AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
Home iLIVER Article
Article Link
Collect
Submit Manuscript
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review | Open Access

The pathophysiological mechanism between hypopituitarism and nonalcoholic fatty liver disease

Gastroenterology Department, The First Affiliated Hospital of China Medical University, No.155 North Nanjing Street, Heping District, Shenyang 110001, Liaoning, China
The 3rd Clinical Department of China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
Show Author Information

Abstract

Nonalcoholic fatty liver disease (NAFLD) is a common chronic metabolic liver disease worldwide. It is closely related to diseases of the cardiovascular system and chronic kidney disease. It can also occur secondary to many other diseases. Current research shows that patients with hypopituitarism have a high risk of developing NAFLD. After the adenohypophysis is dominated by hypothalamic hormones, hormones are secreted to act on the corresponding tissues or organs. It is characterized by a decrease in the thyroid hormone, cortisol, and growth hormone levels. In this review, we analyzed the mechanisms related to NAFLD through thyroid secretion, growth hormone secretion, sex hormone, and prolactin axes in patients with hypopituitarism, which will provide information and a theoretical basis for clinical diagnosis and treatment.

References

[1]

Yeliosof O, Gangat M. Diagnosis and management of hypopituitarism. Curr Opin Pediatr 2019;31(4): 531–6.

[2]

Jasim S, Alahdab F, Ahmed AT, et al. Mortality in adults with hypopituitarism: a systematic review and meta-analysis. Endocrine 2017;56(1): 33–42.

[3]
Heidelbaugh JJ. Endocrinology Update: Hypopituitarism, vol. 451. FP Essent; 2016. p. 25–30.
[4]

Perumpail BJ, Khan MA, Yoo ER, et al. Clinical epidemiology and disease burden of nonalcoholic fatty liver disease. World J Gastroenterol. 2017;23(47): 8263–76.

[5]

Friedman SL, Neuschwander-Tetri BA, Rinella M, et al. Mechanisms of NAFLD development and therapeutic strategies. Nat Med 2018;24(7): 908–22.

[6]

Younossi Z, Anstee QM, Marietti M, et al. Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nat Rev Gastroenterol Hepatol 2018;15(1): 11–20.

[7]

Eslam M, Sanyal AJ, George J. MAFLD: a consensus-driven proposed nomenclature for metabolic associated fatty liver disease. Gastroenterology 2020; 158(7): 1999–2014. e1.

[8]

Lin S, Huang J, Wang M, et al. Comparison of MAFLD and NAFLD diagnostic criteria in real world. Liver Int 2020;40(9): 2082–9.

[9]

Yuan XX, Zhu HJ, Pan H, et al. Clinical characteristics of non-alcoholic fatty liver disease in Chinese adult hypopituitary patients. World J Gastroenterol 2019; 25(14): 1741–52.

[10]

Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-hit pathogenesis of nonalcoholic fatty liver disease (NAFLD). Metabolism 2016;65(8): 1038–48.

[11]

Fang YL, Chen H, Wang CL, et al. Pathogenesis of non-alcoholic fatty liver disease in children and adolescence: from "two hit theory" to "multiple hit model. World J Gastroenterol 2018;24(27): 2974–83.

[12]

Cobbina E, Akhlaghi F. Non-alcoholic fatty liver disease (NAFLD) - pathogenesis, classification, and effect on drug metabolizing enzymes and transporters. Drug Metab Rev 2017;49(2): 197–211.

[13]

Sliz E, Sebert S, Wurtz P, et al. NAFLD risk alleles in PNPLA3, TM6SF2, GCKR and LYPLAL1 show divergent metabolic effects. Hum Mol Genet 2018;27(12): 2214–23.

[14]

Abdelmegeed MA, Ha SK, Choi Y, et al. Role of CYP2E1 in mitochondrial dysfunction and hepatic injury by alcohol and non-alcoholic substances. Curr Mol Pharmacol 2017;10(3): 207–25.

