Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-ndmmz Total loading time: 0 Render date: 2024-05-14T12:20:20.368Z Has data issue: false hasContentIssue false

Chapter 40 - Systemic Disease and the Liver in Children

from Section V - Other Considerations and Issues in Pediatric Hepatology

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

The liver is the largest parenchymal organ in the body and has a complicated circulatory system, with dual afferent blood supply from the portal vein and hepatic artery. Although the liver mass accounts for only 2.5% of adult body weight, it receives nearly 25% of the resting cardiac output and at any given time contains 10–15% of total body blood volume. In addition to serving as an important blood volume reservoir, the liver is also a complex metabolic organ with important functions in biosynthesis, innate immunity, metabolism and toxin/metabolite clearance. By virtue of its size, multiple metabolic functions, and prominent position in the circulatory system, the liver is frequently involved, either as an accomplice or as an innocent bystander, in a range of systemic, circulatory and inflammatory disorders. This chapter reviews hepatic involvement in common, and some uncommon, systemic diseases affecting children and young adults.

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

Brienza, N, et al. Jaundice in critical illness: promoting factors of a concealed reality. Intensive Care Med 2006;32(2):267–74.Google Scholar
Jakob, SM, et al. Effects of systemic arterial hypoperfusion on splanchnic hemodynamics and hepatic arterial buffer response in pigs. Am J Physiol Gastrointest Liver Physiol 2001;280(5):G819–27.Google Scholar
Brienza, N, et al. Effects of PEEP on liver arterial and venous blood flows. Am J Respir Crit Care Med 1995;152(2):504–10.Google Scholar
Parker, GA, Picut, CA. Immune functioning in nonlymphoid organs: the liver. Toxicol Pathol 2011. https://doi.org/10.1177/0192623311428475CrossRefGoogle Scholar
Sander, LE, et al. Hepatic acute-phase proteins control innate immune responses during infection by promoting myeloid-derived suppressor cell function. J Exp Med 2010;207(7):1453–64.Google Scholar
Sakamori, R, et al. Signal transducer and activator of transcription 3 signaling within hepatocytes attenuates systemic inflammatory response and lethality in septic mice. Hepatology 2007;46(5):1564–73.CrossRefGoogle ScholarPubMed
Geier, A, Fickert, P, Trauner, M. Mechanisms of disease: mechanisms and clinical implications of cholestasis in sepsis. Nat Clin Pract Gastroenterol Hepatol 2006;3(10):574–85.Google Scholar
McDonald, B, Kubes, P. Neutrophils and intravascular immunity in the liver during infection and sterile inflammation. Toxicol Pathol 2011. https://doi.org/10.1177/0192623311427570CrossRefGoogle Scholar
Lin, CC, et al. Subacute nonsuppurative cholangitis (cholangitis lenta) in pediatric liver transplant patients. J Pediatr Gastroenterol Nutr 2007;45(2):228–33.Google Scholar
Torous, VF, et al. Cholangitis lenta: a clinicopathologic study of 28 cases. Am J Surg Pathol 2017;41(12):1607–17.Google Scholar
Lefkowitch, JH. Bile ductular cholestasis: an ominous histopathologic sign related to sepsis and “cholangitis lenta.” Hum Pathol 1982;13(1):1924.Google Scholar
Lautt, WW. Regulatory processes interacting to maintain hepatic blood flow constancy: vascular compliance, hepatic arterial buffer response, hepatorenal reflex, liver regeneration, escape from vasoconstriction. Hepatol Res 2007;37(11):891903.CrossRefGoogle ScholarPubMed
Eipel, C, Abshagen, K, Vollmar, B. Regulation of hepatic blood flow: the hepatic arterial buffer response revisited. World J Gastroenterol 2010;16(48):6046–57.CrossRefGoogle ScholarPubMed
