Skip to main content

Advertisement

Log in

Ghrelin and liver disease

  • Published:
Reviews in Endocrine and Metabolic Disorders Aims and scope Submit manuscript

Abstract

Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are two of the most common liver diseases associated with obesity, type 2 diabetes and metabolic syndrome. The prevalence of these conditions are increasingly rising and presently there is not a pharmacological option available in the market. Elucidation of the mechanism of action and the molecular underpinnings behind liver disease could help to better understand the pathophysiology of these illnesses. In this sense, in the last years modulation of the ghrelin system in preclinical animal models emerge as a promising therapeutic tool. In this review, we compile the latest knowledge of the modulation of ghrelin system and its intracellular pathways that regulates lipid metabolism, hepatic inflammation and liver fibrosis. We also describe novel processes implicated in the regulation of liver disease by ghrelin, such as autophagy or dysregulated circadian rhythms. In conclusion, the information displayed in this review support that the ghrelin system could be an appealing strategy for the treatment of liver disease.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

Bibliography

  1. Angulo P. Nonalcoholic fatty liver disease. N Engl J Med. 2002;346(16):1221–31. https://doi.org/10.1056/NEJMra011775.

    Article  CAS  PubMed  Google Scholar 

  2. Pagano G, Pacini G, Musso G, Gambino R, Mecca F, Depetris N, et al. Nonalcoholic steatohepatitis, insulin resistance, and metabolic syndrome: further evidence for an etiologic association. Hepatology. 2002;35(2):367–72. https://doi.org/10.1053/jhep.2002.30690.

    Article  CAS  PubMed  Google Scholar 

  3. Targher G, Day CP, Bonora E. Risk of cardiovascular disease in patients with nonalcoholic fatty liver disease. N Engl J Med. 2010;363(14):1341–50. https://doi.org/10.1056/NEJMra0912063.

    Article  CAS  PubMed  Google Scholar 

  4. Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M. Global epidemiology of nonalcoholic fatty liver disease-meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology. 2016;64(1):73–84. https://doi.org/10.1002/hep.28431.

    Article  PubMed  Google Scholar 

  5. Angulo P, Keach JC, Batts KP, Lindor KD. Independent predictors of liver fibrosis in patients with nonalcoholic steatohepatitis. Hepatology. 1999;30(6):1356–62. https://doi.org/10.1002/hep.510300604.

    Article  CAS  PubMed  Google Scholar 

  6. Baffy G, Brunt EM, Caldwell SH. Hepatocellular carcinoma in non-alcoholic fatty liver disease: an emerging menace. J Hepatol. 2012;56(6):1384–91. https://doi.org/10.1016/j.jhep.2011.10.027.

    Article  PubMed  Google Scholar 

  7. Harmon RC, Tiniakos DG, Argo CK. Inflammation in nonalcoholic steatohepatitis. Expert Rev Gastroenterol Hepatol. 2011;5(2):189–200. https://doi.org/10.1586/egh.11.21.

    Article  PubMed  Google Scholar 

  8. Alkhouri N, Carter-Kent C, Feldstein AE. Apoptosis in nonalcoholic fatty liver disease: diagnostic and therapeutic implications. Expert Rev Gastroenterol Hepatol. 2011;5(2):201–12. https://doi.org/10.1586/egh.11.6.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Koek GH, Liedorp PR, Bast A. The role of oxidative stress in non-alcoholic steatohepatitis. Clinica Chimica Acta; international journal of clinical chemistry. 2011;412(15–16):1297–305. https://doi.org/10.1016/j.cca.2011.04.013.

    Article  CAS  Google Scholar 

  10. Polyzos SA, Kountouras J, Zavos C. The multi-hit process and the antagonistic roles of tumor necrosis factor-alpha and adiponectin in non alcoholic fatty liver disease. Hippokratia. 2009;13(2):127 author reply 8.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Al Massadi O, Lopez M, Tschop M, Dieguez C, Nogueiras R. Current understanding of the hypothalamic ghrelin pathways inducing appetite and adiposity. Trends Neurosci. 2017;40(3):167–80. https://doi.org/10.1016/j.tins.2016.12.003.

    Article  CAS  PubMed  Google Scholar 

  12. Muller TD, Nogueiras R, Andermann ML, Andrews ZB, Anker SD, Argente J, et al. Ghrelin. Mol Metab. 2015;4(6):437–60. https://doi.org/10.1016/j.molmet.2015.03.005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Duerrschmid C, He Y, Wang C, Li C, Bournat JC, Romere C, et al. Asprosin is a centrally acting orexigenic hormone. Nat Med. 2017;23(12):1444–53. https://doi.org/10.1038/nm.4432.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature. 1999;402(6762):656–60. https://doi.org/10.1038/45230.

    Article  CAS  PubMed  Google Scholar 

  15. Seoane LM, Tovar S, Baldelli R, Arvat E, Ghigo E, Casanueva FF, et al. Ghrelin elicits a marked stimulatory effect on GH secretion in freely-moving rats. Eur J Endocrinol. 2000;143(5):R7–9.

