Skip to main content Accessibility help
×
Hostname: page-component-77c89778f8-rkxrd Total loading time: 0 Render date: 2024-07-21T15:45:53.311Z Has data issue: false hasContentIssue false

Chapter 3 - Mechanisms of Bile Formation and the Pathogenesis of Cholestasis

from Section I - Pathophysiology of Pediatric Liver Disease

Published online by Cambridge University Press:  19 January 2021

Frederick J. Suchy
Affiliation:
University of Colorado, Children’s Hospital Colorado, Aurora
Ronald J. Sokol
Affiliation:
University of Colorado, Children’s Hospital Colorado, Aurora
William F. Balistreri
Affiliation:
Cincinnati Children’s Hospital Medical Center, Cincinnati
Jorge A. Bezerra
Affiliation:
Cincinnati Children’s Hospital Medical Center, Cincinnati
Cara L. Mack
Affiliation:
University of Colorado, Children’s Hospital Colorado, Aurora
Benjamin L. Shneider
Affiliation:
Texas Children’s Hospital, Houston
Get access

Summary

Bile formation and flow is an essential physiological function. Adequate bile flow is necessary for innumerable daily functions including digestion, metabolism, growth, development, toxin elimination and adaptation to liver disease. Cholestasis, a slowing or, in its extreme form, cessation of bile flow, is a pathophysiological process that interrupts the necessary functions of bile while biliary constituents accumulate intrahepatically [1–3]. This accretion of bile leads to liver damage from the multiple effects of retained biliary constituents, including various lipids, toxins, and as a principal effector – bile acids [4–6]. These effects of cholestasis, regardless of cause, are amplified and accelerated in the infant, rendering the consequences of unaddressed neonatal cholestasis urgent [1, 7–9]. A molecular understanding of the determinants of bile formation is helpful to link these individual solutes to the responsible genes and functions in normal biology and in cholestasis. Bile formation and flow relies upon membrane transporters of biliary solutes, and their regulators, that function to deliver each of the molecular constituents into bile. The focus of this chapter is to lay the groundwork of current knowledge of the mechanisms of bile formation and to explore normal and pathophysiological processes when these mechanisms are impaired. Moreover, very recent work based upon the knowledge of bile formation has led to new therapeutic targeting, with the goal of filling the serious gap in effective anti-cholestatic therapies.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2021

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Balistreri, WF, Suchy, FJ, Farrell, MK, Heubi, JE. Pathologic versus physiologic cholestasis: elevated serum concentration of a secondary bile acid in the presence of hepatobiliary disease. J Pediatr 1981;98:399402.Google Scholar
Spivak, W, Grand, RJ. General configuration of cholestasis in the newborn. J Pediatr Gastroenterol Nutr 1983;2:381–92.Google Scholar
Yang, T, Khan, GJ, Wu, Z, Wang, X, Zhang, L, Jiang, Z. Bile acid homeostasis paradigm and its connotation with cholestatic liver diseases. Drug Discov Today 2019;24:112–28.CrossRefGoogle ScholarPubMed