Vollmar, B, Menger, MD. The hepatic microcirculation: mechanistic contributions and therapeutic targets in liver injury and repair. Physiol Rev 2009;89(4):1269–339.Google Scholar
Gracia-Sancho, J, Marrone, G, Fernandez-Iglesias, A. Hepatic microcirculation and mechanisms of portal hypertension. Nat Rev Gastroenterol Hepatol 2019;16(4):221–34.CrossRefGoogle ScholarPubMed
Wu, Y, et al. Laser speckle contrast imaging for measurement of hepatic microcirculation during the sepsis: a novel tool for early detection of microcirculation dysfunction. Microvasc Res 2015;97:137–46.Google Scholar
Fan, J, et al. Hemodynamic changes in hepatic sinusoids of hepatic steatosis mice. World J Gastroenterol 2019;25(11):1355–65.Google Scholar
Ebert, EC. Hypoxic liver injury. Mayo Clin Proc 2006;81(9):1232–6.Google Scholar
Jäger, B, et al. Jaundice increases the rate of complications and one-year mortality in patients with hypoxic hepatitis. Hepatology 2012;56(6):2297–304.CrossRefGoogle ScholarPubMed
Peltenburg, HG, et al. Estimation of the fractional catabolic rate constants for the elimination of cytosolic liver enzymes from plasma. Hepatology 1989;10(5):833–9.Google Scholar
Tapper, EB, Sengupta, N, Bonder, A. The incidence and outcomes of ischemic hepatitis: a systematic review with meta-analysis. Am J Med 2015;128(12):1314–21.Google Scholar
Van den Broecke, A, et al. Epidemiology, causes, evolution and outcome in a single-center cohort of 1116 critically ill patients with hypoxic hepatitis. Ann Intensive Care 2018;8(1):15.Google Scholar
Samavati, L, et al. Tumor necrosis factor alpha inhibits oxidative phosphorylation through tyrosine phosphorylation at subunit I of cytochrome c oxidase. J Biol Chem 2008;283(30):21134–44.CrossRefGoogle ScholarPubMed
Horvatits, T, et al. Circulating bile acids predict outcome in critically ill patients. Ann Intensive Care 2017;7(1):48.Google Scholar
Gudnason, HO, Björnsson, ES. Secondary sclerosing cholangitis in critically ill patients: current perspectives. Clin Exp Gastroenterol 2017;10:105–11.Google Scholar
Leonhardt, S, et al. Secondary sclerosing cholangitis in critically ill patients: clinical presentation, cholangiographic features, natural history, and outcome: a series of 16 cases. Medicine 2015;94(49):e2188.Google Scholar
Arcidi, JM Jr., Moore, GW, Hutchins, GM. Hepatic morphology in cardiac dysfunction: a clinicopathologic study of 1000 subjects at autopsy. Am J Pathol 1981;104(2):159–66.Google ScholarPubMed
Matsuda, H, et al. Acute liver dysfunction after modified Fontan operation for complex cardiac lesions. Analysis of the contributing factors and its relation to the early prognosis. J Thorac Cardiovasc Surg 1988;96(2):219–26.CrossRefGoogle Scholar
Camposilvan, S, et al. Liver and cardiac function in the long term after Fontan operation. Ann Thorac Surg 2008;86(1):177–82.Google Scholar
Wu, FM, et al. Liver disease in the patient with Fontan circulation. Congenit Heart Dis 2011;6(3):190201.Google Scholar
Goldberg, DJ, et al. Hepatic fibrosis is universal following Fontan operation, and severity is associated with time from surgery: a liver biopsy and hemodynamic study. J Am Heart Assoc 2017;6(5):pii:e004809. doi:10.1161/JAHA.116.004809Google Scholar
Pike, NA, et al. Clinical profile of the adolescent/adult Fontan survivor. Congenit Heart Dis 2011;6(1):917.Google Scholar
Yang, HK, et al. CT and MR imaging findings of the livers in adults with Fontan palliation: an observational study. Abdom Radiol 2020;45:188202.CrossRefGoogle Scholar