    Article  CAS  PubMed  Google Scholar 

  16. Al Massadi O, Nogueiras R, Dieguez C, Girault JA. Ghrelin and food reward. Neuropharmacology. 2019;148:131–8. https://doi.org/10.1016/j.neuropharm.2019.01.001.

    Article  CAS  PubMed  Google Scholar 

  17. Tokudome T, Otani K, Miyazato M, Kangawa K. Ghrelin and the heart. Peptides. 2019;111:42–6. https://doi.org/10.1016/j.peptides.2018.05.006.

    Article  CAS  PubMed  Google Scholar 

  18. Castellano JM, Tena-Sempere M. Metabolic control of female puberty: potential therapeutic targets. Expert Opin Ther Targets. 2016;20(10):1181–93. https://doi.org/10.1080/14728222.2016.1212015.

    Article  CAS  PubMed  Google Scholar 

  19. Tschop M, Smiley DL, Heiman ML. Ghrelin induces adiposity in rodents. Nature. 2000;407(6806):908–13. https://doi.org/10.1038/35038090.

    Article  CAS  PubMed  Google Scholar 

  20. Seoane LM, Lopez M, Tovar S, Casanueva FF, Senaris R, Dieguez C. Agouti-related peptide, neuropeptide Y, and somatostatin-producing neurons are targets for ghrelin actions in the rat hypothalamus. Endocrinology. 2003;144(2):544–51. https://doi.org/10.1210/en.2002-220795.

    Article  CAS  PubMed  Google Scholar 

  21. Peino R, Baldelli R, Rodriguez-Garcia J, Rodriguez-Segade S, Kojima M, Kangawa K, et al. Ghrelin-induced growth hormone secretion in humans. Eur J Endocrinol. 2000;143(6):R11–4.

    Article  CAS  PubMed  Google Scholar 

  22. Wren AM, Seal LJ, Cohen MA, Brynes AE, Frost GS, Murphy KG, et al. Ghrelin enhances appetite and increases food intake in humans. J Clin Endocrinol Metab. 2001;86(12):5992. https://doi.org/10.1210/jcem.86.12.8111.

    Article  CAS  PubMed  Google Scholar 

  23. Zhao TJ, Liang G, Li RL, Xie X, Sleeman MW, Murphy AJ, et al. Ghrelin O-acyltransferase (GOAT) is essential for growth hormone-mediated survival of calorie-restricted mice. Proc Natl Acad Sci U S A. 2010;107(16):7467–72. https://doi.org/10.1073/pnas.1002271107.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Howard AD, Feighner SD, Cully DF, Arena JP, Liberator PA, Rosenblum CI, et al. A receptor in pituitary and hypothalamus that functions in growth hormone release. Science. 1996;273(5277):974–7.

    Article  CAS  PubMed  Google Scholar 

  25. Sun Y, Wang P, Zheng H, Smith RG. Ghrelin stimulation of growth hormone release and appetite is mediated through the growth hormone secretagogue receptor. Proc Natl Acad Sci U S A. 2004;101(13):4679–84. https://doi.org/10.1073/pnas.0305930101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Guan XM, Yu H, Palyha OC, McKee KK, Feighner SD, Sirinathsinghji DJ, et al. Distribution of mRNA encoding the growth hormone secretagogue receptor in brain and peripheral tissues. Brain Res Mol Brain Res. 1997;48(1):23–9.

    Article  CAS  PubMed  Google Scholar 

  27. Sun Y, Butte NF, Garcia JM, Smith RG. Characterization of adult ghrelin and ghrelin receptor knockout mice under positive and negative energy balance. Endocrinology. 2008;149(2):843–50. https://doi.org/10.1210/en.2007-0271.

    Article  CAS  PubMed  Google Scholar 

  28. Zigman JM, Nakano Y, Coppari R, Balthasar N, Marcus JN, Lee CE, et al. Mice lacking ghrelin receptors resist the development of diet-induced obesity. J Clin Invest. 2005;115(12):3564–72. https://doi.org/10.1172/JCI26002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Hosoda H, Kojima M, Matsuo H, Kangawa K. Ghrelin and des-acyl ghrelin: two major forms of rat ghrelin peptide in gastrointestinal tissue. Biochem Biophys Res Commun. 2000;279(3):909–13. https://doi.org/10.1006/bbrc.2000.4039.

    Article  CAS  PubMed  Google Scholar 

  30. Yang J, Brown MS, Liang G, Grishin NV, Goldstein JL. Identification of the acyltransferase that octanoylates ghrelin, an appetite-stimulating peptide hormone. Cell. 2008;132(3):387–96. https://doi.org/10.1016/j.cell.2008.01.017.