Popper, H. Cholestasis: the future of a past and present riddle. Hepatology 1981;1:187–91.Google Scholar
Hofmann, AF. Biliary secretion and excretion in health and disease: current concepts. Ann Hepatol 2007;6:1527.Google Scholar
Fickert, P, Wagner, M. Biliary bile acids in hepatobiliary injury – What is the link? J Hepatol 2017;67:619–31.CrossRefGoogle ScholarPubMed
Lester, R. Physiologic cholestasis. Gastroenterology 1980;78:864–5.Google Scholar
Fawaz, R, Baumann, U, Ekong, U, Fischler, B, Hadzic, N, Mack, CL, McLin, VA, et al. Guideline for the evaluation of cholestatic jaundice in infants: Joint Recommendations of the North American Society for Pediatric Gastroenterology, Hepatology, and Nutrition and the European Society for Pediatric Gastroenterology, Hepatology, and Nutrition. J Pediatr Gastroenterol Nutr 2017;64:154–68.Google Scholar
Feldman, AG, Sokol, RJ. Neonatal cholestasis: emerging molecular diagnostics and potential novel therapeutics. Nat Rev Gastroenterol Hepatol 2019;16:346–60.Google Scholar
Trauner, M, Fuchs, CD, Halilbasic, E, Paumgartner, G. New therapeutic concepts in bile acid transport and signaling for management of cholestasis. Hepatology 2017;65:13931404.Google Scholar
Tabibian, JH, Masyuk, AI, Masyuk, TV, O’Hara, SP, Larusso, NF. Physiology of cholangiocytes. Compr Physiol 2013;3:541–65.Google Scholar
Dawson, PA, Karpen, SJ. Intestinal transport and metabolism of bile acids. J Lipid Res 2015;56:1085–99.Google Scholar
Jansen, PL, Ghallab, A, Vartak, N, Reif, R, Schaap, FG, Hampe, J, Hengstler, JG. The ascending pathophysiology of cholestatic liver disease. Hepatology 2017;65:722–38.CrossRefGoogle ScholarPubMed
Li, J, Dawson, PA. Animal models to study bile acid metabolism. Biochim Biophys Acta Mol Basis Dis 2019;1865:895911.Google Scholar
Heubi, JE, Setchell, KDR, Bove, KE. Inborn errors of bile acid metabolism. Clin Liver Dis 2018;22:671–87.Google Scholar
Strautnieks, SS, Bull, LN, Knisely, AS, Kocoshis, SA, Dahl, N, Arnell, H, Sokal, E, et al. A gene encoding a liver-specific ABC transporter is mutated in progressive familial intrahepatic cholestasis. Nat Gen 1998;20:233–8.Google Scholar
Borgstroem, B, Lundh, G, Hofmann, A. The site of absorption of conjugated bile salts in man. Gastroenterology 1963;45:229–38.Google ScholarPubMed
Wong, MH, Oelkers, P, Craddock, AL, Dawson, PA. Expression cloning and characterization of the hamster ileal sodium-dependent bile acid transporter. J Biol Chem 1994;269:1340–7.CrossRefGoogle ScholarPubMed
Dawson, PA, Hubbert, M, Haywood, J, Craddock, AL, Zerangue, N, Christian, WV, Ballatori, N. The heteromeric organic solute transporter alpha-beta, Ostalpha-Ostbeta, is an ileal basolateral bile acid transporter. J Biol Chem 2005;280:6960–8.Google Scholar
Hagenbuch, B, Lubbert, H, Stieger, B, Meier, PJ. Expression of the hepatocyte Na+/bile acid cotransporter in Xenopus laevis oocytes. J Biol Chem 1990;265:5357–60.Google Scholar
Orntoft, NW, Munk, OL, Frisch, K, Ott, P, Keiding, S, Sorensen, M. Hepatobiliary transport kinetics of the conjugated bile acid tracer (11)C-CSar quantified in healthy humans and patients by positron emission tomography. J Hepatol 2017;67:321–7.CrossRefGoogle ScholarPubMed