Rutledge, BP, et al. Transplant-amenable hepatocellular carcinoma in a Fontan patient. Case Rep Gastroenterol 2019;13(2):275–9.Google Scholar
Alsaied, T, et al. Relation of magnetic resonance elastography to Fontan failure and portal hypertension. Am J Cardiol 2019;124(9):1454–9.Google Scholar
Chen, H, et al. Budd–Chiari syndrome caused by multiple membranous obstruction of the inferior vena cava in a young man. Ann Vasc Surg 2011;25(8):1139 e57.Google Scholar
Shetty, S, Ghosh, K. Thrombophilic dimension of Budd-Chiari syndrome and portal venous thrombosis–a concise review. Thromb Res 2011;127(6):505–12.Google Scholar
Hernández-Gea, V, et al. Current knowledge in pathophysiology and management of Budd-Chiari syndrome and non-cirrhotic non-tumoral splanchnic vein thrombosis. J Hepatol 2019;71(1):175–99.CrossRefGoogle ScholarPubMed
Qi, X, et al. Review article: the aetiology of primary Budd-Chiari syndrome – differences between the West and China. Aliment Pharmacol Ther 2016;44(11–12):11521167.Google Scholar
Hoekstra, J, et al. Impaired fibrinolysis as a risk factor for Budd-Chiari syndrome. Blood 2010;115(2):388–95.Google Scholar
Zhang, P, et al. Association between JAK2 rs4495487 polymorphism and risk of Budd-Chiari syndrome in China. Gastroenterol Res Pract 2015;2015:807–65.CrossRefGoogle ScholarPubMed
Shukla, A, et al. Budd-Chiari syndrome has different presentations and disease severity during adolescence. Hepatol Int 2018;12(6):560–6.Google Scholar
Coskun, ME, et al. Ruxolitinib treatment in an infant with JAK2+ polycythaemia vera-associated Budd-Chiari syndrome. BMJ Case Rep 2017;2017:pii:bcr-2017-220377. doi: 10.1136/bcr-2017-220377Google Scholar
Karakose, S, et al. Diagnostic value of the JAK2 V617F mutation for latent chronic myeloproliferative disorders in patients with Budd-Chiari syndrome and/or portal vein thrombosis. Turk J Gastroenterol 2015;26(1):42–8.CrossRefGoogle ScholarPubMed
Singh, V, et al. Budd-Chiari syndrome: our experience of 71 patients. J Gastroenterol Hepatol 2000;15(5):550–4.CrossRefGoogle ScholarPubMed
Nobre, S, et al. Primary Budd-Chiari syndrome in children: King’s College Hospital Experience. J Pediatr Gastroenterol Nutr 2017;65(1):93–6.CrossRefGoogle ScholarPubMed
Boozari, B, et al. Ultrasonography in patients with Budd-Chiari syndrome: diagnostic signs and prognostic implications. J Hepatol 2008;49(4):572–80.Google Scholar
Faraoun, SA, et al. Budd-Chiari syndrome: an update on imaging features. Clin Imaging 2016;40(4):637–46.Google Scholar
Wang, L, et al. Diagnosis of Budd-Chiari syndrome: three-dimensional dynamic contrast enhanced magnetic resonance angiography. Abdom Imaging 2011;36(4):399406.CrossRefGoogle ScholarPubMed
Nagral, A, et al. Budd-Chiari syndrome in children: experience with therapeutic radiological intervention. J Pediatr Gastroenterol Nutr 2010;50(1):74–8.Google Scholar
Wang, Q, et al. Angioplasty with versus without routine stent placement for Budd-Chiari syndrome: a randomised controlled trial. Lancet Gastroenterol Hepatol 2019;4(9):686–97.Google Scholar
Sharma, VK, et al. Long-term clinical outcome of Budd-Chiari syndrome in children after radiological intervention. Eur J Gastroenterol Hepatol 2016;28(5):567–75.CrossRefGoogle ScholarPubMed
Koskinas, J, et al. Liver involvement in acute vaso-occlusive crisis of sickle cell disease: prevalence and predisposing factors. Scand J Gastroenterol 2007;42(4):499507.Google Scholar
Allali, S, et al. Hepatobiliary complications in children with sickle cell disease: a retrospective review of medical records from 616 patients. J Clin Med 2019;8(9):pii:E1481. doi: 10.3390/jcm8091481Google Scholar