    Article  CAS  PubMed  Google Scholar 

  31. Gutierrez JA, Solenberg PJ, Perkins DR, Willency JA, Knierman MD, Jin Z, et al. Ghrelin octanoylation mediated by an orphan lipid transferase. Proc Natl Acad Sci U S A. 2008;105(17):6320–5. https://doi.org/10.1073/pnas.0800708105.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Al Massadi O, Tschop MH, Tong J. Ghrelin acylation and metabolic control. Peptides. 2011;32(11):2301–8. https://doi.org/10.1016/j.peptides.2011.08.020.

    Article  CAS  PubMed  Google Scholar 

  33. Kirchner H, Gutierrez JA, Solenberg PJ, Pfluger PT, Czyzyk TA, Willency JA, et al. GOAT links dietary lipids with the endocrine control of energy balance. Nat Med. 2009;15(7):741–5. https://doi.org/10.1038/nm.1997.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Heppner KM, Piechowski CL, Muller A, Ottaway N, Sisley S, Smiley DL, et al. Both acyl and des-acyl ghrelin regulate adiposity and glucose metabolism via central nervous system ghrelin receptors. Diabetes. 2014;63(1):122–31. https://doi.org/10.2337/db13-0414.

    Article  CAS  PubMed  Google Scholar 

  35. Toshinai K, Yamaguchi H, Sun Y, Smith RG, Yamanaka A, Sakurai T, et al. Des-acyl ghrelin induces food intake by a mechanism independent of the growth hormone secretagogue receptor. Endocrinology. 2006;147(5):2306–14. https://doi.org/10.1210/en.2005-1357.

    Article  CAS  PubMed  Google Scholar 

  36. Ariyasu H, Takaya K, Iwakura H, Hosoda H, Akamizu T, Arai Y, et al. Transgenic mice overexpressing des-acyl ghrelin show small phenotype. Endocrinology. 2005;146(1):355–64. https://doi.org/10.1210/en.2004-0629.

    Article  CAS  PubMed  Google Scholar 

  37. Delhanty PJ, Sun Y, Visser JA, van Kerkwijk A, Huisman M, van Ijcken WF, et al. Unacylated ghrelin rapidly modulates lipogenic and insulin signaling pathway gene expression in metabolically active tissues of GHSR deleted mice. PLoS One. 2010;5(7):e11749. https://doi.org/10.1371/journal.pone.0011749.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Al Massadi O, Lear PV, Muller TD, Lopez M, Dieguez C, Tschop MH, et al. Review of novel aspects of the regulation of ghrelin secretion. Curr Drug Metab. 2014;15(4):398–413.

    Article  PubMed  Google Scholar 

  39. Cummings DE, Clement K, Purnell JQ, Vaisse C, Foster KE, Frayo RS, et al. Elevated plasma ghrelin levels in Prader Willi syndrome. Nat Med. 2002;8(7):643–4. https://doi.org/10.1038/nm0702-643.

    Article  CAS  PubMed  Google Scholar 

  40. Tschop M, Weyer C, Tataranni PA, Devanarayan V, Ravussin E, Heiman ML. Circulating ghrelin levels are decreased in human obesity. Diabetes. 2001;50(4):707–9. https://doi.org/10.2337/diabetes.50.4.707.

    Article  CAS  PubMed  Google Scholar 

  41. Shiiya T, Nakazato M, Mizuta M, Date Y, Mondal MS, Tanaka M, et al. Plasma ghrelin levels in lean and obese humans and the effect of glucose on ghrelin secretion. J Clin Endocrinol Metab. 2002;87(1):240–4. https://doi.org/10.1210/jcem.87.1.8129.

    Article  CAS  PubMed  Google Scholar 

  42. Theander-Carrillo C, Wiedmer P, Cettour-Rose P, Nogueiras R, Perez-Tilve D, Pfluger P, et al. Ghrelin action in the brain controls adipocyte metabolism. J Clin Invest. 2006;116(7):1983–93. https://doi.org/10.1172/JCI25811.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Barazzoni R, Bosutti A, Stebel M, Cattin MR, Roder E, Visintin L, et al. Ghrelin regulates mitochondrial-lipid metabolism gene expression and tissue fat distribution in liver and skeletal muscle. Am J Physiol Endocrinol Metab. 2005;288(1):E228–35. https://doi.org/10.1152/ajpendo.00115.2004.

    Article  CAS  PubMed  Google Scholar 

  44. Davies JS, Kotokorpi P, Eccles SR, Barnes SK, Tokarczuk PF, Allen SK, et al. Ghrelin induces abdominal obesity via GHS-R-dependent lipid retention. Mol Endocrinol. 2009;23(6):914–24. https://doi.org/10.1210/me.2008-0432.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Porteiro B, Diaz-Ruiz A, Martinez G, Senra A, Vidal A, Serrano M, et al. Ghrelin requires p53 to stimulate lipid storage in fat and liver. Endocrinology. 2013;154(10):3671–9. https://doi.org/10.1210/en.2013-1176.