[15]

Sinha RA, Bruinstroop E, Singh BK, et al. Nonalcoholic fatty liver disease and hypercholesterolemia: roles of thyroid hormones, metabolites, and agonists. Thyroid 2019;29(9): 1173–91.

[16]

Mullur R, Liu YY, Brent GA. Thyroid hormone regulation of metabolism. Physiol Rev 2014;94(2): 355–82.

[17]

Lugari S, Mantovani A, Nascimbeni F, et al. Hypothyroidism and nonalcoholic fatty liver disease - a chance association? Horm Mol Biol Clin Invest 2018;41(1).

[18]

Chung GE, Kim D, Kim W, et al. Non-alcoholic fatty liver disease across the spectrum of hypothyroidism. J Hepatol 2012;57(1): 150–6.

[19]

Pagadala MR, Zein CO, Dasarathy S, et al. Prevalence of hypothyroidism in nonalcoholic fatty liver disease. Dig Dis Sci 2012;57(2): 528–34.

[20]

Mantovani A, Nascimbeni F, Lonardo A, et al. Association between primary hypothyroidism and nonalcoholic fatty liver disease: a systematic review and meta-analysis. Thyroid 2018;28(10): 1270–84.

[21]

Kim D, Kim W, Joo SK, et al. Subclinical hypothyroidism and low-normal thyroid function are associated with nonalcoholic steatohepatitis and fibrosis. Clin Gastroenterol Hepatol 2018;16(1): 123–131. e1.

[22]

Kim D, Yoo ER, Li AA, et al. Low-normal thyroid function is associated with advanced fibrosis among adults in the United States. Clin Gastroenterol Hepatol 2019;17(11): 2379–81.

[23]

Jaruvongvanich V, Sanguankeo A, Upala S. Nonalcoholic fatty liver disease is not associated with thyroid hormone levels and hypothyroidism: a systematic review and meta-analysis. Eur Thyroid J 2017;6(4): 208–15.

[24]

Martinez EA, Pera G, Arteaga I, et al. Relationship between hypothyroidism and non-alcoholic fatty liver disease in the Spanish population. Med Clin 2020;154(1): 1–6.

[25]

Duntas LH, Brenta G. A renewed focus on the association between thyroid hormones and lipid metabolism. Front Endocrinol (Lausanne) 2018;9: 511.

[26]

Goldberg IJ, Huang LS, Huggins LA, et al. Thyroid hormone reduces cholesterol via a non-LDL receptor-mediated pathway. Endocrinology 2012;153(11): 5143–9.

[27]

Shin DJ, Osborne TF. Thyroid hormone regulation and cholesterol metabolism are connected through Sterol Regulatory Element-Binding Protein-2 (SREBP-2). J Biol Chem 2003;278(36): 34114–8.

[28]

Ouimet M, Barrett TJ, Fisher EA. HDL and reverse cholesterol transport. Circ Res 2019;124(10): 1505–18.

[29]

Pedrelli M, Pramfalk C, Parini P. Thyroid hormones and thyroid hormone receptors: effects of thyromimetics on reverse cholesterol transport. World J Gastroenterol 2010;16(47): 5958–64.

[30]

Babashamsi MM, Koukhaloo SZ, Halalkhor S, et al. ABCA1 and metabolic syndrome; a review of the ABCA1 role in HDL-VLDL production, insulin-glucose homeostasis, inflammation and obesity. Diabetes Metab Syndr 2019;13(2): 1529–34.

[31]

Huuskonen J, Vishnu M, Pullinger CR, et al. Regulation of ATP-binding cassette transporter A1 transcription by thyroid hormone receptor. Biochemistry 2004; 43(6): 1626–32.

[32]

Boutari C, Perakakis N, Mantzoros CS. Association of adipokines with development and progression of nonalcoholic fatty liver disease. Endocrinol Metab (Seoul) 2018;33(1): 33–43.