Ebert, EC, Nagar, M, Hagspiel, KD. Gastrointestinal and hepatic complications of sickle cell disease. Clin Gastroenterol Hepatol 2010;8(6):483–9; quiz e70.Google Scholar
Herrera, JL. Liver in systemic diseases. Clin Liver Dis 2019;23(2):i.Google Scholar
Ahn, H, Li, CS, Wang, W. Sickle cell hepatopathy: clinical presentation, treatment, and outcome in pediatric and adult patients. Pediatr Blood Cancer 2005;45(2):184–90.CrossRefGoogle ScholarPubMed
Mekeel, KL, et al. Liver transplantation in children with sickle-cell disease. Liver Transpl 2007;13(4):505–8.Google Scholar
Emre, S, et al. Liver transplantation in a patient with acute liver failure due to sickle cell intrahepatic cholestasis. Transplantation 2000;69(4):675–6.Google Scholar
Blinder, MA, et al. Successful orthotopic liver transplantation in an adult patient with sickle cell disease and review of the literature. Hematol Rep 2013;5(1):14.Google Scholar
Revel-Vilk, S, et al. The changing face of hepatitis in boys with haemophilia associated with increased prevalence of obesity. Haemophilia 2011;17(4):689–94.Google Scholar
Alvarnas, JC, et al. Acute lymphoblastic leukemia. J Natl Compr Canc Netw 2015;13(10):1240–79.Google Scholar
Denton, CC, et al. Predictors of hepatotoxicity and pancreatitis in children and adolescents with acute lymphoblastic leukemia treated according to contemporary regimens. Pediatr Blood Cancer 2018;65(3). doi: 10.1002/pbc.26891Google Scholar
Hashmi, SK, et al. Incidence and predictors of treatment-related conjugated hyperbilirubinemia during early treatment phases for children with acute lymphoblastic leukemia. Pediatr Blood Cancer 2019;2019:e28063.Google Scholar
Farrow, AC, et al. Serum aminotransferase elevation during and following treatment of childhood acute lymphoblastic leukemia. J Clin Oncol 1997;15(4):1560–6.Google Scholar
Kamal, N, et al. Asparaginase-induced hepatotoxicity: rapid development of cholestasis and hepatic steatosis. Hepatol Int 2019;13(5):641–8.Google Scholar
Aldoss, I, et al. Toxicity profile of repeated doses of PEG-asparaginase incorporated into a pediatric-type regimen for adult acute lymphoblastic leukemia. Eur J Haematol 2016;96(4):375–80.Google Scholar
Schulte, RR, et al. Levocarnitine for asparaginase-induced hepatic injury: a multi-institutional case series and review of the literature. Leuk Lymphoma 2018;59(10):2360–8.Google Scholar
Blackman, A, et al. Levocarnitine and vitamin B complex for the treatment of pegaspargase-induced hepatotoxicity: a case report and review of the literature. J Oncol Pharm Pract 2018;24(5):393–7.Google Scholar
Athale, U, et al. Management of chronic myeloid leukemia in children and adolescents: recommendations from the Children’s Oncology Group CML Working Group. Pediatr Blood Cancer 2019;66(9):e27827.Google Scholar
Pass, AK, et al. Vanishing bile duct syndrome and Hodgkin disease: a case series and review of the literature. J Pediatr Hematol Oncol 2008;30(12):976–80.Google Scholar
Ballonoff, A, et al. Hodgkin lymphoma-related vanishing bile duct syndrome and idiopathic cholestasis: statistical analysis of all published cases and literature review. Acta Oncol 2008;47(5):962–70.Google Scholar
Ghosh, I, Bakhshi, S. Jaundice as a presenting manifestation of pediatric non-Hodgkin lymphoma: etiology, management, and outcome. J Pediatr Hematol Oncol 2010;32(4):e131–5.Google Scholar
McDonald, GB. Hepatobiliary complications of hematopoietic cell transplantation, 40 years on. Hepatology 2010;51(4):1450–60.Google Scholar
Mahadeo, KM, et al. Diagnosis, grading, and treatment recommendations for children, adolescents, and young adults with sinusoidal obstructive syndrome: an international expert position statement. Lancet Haematol 2019. doi: https://doi.org/10.1016/S2352-3026(19)30201-7Google Scholar