    Article  CAS  PubMed  Google Scholar 

  46. Sangiao-Alvarellos S, Vazquez MJ, Varela L, Nogueiras R, Saha AK, Cordido F, et al. Central ghrelin regulates peripheral lipid metabolism in a growth hormone-independent fashion. Endocrinology. 2009;150(10):4562–74. https://doi.org/10.1210/en.2009-0482.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Vousden KH, Ryan KM. p53 and metabolism. Nat Rev Cancer. 2009;9(10):691–700. https://doi.org/10.1038/nrc2715.

    Article  CAS  PubMed  Google Scholar 

  48. Vousden KH, Prives C. Blinded by the light: the growing complexity of p53. Cell. 2009;137(3):413–31. https://doi.org/10.1016/j.cell.2009.04.037.

    Article  CAS  PubMed  Google Scholar 

  49. Berkers CR, Maddocks OD, Cheung EC, Mor I, Vousden KH. Metabolic regulation by p53 family members. Cell Metab. 2013;18(5):617–33. https://doi.org/10.1016/j.cmet.2013.06.019.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Wang X, Zhao X, Gao X, Mei Y, Wu M. A new role of p53 in regulating lipid metabolism. J Mol Cell Biol. 2013;5(2):147–50. https://doi.org/10.1093/jmcb/mjs064.

    Article  PubMed  Google Scholar 

  51. Hallenborg P, Fjaere E, Liaset B, Petersen RK, Murano I, Sonne SB, et al. p53 regulates expression of uncoupling protein 1 through binding and repression of PPARgamma coactivator-1alpha. Am J Physiol Endocrinol Metab. 2016;310(2):E116–28. https://doi.org/10.1152/ajpendo.00119.2015.

    Article  PubMed  Google Scholar 

  52. Al-Massadi O, Porteiro B, Kuhlow D, Kohler M, Gonzalez-Rellan MJ, Garcia-Lavandeira M, et al. Pharmacological and genetic manipulation of p53 in Brown fat at adult but not embryonic stages regulates thermogenesis and body weight in male mice. Endocrinology. 2016;157(7):2735–49. https://doi.org/10.1210/en.2016-1209.

    Article  CAS  PubMed  Google Scholar 

  53. Velasquez DA, Martinez G, Romero A, Vazquez MJ, Boit KD, Dopeso-Reyes IG, et al. The central Sirtuin 1/p53 pathway is essential for the orexigenic action of ghrelin. Diabetes. 2011;60(4):1177–85. https://doi.org/10.2337/db10-0802.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Quinones M, Al-Massadi O, Folgueira C, Bremser S, Gallego R, Torres-Leal L, et al. p53 in AgRP neurons is required for protection against diet-induced obesity via JNK1. Nat Commun. 2018;9(1):3432. https://doi.org/10.1038/s41467-018-05711-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Lopez M, Lage R, Saha AK, Perez-Tilve D, Vazquez MJ, Varela L, et al. Hypothalamic fatty acid metabolism mediates the orexigenic action of ghrelin. Cell Metab. 2008;7(5):389–99. https://doi.org/10.1016/j.cmet.2008.03.006.

    Article  CAS  PubMed  Google Scholar 

  56. Kola B, Hubina E, Tucci SA, Kirkham TC, Garcia EA, Mitchell SE, et al. Cannabinoids and ghrelin have both central and peripheral metabolic and cardiac effects via AMP-activated protein kinase. J Biol Chem. 2005;280(26):25196–201. https://doi.org/10.1074/jbc.C500175200.

    Article  CAS  PubMed  Google Scholar 

  57. Lim CT, Kola B, Feltrin D, Perez-Tilve D, Tschop MH, Grossman AB, et al. Ghrelin and cannabinoids require the ghrelin receptor to affect cellular energy metabolism. Mol Cell Endocrinol. 2013;365(2):303–8. https://doi.org/10.1016/j.mce.2012.11.007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Kola B, Farkas I, Christ-Crain M, Wittmann G, Lolli F, Amin F, et al. The orexigenic effect of ghrelin is mediated through central activation of the endogenous cannabinoid system. PLoS One. 2008;3(3):e1797. https://doi.org/10.1371/journal.pone.0001797.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Senin LL, Al-Massadi O, Folgueira C, Castelao C, Pardo M, Barja-Fernandez S, et al. The gastric CB1 receptor modulates ghrelin production through the mTOR pathway to regulate food intake. PLoS One. 2013;8(11):e80339. https://doi.org/10.1371/journal.pone.0080339.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Osei-Hyiaman D, Liu J, Zhou L, Godlewski G, Harvey-White J, Jeong WI, et al. Hepatic CB1 receptor is required for development of diet-induced steatosis, dyslipidemia, and insulin and leptin resistance in mice. J Clin Invest. 2008;118(9):3160–9. https://doi.org/10.1172/JCI34827.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Despres JP, Golay A, Sjostrom L. Effects of rimonabant on metabolic risk factors in overweight patients with dyslipidemia. N Engl J Med. 2005;353(20):2121–34. https://doi.org/10.1056/NEJMoa044537.

