Journal Home > Volume 1 , Issue 3

Congenital biliary tract malformations are a series of rare but extremely serious diseases that mainly include biliary atresia and biliary hypoplasia (referred to as Alagille syndrome). The rapid progression of biliary atresia and Alagille syndrome results in jaundice, cholestatic liver disease, cirrhosis, and even liver failure. In most cases, supportive or clinically specific therapies cannot achieve satisfactory outcomes. Therefore, liver transplantation (especially living donor liver transplantation) may be required. As many studies have elucidated the role of genetic factors and the molecular mechanism of congenital biliary tract malformations, experimental therapies such as organoid transplantation, cell therapy, and immunotherapy have been proved to be feasible. These advanced methods have shown outstanding advantages, particularly in patients with end-stage biliary tract malformations, surgery failure, and other problems that cannot be solved by conventional therapies. This review article discusses the potential pathogenesis of and promising therapeutic strategies for biliary tract malformations.


menu
Abstract
Full text
Outline
About this article

Advanced therapies for congenital biliary tract malformation: From bench to bedside

Show Author's information Yixuan Shaoa,1Xinyu Yanga,c,d,1Hao Chena,c,dDi Lua,c,dYonggang Huanga,b( )Xiao Xua,c,d ( )
Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
Department of Hernia and Abdominal Wall Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
Institute of Organ Transplantation, Zhejiang University, Hangzhou, 310003, China
Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, China

1 These authors contributed equally to this work.

Abstract

Congenital biliary tract malformations are a series of rare but extremely serious diseases that mainly include biliary atresia and biliary hypoplasia (referred to as Alagille syndrome). The rapid progression of biliary atresia and Alagille syndrome results in jaundice, cholestatic liver disease, cirrhosis, and even liver failure. In most cases, supportive or clinically specific therapies cannot achieve satisfactory outcomes. Therefore, liver transplantation (especially living donor liver transplantation) may be required. As many studies have elucidated the role of genetic factors and the molecular mechanism of congenital biliary tract malformations, experimental therapies such as organoid transplantation, cell therapy, and immunotherapy have been proved to be feasible. These advanced methods have shown outstanding advantages, particularly in patients with end-stage biliary tract malformations, surgery failure, and other problems that cannot be solved by conventional therapies. This review article discusses the potential pathogenesis of and promising therapeutic strategies for biliary tract malformations.

Keywords: Immunotherapy, Biliary tract malformation, Biliary atresia, Alagille syndrome, Pediatric liver transplantation, Organoid

References(106)

[1]

Bezerra JA, Wells RG, Mack CL, et al. Biliary atresia: clinical and research challenges for the twenty-first century. Hepatology 2018;68: 1163–73. https://doi.org/10.1002/hep.29905.

[2]
Diaz-Frias J, Kondamudi NP. Alagille syndrome. In: StatPearls, treasure island (FL). StatPearls Publishing; 2022.
[3]

Lakshminarayanan B, Davenport M. Biliary atresia: a comprehensive review. J Autoimmun 2016;73:1–9. https://doi.org/10.1016/j.jaut.2016.06.005.

[4]

Petersen C, Davenport M. Aetiology of biliary atresia: what is actually known? Orphanet J Rare Dis 2013;8:128. https://doi.org/10.1186/1750-1172-8-128.

[5]

Davit-Spraul A, Baussan C, Hermeziu B, et al. CFC1 gene involvement in biliary atresia with polysplenia syndrome. J Pediatr Gastroenterol Nutr 2008;46:111–2. https://doi.org/10.1097/01.mpg.0000304465.60788.f4.

[6]

Caponcelli E, Knisely AS, Davenport M. Cystic biliary atresia: an etiologic and prognostic subgroup. J Pediatr Surg 2008;43:1619–24. https://doi.org/10.1016/j.jpedsurg.2007.12.058.

[7]

Hill R, Quaglia A, Hussain M, et al. Th-17 cells infiltrate the liver in human biliary atresia and are related to surgical outcome. J Pediatr Surg 2015;50:1297–303. https://doi.org/10.1016/j.jpedsurg.2015.02.005.

[8]

Hartley JL, Davenport M, Kelly DA. Biliary atresia. Lancet 2009;374:1704–13. https://doi.org/10.1016/S0140-6736(09)60946-6.