Ridlon, JM, Harris, SC, Bhowmik, S, Kang, DJ, Hylemon, PB. Consequences of bile salt biotransformations by intestinal bacteria. Gut Microbes 2016;7:2239.CrossRefGoogle ScholarPubMed
Yan, H, Zhong, G, Xu, G, He, W, Jing, Z, Gao, Z, Huang, Y, et al. Sodium taurocholate cotransporting polypeptide is a functional receptor for human hepatitis B and D virus. eLife 2012;1:e00049.CrossRefGoogle ScholarPubMed
Hu, HH, Liu, J, Lin, YL, Luo, WS, Chu, YJ, Chang, CL, Jen, CL, et al. The rs2296651 (S267F) variant on NTCP (SLC10A1) is inversely associated with chronic hepatitis B and progression to cirrhosis and hepatocellular carcinoma in patients with chronic hepatitis B. Gut 2016;65:1514–21.Google Scholar
Li, W, Urban, S. Entry of hepatitis B and hepatitis D virus into hepatocytes: basic insights and clinical implications. J Hepatol 2016;64:S32S40.CrossRefGoogle ScholarPubMed
Dawson, PA. Hepatic bile acid uptake in humans and mice: multiple pathways and expanding potential role for gut-liver signaling. Hepatology 2017;66:1384–6.Google Scholar
Slijepcevic, D, Roscam Abbing, RLP, Katafuchi, T, Blank, A, Donkers, JM, van Hoppe, S, de Waart, DR, et al. Hepatic uptake of conjugated bile acids is mediated by both sodium taurocholate cotransporting polypeptide and organic anion transporting polypeptides and modulated by intestinal sensing of plasma bile acid levels in mice. Hepatology 2017;66:1631–43.Google Scholar
Slijepcevic, D, van de Graaf, SF. Bile acid uptake transporters as targets for therapy. Dig Dis 2017;35:251–8.Google Scholar
Vaz, FM, Paulusma, CC, Huidekoper, H, de Ru, M, Lim, C, Koster, J, Ho-Mok, K, et al. Sodium taurocholate cotransporting polypeptide (SLC10A1) deficiency: conjugated hypercholanemia without a clear clinical phenotype. Hepatology 2015;61:260–7.CrossRefGoogle ScholarPubMed
Karpen, SJ, Dawson, PA. Not all (bile acids) who wander are lost: the first report of a patient with an isolated NTCP defect. Hepatology 2015;61:24–7.Google Scholar
Suchy, FJ, Ananthanarayanan, M. Bile salt excretory pump: biology and pathobiology. J Pediatr Gastroenterol Nutr 2006;43(Suppl1):S1016.Google Scholar
Stieger, B. Role of the bile salt export pump, BSEP, in acquired forms of cholestasis. Drug Metabolism Reviews 2010;42:437–45. http://dx.doi.org/10.3109/03602530903492004Google Scholar
Davit-Spraul, A, Fabre, M, Branchereau, S, Baussan, C, Gonzales, E, Stieger, B, Bernard, O, et al. ATP8B1 and ABCB11 analysis in 62 children with normal gamma-glutamyl transferase progressive familial intrahepatic cholestasis (PFIC): phenotypic differences between PFIC1 and PFIC2 and natural history. Hepatology 2010;51:1645–55.Google Scholar
Groen, A, Romero, MR, Kunne, C, Hoosdally, SJ, Dixon, PH, Wooding, C, Williamson, C, et al. Complementary functions of the flippase ATP8B1 and the floppase ABCB4 in maintaining canalicular membrane integrity. Gastroenterology 2011;141:1927–37.CrossRefGoogle ScholarPubMed
Keitel, V, Nies, A, Brom, M, Hummel-Eisenbeiss, J, Spring, H, Keppler, D. A common Dubin-Johnson syndrome mutation impairs protein maturation and transport activity of MRP2 (ABCC2). Am J Physiol Gastrointest Liver Physiol 2003;284:G165–74.Google Scholar