[33]

Chen Y, Wu X, Wu R, et al. Changes in profile of lipids and adipokines in patients with newly diagnosed hypothyroidism and hyperthyroidism. Sci Rep 2016;6: 26174.

[34]

Lee Y, Yu X, Gonzales F, et al. PPAR alpha is necessary for the lipopenic action of hyperleptinemia on white adipose and liver tissue. Proc Natl Acad Sci U S A 2002; 99(18): 11848–53.

[35]

Pinkney JH, Goodrick SJ, Katz J, et al. Leptin and the pituitary-thyroid axis: a comparative study in lean, obese, hypothyroid and hyperthyroid subjects. Clin Endocrinol (Oxf) 1998;49(5): 583–8.

[36]

Ueki K, Kondo T, Kahn CR. Suppressor of cytokine signaling 1 (SOCS-1) and SOCS- 3 cause insulin resistance through inhibition of tyrosine phosphorylation of insulin receptor substrate proteins by discrete mechanisms. Mol Cell Biol 2004;24(12): 5434–46.

[37]

Rotundo L, Persaud A, Feurdean M, et al. The Association of leptin with severity of non-alcoholic fatty liver disease: a population-based study. Clin Mol Hepatol 2018; 24(4): 392–401.

[38]

La Cava A. Leptin in inflammation and autoimmunity. Cytokine 2017;98: 51–8.

[39]

Martinez-Una M, Lopez-Mancheno Y, Dieguez C, et al. Unraveling the role of leptin in liver function and its relationship with liver diseases. Int J Mol Sci 2020; 21(24).

[40]

Kar K, Sinha S. Variations of adipokines and insulin resistance in primary hypothyroidism. J Clin Diagn Res 2017;11(8): BC07–9.

[41]

Qiu Y, Wang SF, Yu C, et al. Association of circulating adipsin, visfatin, and adiponectin with nonalcoholic fatty liver disease in adults: a case-control study. Ann Nutr Metab 2019;74(1): 44–52.

[42]

Gierej P, Gierej B, Kalinowski P, et al. Expression of resistin in the liver of patients with non-alcoholic fatty liver disease. Pol J Pathol 2017;68(3): 225–33.

[43]

Klose M, Marina D, Hartoft-Nielsen ML, et al. Central hypothyroidism and its replacement have a significant influence on cardiovascular risk factors in adult hypopituitary patients. J Clin Endocrinol Metab 2013;98(9): 3802–10.

[44]

Gronich N, Deftereos SN, Lavi I, et al. Hypothyroidism is a risk factor for newonset diabetes: a cohort study. Diabetes Care 2015;38(9): 1657–64.

[45]

Moeller LC, Dumitrescu AM, Walker RL, et al. Thyroid hormone responsive genes in cultured human fibroblasts. J Clin Endocrinol Metab 2005;90(2): 936–43.

[46]

Maratou E, Hadjidakis DJ, Kollias A, et al. Studies of insulin resistance in patients with clinical and subclinical hypothyroidism. Eur J Endocrinol 2009;160(5): 785–90.

[47]

Tomas E, Lin YS, Dagher Z, et al. Hyperglycemia and insulin resistance: possible mechanisms. Ann N Y Acad Sci 2002;967: 43–51.

[48]

Tritos NA, Klibanski A. Effects of growth hormone on bone. Prog Mol Biol Transl Sci 2016;138: 193–211.

[49]

Press M. Growth hormone and metabolism. Diabetes Metab Rev 1988;4(4): 391–414.

[50]

Nishizawa H, Iguchi G, Murawaki A, et al. Nonalcoholic fatty liver disease in adult hypopituitary patients with GH deficiency and the impact of GH replacement therapy. Eur J Endocrinol 2012;167(1): 67–74.

[51]

Takahashi Y. The role of growth hormone and insulin-like growth factor-I in the liver. Int J Mol Sci 2017;18(7).

