% (130). Nonoperative management of CBD strictures due to chronic pancreatitis includes stricture dilatation with dilating balloons and biliary stenting. Both endoscopic and percutaneous approaches are feasible; selection often depends on local expertise and experience. Endoscopically placed internal stents are preferable to percutaneous techniques due to reduced patient discomfort and less risk of intrahepatic bleeding. Endoscopically placed biliary endoprostheses have been evaluated in several series (Table 4). The endoscopic management of CBD strictures related to chronic pancreatitis often does not provide definitive therapy. Placement of a plastic stent will resolve symptoms of jaundice and cholangitis and will usually cause a reduction in cholestatic liver chemistries, commonly within 2 weeks (124,132–134). However, the alkaline phosphatase does not completely normalize in most cases. Stents are often left in place for 10 to 14 months, being exchanged at
Figure 9 Common bile duct stricture due to chronic pancreatitis.
Page 862 Table 4 Plastic Stenting of Biliary Strictures Secondary to Chronic Pancreatitis
Study Deviere (132)
Clinical improvement
n 19
19
Barthet (124)
19
19
Smits (133)
58
58
Itani (134) Total
a
5 101
NR
Stent dysfunction 18
Median duration of stenting (months)
Permanent stent removal
3
NR
1
3
10
40
16
10
1
4
60
26
6
NR = Not reported. Source: From Ref. 119. a
3 to 4month intervals. Stent migration or clogging occurs in a large number of patients, necessitating these frequent exchanges. Endoscopic stenting may result in complete resolution of the stricture in 15 to 28% of patients (124,132–134). However, most patients either require longterm stenting or surgical bilioenteric bypasses. Selfexpandable metal Wallstents are used extensively in the management of malignant biliary strictures. Their use in benign biliary strictures is much less common but has been reported (135–137). Most patients who received metal stents for benign disease either refused surgery or surgery was felt to be contraindicated for medical reasons. The use of metal stents for biliary stricture due to chronic pancreatitis has been reported in fewer than 30 patients (135–137). Metal stents provided similarly rapid resolution of symptoms of cholangitis and jaundice as well as reductions in cholestatic liver chemistries. After almost 3 years of followup in one study (136), the majority of stents (90%) remained patent. Stents in this study were of short length so as not to preclude future hepaticojejunostomy or choledochojejunostomy. Extensive epithelial hyperplasia between the metal struts causes recurrent obstruction in a small number of cases. Although encouraging, these results should be considered preliminary. The use of metal stents for benign CBD strictures associated with chronic pancreatitis should be discouraged until more data are obtained as to their longterm effectiveness and safety. Predicting the need for decompression in an asymptomatic patient with common duct stricture due to chronic pancreatitis remains difficult. In symptomatic patients, surgical biliary bypass remains the most reliable therapy. Surgical treatment is particularly favored when it is performed at the time of surgery for associated complications related to chronic pancreatitis, such as gastric bypass for duodenal stenosis or pancreatic ductal decompression for chronic pain. Endoscopically or percutaneously placed stents may provide immediate relief and allow time for a decision to be made regarding the most suitable longterm therapy. Nonoperative therapy may also provide time for acceptable pancreatic duct dilatation so that a combined biliary and pancreatic bypass procedure may be performed. Endoscopic or percutaneous biliary drainage provides longterm relief of the biliary stricture in only a minority of patients (119). One potential approach to management would be a trial of endoscopic therapy. If, after 3 to 6 months of stenting, the stricture persists, future management should depend on an assessment of the patient's surgical risks and preferences. In poor surgical candidates, a plastic or selfexpanding metal stent is a reasonable longterm alternative. Asymptomatic patients with a CBD stricture due to chronic pancreatitis pose a particular therapeutic challenge. Potential complications include cholangitis and secondary biliary cirrhosis, but it is difficult to predict which patient will experience these problems. Patients with either a dilated bile duct or a persistent elevation in the alkaline phosphatase (greater than
Page 863
twice the upper limit of normal) are at greater risk for developing complications and may warrant intervention (121,138). C— Primary Sclerosing Cholangitis 1— Etiology and Epidemiology Primary sclerosing cholangitis (PSC) is predominantly a disease of young or middleaged men. Males are affected three times more often than females, and age at diagnosis is typically between 25 and 45 years. Some 75% of patients with PSC have inflammatory bowel disease of the colon, with 87% of these having ulcerative colitis. However, only 5% of patients with inflammatory bowel disease will develop PSC (139). In the past two decades, PSC has been diagnosed with much greater frequency. This is probably not due to an increased incidence of the disease. Rather, it reflects increased clinician awareness of PSC as well as improvements in cholangiographic techniques that facilitate making the diagnosis. PSC is a rare disease, appearing with an incidence of 1 to 6 cases per 100,000 population. Nevertheless, it remains the fourth leading indication for liver transplantation among adults in the United States (140). PSC is an inflammatory disease of the intrahepatic and extrahepatic bile ducts. Inflammation leads to scarring; over time, the bile ducts become irregularly narrowed and obliterated. Despite the striking association between PSC and inflammatory bowel disease, the cause of these ductal lesions remains unknown. It has been postulated that bacteria or toxins enter the portal circulation from the inflamed colon and damage the bile ducts. However, the natural history of PSC argues against a major pathogenic role for the bowel (139). Some patients develop PSC long before they show signs of inflammatory bowel disease, while others develop PSC many years after their ulcerative colitis has been eradicated by proctocolectomy. The most plausible explanation is that certain factors underlying the immune dysregulation responsible for inflammatory bowel disease can also cause immunemediated attacks on large bile ducts (141). Strictures lead to biliary obstruction and cholestasis, which usually progresses to chronic liver disease. Episodes of cholangitis occur when strictured ducts become infected with bacteria, and gallstones may form within the ducts, contributing to obstruction. The natural history of the disease is characterized by intermittent episodes of pruritus, jaundice, and cholangitis. There is an increased risk of cholangiocarcinoma. Most patients eventually develop cirrhosis and require liver transplantation (142,143). 2— Diagnosis The diagnosis of PSC is suggested by the finding of cholestatic liver chemistries in a patient with the appropriate clinical history. Serum antineutrophil cytoplasmic antibodies (ANCA) are positive in only 65% of patients with PSC and are nonspecific (144,145). Liver biopsy is supportive of the diagnosis of PSC but is rarely diagnostic. Visualization of the biliary tract is required to make a definitive diagnosis of PSC. Noninvasive techniques such as ultrasound and computed tomography (CT) may not detect sclerosing cholangitis, as dilatation of ducts upstream from strictures may be absent due to fibrosis of the duct walls. Although magnetic resonance techniques may prove useful in the future, cholangiography is most often obtained via ERCP. Percutaneous cholangiography is technically difficult in PSC because peripheral bile ducts are often fibrotic and nondilated or decreased in number. Cholangiographic findings are characteristic and include multifocal strictures and dilatations, usually involving both intrahepatic and extrahepatic ducts (Fig. 10). In the early stages, the only cholangiographic abnormality may be fine ulcerations of the bile duct, similar to those seen in the colon in early ulcerative colitis (Fig. 11). Diffuse strictures with short intervening segments of normal or dilated bile ducts produce the classic beaded appearance. The gallbladder and cystic duct are involved in as many as 15% of patients (146).
Page 864
Figure 10 Primary sclerosing cholangitis primarily involving the intrahepatic biliary tree.
3— Management Primary sclerosing cholangitis is a progressive disease that ultimately results in liver failure for most patients. Lesions are typically multiple and involve the small intrahepatic radicals as well as large extrahepatic ducts. Intervening for individual strictures may seem futile in the setting of diffuse disease, but when a single extrahepatic lesion can be identified as a flowlimiting ''dominant" stricture, treatment to relieve obstruction at that location may be appropriate. Strictures arising from PSC can be dilated, stented, or bypassed using techniques developed for treating strictures caused by other disorders. However, there are specific features of PSC that should be considered when the best mode of treatment is being selected.