[9]

Garcia-Barcelo M-M, Yeung M-Y, Miao X-P, et al. Genome-wide association study identifies a susceptibility locus for biliary atresia on 10q24.2. Hum Mol Genet 2010;19:2917–25. https://doi.org/10.1093/hmg/ddq196.

[10]

Leyva-Vega M, Gerfen J, Thiel BD, et al. Genomic alterations in biliary atresia suggest region of potential disease susceptibility in 2q37.3. Am J Med Genet 2010; 152A: 886–95. https://doi.org/10.1002/ajmg.a.33332.

[11]

Girard M, Panasyuk G. Genetics in biliary atresia. Curr Opin Gastroenterol 2019; 35:73–81. https://doi.org/10.1097/MOG.0000000000000509.

[12]

Sangkhathat S, Laochareonsuk W, Maneechay W, et al. Variants associated with infantile cholestatic syndromes detected in extrahepatic biliary atresia by whole exome studies: a 20-case series from Thailand. J Pediatr Genet 2018;7:67–73. https://doi.org/10.1055/s-0038-1632395.

[13]
Alagille D, Habib EC, Thomassin N. L'atresie des voies biliaires intrahepatiques avec voies biliaires extrahepatiques permeables chez l'enfant. Paris: Editions Medicales Flammarion; 1969. p. 301–18.
[14]

Fabris L, Fiorotto R, Spirli C, et al. Pathobiology of inherited biliary diseases: a roadmap to understand acquired liver diseases. Nat Rev Gastroenterol Hepatol 2019;16:497–511. https://doi.org/10.1038/s41575-019-0156-4.

[15]

McDaniell R, Warthen DM, Sanchez-Lara PA, et al. NOTCH2 mutations cause Alagille syndrome, a heterogeneous disorder of the notch signaling pathway. Am J Hum Genet 2006;79:169–73. https://doi.org/10.1086/505332.

[16]

Gunadi Null, Kaneshiro M, Okamoto T, et al. Outcomes of liver transplantation for Alagille syndrome after Kasai portoenterostomy: Alagille Syndrome with agenesis of extrahepatic bile ducts at porta hepatis. J Pediatr Surg 2019;54:2387–91. https://doi.org/10.1016/j.jpedsurg.2019.04.022.

[17]

Dědič T, Jirsa M, Keil R, et al. Alagille syndrome mimicking biliary atresia in early infancy. PLoS One 2015;10:e0143939. https://doi.org/10.1371/journal.pone.0143939.

[18]

Rajagopalan R, Gilbert MA, McEldrew DA, et al. Genome sequencing increases diagnostic yield in clinically diagnosed Alagille syndrome patients with previously negative test results. Genet Med 2021;23:323–30. https://doi.org/10.1038/s41436-020-00989-8.

[19]

Abdel Razek AAK, Abdalla A, Elfar R, et al. Assessment of diffusion tensor imaging parameters of hepatic parenchyma for differentiation of biliary atresia from Alagille syndrome. Korean J Radiol 2020;21:1367–73. https://doi.org/10.3348/kjr.2019.0824.

[20]

Fujishiro J, Suzuki K, Watanabe M, et al. Outcomes of Alagille syndrome following the Kasai operation: a systematic review and meta-analysis. Pediatr Surg Int 2018; 34:1073–7. https://doi.org/10.1007/s00383-018-4316-3.

[21]

Emerick KM, Rand EB, Goldmuntz E, et al. Features of Alagille syndrome in 92 patients: frequency and relation to prognosis. Hepatology 1999;29:822–9. https://doi.org/10.1002/hep.510290331.

[22]

Kaye AJ, Rand EB, Munoz PS, et al. Effect of Kasai procedure on hepatic outcome in Alagille syndrome. J Pediatr Gastroenterol Nutr 2010;51:319–21. https://doi.org/10.1097/MPG.0b013e3181df5fd8.

[23]

Kriegermeier A, Green R. Pediatric cholestatic liver disease: review of bile acid metabolism and discussion of current and emerging therapies. Front Med 2020;7: 149. https://doi.org/10.3389/fmed.2020.00149.

[24]

Burns J, Davenport M. Adjuvant treatments for biliary atresia. Transl Pediatr 2020;9:253–65. https://doi.org/10.21037/tp.2016.10.08.