[52]

Matsumoto R, Fukuoka H, Iguchi G, et al. Long-term effects of growth hormone replacement therapy on liver function in adult patients with growth hormone deficiency. Growth Hormone IGF Res 2014;24(5): 174–9.

[53]

Xu L, Xu C, Yu C, et al. Association between serum growth hormone levels and nonalcoholic fatty liver disease: a cross-sectional study. PLoS One 2012;7(8): e44136.

[54]

Bergan-Roller HE, Sheridan MA. The growth hormone signaling system: insights into coordinating the anabolic and catabolic actions of growth hormone. Gen Comp Endocrinol 2018;258: 119–33.

[55]

Ichikawa T, Nakao K, Hamasaki K, et al. Role of growth hormone, insulin-like growth factor 1 and insulin-like growth factor-binding protein 3 in development of non-alcoholic fatty liver disease. Hepatol Int 2007;1(2): 287–94.

[56]

Zaouali MA, Padrissa-Altes S, Ben MI, et al. Insulin like growth factor-1 increases fatty liver preservation in IGL-1 solution. World J Gastroenterol 2010;16(45): 5693–700.

[57]

Adamek A, Kasprzak A. Insulin-like growth factor (IGF) system in liver diseases. Int J Mol Sci 2018;19(5).

[58]

Kim SH, Park MJ. Effects of growth hormone on glucose metabolism and insulin resistance in human. Ann Pediatr Endocrinol Metab 2017;22(3): 145–52.

[59]

Dodington DW, Desai HR, Woo M. JAK/STAT - emerging players in metabolism. Trends Endocrinol Metab 2018;29(1): 55–65.

[60]

Qiu H, Yang JK, Chen C. Influence of insulin on growth hormone secretion, level and growth hormone signalling. Sheng Li Xue Bao 2017;69(5): 541–56.

[61]

Marechal L, Laviolette M, Rodrigue-Way A, et al. The CD36-PPARgamma pathway in metabolic disorders. Int J Mol Sci 2018;19(5).

[62]

Nakamura MT, Yudell BE, Loor JJ. Regulation of energy metabolism by long-chain fatty acids. Prog Lipid Res 2014;53: 124–44.

[63]

Skat-Rordam J, Hojland ID, Lykkesfeldt J, et al. A role of peroxisome proliferatoractivated receptor gamma in non-alcoholic fatty liver disease. Basic Clin Pharmacol Toxicol 2019;124(5): 528–37.

[64]

Pepino MY, Kuda O, Samovski D, et al. Structure-function of CD36 and importance of fatty acid signal transduction in fat metabolism. Annu Rev Nutr 2014;34: 281–303.

[65]

Cai L, Wang Z, Ji A, et al. Scavenger receptor CD36 expression contributes to adipose tissue inflammation and cell death in diet-induced obesity. PLoS One 2012;7(5): e36785.

[66]

Hajri T, Han XX, Bonen A, et al. Defective fatty acid uptake modulates insulin responsiveness and metabolic responses to diet in CD36-null mice. J Clin Invest 2002;109(10): 1381–9.

[67]

Kotzka J, Knebel B, Avci H, et al. Phosphorylation of sterol regulatory elementbinding protein (SREBP)-1a links growth hormone action to lipid metabolism in hepatocytes. Atherosclerosis 2010;213(1): 156–65.

[68]

Shen L, Cui A, Xue Y, et al. Hepatic differentiated embryo-chondrocyte-expressed gene 1 (Dec1) inhibits sterol regulatory element-binding protein-1c (Srebp-1c) expression and alleviates fatty liver phenotype. J Biol Chem 2014;289(34): 23332–42.

[69]

Smith TM, Gilliland K, Clawson GA, et al. IGF-1 induces SREBP-1 expression and lipogenesis in SEB-1 sebocytes via activation of the phosphoinositide 3-kinase/Akt pathway. J Invest Dermatol 2008;128(5): 1286–93.