Figure 11 Primary sclerosing cholangitis with fine ulcerations in common bile duct.
Page 865
Surgery Surgical procedures have been employed to relieve biliary obstruction at a dominant stricture, including creation of proximal bilioenteric anastomosis and resection of the diseased distal duct (147–153). Performance of anastomoses may be technically difficult because of lack of upstream biliary dilation in many patients with PSC, and these open procedures are associated with significant morbidity (147). Nonetheless, one large series reported improved transplantfree survival in precirrhotic patients managed surgically as compared with those treated nonoperatively (153). The authors argued that this benefit was due at least in part to a reduction in the development of cholangiocarcinoma attributed to the removal of the extrahepatic biliary tree. It is difficult to separate out the potential for selection bias in this study, however, and it is likely that less fit candidates were managed nonoperatively. Aside from the technical difficulties and immediate morbidity associated with operation for PSC, there are additional reasons to avoid surgery. Creation of a wide bilioenteric anastomosis allows for chronic reflux of bacteria from the bowel up into the biliary tree, leading to increased potential for bacterial cholangitis. Perhaps most importantly, surgery to the region of the porta hepatis can lead to adhesions or vascular injury that may interfere with or even prohibit liver transplantation (148,149). Thus, surgical management should not be considered firstline therapy for strictures in PSC. Nonoperative Management Whenever biliary intervention is undertaken in a patient with PSC, there are risks for bacterial contamination upstream of intrahepatic strictures, injury to blood vessels or bile ducts affecting subsequent transplantability, and hepatic decompensation due to underlying liver disease. In the absence of symptoms such as pruritus, jaundice, or active cholangitis, the best approach may be to avoid invasive interventions altogether. Once symptoms appear, however, intervention is usually required and the least invasive technique is preferred. In most circumstances the best initial approach is with ERCP. Endoscopic biliary access is achieved as for ERCP performed for other indications. Sphincterotomy should be avoided when possible, as PSC patients are at increased risk for duodenobiliary reflux, leading to bacterial cholangitis. Strictures may be quite tight and fibrotic, and flexible hydrophilic guidewires are generally required to cross these lesions. To reduce the risk of introducing infection, special care must be taken to avoid overinjection of contrast, particularly into intrahepatic ducts or areas of diffuse stricturing. Isolated dominant strictures of the extrahepatic ducts are best treated by endoscopic balloon dilatation (Fig. 12). In contrast to strictures caused by malignancy or operative injury, strictures in PSC are caused by deposition of scar tissue from chronic inflammation. This situation is analogous to that of peptic strictures of the esophagus, for which dilation by balloon or bouginage is usually sufficient to maintain luminal patency for relatively long periods. Similarly, dilation of biliary strictures in PSC usually results in a significant period of patency without the need for stenting. It may be necessary to begin with a rigid step dilator, but balloon dilation should follow in order to achieve sufficient diameter for a sustained response. Restenosis may occur, particularly if the periductal inflammation continues unchecked, but this process typically takes many months. Plastic stents may be left to assure patency following stricture dilation or to prevent restenosis. They must be exchanged every 3 to 6 months, however, and because they keep the sphincter of Oddi open and facilitate reflux, they may increase the risk of bacterial cholangitis. Some authors have routinely used stents in PSC (154– 160), but we prefer to reserve them for cases of particularly tight stenoses or when there is poor drainage of contrast following dilation. Several published series describe potential benefits of endoscopic biliary dilatation or stent insertion for PSC (154–159,161,162). Although results have been reproducible, each study has been relatively small and lacked a randomized control group. Reports have highlighted reduced hospitalizations for cholangitis, improvement in laboratory parameters, improvement in the radiographic appearance of the bile ducts, and symptomatic improvement in patients undergoing endoscopic therapy. Common to all series of endoscopic therapy is the finding that several sessions of treatment may be necessary before an adequate response is achieved.
Page 866
Figure 12 a. "Dominant" stricture in common hepatic duct in patient with primary sclerosing cholangitis. b. Endoscopic balloon dilation of dominant stricture. c. Cholangiographic appearance after dilation.
Percutaneous transhepatic cholangiography (PTC) may be attempted if ERCP is unsuccessful. PTC can be technically difficult in sclerosing cholangitis and is associated with a higher risk of complications. Once access is achieved, similar methods of stenting and balloon dilation are appropriate. If repeated sessions are necessary, a combined endoscopicpercutaneous technique may be preferable. Despite the lack of a randomized, controlled trial, it is apparent that many patients with dominant strictures experience symptomatic improvement following dilation or stenting. It remains unclear whether this treatment affects the natural history of PSC and whether such intervention is warranted in the asymptomatic patient. Extrahepatic biliary intervention is unlikely to have beneficial effects upon the sclerotic process affecting smaller intrahepatic ducts. Thus, it is doubtful that such therapy can prevent liver failure or forestall the need for transplantation indefinitely. Perhaps by alleviating extrahepatic obstruction, endoscopic or percutaneous therapy can decrease the deleterious effects of subclinical cholestasis and slow the progression to cirrhosis, but data are currently lacking.
Page 867
Figure 12 Continued.
VII— Conclusion The management of both malignant and benign biliary strictures should involve the coordinated efforts of an endoscopist, interventional radiologist, and surgeon. Surgical bypass of strictures remains an important option in the management of both benign and malignant strictures. However, continued advances in percutaneous and endoscopic techniques provide valuable alternatives in the treatment of these patients. Nonoperative techniques are also important in treating acute problems, such as cholangitis, prior to definitive management. If expertise in particular endoscopic or surgical techniques is not available, one should consider transferring the patient to a center where these options are accessible. Which management option is optimal depends on the clinical situation and patient characteristics. References 1. Gregg J, CarrLocke D, Gallagher M. Importance of common bile duct stricture associated with chronic pancreatitis. Am J Surg 1981; 141:199–203. 2. Warshaw AL, Schapiro RH, Ferrucci JT, Galdabini JJ. Persistent obstructive jaundice, cholangitis, and biliary cirrhosis due to common bile duct stenosis in chronic pancreatitis. Gastroenterology 1976; 70:562–567. 3. Ishizaki Y, Wakayama T, Okada Y, Kobayashi T. Magnetic resonance cholangiography for evaluation of obstructive jaundice. Am J Gastroenterol 1993; 88:2072–2077. 4. Bearcroft PW, Lomas DJ. Magnetic resonance cholangiopancreatography. Gut 1997; 41:135–137. 5. Kay CL, Aabakken LE, Tarnasky P, et al. Magnetic resonance cholangiopancreatography: a problem solving modality. Gastrointest Endosc 1997; 46:363–366. 6. Landis S, Murray T, Bolden S, Wingo P. Cancer Statistics, 1998. CA 1998; 48:6–30. 7. Lillemoe K, Sauter P, Pitt H, Yeo C, Cameron J. Current status of surgical palliation of periampullary carcinoma. Surg Gynecol Obstet 1993; 176:1–10. 8. Smith AC, Dowsett JF, Russell RC, Hatfield AR, Cotton PB. Randomised trial of endoscopic stenting versus surgical bypass in malignant low bile duct obstruction. Lancet 1994; 344:1655–1660.