Corpechot, C, Barbu, V, Chazouilleres, O, Broue, P, Girard, M, Roquelaure, B, Chretien, Y, et al. Genetic contribution of ABCC2 to Dubin-Johnson syndrome and inherited cholestatic disorders. Liver Int 2020;40:163–74.CrossRefGoogle ScholarPubMed
Lazaridis, KN, LaRusso, NF. The cholangiopathies. Mayo Clin Proc 2015;90:791800.Google Scholar
Banales, JM, Huebert, RC, Karlsen, T, Strazzabosco, M, LaRusso, NF, Gores, GJ. Cholangiocyte pathobiology. Nat Rev Gastroenterol Hepatol 2019;16:269–81.Google Scholar
Fabris, L, Fiorotto, R, Spirli, C, Cadamuro, M, Mariotti, V, Perugorria, MJ, Banales, JM, et al. Pathobiology of inherited biliary diseases: a roadmap to understand acquired liver diseases. Nat Rev Gastroenterol Hepatol 2019;16:497511.Google Scholar
Beuers, U, Maroni, L, Elferink, RO. The biliary HCO(3)(-) umbrella: experimental evidence revisited. Curr Opin Gastroenterol 2012;28:253–7.Google Scholar
Maillette de Buy Wenniger, LJ, Hohenester, S, Maroni, L, van Vliet, SJ, Oude Elferink, RP, Beuers, U. The cholangiocyte glycocalyx stabilizes the “biliary HCO3 umbrella”: an integrated line of defense against toxic bile acids. Dig Dis 2015;33:397407.Google Scholar
Masyuk, AI, Masyuk, TV, LaRusso, NF. Cholangiocyte primary cilia in liver health and disease. Dev Dyn 2008;237:2007–12.Google Scholar
Larusso, NF, Masyuk, TV. The role of cilia in the regulation of bile flow. Dig Dis 2011;29:612.Google Scholar
Perugorria, MJ, Masyuk, TV, Marin, JJ, Marzioni, M, Bujanda, L, LaRusso, NF, Banales, JM. Polycystic liver diseases: advanced insights into the molecular mechanisms. Nat Rev Gastroenterol Hepatol 2014;11:750–61.Google Scholar
Ghallab, A, Hofmann, U, Sezgin, S, Vartak, N, Hassan, R, Zaza, A, Godoy, P, et al. Bile microinfarcts in cholestasis are initiated by rupture of the apical hepatocyte membrane and cause shunting of bile to sinusoidal blood. Hepatology 2019;69:666–83.CrossRefGoogle ScholarPubMed
Watkins, JB, Ingall, D, Szczepanik, P, Klein, PD, Lester, R. Bile-salt metabolism in the newborn. Measurement of pool size and synthesis by stable isotope technic. N Engl J Med 1973;288:431–4.Google Scholar
Suchy, FJ, Balistreri, WF, Heubi, JE, Searcy, JE, Levin, RS. Physiologic cholestasis: elevation of the primary serum bile acid concentrations in normal infants. Gastroenterology 1981;80:1037–41.Google Scholar
Balistreri, WF, Heubi, JE, Suchy, FJ. Immaturity of the enterohepatic circulation in early life: factors predisposing to “physiologic” maldigestion and cholestasis. J Pediatr Gastroenterol Nutr 1983;2:346–54.Google Scholar
Khandekar, G, Llewellyn, J, Kriegermeier, A, Waisbourd-Zinman, O, Johnson, N, Du, Y, Giwa, R, et al. Coordinated development of the mouse extrahepatic bile duct: implications for neonatal susceptibility to biliary injury. J Hepatol 2020;72:135–45.Google Scholar
Karpen, SJ, Trauner, M. The new therapeutic frontier–nuclear receptors and the liver. J Hepatol 2010;52:455–62.Google Scholar
Wagner, M, Zollner, G, Trauner, M. Nuclear receptors in liver disease. Hepatology 2011;53:1023–34.Google Scholar