    Article  CAS  PubMed  Google Scholar 

  62. Nogueiras R, Veyrat-Durebex C, Suchanek PM, Klein M, Tschop J, Caldwell C, et al. Peripheral, but not central, CB1 antagonism provides food intake-independent metabolic benefits in diet-induced obese rats. Diabetes. 2008;57(11):2977–91. https://doi.org/10.2337/db08-0161.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Kola B, Wittman G, Bodnar I, Amin F, Lim CT, Olah M, et al. The CB1 receptor mediates the peripheral effects of ghrelin on AMPK activity but not on growth hormone release. FASEB J. 2013;27(12):5112–21. https://doi.org/10.1096/fj.13-232918.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Xu G, Li Y, An W, Li S, Guan Y, Wang N, et al. Gastric mammalian target of rapamycin signaling regulates ghrelin production and food intake. Endocrinology. 2009;150(8):3637–44. https://doi.org/10.1210/en.2009-0372.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Martins L, Fernandez-Mallo D, Novelle MG, Vazquez MJ, Tena-Sempere M, Nogueiras R, et al. Hypothalamic mTOR signaling mediates the orexigenic action of ghrelin. PLoS One. 2012;7(10):e46923. https://doi.org/10.1371/journal.pone.0046923.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Li Z, Xu G, Qin Y, Zhang C, Tang H, Yin Y, et al. Ghrelin promotes hepatic lipogenesis by activation of mTOR-PPARgamma signaling pathway. Proc Natl Acad Sci U S A. 2014;111(36):13163–8. https://doi.org/10.1073/pnas.1411571111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Ferre P. The biology of peroxisome proliferator-activated receptors: relationship with lipid metabolism and insulin sensitivity. Diabetes. 2004;53(Suppl 1):S43–50. https://doi.org/10.2337/diabetes.53.2007.s43.

    Article  CAS  PubMed  Google Scholar 

  68. Timchenko NA. Aging and liver regeneration. Trends Endocrinol Metab. 2009;20(4):171–6. https://doi.org/10.1016/j.tem.2009.01.005.

    Article  CAS  PubMed  Google Scholar 

  69. Guillory B, Jawanmardi N, Iakova P, Anderson B, Zang P, Timchenko NA et al. Ghrelin deletion protects against age-associated hepatic steatosis by downregulating the C/EBPalpha-p300/DGAT1 pathway. Aging cell. 2018;17(1). https://doi.org/10.1111/acel.12688.

  70. Park EA, Gurney AL, Nizielski SE, Hakimi P, Cao Z, Moorman A, et al. Relative roles of CCAAT/enhancer-binding protein beta and cAMP regulatory element-binding protein in controlling transcription of the gene for phosphoenolpyruvate carboxykinase (GTP). J Biol Chem. 1993;268(1):613–9.

    CAS  PubMed  Google Scholar 

  71. Timchenko LT, Salisbury E, Wang GL, Nguyen H, Albrecht JH, Hershey JW, et al. Age-specific CUGBP1-eIF2 complex increases translation of CCAAT/enhancer-binding protein beta in old liver. J Biol Chem. 2006;281(43):32806–19. https://doi.org/10.1074/jbc.M605701200.

    Article  CAS  PubMed  Google Scholar 

  72. Jin J, Iakova P, Breaux M, Sullivan E, Jawanmardi N, Chen D, et al. Increased expression of enzymes of triglyceride synthesis is essential for the development of hepatic steatosis. Cell Rep. 2013;3(3):831–43. https://doi.org/10.1016/j.celrep.2013.02.009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Jin J, Valanejad L, Nguyen TP, Lewis K, Wright M, Cast A, et al. Activation of CDK4 triggers development of non-alcoholic fatty liver disease. Cell Rep. 2016;16(3):744–56. https://doi.org/10.1016/j.celrep.2016.06.019.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Barazzoni R, Semolic A, Cattin MR, Zanetti M, Guarnieri G. Acylated ghrelin limits fat accumulation and improves redox state and inflammation markers in the liver of high-fat-fed rats. Obesity (Silver Spring). 2014;22(1):170–7. https://doi.org/10.1002/oby.20454.

    Article  CAS  Google Scholar 

  75. Cetin E, Kanbur M, Cetin N, Eraslan G, Atasever A. Hepatoprotective effect of ghrelin on carbon tetrachloride-induced acute liver injury in rats. Regul Pept. 2011;171(1–3):1–5. https://doi.org/10.1016/j.regpep.2011.05.010.

    Article  CAS  PubMed  Google Scholar 

  76. Li Y, Hai J, Li L, Chen X, Peng H, Cao M, et al. Administration of ghrelin improves inflammation, oxidative stress, and apoptosis during and after non-alcoholic fatty liver disease development. Endocrine. 2013;43(2):376–86. https://doi.org/10.1007/s12020-012-9761-5.