Page 868
9. Baker AR, Neoptolemos JP, CarrLocke DL, Fossard DP. Sump syndrome following choledochoduodenostomy and its endoscopic treatment. Br J Surg 1985; 72:433–435. 10. Khan TF, Sherazi ZA, Muniandy S, Mumtaz M. Recurrent pyogenic cholangitis: "sump syndrome" following choledochoduodenostomy. Trop Doct 1997; 27:51– 52. 11. Morrissey PE, Burns GA, Cohn SM. Sump syndrome complicating RouxenY hepaticojejunostomy: case report and review of the literature. Surgery 1996; 119:403–405. 12. Speer AG, Cotton PB, Russell RC, et al. Randomised trial of endoscopic versus percutaneous stent insertion in malignant obstructive jaundice. Lancet 1987; 2:57–62. 13. Loperfido S, Angelini G, Benedetti G, et al. Major early complications from diagnostic and therapeutic ERCP: a prospective multicenter study (see comments). Gastrointest Endosc 1998; 48:1–10. 14. Miller DL, Haines GA, Schmetz MA, Ghosh BC. Percutaneous biliary stents for palliation of hilar malignancies. J Surg Oncol 1996; 62:226–227. 15. Chang W, Kortan P, Haber G. Outcome in patients with bifurcation tumors who undergo unilateral versus bilateral hepatic duct drainage. Gastrointest Endosc 1998; 47:354–362. 16. Libby E, Leung J. Prevention of biliary stent clogging: a clinical review. Am J Gastroenterol 1996; 91:1301–1308. 17. Speer AG, Cotton PB, MacRae KD. Endoscopic management of malignant biliary obstruction: stents of 10 French gauge are preferable to stents of 8 French gauge. Gastrointest Endosc 1988; 34:412–417. 18. Matsuda Y, Shimakura K, Akamatsu T. Factors affecting the patency of stents in malignant biliary obstructive disease: univariate and multivariate analysis. Am J Gastroenterol 1991; 86:843–849. 19. Davids PH, Groen AK, Rauws EA, Tytgat GN, Huibregtse K. Randomised trial of selfexpanding metal stents versus polyethylene stents for distal malignant biliary obstruction. Lancet 1992; 340:1488–1492. 20. Shepherd HA, Royle G, Ross AP, Diba A, Arthur M, ColinJones D. Endoscopic biliary endoprosthesis in the palliation of malignant obstruction of the distal common bile duct: a randomized trial. Br J Surg 1988; 75:1166–1168. 21. Knyrim K, Wagner HJ, Pausch J, Vakil N. A prospective, randomized, controlled trial of metal stents for malignant obstruction of the common bile duct. Endoscopy 1993; 25:207–212. 22. Sung J, Lai C, Chung S. Omitting side holes in biliary stents does not improve drainage of the obstructed biliary system: a prospective randomized trial. Gastrointest Endosc 1994; 40:321–325. 23. Barrioz T, Ingrand P, Besson I, de Ledinghen V, Silvain C, Beauchant M. Randomised trial of prevention of biliary stent occlusion by ursodeoxycholic acid plus norfloxacin (see comments). Lancet 1994; 344:581–582. 24. Ghosh S, Palmer K. Prevention of biliary stent occlusion using cyclical antibiotics and ursodeoxycholic acid. Gut 1994; 35:1757–1759. 25. Chae FH, Stiegmann GV. Current laparoscopic gastrointestinal surgery. Gastrointest Endosc 1998; 47:500–511. 26. Gudjonsson B. Cancer of the pancreas: 50 years of surgery. Cancer 1987; 60:2284–2303. 27. Sarr MG, Cameron JL. Surgical palliation of unresectable carcinoma of the pancreas. World J Surg 1984; 8:906–918. 28. Ryan ME. Cytologic brushings of ductal lesions during ERCP. Gastrointest Endosc 1991; 37:139–142. 29. Scudera PL, Koizumi J, Jacobson IM. Brush cytology evaluation of lesions encountered during ERCP. Gastrointest Endosc 1990; 36:281–284. 30. Schoefl R, Haefner M, Wrba F, et al. Forceps biopsy and brush cytology during endoscopic retrograde cholangiopancreatography for the diagnosis of biliary stenoses. Scand J Gastroenterol 1997; 32:363–368.
Page 869
31. McGuire DE, Venu RP, Brown RD, Etzkorn KP, Glaws WR, AbuHammour A. Brush cytology for pancreatic carcinoma: an analysis of factors influencing results. Gastrointest Endosc 1996; 44:300–304. 32. Warshaw A, Gu Z, Wittenberg J, Waltman A. Preoperative staging and assessment of resectability of pancreatic cancer. Arch Surg 1990; 125:230–233. 33. Cotton P. Nonsurgical palliation of jaundice in pancreatic cancer. Surg Clin North Am 1989; 69:613–627. 34. Speer AG, Cotton PB. Endoscopic treatment of pancreatic cancer. Int J Pancreatol 1988; 3:S147–S158. 35. Andersen JR, Sorensen SM, Kruse A, Rokkjaer M, Matzen P. Randomised trial of endoscopic endoprosthesis versus operative bypass in malignant obstructive jaundice. Gut 1989; 30:1132–1135. 36. Bornman PC, HarriesJones EP, Tobias R, Van Stiegmann G, Terblanche J. Prospective controlled trial of transhepatic biliary endoprosthesis versus bypass surgery for incurable carcinoma of head of pancreas. Lancet 1986; 1:69–71. 37. Dowsett J, Russell R, Hatfield A, et al. Malignant obstructive jaundice: a prospective randomized trial of bypass surgery versus endoscopic stenting. Gastroenterology 1989; 96:128A. 38. Brandabur J, Kozarek R, Ball T, et al. Nonoperative versus operative treatment of obstructive jaundice in pancreatic cancer: cost and survival analysis. Am J Gastroenterol 1988; 83:1132–1138. 39. Huibregtse K, Katon RM, Coene PP, Tytgat GN. Endoscopic palliative treatment in pancreatic cancer. Gastrointest Endosc 1986; 32:334–338. 40. Rhodes M, Nathanson L, Fielding G. Laparoscopic biliary and gastric bypass: a useful adjunct in the treatment of carcinoma of the pancreas. Gut 1995; 36:778– 780. 41. Feretis C, Benakis P, Dimopoulos C, Manouras A, Tsimbloulis B, Apostolidis N. Duodenal obstruction caused by pancreatic head carcinoma: palliation with self expandable endoprostheses. Gastrointest Endosc 1997; 46:161–165. 42. Freeman ML, Cass OW. Interlocking expandable metal stents for simultaneous treatment of malignant biliary and duodenal obstruction (letter). Gastrointest Endosc 1996; 44:98–99. 43. Howden CW, Woods BL. Selfexpanding metal stents for palliative treatment of malignant biliary and duodenal stenoses. Gastrointest Endosc 1995; 42:104– 105. 44. CarrLocke D, Ball T, Connors P, et al. Multicenter, Randomized trial of Wallstent biliary endoprosthesis versus plastic stents. Gastrointest Endosc 1993; 39:A310. 45. Cho Y, Lee Y, Kim J, et al. Plastic endoprosthesis versus metal stents in the palliative treatment of malignant biliary obstruction (abstr). Gastrointest Endosc 1997; 45:AB126. 46. Lammer J, Hausegger KA, Fluckiger F, et al. Common bile duct obstruction due to malignancy: treatment with plastic versus metal stents. Radiology 1996; 201:167–172. 47. Wagner HJ, Knyrim K, Vakil N, Klose KJ. Plastic endoprostheses versus metal stents in the palliative treatment of malignant hilar biliary obstruction: a prospective and randomized trial. Endoscopy 1993; 25:213–218. 48. Prat F, Chapat O, Ducot B, et al. A randomized trial of endoscopic drainage methods for inoperable malignant strictures of the common bile duct. Gastrointest Endosc 1998; 47:1–7. 49. Alexander F, Rossi RL, O'Bryan M, Khettry U, Braasch JW, Watkins E Jr. Biliary carcinoma: a review of 109 cases. Am J Surg 1984; 147:503–509. 50. Bismuth H, Castaing D, Traynor O. Resection or palliation: priority of surgery in the treatment of hilar cancer. World J Surg 1988; 12:39–47. 51. Blumgart LH, Hadjis NS, Benjamin IS, Beazley R. Surgical approaches to cholangiocarcinoma at confluence of hepatic ducts. Lancet 1984; 1:66–70. 52. Boerma EJ. Research into the results of resection of hilar bile duct cancer. Surgery 1990; 108:572–580.
