Endocrinol Metab.  2021 Dec;36(6):1151-1160. 10.3803/EnM.2021.1331.

Serotonergic Regulation of Hepatic Energy Metabolism

Affiliations
  • 1Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
  • 2Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea

Abstract

The liver is a vital organ that regulates systemic energy metabolism and many physiological functions. Nonalcoholic fatty liver disease (NAFLD) is the commonest cause of chronic liver disease and end-stage liver failure. NAFLD is primarily caused by metabolic disruption of lipid and glucose homeostasis. Serotonin (5-hydroxytryptamine [5-HT]) is a biogenic amine with several functions in both the central and peripheral systems. 5-HT functions as a neurotransmitter in the brain and a hormone in peripheral tissues to regulate systemic energy homeostasis. Several recent studies have proposed various roles of 5-HT in hepatic metabolism and inflammation using tissue-specific knockout mice and 5-HT-receptor agonists/antagonists. This review compiles the most recent research on the relationship between 5-HT and hepatic metabolism, and the role of 5-HT signaling as a potential therapeutic target in NAFLD.

Keyword

Serotonin; Liver; Non-alcoholic fatty liver disease

Figure

  • Fig. 1 5-Hydroxytryptamine (5-HT) receptors and signaling pathways. The 5-HT1 and 5-HT5 receptors are Gi/Go-protein coupled receptors that inhibit adenylate cyclase (AC) and thus suppress the cyclic adenosine monophosphate (cAMP) downstream pathways. 5-HT2 receptors are Gq/G11-protein coupled receptors that activate phospholipase C, resulting in the activation of the inositol triphosphate (IP3) and diacylglycerol (DAG) downstream pathways. The only ligand-gated ion channel that can regulate membrane potential is the 5-HT3 receptor. The Gs-protein coupled receptors 5-HT4, 5-HT6, and 5-HT7 activate AC.

  • Fig. 2 Metabolic functions of 5-hydroxytryptamine (5-HT) in liver. Most peripheral 5-HTs are derived from enterochromaffin cells of the gut. 5-HT regulates hepatic fibrosis in hepatic stellate cells (HSCs) by activating HSC-produced transforming growth factor (TGF)-signaling. Through the 5-HT2B receptor, 5-HT promotes gluconeogenesis in hepatocytes by increasing the activity of fructose 1,6-bisphosphatase (FBPase) and glucose 6-phosphatase (G6pase). 5-HT2B receptor signaling also inhibits glucose uptake by promoting the breakdown of glucose transporter 2 (GLUT2). The activation of the sterol-regulatory-element-binding protein 1 (SREBP1) signaling pathway by the 5-HT2A receptor increases lipogenesis in the liver. ERK1/2, extracellular signal–regulated protein kinase.