[25]

Emerick KM, Elias MS, Melin-Aldana H, et al. Bile composition in Alagille syndrome and PFIC patients having partial external biliary diversion. BMC Gastroenterol 2008;8:47. https://doi.org/10.1186/1471-230X-8-47.

[26]

Valamparampil JJ, Reddy MS, Shanmugam N, et al. Living donor liver transplantation in Alagille syndrome-Single center experience from south Asia. Pediatr Transplant 2019;23:e13579. https://doi.org/10.1111/petr.13579.

[27]

Sundaram SS, Mack CL, Feldman AG, et al. Biliary atresia: indications and timing of liver transplantation and optimization of pretransplant care. Liver Transpl 2017;23:96–109. https://doi.org/10.1002/lt.24640.

[28]

Shneider BL, Brown MB, Haber B, et al. A multicenter study of the outcome of biliary atresia in the United States, 1997 to 2000. J Pediatr 2006;148:467–74. https://doi.org/10.1016/j.jpeds.2005.12.054.

[29]

Hadžić N, Quaglia A, Portmann B, et al. Hepatocellular carcinoma in biliary atresia: king's College Hospital experience. J Pediatr 2011;159:617–622.e1. https://doi.org/10.1016/j.jpeds.2011.03.004.

[30]

Lampela H, Kosola S, Heikkilä P, et al. Native liver histology after successful portoenterostomy in biliary atresia. J Clin Gastroenterol 2014;48:721–8. https://doi.org/10.1097/MCG.0000000000000013.

[31]

Kerola A, Lampela H, Lohi J, et al. Molecular signature of active fibrogenesis prevails in biliary atresia after successful portoenterostomy. Surgery 2017;162: 548–56. https://doi.org/10.1016/j.surg.2017.04.013.

[32]

Fanna M, Masson G, Capito C, et al. Management of biliary atresia in France 1986 to 2015: long-term results. J Pediatr Gastroenterol Nutr 2019;69:416–24. https://doi.org/10.1097/MPG.0000000000002446.

[33]

Pakarinen MP, Johansen LS, Svensson JF, et al. Outcomes of biliary atresia in the Nordic countries - a multicenter study of 158 patients during 2005-2016. J Pediatr Surg 2018;53:1509–15. https://doi.org/10.1016/j.jpedsurg.2017.08.048.

[34]

El-Araby HA, Saber MA, Radwan NM, et al. SOX9 in biliary atresia: new insight for fibrosis progression. Hepatobiliary Pancreat Dis Int 2021;20:154–62. https://doi.org/10.1016/j.hbpd.2020.12.007.

[35]

Kasahara M, Umeshita K, Sakamoto S, et al. Liver transplantation for biliary atresia: a systematic review. Pediatr Surg Int 2017;33:1289–95. https://doi.org/10.1007/s00383-017-4173-5.

[36]

Cortes-Cerisuelo M, Boumpoureka C, Cassar N, et al. Liver transplantation for biliary atresia in adulthood: single-centre surgical experience. J Clin Med 2021;10: 4969. https://doi.org/10.3390/jcm10214969.

[37]

Yoeli D, Choudhury RA, Sundaram SS, et al. Primary vs. salvage liver transplantation for biliary atresia: a retrospective cohort study. J Pediatr Surg 2022;S0022–3468(22): 4–5. https://doi.org/10.1016/j.jpedsurg.2021.12.027.0000.

[38]

Kamath BM, Ye W, Goodrich NP, et al. Outcomes of childhood cholestasis in Alagille syndrome: results of a multicenter observational study. Hepatol Commun 2020;4:387–98. https://doi.org/10.1002/hep4.1468.

[39]
Vandriel S, Wang J-S, Li L, et al. Clinical features and outcomes in an international cohort of 731 Alagille syndrome patients from 19 countries. 2019. https://doi.org/10.1002/hep.30940.
[40]

Superina R. Biliary atresia and liver transplantation: results and thoughts for primary liver transplantation in select patients. Pediatr Surg Int 2017;33: 1297–304. https://doi.org/10.1007/s00383-017-4174-4.