Page 870
53. Scapa E, Peer A, Witz E, Eshchar J. "Rendezvous" procedure (RVP) for obstructive jaundice. Surg Laparosc Endosc 1994; 4:82–85. 54. Deviere J, Baize M, de Toeuf J, Cremer M. Longterm followup of patients with hilar malignant stricture treated by endoscopic internal biliary drainage. Gastrointest Endosc 1988; 34:95–101. 55. Polydorou AA, Cairns SR, Dowsett JF, et al. Palliation of proximal malignant biliary obstruction by endoscopic endoprosthesis insertion. Gut 1991; 32:685–689. 56. Erickson B, Wilson JF. Clinical indications for brachytherapy. J Surg Oncol 1997; 65:218–227. 57. Leung JT, Kuan R. Intraluminal brachytherapy in the treatment of bile duct carcinomas. Australas Radiol 1997; 41:151–154. 58. Rosenquist H, Myrin S. Operative injury to the bile ducts. Acta Chir Scand 1960; 119:92–107. 59. Roslyn JJ, Binns GS, Hughes EF, SaundersKirkwood K, Zinner MJ, Cates JA. Open cholecystectomy: a contemporary analysis of 42,474 patients. Ann Surg 1993; 218:129–137. 60. Airan M, Appel M, Berci G, et al. Retrospective and prospective multiinstitutional laparoscopic cholecystectomy study organized by the Society of American Gastrointestinal Endoscopic Surgeons (see comments). Surg Endosc 1992; 6:169–176; discussion 177–178. 61. Deziel DJ, Millikan KW, Economou SG, Doolas A, Ko ST, Airan MC. Complications of laparoscopic cholecystectomy: a national survey of 4,292 hospitals and an analysis of 77,604 cases. Am J Surg 1993; 165:9–14. 62. RegolyMerei J, Ihasz M, Szeberin Z, Sandor J, Mate M. Biliary tract complications in laparoscopic cholecystectomy: a multicenter study of 148 biliary tract injuries in 26,440 operations. Surg Endosc 1998; 12:294–300. 63. Davidoff AM, Pappas TN, Murray EA, et al. Mechanisms of major biliary injury during laparoscopic cholecystectomy. Ann Surg 1992; 215:196–202. 64. Raute M, Podlech P, Jaschke W, Manegold BC, Trede M, Chir B. Management of bile duct injuries and strictures following cholecystectomy. World J Surg 1993; 17:553–562. 65. Gouma DJ, Go PM. Bile duct injury during laparoscopic and conventional cholecystectomy. J Am Coll Surg 1994; 178:229–233. 66. McMahon AJ, Fullarton G, Baxter JN, O'Dwyer PJ. Bile duct injury and bile leakage in laparoscopic cholecystectomy. Br J Surg 1995; 82:307–313. 67. Frattaroli FM, Reggio D, Guadalaxara A, Illomei G, Pappalardo G. Benign biliary strictures: a review of 21 years of experience. J Am Coll Surg 1996; 183:506– 513. 68. Davids PH, Rauws EA, Coene PP, Tytgat GN, Huibregtse K. Endoscopic stenting for postoperative biliary strictures. Gastrointest Endosc 1992; 38:12–18. 69. Davids PH, Tanka AK, Rauws EA, et al. Benign biliary strictures: surgery or endoscopy? Ann Surg 1993; 217:237–243. 70. Innes JT, Ferrara JJ, Carey LC. Biliary reconstruction without transanastomotic stent. Am Surg 1988; 54:27–30. 71. Genest J, Nanos E, GrundfestBroniatowski S, Vogt D, Hermann R. Benign biliary strictures: an analytic review (1970–1984). Surgery 1986; 99:409–413. 72. Chapman WC, Halevy A, Blumgart LH, Benjamin IS. Postcholecystectomy bile duct strictures: management and outcome in 130 patients. Arch Surg 1995; 130:597–602; discussion 602–604. 73. Pitt HA, Miyamoto T, Parapatis SK, Tompkins RK, Longmire WP Jr. Factors influencing outcome in patients with postoperative biliary strictures. Am J Surg 1982; 144:14–21. 74. Pitt HA, Kaufman SL, Coleman J, White RI, Cameron JL. Benign postoperative biliary strictures: operate or dilate? Ann Surg 1989; 210:417–425; discussion 426–427. 75. Pellegrini CA, Thomas MJ, Way LW. Recurrent biliary stricture: patterns of recurrence and outcome of surgical therapy. Am J Surg 1984; 147:175–180.
Page 871
76. Lillemoe K, Martin S, Cameron J, et al. Major bile duct injuries during laparoscopic cholecystectomy. Ann Surg 1997; 225:459–471. 77. Moore AV Jr, Illescas FF, Mills SR, et al. Percutaneous dilation of benign biliary strictures. Radiology 1987; 163:625–628. 78. Mueller PR, vanSonnenberg E, Ferrucci JT Jr, et al. Biliary stricture dilatation: multicenter review of clinical management in 73 patients. Radiology 1986; 160:17– 22. 79. Vogel SB, Howard RJ, Caridi J, Hawkins IF Jr. Evaluation of percutaneous transhepatic balloon dilatation of benign biliary strictures in highrisk patients. Am J Surg 1985; 149:73–79. 80. Lillemoe KD. Benign postoperative bile duct strictures. Baillieres Clin Gastroenterol 1997; 11:749–779. 81. Williams HJ, Jr., Bender CE, May GR. Benign postoperative biliary strictures: dilation with fluoroscopic guidance. Radiology 1987; 163:629–634. 82. Salomonowitz E, CastanedaZuniga WR, Lund G, et al. Balloon dilatation of benign biliary strictures. Radiology 1984; 151:613–616. 83. Trambert JJ, Bron KM, Zajko AB, Starzl TE, Iwatsuki S. Percutaneous transhepatic balloon dilatation of benign biliary strictures. AJR 1987; 149:945–948. 84. Gallacher DJ, Kadir S, Kaufman SL, et al. Nonoperative management of benign postoperative biliary strictures. Radiology 1985; 156:625–629. 85. Bonnel DH, Liguory CL, Lefebvre JF, Cornud FE. Placement of metallic stents for treatment of postoperative biliary strictures: longterm outcome in 25 patients. AJR 1997; 169:1517–1522. 86. Dumonceau JM, Deviere J, Delhaye M, Baize M, Cremer M. Plastic and metal stents for postoperative benign bile duct strictures: the best and the worst (see comments). Gastrointest Endosc 1998; 47:8–17. 87. Foutch PG, Sivak MV Jr. Therapeutic endoscopic balloon dilatation of the extrahepatic biliary ducts. Am J Gastroenterol 1985; 80:575–580. 88. Geenen DJ, Geenen JE, Hogan WJ, et al. Endoscopic therapy for benign bile duct strictures. Gastrointest Endosc 1989; 35:367–371. 89. Berkelhammer C, Kortan P, Haber GB. Endoscopic biliary prostheses as treatment for benign postoperative bile duct strictures. Gastrointest Endosc 1989; 35:95–101. 90. Doctor N, Dooley JS, Dick R, Watkinson A, Rolles K, Davidson BR. Multidisciplinary approach to biliary complications of laparoscopic cholecystectomy. Br J Surg 1998; 85:627–632. 91. Popescu I, Sheiner P, Mor E, et al. Biliary complications in 400 cases of liver transplantation. Mt Sinai J Med 1994; 61:57–62. 92. O'Connor TP, Lewis WD, Jenkins RL. Biliary tract complications after liver transplantation. Arch Surg 1995; 130:312–317. 93. Rossi G, Lucianetti A, Gridelli B, et al. Biliary tract complications in 224 orthotopic liver transplantations. Transplant Proc 1994; 26:3626–3628. 94. Sossenheimer M, Slivka A, CarrLocke D. Management of extrahepatic biliary disease after orthotopic liver transplantation: review of the literature and results of a multicenter survey. Endoscopy 1996; 28:565–571. 95. Northover J, Terblanche J. Bile duct blood supply: its importance in human liver transplantation. Transplantation 1978; 26:67–69. 96. Klein AS, Savader S, Burdick JF, et al. Reduction of morbidity and mortality from biliary complications after liver transplantation. Hepatology 1991; 14:818–823. 97. Sankary HN, McChesney L, Frye E, Cohn S, Foster P, Williams J. A simple modification in operative technique can reduce the incidence of nonanastomotic biliary strictures after orthotopic liver transplantation. Hepatology 1995; 21:63–69. 98. Wozney P, Zajko AB, Bron KM, Point S, Starzl TE. Vascular complications after liver transplantation: a 5year experience. AJR 1986; 147:657–663. 99. Saxena R, Tokat Y, Soin AS, Rasmussen A, Jamieson NV, Calne R. Relationship between
Page 872