Corbacioglu, S, et al. Diagnosis and severity criteria for sinusoidal obstruction syndrome/veno-occlusive disease in pediatric patients: a new classification from the European society for blood and marrow transplantation. Bone Marrow Transplant 2018;53(2):138–45.Google Scholar
McCarville, MB, et al. Hepatic veno-occlusive disease in children undergoing bone-marrow transplantation: usefulness of sonographic findings. Pediatr Radiol 2001;31(2):102–5.CrossRefGoogle ScholarPubMed
Shulman, HM, et al. Utility of transvenous liver biopsies and wedged hepatic venous pressure measurements in sixty marrow transplant recipients. Transplantation 1995;59(7):1015–22.Google Scholar
Cheuk, DK, et al. Interventions for prophylaxis of hepatic veno-occlusive disease in people undergoing haematopoietic stem cell transplantation. Cochrane Database Syst Rev 2015;5:CD009311.Google Scholar
Tay, J, et al. Systematic review of controlled clinical trials on the use of ursodeoxycholic acid for the prevention of hepatic veno-occlusive disease in hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2007;13(2):206–17.Google Scholar
Corbacioglu, S, et al. Defibrotide for prophylaxis of hepatic veno-occlusive disease in paediatric haemopoietic stem-cell transplantation: an open-label, phase 3, randomised controlled trial. Lancet 2012;379(9823):1301–9. doi: 10.1016/S0140-6736(11)61938-7Google Scholar
Bajwa, RPS, et al. Consensus Report by Pediatric Acute Lung Injury and Sepsis Investigators and Pediatric Blood and Marrow Transplantation Consortium Joint Working Committees: Supportive Care Guidelines for Management of Veno-Occlusive Disease in Children and Adolescents, Part 1: Focus on Investigations, Prophylaxis, and Specific Treatment. Biol Blood Marrow Transplant 2017;23(11):1817–25.Google Scholar
Mahadeo, KM, et al. Consensus Report by the Pediatric Acute Lung Injury and Sepsis Investigators and Pediatric Blood and Marrow Transplant Consortium Joint Working Committees on Supportive Care Guidelines for Management of Veno-Occlusive Disease in Children and Adolescents: Part 2-Focus on Ascites, Fluid and Electrolytes, Renal, and Transfusion Issues. Biol Blood Marrow Transplant 2017;23(12):2023–33.Google Scholar
Coppell, JA, et al. Hepatic veno-occlusive disease following stem cell transplantation: incidence, clinical course, and outcome. Biol Blood Marrow Transplant 2010;16(2):157–68.Google Scholar
Pidala, J. Graft-vs-host disease following allogeneic hematopoietic cell transplantation. Cancer Control 2011;18(4):268–76.CrossRefGoogle ScholarPubMed
Matsukuma, KE, et al. Diagnosis and differential diagnosis of hepatic graft versus host disease (GVHD). J Gastrointest Oncol 2016;7(Suppl. 1):S2131.Google Scholar
Ruggiu, M, et al. Utility and safety of liver biopsy in patients with undetermined liver blood test anomalies after allogeneic hematopoietic stem cell transplantation: a monocentric retrospective cohort study. Biol Blood Marrow Transplant 2018;24(12):2523–31.Google Scholar
Oshrine, B, Lehmann, LE, Duncan, CN. Safety and utility of liver biopsy after pediatric hematopoietic stem cell transplantation. J Pediatr Hematol Oncol 2011;33(3):e92–7.Google Scholar
Salomao, M, et al. Histopathology of graft-vs-host disease of gastrointestinal tract and liver: an update. Am J Clin Pathol 2016;145(5):591603.Google Scholar
Shulman, HM, et al. Histopathologic diagnosis of chronic graft-versus-host disease: National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: II. Pathology Working Group Report. Biol Blood Marrow Transplant 2006;12(1):3147.Google Scholar