[41]

Utterson EC, Shepherd RW, Sokol RJ, et al. Biliary atresia: clinical profiles, risk factors, and outcomes of 755 patients listed for liver transplantation. J Pediatr 2005;147:180–5. https://doi.org/10.1016/j.jpeds.2005.04.073.

[42]

Zhang R, Zhu Z-J, Sun L-Y, et al. Outcomes of liver transplantation using pediatric deceased donor livers: a single-center analysis of 102 donors. Chin Med J (Engl) 2018;131:677–83. https://doi.org/10.4103/0366-6999.226901.

[43]

Arnon R, Annunziato RA, D'Amelio G, et al. Liver transplantation for biliary atresia: is there a difference in outcome for infants? J Pediatr Gastroenterol Nutr 2016;62:220–5. https://doi.org/10.1097/MPG.0000000000000986.

[44]

Raices M, Czerwonko ME, Ardiles V, et al. Short- and long-term outcomes after live-donor transplantation with hyper-reduced liver grafts in low-weight pediatric recipients. J Gastrointest Surg 2019;23:2411–20. https://doi.org/10.1007/s11605-019-04188-y.

[45]

Cha DJ, Alfrey EJ, Desai DM, et al. Increased risk of vascular thrombosis in pediatric liver transplant recipients with thrombophilia. J Surg Res 2015;199: 671–5. https://doi.org/10.1016/j.jss.2015.07.043.

[46]

Kanazawa H, Sakamoto S, Fukuda A, et al. Living-donor liver transplantation with hyperreduced left lateral segment grafts: a single-center experience. Transplantation 2013;95:750–4. https://doi.org/10.1097/TP.0b013e31827a93b4.

[47]

Alexopoulos SP, Nekrasov V, Cao S, et al. Effects of recipient size and allograft type on pediatric liver transplantation for biliary atresia. Liver Transpl 2017;23: 221–33. https://doi.org/10.1002/lt.24675.

[48]

Goldaracena N, Echeverri J, Kehar M, et al. Pediatric living donor liver transplantation with large-for-size left lateral segment grafts. Am J Transplant 2020;20:504–12. https://doi.org/10.1111/ajt.15609.

[49]

Ziogas IA, Ye F, Zhao Z, et al. Mortality determinants in children with biliary atresia awaiting liver transplantation. J Pediatr 2021;228:177–82. https://doi.org/10.1016/j.jpeds.2020.09.005.

[50]

Kamath BM, Yin W, Miller H, et al. Outcomes of liver transplantation for patients with Alagille syndrome: the studies of pediatric liver transplantation experience. Liver Transpl 2012;18:940–8. https://doi.org/10.1002/lt.23437.

[51]

Kim WR, Lake JR, Smith JM, et al. OPTN/SRTR 2016 annual data report: liver. Am J Transplant 2018;18(Suppl 1): 172–253. https://doi.org/10.1111/ajt.14559.

[52]

Lin T-S, Co JS, Chen C-L, et al. Optimizing biliary outcomes in living donor liver transplantation: evolution towards standardization in a high-volume center. Hepatobiliary Pancreat Dis Int 2020;19:324–7. https://doi.org/10.1016/j.hbpd.2020.06.012.

[53]

Fisher RA. Living donor liver transplantation: eliminating the wait for death in end-stage liver disease? Nat Rev Gastroenterol Hepatol 2017;14:373–82. https://doi.org/10.1038/nrgastro.2017.2.

[54]

Levitsky J, Kaneku H, Jie C, et al. Donor-specific HLA antibodies in living versus deceased donor liver transplant recipients. Am J Transplant 2016;16:2437–44. https://doi.org/10.1111/ajt.13757.

[55]

Goldaracena N, Barbas AS. Living donor liver transplantation. Curr Opin Organ Transplant 2019;24:131–7. https://doi.org/10.1097/MOT.0000000000000610.

[56]

Kasahara M, Umeshita K, Sakamoto S, et al. Living donor liver transplantation for biliary atresia: an analysis of 2085 cases in the registry of the Japanese Liver Transplantation Society. Am J Transplant 2018;18:659–68. https://doi.org/10.1111/ajt.14489.

[57]

Toso C, Ris F, Mentha G, et al. Potential impact of in situ liver splitting on the number of available grafts. Transplantation 2002;74:222–6. https://doi.org/10.1097/00007890-200207270-00013.