patterns of hepatobiliary vascular supply and biliary complications in liver transplantation: an anatomical and clinical analysis. Transplant Proc 1995; 27:1199– 1200. 100. Sheng R, Zajko AB, Campbell WL, AbuElmagd K. Biliary strictures in hepatic transplants: prevalence and types in patients with primary sclerosing cholangitis vs those with other liver diseases. AJR 1993; 161:297–300. 101. Orons PD, Sheng R, Zajko AB. Hepatic artery stenosis in liver transplant recipients: prevalence and cholangiographic appearance of associated biliary complications. AJR 1995; 165:1145–1149. 102. Drazan K, Shaked A, Olthoff KM, et al. Etiology and management of symptomatic adult hepatic artery thrombosis after orthotopic liver transplantation (OLT). Am Surg 1996; 62:237–240. 103. SanchezUrdazpal L, Gores GJ, Ward EM, et al. Diagnostic features and clinical outcome of ischemictype biliary complications after liver transplantation. Hepatology 1993; 17:605–609. 104. Kowdley KV, Fawaz KA, Kaplan MM. Extrahepatic biliary stricture associated with cytomegalovirus in a liver transplant recipient. Transplant Int 1996; 9:161– 163. 105. Gholson CF, Zibari G, McDonald JC. Endoscopic diagnosis and management of biliary complications following orthotopic liver transplantation. Dig Dis Sci 1996; 41:1045–1053. 106. Kuo PC, Lewis WD, Stokes K, Pleskow D, Simpson MA, Jenkins RL. A comparison of operation, endoscopic retrograde cholangiopancreatography, and percutaneous transhepatic cholangiography in biliary complications after hepatic transplantation. J Am Coll Surg 1994; 179:177–181. 107. Greif F, Bronsther OL, Van Thiel DH, et al. The incidence, timing, and management of biliary tract complications after orthotopic liver transplantation. Ann Surg 1994; 219:40–45. 108. Stratta RJ, Wood RP, Langnas AN, et al. Diagnosis and treatment of biliary tract complications after orthotopic liver transplantation. Surgery 1989; 106:675– 683; discussion 683–684. 109. Lerut J, Gordon RD, Iwatsuki S, et al. Biliary tract complications in human orthotopic liver transplantation. Transplantation 1987; 43:47–51. 110. Rizk RS, McVicar JP, Emond MJ, et al. Endoscopic management of biliary strictures in liver transplant recipients: effect on patient and graft survival. Gastrointest Endosc 1998; 47:128–135. 111. Bourgeois N, Deviere J, Yeaton P, et al. Diagnostic and therapeutic endoscopic retrograde cholangiography after liver transplantation. Gastrointest Endosc 1995; 42:527–534. 112. Rossi AF, Grosso C, Zanasi G, et al. Longterm efficacy of endoscopic stenting in patients with stricture of the biliary anastomosis after orthotopic liver transplantation. Endoscopy 1998; 30:360–366. 113. Sherman S, Jamidar P, Shaked A, Kendall BJ, Goldstein LI, Busuttil RW. Biliary tract complications after orthotopic liver transplantation: endoscopic approach to diagnosis and therapy. Transplantation 1995; 60:467–470. 114. Wisloff F JJ, Osnes M. Stenosis of the common bile duct in chronic pancreatitis. Br J Surg 1982; 69:52–54. 115. Frey CF, Suzuki M, Isaji S. Treatment of chronic pancreatitis complicated by obstruction of the common bile duct or duodenum. World J Surg 1990; 14:59–69. 116. Aranha GV, Prinz RA, Freeark RJ, Greenlee HB. The spectrum of biliary tract obstruction from chronic pancreatitis. Arch Surg 1984; 119:595–600. 117. Schulte WJ LA, Condon RE, Unger GF, Geenen JE, DeCosse JJ. Chronic pancreatitis: a cause of biliary stricture. Surgery 1977; 82:303–309. 118. Petrozza JA, Dutta SK, Latham PS, Iber FL, Gadacz TR. Prevalence and natural history of distal common bile duct stenosis in alcoholic paancreatitis. Dig Dis Sci 1984; 29:890–895.
Page 873
119. Ng C, Huibregtse K. The role of endoscopic therapy in chronic pancreatitis—induced common bile duct strictures. Gastrointest Endosc Clin North Am 1998; 8:181–193. 120. Snape WJ LW, Trotman BW, Marin GA, Czaja AJ. Marked alkaline phosphatase elevation with partial common bile duct obstruction due to calcific pancreatitis. Gastroenterology 1976; 70:70–73. 121. Stahl TJ, Allen MO, Ansel HJ, Vennes JA. Partial biliary obstruction caused by chronic pancreatitis: an appraisal of indications for surgical biliary drainage. Ann Surg 1988; 207:26–32. 122. Afroudakis A, Kaplowitz N. Liver histopathology in chronic common bile duct stenosis due to chronic alcoholic pancreatitis. Hepatology 1981; 1:65–72. 123. Yadegar J, Williams RA, Passaro E Jr, Wilson SE. Common duct stricture from chronic pancreatitis. Arch Surg 1980; 115:582–586. 124. Barthet M, Bernard JP, Duval JL, Affriat C, Sahel J. Biliary stenting in benign biliary stenosis complicating chronic calcifying pancreatitis. Endoscopy 1994; 26:569–572. 125. Schlauch D, Kohler B, Riemann JF. Doubleductsign—is it always cancer? (letter). Endoscopy 1993; 25:489–490. 126. Eckhauser FE, Knol JA, Strodel WE, Achem S, Nostrant T. Common bile duct strictures associated with chronic pancreatitis. Am Surg 1983; 49:350–358. 127. Byrne RL, Gompertz RH, Venables CW. Surgery for chronic pancreatitis: a review of 12 years experience. Ann R Coll Surg Engl 1997; 79:405–409. 128. Kozicki I, Bielecki K, Kawalski A, Krolicki L. Repeated reconstruction for recurrent benign bile duct stricture. Br J Surg 1994; 81:677–679. 129. Warren KW, Christophi C, Armendari ZR. The evolution and current prospectives of the treatment of benign bile duct strictures: a review. Surgical Gastroenterol 1982; 1:141–154. 130. Rothlin MA, Lopfe M, Schlumpf R, Largiader F. Longterm results of hepaticojejunostomy for benign lesions of the bile ducts. Am J Surg 1998; 175:22–26. 131. Schweizer WP, Matthews JB, Baer HU, et al. Combined surgical and interventional radiological approach for complex benign biliary tract obstruction. Br J Surg 1991; 78:559–563. 132. Deviere J, Devaere S, Baize M, Cremer M. Endoscopic biliary drainage in chronic pancreatitis. Gastrointest Endosc 1990; 36:96–100. 133. Smits M, Rauws EAJ, van Gulik TM, Gouma DJ, Tytgat NJ, Huibregtse K. Longterm results of endoscopic stenting and surgical drainage for biliary stricture due to chronic pancreatitis. Br J Surg 1996; 83:764–768. 134. Itani KM, Taylor TV. The challenge of therapy for pancreatitisrelated common bile duct stricture. Am J Surg 1995; 170:543–546. 135. O'Brien SM, Hatfield AR, Craig PI, Williams SP. A 5year followup selfexpanding metal stents in the endoscopic management of patients with benign bile duct strictures. Eur J Gastroenterol Hepatol 1998; 10:141–145. 136. Deviere J, Cremer M, Baize M, Love J, Sugai B, Vandermeeren A. Management of common bile duct stricture caused by chronic pancreatitis with metal mesh selfexpandable stents. Gut 1994; 35:122–126. 137. Hausegger KA, Kugler C, Uggowitzer M, et al. Benign biliary obstruction: is treatment with the Wallstent advisable? Radiology 1996; 200:437–441. 138. Scott J, Summerfield JA, Elias E, Dick R, Sherlock S. Chronic pancreatitis: a cause of cholestasis. Gut 1977; 18:196–201. 139. Lee Y, Kaplan M. Primary sclerosing cholangitis. N Engl Med 1995; 332:924–933. 140. Belle SH, Beringer KC, Detre KM. An update on liver transplantation in the United States: recipient characteristics and outcome. Clin Transplant 1995:19–33. 141. Kaplan MM. Toward better treatment of primary sclerosing cholangitis (editorial; comment). N Engl J Med 1997; 336:719–721. 142. Klompmaker IJ, Haagsma EB, Verwer R, Jansen PL, Sloofe MJ. Primary sclerosing cholangitis and liver transplantation. Scand J Gastroenterol Suppl 1996; 218:98–102.