    Article  CAS  PubMed  Google Scholar 

  77. Ezquerro S, Mocha F, Fruhbeck G, Guzman-Ruiz R, Valenti V, Mugueta C, et al. Ghrelin reduces TNF-alpha-induced human hepatocyte apoptosis, autophagy, and Pyroptosis: role in obesity-associated NAFLD. J Clin Endocrinol Metab. 2019;104(1):21–37. https://doi.org/10.1210/jc.2018-01171.

    Article  PubMed  Google Scholar 

  78. Chorny A, Anderson P, Gonzalez-Rey E, Delgado M. Ghrelin protects against experimental sepsis by inhibiting high-mobility group box 1 release and by killing bacteria. J Immunol. 2008;180(12):8369–77. https://doi.org/10.4049/jimmunol.180.12.8369.

    Article  CAS  PubMed  Google Scholar 

  79. Qin Y, Li Z, Wang Z, Li Y, Zhao J, Mulholland M, et al. Ghrelin contributes to protection of hepatocellular injury induced by ischaemia/reperfusion. Liver Int. 2014;34(4):567–75. https://doi.org/10.1111/liv.12286.

    Article  CAS  PubMed  Google Scholar 

  80. Kasimay O, Iseri SO, Barlas A, Bangir D, Yegen C, Arbak S, et al. Ghrelin ameliorates pancreaticobiliary inflammation and associated remote organ injury in rats. Hepatol Res. 2006;36(1):11–9. https://doi.org/10.1016/j.hepres.2006.06.009.

    Article  CAS  PubMed  Google Scholar 

  81. Huang CX, Yuan MJ, Huang H, Wu G, Liu Y, Yu SB, et al. Ghrelin inhibits post-infarct myocardial remodeling and improves cardiac function through anti-inflammation effect. Peptides. 2009;30(12):2286–91. https://doi.org/10.1016/j.peptides.2009.09.004.

    Article  CAS  PubMed  Google Scholar 

  82. Gonzalez-Rey E, Chorny A, Delgado M. Therapeutic action of ghrelin in a mouse model of colitis. Gastroenterology. 2006;130(6):1707–20. https://doi.org/10.1053/j.gastro.2006.01.041.

    Article  CAS  PubMed  Google Scholar 

  83. Moreno M, Chaves JF, Sancho-Bru P, Ramalho F, Ramalho LN, Mansego ML, et al. Ghrelin attenuates hepatocellular injury and liver fibrogenesis in rodents and influences fibrosis progression in humans. Hepatology. 2010;51(3):974–85. https://doi.org/10.1002/hep.23421.

    Article  CAS  PubMed  Google Scholar 

  84. Mao Y, Zhang S, Yu F, Li H, Guo C, Fan X. Ghrelin attenuates liver fibrosis through regulation of TGF-beta1 expression and autophagy. Int J Mol Sci. 2015;16(9):21911–30. https://doi.org/10.3390/ijms160921911.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Kabil NN, Seddiek HA, Yassin NA, Gamal-Eldin MM. Effect of ghrelin on chronic liver injury and fibrogenesis in male rats: possible role of nitric oxide. Peptides. 2014;52:90–7. https://doi.org/10.1016/j.peptides.2013.11.022.

    Article  CAS  PubMed  Google Scholar 

  86. Golestan Jahromi M, Nabavizadeh F, Vahedian J, Nahrevanian H, Dehpour AR, Zare-Mehrjardi A. Protective effect of ghrelin on acetaminophen-induced liver injury in rat. Peptides. 2010;31(11):2114–7. https://doi.org/10.1016/j.peptides.2010.08.009.

    Article  CAS  PubMed  Google Scholar 

  87. Ezquerro S, Fruhbeck G, Rodriguez A. Ghrelin and autophagy. Curr Opin Clin Nutr Metab Care. 2017;20(5):402–8. https://doi.org/10.1097/MCO.0000000000000390.

    Article  PubMed  Google Scholar 

  88. Song YM, Lee YH, Kim JW, Ham DS, Kang ES, Cha BS, et al. Metformin alleviates hepatosteatosis by restoring SIRT1-mediated autophagy induction via an AMP-activated protein kinase-independent pathway. Autophagy. 2015;11(1):46–59. https://doi.org/10.4161/15548627.2014.984271.