[58]

Mogul DB, Luo X, Bowring MG, et al. Fifteen-year trends in pediatric liver transplants: split, whole deceased, and living donor grafts. J Pediatr 2018;196: 148–153.e2. https://doi.org/10.1016/j.jpeds.2017.11.015.

[59]

Mogul DB, Luo X, Garonzik-Wang J, et al. Expansion of the liver donor supply through greater use of split-liver transplantation: identifying optimal recipients. Liver Transpl 2019;25:119–27. https://doi.org/10.1002/lt.25340.

[60]

Marques HP, Barros I, Li J, et al. Current update in domino liver transplantation. Int J Surg 2020;82S: 163–8. https://doi.org/10.1016/j.ijsu.2020.03.017.

[61]

Herden U, Li J, Fischer L, et al. The first case of domino-split-liver transplantation in maple syrup urine disease. Pediatr Transplant 2017;21. https://doi.org/10.1111/petr.12993.

[62]

Shimizu S, Sakamoto S, Fukuda A, et al. Surgical technique and the long-term outcomes of pediatric living donor domino liver transplantation from patients with maple syrup urine disease. Pediatr Transplant 2022;26:e14174. https://doi.org/10.1111/petr.14174.

[63]

Macías-Rosales R, Larrosa-Haro A, Ortíz-Gabriel G, et al. Effectiveness of enteral versus oral nutrition with a medium-chain triglyceride formula to prevent malnutrition and growth impairment in infants with biliary atresia. J Pediatr Gastroenterol Nutr 2016; 62:101–9. https://doi.org/10.1097/MPG.0000000000000909.

[64]

Kamath BM, Baker A, Houwen R, et al. Systematic review: the epidemiology, natural history, and burden of Alagille syndrome. J Pediatr Gastroenterol Nutr 2018;67:148–56. https://doi.org/10.1097/MPG.0000000000001958.

[65]

McGahan RK, Tang JE, Iyer MH, et al. Combined liver kidney transplant in adult patient with Alagille syndrome and pulmonary hypertension. Semin CardioThorac Vasc Anesth 2021;25:191–5. https://doi.org/10.1177/10892532211008742.

[66]

Khanna R, Verma SK. Pediatric hepatocellular carcinoma. World J Gastroenterol 2018;24:3980–99. https://doi.org/10.3748/wjg.v24.i35.3980.

[67]

Boster JM, Feldman AG, Mack CL, et al. Malnutrition in biliary atresia: assessment, management, and outcomes. Liver Transpl 2022;28:483–92. https://doi.org/10.1002/lt.26339.

[68]

Suchy FJ, Sokol RJ, Balistreri WF, editors. Liver disease in children. 4th ed. Cambridge: Cambridge University Press; 2014. https://doi.org/10.1017/CBO9781139012102.

[69]

Feldman AG, Sokol RJ. Neonatal cholestasis: emerging molecular diagnostics and potential novel therapeutics. Nat Rev Gastroenterol Hepatol 2019;16:346–60. https://doi.org/10.1038/s41575-019-0132-z.

[70]

Mohammad S, Grimberg A, Rand E, et al. Long-term linear growth and puberty in pediatric liver transplant recipients. J Pediatr 2013;163:1354–60. https://doi.org/10.1016/j.jpeds.2013.06.039. e1-7.

[71]

Mitchell E, Gilbert M, Loomes KM. Alagille syndrome. Clin Liver Dis 2018;22: 625–41. https://doi.org/10.1016/j.cld.2018.06.001.

[72]

Leung DH, Sorensen LG, Ye W, et al. Neurodevelopmental outcomes in children with inherited liver disease and native liver. J Pediatr Gastroenterol Nutr 2022;74: 96–103. https://doi.org/10.1097/MPG.0000000000003337.

[73]

Tambucci R, de Magnée C, Szabo M, et al. Sequential treatment of biliary atresia with kasai hepatoportoenterostomy and liver transplantation: benefits, risks, and outcome in 393 children. Front Pediatr 2021;9:697581. https://doi.org/10.3389/fped.2021.697581.

[74]

Sato K, Zhang W, Safarikia S, et al. Organoids and spheroids as models for studying cholestatic liver injury and cholangiocarcinoma. Hepatology 2021;74: 491–502. https://doi.org/10.1002/hep.31653.