Arab, JP, Karpen, SJ, Dawson, PA, Arrese, M, Trauner, M. Bile acids and nonalcoholic fatty liver disease: molecular insights and therapeutic perspectives. Hepatology 2017;65:350–62.Google Scholar
Karpen, SJ, Trauner, M. The new therapeutic frontier–nuclear receptors and the liver. J Hepatol 2010;52:455–62.CrossRefGoogle ScholarPubMed
Keitel, V, Droge, C, Haussinger, D. Targeting FXR in cholestasis. Handb Exp Pharmacol 2019;256:299324.Google Scholar
Keitel, V, Haussinger, D. Role of TGR5 (GPBAR1) in liver disease. Semin Liver Dis 2018;38:333–9.Google Scholar
Nevens, F, Andreone, P, Mazzella, G, Strasser, SI, Bowlus, C, Invernizzi, P, Drenth, JP, et al. A placebo-controlled trial of obeticholic acid in primary biliary cholangitis. N Engl J Med 2016;375:631–43.Google Scholar
Chen, XQ, Wang, LL, Shan, QW, Tang, Q, Deng, YN, Lian, SJ, Yun, X. A novel heterozygous NR1H4 termination codon mutation in idiopathic infantile cholestasis. World J Pediatr 2012;8:6771.Google Scholar
Gomez-Ospina, N, Potter, CJ, Xiao, R, Manickam, K, Kim, MS, Kim, KH, Shneider, BL, et al. Mutations in the nuclear bile acid receptor FXR cause progressive familial intrahepatic cholestasis. Nat Commun 2016;7:10713.Google Scholar
Karlsen, TH, Lammert, F, Thompson, RJ. Genetics of liver disease: from pathophysiology to clinical practice. J Hepatol 2015;62:S6S14.Google Scholar
Nicolaou, M, Andress, EJ, Zolnerciks, JK, Dixon, PH, Williamson, C, Linton, KJ. Canalicular ABC transporters and liver disease. J Pathol 2012;226:300–15.Google Scholar
Grammatikopoulos, T, Sambrotta, M, Strautnieks, S, Foskett, P, Knisely, AS, Wagner, B, Deheragoda, M, et al. Mutations in DCDC2 (doublecortin domain containing protein 2) in neonatal sclerosing cholangitis. J Hepatol 2016;65:1179–87.Google Scholar
Maddirevula, S, Alhebbi, H, Alqahtani, A, Algoufi, T, Alsaif, HS, Ibrahim, N, Abdulwahab, F, et al. Identification of novel loci for pediatric cholestatic liver disease defined by KIF12, PPM1F, USP53, LSR, and WDR83OS pathogenic variants. Genet Med 2019;21:1164–72.Google Scholar
Unlusoy Aksu, A, Das, SK, Nelson-Williams, C, Jain, D, Ozbay Hosnut, F, Evirgen Sahin, G, Lifton, RP, et al. Recessive mutations in KIF12 cause high gamma-glutamyltransferase cholestasis. Hepatol Commun 2019;3:471–7.CrossRefGoogle ScholarPubMed
Sultan, M, Rao, A, Elpeleg, O, Vaz, FM, Abu Libdeh, BY, Karpen, SJ, Dawson, PA. Organic solute transporter-beta (SLC51B) deficiency in two brothers with congenital diarrhea and features of cholestasis. Hepatology 2018;68:590–98.Google Scholar
Berauer, JP, Mezina, AI, Okou, DT, Sabo, A, Muzny, DM, Gibbs, RA, Hegde, MR, et al. Identification of polycystic kidney disease 1 like 1 gene variants in children with biliary atresia splenic malformation syndrome. Hepatology 2019;70:899910.CrossRefGoogle ScholarPubMed
Shagrani, M, Burkholder, J, Broering, D, Abouelhoda, M, Faquih, T, El-Kalioby, M, Subhani, SN, et al. Genetic profiling of children with advanced cholestatic liver disease. Clin Genet 2017;92:5261.CrossRefGoogle ScholarPubMed
Nicastro, E, D’Antiga, L. Next generation sequencing in pediatric hepatology and liver transplantation. Liver Transpl 2018;24:282–93.Google Scholar
Osler, W. (1901). Principles and Practice of Medicine (p. 550). New York: Appleton and Company.Google Scholar