Page 874
143. Goss JA, Shackleton CR, Farmer DG, et al. Orthotopic liver transplantation for primary sclerosing cholangitis: a 12year single center experience. Ann Surg 1997; 225:472–481; discussion 481–483. 144. Roozendaal C, Van Milligen de Wit AW, Haagsma EB, et al. Antineutrophil cytoplasmic antibodies in primary sclerosing cholangitis: defined specificities may be associated with distinct clinical features. Am J Med 1998; 105:393–399. 145. Seibold F, Weber P, Schoning A, Mork H, Goppel S, Scheurlen M. Neutrophil antibodies (pANCA) in chronic liver disease and inflammatory bowel disease: do they react with different antigens? Eur J Gastroenterol Hepatol 1996; 8:1095–1100. 146. Brandt DJ, MacCarty RL, Charboneau JW, LaRusso NF, Wiesner RH, Ludwig J. Gallbladder disease in patients with primary sclerosing cholangitis. AJR 1988; 150:571–574. 147. Eckhauser FE, Colleti LM, Knol JA. The changing role of surgery for sclerosing cholangitis. Dig Dis 1996; 14:180–191. 148. Ismail T, Angrisani L, Powell JE, et al. Primary sclerosing cholangitis: surgical options, prognostic variables and outcome. Br J Surg 1991; 78:564–567. 149. Farges O, Malassagne B, Sebagh M, Bismuth H. Primary sclerosing cholaangitis: liver transplantation or biliary surgery. Surgery 1995; 117:146–155. 150. Lemmer ER, Bornman PC, Krige JE, et al. Primary sclerosing cholangitis: requiem for biliary drainage operations? Arch Surg 1994; 129:723–728. 151. Lillemoe KD, Cameron JL. Surgical approaches to primary sclerosing cholangitis. Semin Liver Dis 1991; 11:49–55. 152. Myburgh JA. Surgical biliary drainage in primary sclerosing cholangitis: the role of the HeppCouinaud approach. Arch Surg 1994; 129:1057–1062. 153. Ahrendt SA, Pitt HA, Kalloo AN, et al. Primary sclerosing cholangitis: resect, dilate, or transplant? Ann Surg 1998; 227:412–423. 154. Gaing AA, Geders JM, Cohen SA, Siegel JH. Endoscopic management of primary sclerosing cholangitis: review, and report of an open series. Am J Gastroenterol 1993; 88:2000–2008. 155. van Milligen de Wit AW, van Bracht J, Rauws EA, Jones EA, Tytgat GN, Huibregtse K. Endoscopic stent therapy for dominant extrahepatic bile duct strictures in primary sclerosing cholangitis. Gastrointest Endosc 1996; 44:293–299. 156. van Milligen de Wit AW, Rauws EA, van Bracht J, et al. Lack of complications following shortterm stent therapy for extrahepatic bile duct strictures in primary sclerosing cholangitis. Gastrointest Endosc 1997; 46:344–347. 157. Johnson GK, Geenen JE, Venu RP, Schmalz MJ, Hogan WJ. Endoscopic treatment of biliary tract strictures in sclerosing cholangitis: a larger series and recommendations for treatment. Gastrointest Endosc 1991; 37:38–43. 158. Lee JG, Schutz SM, England RE, Leung JW, Cotton PB. Endoscopic therapy of sclerosing cholangitis. Hepatology 1995; 21:661–667. 159. Wagner S, Gebel M, Meier P, et al. Endoscopic management of biliary tract strictures in primary sclerosing cholangitis (see comments). Endoscopy 1996; 28:546–551. 160. Cotton PB, Nickl N. Endoscopic and radiologic approaches to therapy in primary sclerosing cholangitis. Semin Liver Dis 1991; 11:40–48. 161. Lombard M, Farrant M, Karani J, Westaby D, Williams R. Improving biliaryenteric drainage in primary sclerosing cholangitis: experience with endoscopic methods. Gut 1991; 32:1364–1368. 162. Stiehl A, Rudolph G, Sauer P, et al. Efficacy of ursodeoxycholic acid treatment and endoscopic dilation of major duct stenoses in primary sclerosing cholangitis: an 8year prospective study. J Hepatol 1997; 26:560–566.