Jamil, MO, Mineishi, S. State-of-the-art acute and chronic GVHD treatment. Int J Hematol 2015;101(5):452–66.Google Scholar
Cutler, CS, Koreth, J, Ritz, J. Mechanistic approaches for the prevention and treatment of chronic GVHD. Blood 2017;129(1):22–9.Google Scholar
Barshes, NR, et al. Liver transplantation for severe hepatic graft-versus-host disease. An analysis of aggregate survival data. Liver Transplantation 2005;11(5):525–31.Google Scholar
Zeidan, AM, et al. Successful treatment of severe refractory hepatic graft-versus-host disease by cadaveric liver transplant. Leuk Lymphoma 2013;54(12):2756–9.Google Scholar
Tomita, Y, et al. High incidence of fatty liver and insulin resistance in long-term adult survivors of childhood SCT. Bone Marrow Transplant 2011;46(3):416–25.Google Scholar
Behrens, EM, et al. Evaluation of the presentation of systemic onset juvenile rheumatoid arthritis: data from the Pennsylvania Systemic Onset Juvenile Arthritis Registry (PASOJAR). J Rheumatol 2008;35(2):343–8.Google Scholar
Lee, JJY, Schneider, R. Systemic juvenile idiopathic arthritis. Pediatr Clin North Am 2018;65(4):691709.CrossRefGoogle ScholarPubMed
Takahashi, A, et al. Clinical features of liver dysfunction in collagen diseases. Hepatol Res 2010;40(11):1092–7.Google Scholar
Hashkes, PJ, et al. The relationship of hepatotoxic risk factors and liver histology in methotrexate therapy for juvenile rheumatoid arthritis. J Peds 1999;134:4752.Google Scholar
Wood, PR, Caplan, L. Drug-induced gastrointestinal and hepatic disease associated with biologic and nonbiologic disease-modifying antirheumatic drugs. Rheum Dis Clin North Am 2018;44(1):2943.CrossRefGoogle ScholarPubMed
Kaplowitz, N, DeLeve, LD. (2007). Drug-Induced Liver Disease, 2nd edn., (pp. xv, 8081). New York: Informa Healthcare.Google Scholar
Shimizu, M, et al. Distinct cytokine profiles of systemic-onset juvenile idiopathic arthritis-associated macrophage activation syndrome with particular emphasis on the role of interleukin-18 in its pathogenesis. Rheumatology 2010;49(9):1645–53.Google Scholar
Minoia, F, et al. Clinical features, treatment, and outcome of macrophage activation syndrome complicating systemic juvenile idiopathic arthritis: a multinational, multicenter study of 362 patients. Arthritis Rheumatol 2014;66(11):3160–9.CrossRefGoogle ScholarPubMed
Ebert, EC, Hagspiel, KD. Gastrointestinal and hepatic manifestations of rheumatoid arthritis. Dig Dis Sci 2011;56(2):295302.Google Scholar
Harry, O, Yasin, S, Brunner, H. Childhood-onset systemic lupus erythematosus: a review and update. J Pediatr 2018;196:22–30.e2.Google Scholar
Gebreselassie, A, Aduli, F, Howell, CD. Rheumatologic diseases and the liver. Clin Liver Dis 2019;23(2):247–61.Google Scholar
Ohira, H, et al. High frequency of anti-ribosomal P antibody in patients with systemic lupus erythematosus-associated hepatitis. Hepatol Res 2004;28(3):137–9.Google Scholar
Vanoni, F, et al. Neonatal systemic lupus erythematosus syndrome: a comprehensive review. Clin Rev Allergy Immunol 2017;53(3):469–76.CrossRefGoogle ScholarPubMed
Lee, LA, Sokol, RJ, Buyon, JP. Hepatobiliary disease in neonatal lupus: prevalence and clinical characteristics in cases enrolled in a national registry. Pediatrics 2002;109(1):E11.Google Scholar
Eladawy, M, et al. Abnormal liver panel in acute kawasaki disease. Pediatr Infect Dis J 2011;30(2):141–4.Google Scholar
Wang, Y, et al. Unique molecular patterns uncovered in Kawasaki disease patients with elevated serum gamma glutamyl transferase levels: implications for intravenous immunoglobulin responsiveness. PLoS One 2016;11(12):e0167434.Google Scholar