[70]

Belkaid A, Duguay SR, Ouellette RJ, et al. 17beta-estradiol induces stearoyl-CoA desaturase-1 expression in estrogen receptor-positive breast cancer cells. BMC Cancer 2015;15: 440.

[71]

Shoham Z, Schachter M. Estrogen biosynthesis–regulation, action, remote effects, and value of monitoring in ovarian stimulation cycles. Fertil Steril 1996;65(4): 687–701.

[72]

Bienenfeld A, Azarchi S, Lo SK, et al. Androgens in women: androgen-mediated skin disease and patient evaluation. J Am Acad Dermatol 2019;80(6): 1497–506.

[73]

Santen RJ, Simpson E. History of estrogen: its purification, structure, synthesis, biologic actions, and clinical implications. Endocrinology 2019;160(3): 605–25.

[74]

O'Reilly MW, Reulen RC, Gupta S, et al. ACTH and gonadotropin deficiencies predict mortality in patients treated for nonfunctioning pituitary adenoma: longterm follow-up of 519 patients in two large European centres. Clin Endocrinol (Oxf) 2016;85(5): 748–56.

[75]

Hagymasi K, Reismann P, Racz K, et al. [Role of the endocrine system in the pathogenesis of non-alcoholic fatty liver disease]. Orv Hetil 2009;150(48): 2173–81.

[76]

Zhu L, Brown WC, Cai Q, et al. Estrogen treatment after ovariectomy protects against fatty liver and may improve pathway-selective insulin resistance. Diabetes 2013;62(2): 424–34.

[77]

Marino L, Jornayvaz FR. Endocrine causes of nonalcoholic fatty liver disease. World J Gastroenterol 2015;21(39): 11053–76.

[78]

Shen M, Kumar SP, Shi H. Estradiol regulates insulin signaling and inflammation in adipose tissue. Horm Mol Biol Clin Invest 2014;17(2): 99–107.

[79]

Palmisano BT, Zhu L, Stafford JM. Role of estrogens in the regulation of liver lipid metabolism. Adv Exp Med Biol 2017;1043: 227–56.

[80]

Lundholm L, Zang H, Hirschberg AL, et al. Key lipogenic gene expression can be decreased by estrogen in human adipose tissue. Fertil Steril 2008;90(1): 44–8.

[81]

Kim DK, Kim YH, Lee JH, et al. Estrogen-related receptor gamma controls sterol regulatory element-binding protein-1c expression and alcoholic fatty liver. Biochim Biophys Acta Mol Cell Biol Lipids 2019;1864(12): 158521.

[82]

Santos RS, Frank AP, Fatima LA, et al. Activation of estrogen receptor alpha induces beiging of adipocytes. Mol Metabol 2018;18: 51–9.

[83]

Luo S, Hao L, Li X, et al. Adipose tissue-derived stem cells treated with estradiol enhance survival of autologous fat transplants. Tohoku J Exp Med 2013;231(2): 101–10.

[84]

Jeong S, Han M, Lee H, et al. Effects of fenofibrate on high-fat diet-induced body weight gain and adiposity in female C57BL/6J mice. Metabolism 2004;53(10): 1284–9.

[85]

Rettberg JR, Yao J, Brinton RD. Estrogen: a master regulator of bioenergetic systems in the brain and body. Front Neuroendocrinol 2014;35(1): 8–30.

[86]

Huang CF, Yang CY, Chan DC, et al. Arsenic exposure and glucose intolerance/ insulin resistance in estrogen-deficient female mice. Environ Health Perspect 2015;123(11): 1138–44.

[87]

Suba Z, Kasler M. [Interactions of insulin and estrogen in the regulation of cell proliferation and carcinogenesis]. Orv Hetil 2012;153(4): 125–36.

[88]

Lee C, Kim J, Jung Y. Potential therapeutic application of estrogen in gender disparity of nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. Cells 2019;8(10).

[89]

Polyzos SA, Mousiolis A, Mintziori G, et al. Nonalcoholic fatty liver disease in males with low testosterone concentrations. Diabetes Metab Syndr 2020;14(5): 1571–7.