    Article  CAS  PubMed  Google Scholar 

  89. Chang E, Kim L, Park SE, Rhee EJ, Lee WY, Oh KW, et al. Ezetimibe improves hepatic steatosis in relation to autophagy in obese and diabetic rats. World J Gastroenterol. 2015;21(25):7754–63. https://doi.org/10.3748/wjg.v21.i25.7754.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Ezquerro S, Mendez-Gimenez L, Becerril S, Moncada R, Valenti V, Catalan V, et al. Acylated and desacyl ghrelin are associated with hepatic lipogenesis, beta-oxidation and autophagy: role in NAFLD amelioration after sleeve gastrectomy in obese rats. Sci Rep. 2016;6:39942. https://doi.org/10.1038/srep39942.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Zhang Y, Fang F, Goldstein JL, Brown MS, Zhao TJ. Reduced autophagy in livers of fasted, fat-depleted, ghrelin-deficient mice: reversal by growth hormone. Proc Natl Acad Sci U S A. 2015;112(4):1226–31. https://doi.org/10.1073/pnas.1423643112.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Zhang S, Mao Y, Fan X. Inhibition of ghrelin o-acyltransferase attenuated lipotoxicity by inducing autophagy via AMPK-mTOR pathway. Drug Des Devel Ther. 2018;12:873–85. https://doi.org/10.2147/DDDT.S158985.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Takahashi JS, Hong HK, Ko CH, McDearmon EL. The genetics of mammalian circadian order and disorder: implications for physiology and disease. Nat Rev Genet. 2008;9(10):764–75. https://doi.org/10.1038/nrg2430.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Berson DM, Dunn FA, Takao M. Phototransduction by retinal ganglion cells that set the circadian clock. Science. 2002;295(5557):1070–3. https://doi.org/10.1126/science.1067262.

    Article  CAS  PubMed  Google Scholar 

  95. Reinke H, Asher G. Circadian clock control of liver metabolic functions. Gastroenterology. 2016;150(3):574–80. https://doi.org/10.1053/j.gastro.2015.11.043.

    Article  PubMed  Google Scholar 

  96. Stow LR, Gumz ML. The circadian clock in the kidney. J Am Soc Nephrol. 2011;22(4):598–604. https://doi.org/10.1681/ASN.2010080803.

    Article  CAS  PubMed  Google Scholar 

  97. Zvonic S, Ptitsyn AA, Conrad SA, Scott LK, Floyd ZE, Kilroy G, et al. Characterization of peripheral circadian clocks in adipose tissues. Diabetes. 2006;55(4):962–70. https://doi.org/10.2337/diabetes.55.04.06.db05-0873.

    Article  CAS  PubMed  Google Scholar 

  98. Dibner C, Schibler U, Albrecht U. The mammalian circadian timing system: organization and coordination of central and peripheral clocks. Annu Rev Physiol. 2010;72:517–49. https://doi.org/10.1146/annurev-physiol-021909-135821.

    Article  CAS  PubMed  Google Scholar 

  99. Damiola F, Le Minh N, Preitner N, Kornmann B, Fleury-Olela F, Schibler U. Restricted feeding uncouples circadian oscillators in peripheral tissues from the central pacemaker in the suprachiasmatic nucleus. Genes Dev. 2000;14(23):2950–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Stokkan KA, Yamazaki S, Tei H, Sakaki Y, Menaker M. Entrainment of the circadian clock in the liver by feeding. Science. 2001;291(5503):490–3. https://doi.org/10.1126/science.291.5503.490.

    Article  CAS  PubMed  Google Scholar 

  101. Kohsaka A, Laposky AD, Ramsey KM, Estrada C, Joshu C, Kobayashi Y, et al. High-fat diet disrupts behavioral and molecular circadian rhythms in mice. Cell Metab. 2007;6(5):414–21. https://doi.org/10.1016/j.cmet.2007.09.006.

    Article  CAS  PubMed  Google Scholar 

  102. Wang Q, Yin Y, Zhang W. Ghrelin Restores the Disruption of the Circadian Clock in Steatotic Liver. Int J Mol Sci. 2018;19(10). https://doi.org/10.3390/ijms19103134.

  103. Castaneda TR, Tong J, Datta R, Culler M, Tschop MH. Ghrelin in the regulation of body weight and metabolism. Front Neuroendocrinol. 2010;31(1):44–60. https://doi.org/10.1016/j.yfrne.2009.10.008.

    Article  CAS  PubMed  Google Scholar 

  104. De Vriese C, Gregoire F, Lema-Kisoka R, Waelbroeck M, Robberecht P, Delporte C. Ghrelin degradation by serum and tissue homogenates: identification of the cleavage sites. Endocrinology. 2004;145(11):4997–5005. https://doi.org/10.1210/en.2004-0569.

    Article  CAS  PubMed  Google Scholar 

  105. Randell EW, Mathews MS, Zhang H, Seraj JS, Sun G. Relationship between serum butyrylcholinesterase and the metabolic syndrome. Clin Biochem. 2005;38(9):799–805. https://doi.org/10.1016/j.clinbiochem.2005.04.008.

    Article  CAS  PubMed  Google Scholar 

  106. Santarpia L, Grandone I, Contaldo F, Pasanisi F. Butyrylcholinesterase as a prognostic marker: a review of the literature. J Cachexia Sarcopenia Muscle. 2013;4(1):31–9. https://doi.org/10.1007/s13539-012-0083-5.

    Article  PubMed  Google Scholar 

  107. Alcantara VM, Rodrigues LC, Oliveira LC, Chautard-Freire-Maia EA. Association of the CHE2 locus with body mass index and butyrylcholinesterase activity. Hum Biol. 2001;73(4):587–95.

    Article  CAS  PubMed  Google Scholar 

  108. Chautard-Freire-Maia EA, Primo-Parmo SL, Picheth G, Lourenco MA, Vieira MM. The C5 isozyme of serum cholinesterase and adult weight. Hum Hered. 1991;41(5):330–9. https://doi.org/10.1159/000154021.