Kim, BY, et al. Non-responders to intravenous immunoglobulin and coronary artery dilatation in Kawasaki disease: predictive parameters in Korean children. Korean Circ J 2016;46(4):542–9.Google Scholar
Grammatikopoulos, T, et al. Reduced hepatocellular expression of canalicular transport proteins in infants with neonatal cholestasis and congenital hypopituitarism. J Pediatr 2018;200:181–7.Google Scholar
Malik, R, Hodgson, H. The relationship between the thyroid gland and the liver. QJM 2002;95(9):559–69.Google Scholar
Eshraghian, A, Hamidian Jahromi, A. Non-alcoholic fatty liver disease and thyroid dysfunction: a systematic review. World J Gastroenterol 2014;20(25):8102–9.CrossRefGoogle ScholarPubMed
Elder, CJ, Natarajan, A. Mauriac syndrome–a modern reality. J Pediatr Endocrinol Metab 2010;23(3):311–13.Google Scholar
Sumida, Y, Yoneda, M. Glycogen hepatopathy: an under-recognized hepatic complication of uncontrolled type 1 diabetes mellitus. Intern Med 2018;57(8):1063–4.CrossRefGoogle ScholarPubMed
Rubio-Tapia, A, Murray, JA. The liver and celiac disease. Clin Liver Dis 2019;23(2):167–76.Google Scholar
Mounajjed, T, et al. The liver in celiac disease: clinical manifestations, histologic features, and response to gluten-free diet in 30 patients. Am J Clin Pathol 2011;136(1):128–37.Google Scholar
Finn, PF, Dice, JF. Proteolytic and lipolytic responses to starvation. Nutrition 2006;22(7–8):830–44.Google Scholar
Grover, Z, Ee, LC. Protein energy malnutrition. Pediatr Clin North Am 2009;56(5):1055–68.Google Scholar
Fong, HF, et al. Prevalence and predictors of abnormal liver enzymes in young women with anorexia nervosa. J Pediatr 2008;153(2):247–53.Google Scholar
Rautou, PE, et al. Acute liver cell damage in patients with anorexia nervosa: a possible role of starvation-induced hepatocyte autophagy. Gastroenterology 2008;135(3):840–8, 848.e1–3.Google Scholar
Nadelson, AC, et al. Expanding the differential diagnosis for transaminitis in patients with anorexia nervosa. J Gen Intern Med 2017;32(4):486–9.CrossRefGoogle ScholarPubMed
De Caprio, C, et al. Severe acute liver damage in anorexia nervosa: two case reports. Nutrition 2006;22(5):572–5.CrossRefGoogle ScholarPubMed
Rosen, E, et al. Hepatic complications of anorexia nervosa. Dig Dis Sci 2017;62(11):2977–81.Google Scholar
Navarro, VJ, et al. Liver injury from herbal and dietary supplements. Hepatology 2017;65(1):363–73.Google Scholar
García-Cortés, M, et al. Hepatotoxicity by dietary supplements: a tabular listing and clinical characteristics.Int J Mol Sci 2016;17(4):537.Google Scholar
Dulai, PS, Rothstein, RI. Disseminated sarcoidosis presenting as granulomatous gastritis: a clinical review of the gastrointestinal and hepatic manifestations of sarcoidosis. J Clin Gastroenterol 2012;46(5):367–74. doi: 10.1097/MCG.0b013e318247106bGoogle Scholar
Kumar, M, Herrera, JL. Sarcoidosis and the liver. Clin Liver Dis 2019;23(2):331–43.Google Scholar
Cremers, J, et al. Liver-test abnormalities in sarcoidosis. Eur J Gastroenterol Hepatol 2012;24(1):1724.Google Scholar
Vuppalanchi, R, et al. Effects of liver biopsy sample length and number of readings on sampling variability in nonalcoholic Fatty liver disease. Clin Gastroenterol Hepatol 2009;7(4):481–6.Google Scholar
Patnaik, MM, Kamath, PS, Simonetto, DA. Hepatic manifestations of telomere biology disorders. J Hepatol 2018;69(3):736–43.Google Scholar
Mangaonkar, AA, Patnaik, MM. Short telomere syndromes in clinical practice: bridging bench and bedside. Mayo Clin Proc 2018;93(7):904–16.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org 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
×