[90]

Jaruvongvanich V, Sanguankeo A, Riangwiwat T, et al. Testosterone, sex hormone-binding globulin and nonalcoholic fatty liver disease: a systematic review and meta-analysis. Ann Hepatol 2017;16(3): 382–94.

[91]

Haider A, Gooren LJ, Padungtod P, et al. Improvement of the metabolic syndrome and of non-alcoholic liver steatosis upon treatment of hypogonadal elderly men with parenteral testosterone undecanoate. Exp Clin Endocrinol Diabetes 2010; 118(3): 167–71.

[92]

Kim S, Kwon H, Park JH, et al. A low level of serum total testosterone is independently associated with nonalcoholic fatty liver disease. BMC Gastroenterol 2012;12: 69.

[93]

Volzke H, Aumann N, Krebs A, et al. Hepatic steatosis is associated with low serum testosterone and high serum DHEAS levels in men. Int J Androl 2010;33(1): 45–53.

[94]

Woodcock T, Barker P, Daniel S, et al. Guidelines for the management of glucocorticoids during the peri-operative period for patients with adrenal insufficiency: guidelines from the association of anaesthetists, the royal college of physicians and the society for endocrinology UK. Anaesthesia 2020;75(5): 654–63.

[95]

Karaca Z, Hacioglu A, Kelestimur F. Neuroendocrine changes after aneurysmal subarachnoid haemorrhage. Pituitary 2019;22(3): 305–21.

[96]

Arnadottir M, Dallongeville J, Nilsson-Ehle P, et al. Effects of short-term treatment with corticotropin on the serum apolipoprotein pattern. Scand J Clin Lab Invest 2001;61(4): 301–6.

[97]

He X, Xue P, Xu X, et al. Short-term administration of ACTH improves plasma lipid profile and renal function in kidney transplant patients. Transplant Proc 2006; 38(5): 1371–4.

[98]

Berg AL, Rafnsson AT, Johannsson M, et al. The effects of adrenocorticotrophic hormone and an equivalent dose of cortisol on the serum concentrations of lipids, lipoproteins, and apolipoproteins. Metabolism 2006;55(8): 1083–7.

[99]

Xu N, Ekstrom U, Nilsson-Ehle P. ACTH decreases the expression and secretion of apolipoprotein B in HepG2 cell cultures. J Biol Chem 2001;276(42): 38680–4.

[100]

Skoog M, Berggren-Soderlund M, Nilsson-Ehle P, et al. Lipid synthesis and secretion in HepG2 cells is not affected by ACTH. Lipids Health Dis 2010;9: 48.

[101]

Werner B, Annette K, Christoph W. Corticotropin increases the receptor-specific uptake of native low-density lipoprotein (LDL)—but not of oxidized LDL and native or oxidized lipoprotein(a)[LP(a)]—in HEPG2 cells: No evidence for Lp(a) catabolism via the LDL-receptor. Metabolism 1997;46(7).

[102]

Shao S, Yao Z, Lu J, et al. Ablation of prolactin receptor increases hepatic triglyceride accumulation. Biochem Biophys Res Commun 2018;498(3): 693–9.

[103]

Zhang P, Ge Z, Wang H, et al. Prolactin improves hepatic steatosis via CD36 pathway. J Hepatol 2018;68(6): 1247–55.

iLIVER
Pages 65-71
Cite this article:
Zhang X, Tian H, Li Y. The pathophysiological mechanism between hypopituitarism and nonalcoholic fatty liver disease. iLIVER, 2022, 1(1): 65-71. https://doi.org/10.1016/j.iliver.2022.02.004

1128

Views

2

Crossref

Altmetrics

Received: 14 October 2021
Revised: 16 February 2022
Accepted: 16 February 2022
Published: 10 March 2022
© 2022 The Author(s). Published by Elsevier Ltd on behalf of Tsinghua University Press.

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

Return