[75]

Shiota J, Samuelson LC, Razumilava N. Hepatobiliary organoids and their applications for studies of liver health and disease: are we there yet? Hepatology 2021;74:2251–63. https://doi.org/10.1002/hep.31772.

[76]

Huch M, Gehart H, van Boxtel R, et al. Long-term culture of genome-stable bipotent stem cells from adult human liver. Cell 2015;160:299–312. https://doi.org/10.1016/j.cell.2014.11.050.

[77]

Amarachintha SP, Mourya R, Ayabe H, et al. Biliary organoids uncover delayed epithelial development and barrier function in biliary atresia. Hepatology 2022; 75:89–103. https://doi.org/10.1002/hep.32107.

[78]

Rimland CA, Tilson SG, Morell CM, et al. Regional differences in human biliary tissues and corresponding in vitro-derived organoids. Hepatology 2021;73: 247–67. https://doi.org/10.1002/hep.31252.

[79]

Kurial SNT, Willenbring H. Emerging cell therapy for biliary diseases. Science 2021;371:786–7. https://doi.org/10.1126/science.abg3179.

[80]

Takebe T, Sekine K, Enomura M, et al. Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature 2013;499:481–4. https://doi.org/10.1038/nature12271.

[81]

Sampaziotis F, Justin AW, Tysoe OC, et al. Reconstruction of the mouse extrahepatic biliary tree using primary human extrahepatic cholangiocyte organoids. Nat Med 2017;23:954–63. https://doi.org/10.1038/nm.4360.

[82]

Smith Q, Bays J, Li L, et al. Directing cholangiocyte morphogenesis in natural biomaterial scaffolds. Adv Sci (Weinh) 2022;9:e2102698. https://doi.org/10.1002/advs.202102698.

[83]

Schaub JR, Huppert KA, Kurial SNT, et al. De novo formation of the biliary system by TGFβ-mediated hepatocyte transdifferentiation. Nature 2018;557:247–51. https://doi.org/10.1038/s41586-018-0075-5.

[84]

Sampaziotis F, Muraro D, Tysoe OC, et al. Cholangiocyte organoids can repair bile ducts after transplantation in the human liver. Science 2021;371:839–46. https://doi.org/10.1126/science.aaz6964.

[85]

Nunes de Carvalho S, Helal-Neto E, de Andrade DC, et al. Bone marrow mononuclear cell transplantation increases metalloproteinase-9 and 13 and decreases tissue inhibitors of metalloproteinase-1 and 2 expression in the liver of cholestatic rats. Cells Tissues Organs 2013;198:139–48. https://doi.org/10.1159/000353215.

[86]

Kim M-D, Kim S-S, Cha H-Y, et al. Therapeutic effect of hepatocyte growth factorsecreting mesenchymal stem cells in a rat model of liver fibrosis. Exp Mol Med 2014;46:e110. https://doi.org/10.1038/emm.2014.49.

[87]

Pinheiro D, Leirós L, Dáu JBT, et al. Cytokines, hepatic cell profiling and cell interactions during bone marrow cell therapy for liver fibrosis in cholestatic mice. PLoS One 2017;12:e0187970. https://doi.org/10.1371/journal.pone.0187970.

[88]

Watanabe Y, Tsuchiya A, Terai S. The development of mesenchymal stem cell therapy in the present, and the perspective of cell-free therapy in the future. Clin Mol Hepatol 2021;27:70–80. https://doi.org/10.3350/cmh.2020.0194.

[89]

Nguyen TL, Nguyen HP, Ngo DM, et al. Autologous bone marrow mononuclear cell infusion for liver cirrhosis after the Kasai operation in children with biliary atresia. Stem Cell Res Ther 2022;13:108. https://doi.org/10.1186/s13287-022-02762-x.

[90]

Brindley SM, Lanham AM, Karrer FM, et al. Cytomegalovirus-specific T-cell reactivity in biliary atresia at the time of diagnosis is associated with deficits in regulatory T cells. Hepatology 2012;55:1130–8. https://doi.org/10.1002/hep.24807.