Reference

1. Abai B. StatPearls. Treasure Island: StatPearls Publishing;2021. Chapter, Physiology, liver. https://www.ncbi.nlm.nih.gov/books/NBK535438 .
2. Fotbolcu H, Zorlu E. Nonalcoholic fatty liver disease as a multi-systemic disease. World J Gastroenterol. 2016; 22:4079–90.
Article
3. Ferguson D, Finck BN. Emerging therapeutic approaches for the treatment of NAFLD and type 2 diabetes mellitus. Nat Rev Endocrinol. 2021; 17:484–95.
Article
4. Byrne CD, Targher G. NAFLD: a multisystem disease. J Hepatol. 2015; 62(1 Suppl):S47–64.
Article
5. Chao HW, Chao SW, Lin H, Ku HC, Cheng CF. Homeostasis of glucose and lipid in non-alcoholic fatty liver disease. Int J Mol Sci. 2019; 20:298.
Article
6. Pei K, Gui T, Kan D, Feng H, Jin Y, Yang Y, et al. An overview of lipid metabolism and nonalcoholic fatty liver disease. Biomed Res Int. 2020; 2020:4020249.
Article
7. Oh CM, Park S, Kim H. Serotonin as a new therapeutic target for diabetes mellitus and obesity. Diabetes Metab J. 2016; 40:89–98.
Article
8. Yabut JM, Crane JD, Green AE, Keating DJ, Khan WI, Steinberg GR. Emerging roles for serotonin in regulating metabolism: new implications for an ancient molecule. Endocr Rev. 2019; 40:1092–107.
Article
9. Badawy AA. Kynurenine pathway of tryptophan metabolism: regulatory and functional aspects. Int J Tryptophan Res. 2017; 10:1178646917691938.
Article
10. Nakamura K, Hasegawa H. Developmental role of tryptophan hydroxylase in the nervous system. Mol Neurobiol. 2007; 35:45–54.
Article
11. Prah A, Purg M, Stare J, Vianello R, Mavri J. How monoamine oxidase a decomposes serotonin: an empirical valence bond simulation of the reactive step. J Phys Chem B. 2020; 124:8259–65.
Article
12. Gauer F, Craft CM. Circadian regulation of hydroxyindole-O-methyltransferase mRNA levels in rat pineal and retina. Brain Res. 1996; 737:99–109.
Article
13. Pissios P, Maratos-Flier E. More than satiety: central serotonin signaling and glucose homeostasis. Cell Metab. 2007; 6:345–7.
Article
14. van Galen KA, Ter Horst KW, Serlie MJ. Serotonin, food intake, and obesity. Obes Rev. 2021; 22:e13210.
15. Namkung J, Kim H, Park S. Peripheral serotonin: a new player in systemic energy homeostasis. Mol Cells. 2015; 38:1023–8.
Article
16. Vandenbussche N, Goadsby PJ. Lorcaserin safety in overweight or obese patients. N Engl J Med. 2019; 380:99.
Article
17. Sharretts J, Galescu O, Gomatam S, Andraca-Carrera E, Hampp C, Yanoff L. Cancer risk associated with lorcaserin: the FDA’s review of the CAMELLIA-TIMI 61 Trial. N Engl J Med. 2020; 383:1000–2.
Article
18. Cano G, Passerin AM, Schiltz JC, Card JP, Morrison SF, Sved AF. Anatomical substrates for the central control of sympathetic outflow to interscapular adipose tissue during cold exposure. J Comp Neurol. 2003; 460:303–26.
Article
19. Madden CJ, Morrison SF. Endogenous activation of spinal 5-hydroxytryptamine (5-HT) receptors contributes to the thermoregulatory activation of brown adipose tissue. Am J Physiol Regul Integr Comp Physiol. 2010; 298:R776–83.
Article
20. McGlashon JM, Gorecki MC, Kozlowski AE, Thirnbeck CK, Markan KR, Leslie KL, et al. Central serotonergic neurons activate and recruit thermogenic brown and beige fat and regulate glucose and lipid homeostasis. Cell Metab. 2015; 21:692–705.
Article
21. Berglund ED, Liu C, Sohn JW, Liu T, Kim MH, Lee CE, et al. Serotonin 2C receptors in pro-opiomelanocortin neurons regulate energy and glucose homeostasis. J Clin Invest. 2013; 123:5061–70.
Article
22. Könner AC, Janoschek R, Plum L, Jordan SD, Rother E, Ma X, et al. Insulin action in AgRP-expressing neurons is required for suppression of hepatic glucose production. Cell Metab. 2007; 5:438–49.
Article
23. Chandran S, Guo T, Tolliver T, Chen W, Murphy DL, McPherron AC. Effects of serotonin on skeletal muscle growth. BMC Proc. 2012; 6(Suppl 3):O3.
Article
24. Oh CM, Namkung J, Go Y, Shong KE, Kim K, Kim H, et al. Regulation of systemic energy homeostasis by serotonin in adipose tissues. Nat Commun. 2015; 6:6794.
Article
25. Park H, Oh CM, Park J, Park H, Cui S, Kim HS, et al. Deletion of the serotonin receptor type 3A in mice leads to sudden cardiac death during pregnancy. Circ J. 2015; 79:1807–15.
Article
26. Kim H, Toyofuku Y, Lynn FC, Chak E, Uchida T, Mizukami H, et al. Serotonin regulates pancreatic beta cell mass during pregnancy. Nat Med. 2010; 16:804–8.
Article