    Article  CAS  PubMed  Google Scholar 

  109. Hashim Y, Shepherd D, Wiltshire S, Holman RR, Levy JC, Clark A, et al. Butyrylcholinesterase K variant on chromosome 3 q is associated with type II diabetes in white Caucasian subjects. Diabetologia. 2001;44(12):2227–30. https://doi.org/10.1007/s001250100033.

    Article  CAS  PubMed  Google Scholar 

  110. Lima JK, Leite N, Turek LV, Souza RL, da Silva TL, Osiecki AC, et al. 1914G variant of BCHE gene associated with enzyme activity, obesity and triglyceride levels. Gene. 2013;532(1):24–6. https://doi.org/10.1016/j.gene.2013.08.068.

    Article  CAS  PubMed  Google Scholar 

  111. Iwasaki T, Yoneda M, Nakajima A, Terauchi Y. Serum butyrylcholinesterase is strongly associated with adiposity, the serum lipid profile and insulin resistance. Intern Med. 2007;46(19):1633–9.

    Article  PubMed  Google Scholar 

  112. Chen VP, Gao Y, Geng L, Stout MB, Jensen MD, Brimijoin S. Butyrylcholinesterase deficiency promotes adipose tissue growth and hepatic lipid accumulation in male mice on high-fat diet. Endocrinology. 2016;157(8):3086–95. https://doi.org/10.1210/en.2016-1166.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Barnett BP, Hwang Y, Taylor MS, Kirchner H, Pfluger PT, Bernard V, et al. Glucose and weight control in mice with a designed ghrelin O-acyltransferase inhibitor. Science. 2010;330(6011):1689–92. https://doi.org/10.1126/science.1196154.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Abegg K, Bernasconi L, Hutter M, Whiting L, Pietra C, Giuliano C, et al. Ghrelin receptor inverse agonists as a novel therapeutic approach against obesity-related metabolic disease. Diabetes Obes Metab. 2017;19(12):1740–50. https://doi.org/10.1111/dom.13020.

    Article  CAS  PubMed  Google Scholar 

  115. Perez-Tilve D, Gonzalez-Matias L, Alvarez-Crespo M, Leiras R, Tovar S, Dieguez C, et al. Exendin-4 potently decreases ghrelin levels in fasting rats. Diabetes. 2007;56(1):143–51. https://doi.org/10.2337/db05-0996.

    Article  CAS  PubMed  Google Scholar 

  116. Zorrilla EP, Iwasaki S, Moss JA, Chang J, Otsuji J, Inoue K, et al. Vaccination against weight gain. Proc Natl Acad Sci U S A. 2006;103(35):13226–31. https://doi.org/10.1073/pnas.0605376103.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Shearman LP, Wang SP, Helmling S, Stribling DS, Mazur P, Ge L, et al. Ghrelin neutralization by a ribonucleic acid-SPM ameliorates obesity in diet-induced obese mice. Endocrinology. 2006;147(3):1517–26. https://doi.org/10.1210/en.2005-0993.

    Article  CAS  PubMed  Google Scholar 

  118. Ge X, Yang H, Bednarek MA, Galon-Tilleman H, Chen P, Chen M, et al. LEAP2 is an endogenous antagonist of the ghrelin receptor. Cell Metab. 2018;27(2):461–9 e6. https://doi.org/10.1016/j.cmet.2017.10.016.

    Article  CAS  PubMed  Google Scholar 

  119. Al-Massadi O, Muller T, Tschop M, Dieguez C, Nogueiras R. Ghrelin and LEAP-2: rivals in energy metabolism. Trends Pharmacol Sci. 2018;39(8):685–94. https://doi.org/10.1016/j.tips.2018.06.004.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work is supported by grants of Xunta (MQ: 2018-PG013). Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición (CIBERobn). Western Norway Regional Health Authority (Helse Vest RHF), CIBERobn is an initiative of the Instituto de Salud Carlos III (ISCIII) of Spain which is supported by FEDER funds. M.Q. is a recipient of a Postdoctoral contract from Galician Government (Xunta de Galicia ED481B2018/004). The figures were generated by using materials from Servier Medical Art (Servier) under consideration of a Creative Commons Attribution 3.0 Unported License.

Author information

Authors and Affiliations

Authors

Contributions

OA-M conceptualized the article, MQ performed the literature search and data analysis, MQ, JF and OA-M drafted and/or critically revised the work.

Corresponding author

Correspondence to Omar Al-Massadi.

Ethics declarations

Conflict of interest

The authors declare that they have no conflicts of interest in the authorship or publication of this work.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Quiñones, M., Fernø, J. & Al-Massadi, O. Ghrelin and liver disease. Rev Endocr Metab Disord 21, 45–56 (2020). https://doi.org/10.1007/s11154-019-09528-6

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11154-019-09528-6

Keywords

Navigation