Kosters, A, Karpen, SJ. The role of inflammation in cholestasis: clinical and basic aspects. Semin Liver Dis 2010;30:186–94.Google Scholar
Dunham, EC. Septicemia in the new-born. Am J Dis Child 1933;45:229–53.Google Scholar
Utili, R, Abernathy, C, Zimmerman, H. Endotoxin effects on the liver. Life Sci 1977;20:553–68.Google Scholar
Tacke, F, Luedde, T, Trautwein, C. Inflammatory pathways in liver homeostasis and liver injury. Clin Rev Allergy Immunol 2009;36:412.Google Scholar
Strnad, P, Tacke, F, Koch, A, Trautwein, C. Liver – guardian, modifier and target of sepsis. Nat Rev Gastroenterol Hepatol 2017;14:5566.Google Scholar
Moshage, H. Cytokines and the hepatic acute phase response. J Pathol 1997;181:257–66.Google Scholar
Padda, MS, Sanchez, M, Akhtar, AJ, Boyer, JL. Drug-induced cholestasis. Hepatology 2011;53:1377–87.CrossRefGoogle ScholarPubMed
Hoofnagle, JH, Bjornsson, ES. Drug-induced liver injury – types and phenotypes. N Engl J Med 2019;381:264–73.Google Scholar
Woolbright, BL, Jaeschke, H. Mechanisms of inflammatory liver injury and drug-induced hepatotoxicity. Curr Pharmacol Rep 2018;4:346–57.CrossRefGoogle ScholarPubMed
Kearns, G, Abdel-Rahman, S, Alander, S, Blowey, D, Leeder, J, Kauffman, R. Developmental pharmacology – drug disposition, action, and therapy in infants and children. N Engl J Med 2003;349:1157–67.CrossRefGoogle ScholarPubMed
Zollner, G, Wagner, M, Trauner, M. Nuclear receptors as drug targets in cholestasis and drug-induced hepatotoxicity. Pharmacol Ther 2010;126:228–43.Google Scholar
Trauner, M, Baghdasaryan, A, Claudel, T, Fickert, P, Halilbasic, E, Moustafa, T, Zollner, G. Targeting nuclear bile acid receptors for liver disease. Dig Dis 2011;29:98102.Google Scholar
Carter, BA, Karpen, SJ. Intestinal failure-associated liver disease: management and treatment strategies past, present, and future. Semin Liver Dis 2007;27:251–8.Google Scholar
Lee, WS, Sokol, RJ. Intestinal microbiota, lipids, and the pathogenesis of intestinal failure-associated liver disease. J Pediatr 2015;167:519–26.Google Scholar
El Kasmi, KC, Vue, PM, Anderson, AL, Devereaux, MW, Ghosh, S, Balasubramaniyan, N, Fillon, SA, et al. Macrophage-derived IL-1beta/NF-kappaB signaling mediates parenteral nutrition-associated cholestasis. Nat Commun 2018;9:1393.Google Scholar
Rochling, FA, Catron, HA. Intestinal failure-associated liver disease: causes, manifestations and therapies. Curr Opin Gastroenterol 2019;35:126–33.CrossRefGoogle ScholarPubMed
Pironi, L, Sasdelli, AS. Intestinal failure-associated liver disease. Clin Liver Dis 2019;23:279–91.Google Scholar
Josephson, J, Turner, JM, Field, CJ, Wizzard, PR, Nation, PN, Sergi, C, Ball, RO, et al. Parenteral soy oil and fish oil emulsions: impact of dose restriction on bile flow and brain size of parenteral nutrition-fed neonatal piglets. JPEN J Parenter Enteral Nutr 2015;39:677–87.Google Scholar
Koelfat, KVK, Schaap, FG, Hodin, C, Visschers, RGJ, Svavarsson, BI, Lenicek, M, Shiri-Sverdlov, R, et al. Parenteral nutrition dysregulates bile salt homeostasis in a rat model of parenteral nutrition-associated liver disease. Clin Nutr 2017;36:1403–10.Google Scholar