27. Li Z, Chalazonitis A, Huang YY, Mann JJ, Margolis KG, Yang QM, et al. Essential roles of enteric neuronal serotonin in gastrointestinal motility and the development/survival of enteric dopaminergic neurons. J Neurosci. 2011; 31:8998–9009.
Article
28. Shajib MS, Khan WI. The role of serotonin and its receptors in activation of immune responses and inflammation. Acta Physiol (Oxf). 2015; 213:561–74.
Article
29. Hoffman JM, Tyler K, MacEachern SJ, Balemba OB, Johnson AC, Brooks EM, et al. Activation of colonic mucosal 5-HT(4) receptors accelerates propulsive motility and inhibits visceral hypersensitivity. Gastroenterology. 2012; 142:844–54.
Article
30. Liu MT, Kuan YH, Wang J, Hen R, Gershon MD. 5-HT4 receptor-mediated neuroprotection and neurogenesis in the enteric nervous system of adult mice. J Neurosci. 2009; 29:9683–99.
Article
31. Kwon YH, Wang H, Denou E, Ghia JE, Rossi L, Fontes ME, et al. Modulation of gut microbiota composition by serotonin signaling influences intestinal immune response and susceptibility to colitis. Cell Mol Gastroenterol Hepatol. 2019; 7:709–28.
Article
32. Yano JM, Yu K, Donaldson GP, Shastri GG, Ann P, Ma L, et al. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell. 2015; 161:264–76.
Article
33. Ohara-Imaizumi M, Kim H, Yoshida M, Fujiwara T, Aoyagi K, Toyofuku Y, et al. Serotonin regulates glucose-stimulated insulin secretion from pancreatic β cells during pregnancy. Proc Natl Acad Sci U S A. 2013; 110:19420–5.
Article
34. Kim K, Oh CM, Ohara-Imaizumi M, Park S, Namkung J, Yadav VK, et al. Functional role of serotonin in insulin secretion in a diet-induced insulin-resistant state. Endocrinology. 2015; 156:444–52.
Article
35. Moon JH, Kim YG, Kim K, Osonoi S, Wang S, Saunders DC, et al. Serotonin regulates adult β-cell mass by stimulating perinatal β-cell proliferation. Diabetes. 2020; 69:205–14.
Article
36. Sumara G, Sumara O, Kim JK, Karsenty G. Gut-derived serotonin is a multifunctional determinant to fasting adaptation. Cell Metab. 2012; 16:588–600.
Article
37. Savelieva KV, Zhao S, Pogorelov VM, Rajan I, Yang Q, Cullinan E, et al. Genetic disruption of both tryptophan hydroxylase genes dramatically reduces serotonin and affects behavior in models sensitive to antidepressants. PLoS One. 2008; 3:e3301.
Article
38. Gutknecht L, Araragi N, Merker S, Waider J, Sommerlandt FM, Mlinar B, et al. Impacts of brain serotonin deficiency following Tph2 inactivation on development and raphe neuron serotonergic specification. PLoS One. 2012; 7:e43157.
Article
39. Shong KE, Oh CM, Namkung J, Park S, Kim H. Serotonin regulates de novo lipogenesis in adipose tissues through serotonin receptor 2A. Endocrinol Metab (Seoul). 2020; 35:470–9.
Article
40. Crane JD, Palanivel R, Mottillo EP, Bujak AL, Wang H, Ford RJ, et al. Inhibiting peripheral serotonin synthesis reduces obesity and metabolic dysfunction by promoting brown adipose tissue thermogenesis. Nat Med. 2015; 21:166–72.
Article
41. Omenetti A, Yang L, Gainetdinov RR, Guy CD, Choi SS, Chen W, et al. Paracrine modulation of cholangiocyte serotonin synthesis orchestrates biliary remodeling in adults. Am J Physiol Gastrointest Liver Physiol. 2011; 300:G303–15.
Article
42. Ruddell RG, Mann DA, Ramm GA. The function of serotonin within the liver. J Hepatol. 2008; 48:666–75.
Article
43. Richardson PD, Withrington PG. A comparison of the effects of bradykinin, 5-hydroxytryptamine and histamine on the hepatic arterial and portal venous vascular beds of the dog: histamine H1 and H2-receptor populations. Br J Pharmacol. 1977; 60:123–33.
Article
44. Cummings SA, Groszmann RJ, Kaumann AJ. Hypersensitivity of mesenteric veins to 5-hydroxytryptamine- and ketanserin-induced reduction of portal pressure in portal hypertensive rats. Br J Pharmacol. 1986; 89:501–13.
Article
45. Cummings JL, Cilento EV, Reilly FD. Hepatic microvascular regulatory mechanisms. XII. Effects of 5-HT2-receptor blockade on serotonin-induced intralobular hypoperfusion. Int J Microcirc Clin Exp. 1993; 13:99–112.
46. Kulinskii AS, Saratikov AS, Vstavskaia IuA, Udovitsina TI. Effect of substances altering the metabolism of endogenous serotonin on mitotic activity in the regenerating liver of mice. Farmakol Toksikol. 1983; 46:92–5.
47. Kulinskii VI, Saratikov AS, Vstavskaia IuA, Udovitsina TI. Receptors mediating serotonin-stimulating liver regeneration in mice. Biull Eksp Biol Med. 1983; 95:89–91.