[91]

Mitchell P, Afzali B, Lombardi G, et al. The T helper 17-regulatory T cell axis in transplant rejection and tolerance. Curr Opin Organ Transplant 2009;14:326–31. https://doi.org/10.1097/MOT.0b013e32832ce88e.

[92]

Lee GR. The balance of Th17 versus Treg cells in autoimmunity. Int J Mol Sci 2018;19:E730. https://doi.org/10.3390/ijms19030730.

[93]

Ni X, Wang Q, Gu J, et al. Clinical and basic research progress on treg-induced immune tolerance in liver transplantation. Front Immunol 2021;12:535012. https://doi.org/10.3389/fimmu.2021.535012.

[94]

Magee CN, Murakami N, Borges TJ, et al. Notch-1 inhibition promotes immune regulation in transplantation via treg-dependent mechanisms. Circulation 2019; 140:846–63. https://doi.org/10.1161/CIRCULATIONAHA.119.040563.

[95]

Feldman AG, Mack CL. Biliary atresia: cellular dynamics and immune dysregulation. Semin Pediatr Surg 2012;21:192–200. https://doi.org/10.1053/j.sempedsurg.2012.05.003.

[96]

Shen W-J, Chen G, Wang M, et al. Liver fibrosis in biliary atresia. World J Pediatr 2019;15:117–23. https://doi.org/10.1007/s12519-018-0203-1.

[97]
CD4þT Cell Subset Profiling in Biliary Atresia Reveals ICOS– Regulatory T Cells as a Favorable Prognostic Factor - PMC n.d. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6637302/(accessed July 15, 2022).
[98]

Kageyama S, Kadono K, Hirao H, et al. Ischemia-reperfusion injury in allogeneic liver transplantation: a role of CD4 T cells in early allograft injury. Transplantation 2021;105. https://doi.org/10.1097/TP.0000000000003488.1989–97.

[99]

Wang J, Xu Y, Chen Z, et al. Liver immune profiling reveals pathogenesis and therapeutics for biliary atresia. Cell 2020;183:1867–1883.e26. https://doi.org/10.1016/j.cell.2020.10.048.

[100]

Jin Y, Li C, Xu D, et al. Jagged 1-mediated myeloid Notch 1 signaling activates HSF1/Snail and controls NLRP3 inflammasome activation in liver inflammatory injury. Cell Mol Immunol 2020;17:1245–56. https://doi.org/10.1038/s41423-019-0318-x.

[101]

Bai H, Wen J, Gong J-P, et al. Blockade of the Notch 1/Jagged 1 pathway in Kupffer cells aggravates ischemia-reperfusion injury of orthotopic liver transplantation in mice. Autoimmunity 2019;52:176–84. https://doi.org/10.1080/08916934.2019.1637424.

[102]

Tilib Shamoun S, Le Friec G, Spinner N, et al. Immune dysregulation in Alagille syndrome: a new feature of the evolving phenotype. Clin Res Hepatol Gastroenterol 2015;39:566–9. https://doi.org/10.1016/j.clinre.2015.02.003.

[103]

Mitra A, Shanthalingam S, Sherman HL, et al. CD28 signaling drives notch ligand expression on CD4 T cells. Front Immunol 2020;11:735. https://doi.org/10.3389/fimmu.2020.00735.

[104]

Garis M, Garrett-Sinha LA. Notch signaling in B cell immune responses. Front Immunol 2021;11:609324. https://doi.org/10.3389/fimmu.2020.609324.

[105]

Gonzales E, Hardikar W, Stormon M, et al. Efficacy and safety of maralixibat treatment in patients with Alagille syndrome and cholestatic pruritus (ICONIC): a randomised phase 2 study. Lancet 2021;398:1581–92. https://doi.org/10.1016/S0140-6736(21)01256-3.

[106]

Yang X, Lu D, Wang R, et al. Single-cell profiling reveals distinct immune phenotypes that contribute to ischaemia-reperfusion injury after steatotic liver transplantation. Cell Prolif 2021;54:e13116. https://doi.org/10.1111/cpr.13116.

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 22 June 2022
Revised: 19 July 2022
Accepted: 02 August 2022
Published: 30 August 2022
Issue date: September 2022

Copyright

© 2022 The Author(s). Published by Elsevier Ltd on behalf of Tsinghua University Press.

Acknowledgements

None.

Rights and permissions

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return