Page 875
Index A Acidification of bile, 325 Acromegaly, 298–300 gallstones, 298 Acute acalculous cholecystitis, 593 clinical presentation, 601 HIDA scanning, 603–604 pathology, 597 risk factors, 597–601 treatment, 605–606 ultrasound findings, 602–603 Acute bacterial cholangitis, pathogenesis, 773–776 Acute cholangitis, antibiotic therapy, 782–786 biliary drainage, 786–788 clinical features, 778 differential diagnosis, 780 endoscopy, 786–787 etiology, 776–778 intrabiliary pressure, 775 lab tests, 778–779 microbiology, 781–782 pathology, 778 percutaneous transhepatic drainage, 787 radiology, 779 surgery, 788 Adenomyomatosis, 609 Adjuvant litholytic therapy, 536 Adult idiopathic ductopenia, 713–714 Age and gallstones, 369 AIDS cholangiopathy, 802–805 pathogenesis, 803 treatment, 805 AminopeptidaseN, 238, 341 Ampullary carcinoma, 756, 767–769 cholangiography, 759–762 clinical features, 757 endoscopic ultrasound, 761–762 epidemiology, 756 local excision, 763–766 palliation, 767 pancreaticoduodenectomy, 766 pathology, 755–756 radiology, 757–759 staging, 763 Anhydrous cholesterol, 187–188 Animal models, prostaglandins, 282 of PSC, 681 Anionic peptide fraction, 333–337 binding to cholesterol vesicles, 340 Anomalous pancreaticobiliary ductal junction, 626, 647 Antineutrophil cystoplasmic antibodies, 678 Apolipoproteins, 237–238, 242 Ascaris lumbricoides, 793, 799–800 Autoimmune cholangitis, 719 B Bacterial biofilm, 775 Bactibilia, 774–775 Benign biliary strictures, 853–863 Bile acids, diurnal variation, 254 Bile acid synthesis, inborn errors, 174 Bile duct development, 705
Page 876
Bile duct injuries, classification, 824–825 early repair, 828 endoscopic approach, 834–836 mechanisms, 825–828 medicolegal issues, 837–839 radiological approach, 833–834 surgical management, 828–833 Bile duct ischemia, 854 Bile duct stones, classification, 567–568 clinical presentation, 568 gallbladder in sim, 583 percutaneous approach, 576–577 and pregnancy, 584 postcholecystectomy, 582 Ttubes, 582 Bile duct strictures, postliver transplant, 857–859 Bile duct transection, 829 Bile duct units, 101–104 Bile ducts, anatomy, 99–100 candidal infection, 792 in liver transplantation, 708–713 Bile salt biology, 169 Bile salt secretion, 70–71 Bile salt synthesis, 169–172 Biliary atresia, 640–646 clinical patterns, 642–643 differential diagnosis, 642 hepatic transplantation, 645–646 pathogenesis, 640–641 surgery, 644 Biliary enteric anastomosis, 830 Biliary glycoprotein, 680–681 Biliary lipids, physical chemistry, 135–137 Biliary proteins, 235–236 secretion, 33 Biliary secretion, regulation, 112–118 Biliary sludge, 213 Biliary stents, 848–850 prophylactic antibiotics, 792 Biliary strictures, diagnosis, 843–844 etiology, 844 treatment, 845–847 Biliary surgery, antibiotic prophylaxis, 789–790 Biliary tract embryology, 639 Bilirubin, absorption, 31 Biomineralization, 319, 330–332 calcium salts, 331 cholesterol, 331 Black pigment gallstones, 149, 152–153 Brown pigment gallstones, 149–150, 156 C Calcium, 200 bicarbonate, 148, 152–153 and bile salts, 325 bilirubinate, 327–328 binding protein, 333–337 carbonate, 154, 327 effect on calcium, 340 effect on cholesterol crystallization and growth, 338 electrodes, 323–324 hydroxyapatite, 148, 154 ionized, 322 phosphase, 327 salts, 155, 319–324 solubility product, 324 total, 322, 324 Calcium complex, formation constant, 324 Calcium salt precipitations, 329 Calcium salt saturation, 325 Canalicular lipids, 67–69 Canals of Hering, 66–67 Caroli's disease, 653–654 Cavitation, 528 CFTR mutations, 389 Charcot's triad, 568 Chenodeoxycholic acid, 172 Chloride channels, 108–109 Cholangiocarcinoma, 651, 671–672 adjuvant therapy, 747–749 chemoradiation, 748 chemotherapy, 747 cytology, 732 diagnosis, 728–732 etiology and risk factors, 726–727 incidence, 725 nonoperative palliation, 739 palliative therapy, 738–742 pathology, 727 staging, 733–734 surgery, 734–738 survival, 742–747 Cholangitis (see Acute cholangitis) Cholecystitis, and HIV, 600 and immunosuppression, 594 Cholecystokinin, 253, 277
Page 877
Cholecystokinin receptors, 277–278 Choledochal cysts, 646–654 classification, 648 clinical features, 647–648 imaging, 647 pathogenesis, 646–647 treatment, 650–651 Choledochocele, 652 Choledocholithiasis and laparoscopic cholecystectomy, 577–580 Cholescintigraphy, 256–257 Cholestasis, 80–82 Cholesterol, 73–74 7a hydroxylase, 171 biology, 165–169 CCK receptor, 283 critical nucleus, 192 crystallization, 185 proteins, 240–244 esters, 166 filaments, 188–189 homeostasis, 176 metabolism, 133, 175–176 monohydrate, 186–188 monomers, 199 nucleation, 236 inhibitors, 237–238 mechanism, 241–242 promotors, 238–240 time, 236 precipitation, 329 saturation index, 191, 199 secretion, 79–80 solubility, 362–363 structures, 194–195 synthesis, 166 Cholesterolosis, 609 Cholic acid, 172 Chronic acalculous cholecystitis, 608–612 CCK stimulation tests, 611–612 clinical features, 610 pathology, 609 Chronic ductopenic rejection, 711–712 Chronic pancreatitis, 860–863 endoscopic stents, 861–862 Clonorchis sinensis, 793, 801 Common bile duct structure, 860 Common bile duct surgery, 575 Common duct stones, endoscopic therapy, 571–573 Concanavalin Abinding fraction, 238–240, 243 Crystal growth, 200–201, 214 Crystal growth assay, 193 Cystic fibrosis, 24–25 bile acid loss, 390 bile acid metabolism, 396 bile duct mucus, 389–390 cholecystectomy, 399 clinical features, 391 diagnosis, 392 gene therapy, 401 genetics, 387–389 imaging, 392–394 liver pathology, 395–397 liver transplantation, 400–401 prevalence, 388 transmembrane conductance regulator, 388 ursodeoxycholic acid, 399 D Deoxycholate, 596 absorption and pH, 307 CS1, 304 effect on cholesterol secretion, 303 feeding and biliary cholesterol saturation, 304–305 formation rate, 302 and phosphatidylcholine, 304 levels, 303 role in gallstone formation, 308 Diabetes mellitus, 370 Ductopenia, biopsy findings, 705–708 druginduced, 717 in infancy and childhood, 715 Duodenal perforation, 574 E Echinococcal cholangitis, 802 Eicosanoids, 34 Electromagnetic system, 529 Electrolyte secretion, 29–30 Electrolyte transport, 105–110 calcium, 27–29 cAMP, 27 hormones, 27 Electrolyte transporter, regulation, 26 Electrolyte secretion, 29–30 Emphysematous cholecystitis, 607 Empyema of the gallbladder, 608 Endoscopic balloon dilation of sphincter, 574 Endoscopic retrograde cholangiopancreatography, 569–570
Page 878