Mutanen, A, Lohi, J, Heikkila, P, Jalanko, H, Pakarinen, MP. Liver inflammation relates to decreased canalicular bile transporter expression in pediatric onset intestinal failure. Ann Surg 2018:268(2):332–9.CrossRefGoogle ScholarPubMed
Lam, CKL, Church, PC, Haliburton, B, Chambers, K, Martincevic, I, Vresk, L, Courtney-Martin, G, et al. Long-term exposure of children to a mixed lipid emulsion is less hepatotoxic than soybean-based lipid emulsion. J Pediatr Gastroenterol Nutr 2018;66:501–4.Google Scholar
Carter, BA, Taylor, OA, Prendergast, DR, Zimmerman, TL, Von Furstenberg, R, Moore, DD, Karpen, SJ. Stigmasterol, a soy lipid-derived phytosterol, is an antagonist of the bile acid nuclear receptor FXR. Pediatr Res 2007;62:301–6.Google Scholar
Isaac, DM, Alzaben, AS, Mazurak, VC, Yap, J, Wizzard, PR, Nation, PN, Zhao, YY, et al. Mixed lipid, fish oil and soybean oil parenteral lipids impact cholestasis, hepatic phytosterol and lipid composition. J Pediatr Gastroenterol Nutr 2019;68:861–7.CrossRefGoogle ScholarPubMed
Rochling, FA, Catron, HA. Intestinal failure-associated liver disease: causes, manifestations and therapies. Curr Opin Gastroenterol 2019;35:126–33.Google Scholar
Fuchs, CD, Schwabl, P, Reiberger, T, Trauner, M. Liver capsule: FXR agonists against liver disease. Hepatology 2016;64:1773.CrossRefGoogle ScholarPubMed
Pradhan-Sundd, T, Zhou, L, Vats, R, Jiang, A, Molina, L, Singh, S, Poddar, M, et al. Dual catenin loss in murine liver causes tight junctional deregulation and progressive intrahepatic cholestasis. Hepatology 2018;67:2320–37.Google Scholar
Pradhan-Sundd, T, Monga, SP. Blood-bile barrier: morphology, regulation, and pathophysiology. Gene Expr 2019;19:6987.Google Scholar
Hofmann, AF. Inappropriate ileal conservation of bile acids in cholestatic liver disease: homeostasis gone awry. Gut 2003;52:1239–41.Google Scholar
Miethke, AG, Zhang, W, Simmons, J, Taylor, AE, Shi, T, Shanmukhappa, SK, Karns, R, et al. Pharmacological inhibition of apical sodium-dependent bile acid transporter changes bile composition and blocks progression of sclerosing cholangitis in multidrug resistance 2 knockout mice. Hepatology 2016;63:512–23.Google Scholar
Fuchs, CD, Paumgartner, G, Mlitz, V, Kunczer, V, Halilbasic, E, Leditznig, N, Wahlstrom, A, et al. Colesevelam attenuates cholestatic liver and bile duct injury in Mdr2(-/-) mice by modulating composition, signalling and excretion of faecal bile acids. Gut 2018;67:1683–91.Google Scholar
Poupon, R. ASBT inhibitors in cholangiopathies – Good for mice, good for men? J Hepatol 2016;64:537–8.Google Scholar
Karpen, SJ. Novel bile acid therapies for liver disease. Gastroenterol Hepatol 2018;14:117–19.Google ScholarPubMed
Hakim, A, Zhang, X, DeLisle, A, Oral, EA, Dykas, D, Drzewiecki, K, Assis, DN, et al. Clinical utility of genomic analysis in adults with idiopathic liver disease. J Hepatol 2019;70:1214–21.Google Scholar
Schaap, FG, Trauner, M, Jansen, PL. Bile acid receptors as targets for drug development. Nat Rev Gastroenterol Hepatol 2014;11:5567.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure [email protected] is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×