Endoscopic retrograde gallbladder cannulation, 559 Endoscopic sphincterotomy, 571 complications, 573–574 Endothelins, 280 Enteric nervous system, 1 Enterohepatic circulation, 254 Epithelium, acid secretion, 23–24 basolateral membrane, 24 cell culture, 22 morphology, 22 water absorption, 23 Equilibriumphase diagram, 190–191 Ethyl propionate, 559 Extracorporeal shockwave lithotripsy, 523 F Familial adenomatous polyposis, 756 Fasciola hepatica, 802 Fat malabsorption, 398 Fatty acid salts, 156 Fatty acylates, 328 Fecal bile acids, 396 Fluid absorption, 25–26 Free calcium, 320 G Gallbladder, chemical injury, 595–596 Gallbladder cancer, adjuvant therapy, 634–635 association with gallstones, 626 clinical features, 632 epidemiology, 625 histology, 628 lymphatic drainage, 627 natural history, 629 pathology, 626 staging, 629–631 surgery, 632–634 Gallbladder dysmotility, cholecystokinin, 281–282 cholecystokinin receptor, 282 etiology, 281 fasting, 281 gallstone pathogenesis, 280–282 pregnancy, 281 Gallbladder embryology, 275 Gallbladder emptying, 259 animal models, 280 duodenal perfusion, 257–258 ejection fraction, 262 intraindividual variability, 260 methodology, 261 [Gallbladder emptying] ultrasonography, 255–256 vesodeoxycholic acid, 265 Gallbladder filling, 258 Gallbladder gangrene, 607 Gallbladder infections, 594–595 Gallbladder ischemia, 593–594 Gallbladder mixing, 259–260 Gallbladder motility, 364 abnormal gallbladder motility, 266 control, 252–253 fasting gallbladder motility, 276 intrinsic neurotransmitters, 253 neural control, 252–253 Gallbladder mucin, 363–364 Gallbladder neuronal innervation, 275 Gallbladder obstruction, 596 Gallbladder perforation, 608 Gallbladder sludge, 365 Gallbladder storage, 258 Gallbladder torsion, 594 Gallbladder washout, 264 Gallstone composition, 317–319 Gallstone content, 186 Gallstone disease, mucin gene expression, 223 Gallstone epidemiology, 127–128 Gallstone growth, 235–244 Gallstone pancreatitis, 581–582 Gallstone prevention, aspirin, 373 cholecystokinin, 374 lowcalorie diets, 371 prokinetics, 284 ursodeoxycholic acid, 371–372 vitamin C, 371 weight reduction rate, 370 Gallstones, and exercise, 369 highrisk population, 365 large bowel transit time, 306–307 risk factors, 128–133 symptomatic, 522 Ganglia, 10, 12–15 CCK, 13 celiac, 9 duodenal, 12 EPSP, 13 regulation, 15 sensory, 10 sympathetic, 9 vagal, 13 Gastrin, 253
Page 879
GibbsDonnan equilibrium, 322–323 Glycan, 211 Glucose transport, 111 Graft versus host disease, 713 H Hepatic osteodystrophy, 673 Hepatic sarcoidosis, 715 Hepaticojejunostomies, 832 Hepatocyte, morphology, 65–66 Hepatolithiasis, 792–799 epidemiology, 793 pathogenesis, 793–794 Highdensity lipoproteins, 169 Hilar malignant biliary obstruction, 852–853 Hormones, CCK, 12 HMGCoA reductase, 166, 167 inhibitors, 373 Human mucin genes, 215–219 I Immunoglobulins, 33–34, 238–239 Intracorporeal lithotripsy, 539–540, 572 Ion transporters, 105–107 Ischemic cholangiopathy, 713 J Jaundice, 568 K Kasai procedure, 644 L Laparoscopic common bile duct exploration, 576 Large bowel transit, deoxycholate levels, 301 Leiomyopathy, 49 Liesegang rings, 151 Lipid absorption, 30–31 Lipid secretion, 75–80 Lipoprotein X, 85 Lithotripsy, bile duct stones, 538–541 cost effectiveness, 538 gallstone recurrence, 537–538 stone composition, 531–532 stone number, 532 stone volume, 532 success rate, 533–536 Liver fluke cholangitis, 800–802 Lowdensity lipoproteins, 168 M Malignant biliary strictures, 847–853 distal obstruction, 847–852 MDR 1 transporter, 25 Mechanisms of crystallization, 194 Metallic stents, 850 Methyl tertbutyl ether, 548–549 Micelles, 189 Microgallbladder, 390 Microprocessorassisted solvent transfer system, 554 Migrating myoelectric complex, 12 Mineral nucleation, 345 Mirizzi's syndrome, 780 Monooctanoin, 548 dissolution of bile duct stones, 548 Motilin, 253 MUC1, 215 MUC3, 217–218 MUC5B, structural organization, 219–221 Mucin, 243 bile salts, 32 cAMP, 32 cFTR, 32 domains, 214, 222 glycans, 212 hypersecretion, 213 matrix, 201 and nucleation, 214 polymerization, 211 protein backbone, 212 primary structure, 212 secretion, 223–225 side chains, 212 structure, 322 Mucus gel, 332, 333 calcium and cholesterol precipitation, 333 layer, 213 Mucus hypersecretion, 332 Mucus layer, 211 Multilamellar vesicles, 197 Muscle contraction, 40–47 bile salts, 51–53 CCK, 43–44 cholesterol, 48–50 G proteins, 45 hormones, 46–47 inflammation, 51 muscarinic receptors, 41 neuronal, 51 tone, 41 Muscle relaxation, 47–48 MTBE, solvent delivery, 550–551
Page 880
N Nasobiliary stent, 573 Neural plexus, serosa, 2 submucosal, 2 Neurons, 1, 3, 6, 7, 8 action potential, 7 adaptive, 8 electrical properties, 6 parasympathetic, 3, 8–9 sinapase, 8 ultrastructure, 3 vagal, 8–9 Neurotransmitters, 3, 6, 9, 10, 11 acetylcholine, 9 CGRP, 10 cholinergic, 3 norepinephrine, 9 neuropeptide Y, 3 PACAP, 3 regulation, 6 substance P, 10 vasoactive intestinal peptide, 3 Nitric oxide, 253, 279 Nitric oxide synthase, 4 Nonsteroidal antiinflammatory drugs, 282 Nucleation, 138–139, 363 Nucleation time, 192 O Obesity, 366 Octreotide, 298–300 cholesterol content of gallstones, 299 effect on gallbladder emptying, 299 Octreotide gallstones, biliary lipids, 299 cholesterol supersaturation, 300 deoxycholate, 300 Opisthorchis viverrini, 801 Oral cholecystography, 255 Oral dissolution therapy, 522 Oriental cholangiohepatitis, 792–799 P Pancreatic carcinoma, 847, 847 Pancreatitis, 568 Pathways of cholesterol crystallization, 195–198 Percutaneous gallbladder endoscopy, 557 Percutaneous topical gallstone dissolution, 549–550 Piezoelectric system, 529 Pigment gallstones, 364 epidemiology, 147–150 Phosphatidylcholine, 200 Phosphatidylcholine secretion, 75–79 Phospholipids, 71–73, 176 Phospholipid synthesis, 176–178 Pituitary adenylate cyclase activating polypeptide, 279–280 Portal hypertension, 400 Postoperative biliary stricture, 853–857 Postprandial gallbladder motility, endocrine, 277 neuronal, 277 Postprandial gallbladder refilling, 263–264 Post topical dissolution imaging, 552–553 Posttraumatic cholecystitis, 599 Pregnancy and sex steroid hormones, 367–368 Prevention of gallstones, 369–375 Primary biliary cirrhosis, 718 Primary sclerosing cholangitis, 863–868 Prostaglandins, 13, 34–36, 223, 254, 278–279 gallbladder contraction, 13 gallstone formation, 34 inflammation, 34 PGE2, 13, 34 water absorption, 34 Protein crystallization, 155 R Rapid weight loss, 366–377 Recurrent cholangitis, maintenance antibiotics, 791 Recurrent pyogenic cholangitis, 792–799 endoscopic therapy, 796–797 pathology, 794 radiology, 794–795 surgery, 797–799 treatment, 795–799 Reynolds' pentad, 773 S Sclerosing cholangitis, and autoimmunity, 677–679 and cholangiocarcinoma, 666 and inflammatory bowel disease, 659–661 autoantibodies, 664 cholangiography, 667 clinical features, 662–664 complications, 670–671 cytokines, 679–680 diagnostic criteria, 666–668 differential diagnosis, 676 dominant stricture, 673, 864 etiology, 677–682 liver transplantation, 685–687 medical treatment, 683–685
Page 881
[Sclerosing cholangitis] natural history, 668–670 pathology, 664–665 prognostic models, 675 risk factors, 661 small duct disease, 667 therapy for complications, 687–689 treatment, 682–687 ursodeoxycholic acid, 684–685 Secretin, 113 Sensory neurons, 4 CGRP, 4 SP, 4 Serum deoxycholate, 302 Shock wave lithotripsy, morbidity and mortality, 536–537 Shock waves, 528–531 SmithLemliOpitz syndrome, 172–173 Smooth muscle cells, 39–40 cholesterol, 50–51 contraction, 39 morphology, 39 Slow transit constipation, 305 Small bowel disease, 369 Sodium transport, 23 Solvent delivery systems, 553–555 Somatostatin, 368 Sparkgap system, 529 Sphincter of Oddi, 11–12 reflex relaxation, 11 Spinal cord injury, 367 Steatorrhea, 673 Sterol 27 hydroxylase, 171 Sterol regulatory element, 166, 168 Sterol regulatory element binding protein, 166, 168 Stone matrix, 213 T Topical dissolution therapy, 524 CT index of gallstones, 555–556 gallstone recurrence, 560 patient selection, 551 Tumor necrosis factor, 224 U Unilamellar vesicles, 198 Ursodeoxycholic acid, 371–372, 536 V Vasoactive intestinal peptide, 253, 279 Vesicle trafficking, 69 Vesicular cholesterol, 191 W White bile, 30