48. Papadimas GK, Tzirogiannis KN, Mykoniatis MG, Grypioti AD, Manta GA, Panoutsopoulos GI. The emerging role of serotonin in liver regeneration. Swiss Med Wkly. 2012; 142:w13548.
Article
49. Lesurtel M, Graf R, Aleil B, Walther DJ, Tian Y, Jochum W, et al. Platelet-derived serotonin mediates liver regeneration. Science. 2006; 312:104–7.
Article
50. Ebrahimkhani MR, Oakley F, Murphy LB, Mann J, Moles A, Perugorria MJ, et al. Stimulating healthy tissue regeneration by targeting the 5-HT2B receptor in chronic liver disease. Nat Med. 2011; 17:1668–73.
Article
51. Fang Y, Liu C, Shu B, Zhai M, Deng C, He C, et al. Axis of serotonin: pERK-YAP in liver regeneration. Life Sci. 2018; 209:490–7.
Article
52. Starlinger P, Pereyra D, Hackl H, Ortmayr G, Braunwarth E, Santol J, et al. Consequences of perioperative serotonin reuptake inhibitor treatment during hepatic surgery. Hepatology. 2021; 73:1956–66.
Article
53. Moore MC, DiCostanzo CA, Dardevet D, Lautz M, Farmer B, Neal DW, et al. Portal infusion of a selective serotonin reuptake inhibitor enhances hepatic glucose disposal in conscious dogs. Am J Physiol Endocrinol Metab. 2004; 287:E1057–63.
Article
54. Haub S, Ritze Y, Ladel I, Saum K, Hubert A, Spruss A, et al. Serotonin receptor type 3 antagonists improve obesity-associated fatty liver disease in mice. J Pharmacol Exp Ther. 2011; 339:790–8.
Article
55. Choi W, Namkung J, Hwang I, Kim H, Lim A, Park HJ, et al. Serotonin signals through a gut-liver axis to regulate hepatic steatosis. Nat Commun. 2018; 9:4824.
Article
56. Ruddell RG, Oakley F, Hussain Z, Yeung I, Bryan-Lluka LJ, Ramm GA, et al. A role for serotonin (5-HT) in hepatic stellate cell function and liver fibrosis. Am J Pathol. 2006; 169:861–76.
Article
57. Kim DC, Jun DW, Kwon YI, Lee KN, Lee HL, Lee OY, et al. 5-HT2A receptor antagonists inhibit hepatic stellate cell activation and facilitate apoptosis. Liver Int. 2013; 33:535–43.
Article
58. Choi WG, Choi W, Oh TJ, Cha HN, Hwang I, Lee YK, et al. Inhibiting serotonin signaling through HTR2B in visceral adipose tissue improves obesity related insulin resistance. J Clin Invest. 2021. Oct. 7. [Epub]. https://doi.org/10.1172/JCI145331 .
Article
59. Wegermann K, Howe C, Henao R, Wang Y, Guy CD, Abdelmalek MF, et al. Serum bile acid, vitamin E, and serotonin metabolites are associated with future liver-related events in nonalcoholic fatty liver disease. Hepatol Commun. 2021; 5:608–17.
Article
60. Binetti J, Bertran L, Riesco D, Aguilar C, Martinez S, Sabench F, et al. Deregulated serotonin pathway in women with morbid obesity and NAFLD. Life (Basel). 2020; 10:245.
Article
61. Pantano L, Agyapong G, Shen Y, Zhuo Z, Fernandez-Albert F, Rust W, et al. Molecular characterization and cell type composition deconvolution of fibrosis in NAFLD. Sci Rep. 2021; 11:18045.
Article
62. Horst DA, Grace ND, LeCompte PM. Prolonged cholestasis and progressive hepatic fibrosis following imipramine therapy. Gastroenterology. 1980; 79:550–4.
Article
63. Le Bricquir Y, Larrey D, Blanc P, Pageaux GP, Michel H. Tianeptine: an instance of drug-induced hepatotoxicity predicted by prospective experimental studies. J Hepatol. 1994; 21:771–3.
64. Levine B, Jenkins AJ, Queen M, Jufer R, Smialek JE. Distribution of venlafaxine in three postmortem cases. J Anal Toxicol. 1996; 20:502–5.
Article
65. Vuppalanchi R, Hayashi PH, Chalasani N, Fontana RJ, Bonkovsky H, Saxena R, et al. Duloxetine hepatotoxicity: a case-series from the drug-induced liver injury network. Aliment Pharmacol Ther. 2010; 32:1174–83.
Article
66. Ayyash A, Holloway AC. Fluoxetine-induced hepatic lipid accumulation is linked to elevated serotonin production. Can J Physiol Pharmacol. 2021; 99:983–8.
Article
67. Pan SJ, Tan YL, Yao SW, Xin Y, Yang X, Liu J, et al. Fluoxetine induces lipid metabolism abnormalities by acting on the liver in patients and mice with depression. Acta Pharmacol Sin. 2018; 39:1463–72.
Article
68. Tuccinardi D, Farr OM, Upadhyay J, Oussaada SM, Mathew H, Paschou SA, et al. Lorcaserin treatment decreases body weight and reduces cardiometabolic risk factors in obese adults: a six-month, randomized, placebo-controlled, double-blind clinical trial. Diabetes Obes Metab. 2019; 21:1487–92.
Article
69. Cerpa V, Gonzalez A, Richerson GB. Diphtheria toxin treatment of Pet-1-Cre floxed diphtheria toxin receptor mice disrupts thermoregulation without affecting respiratory chemoreception. Neuroscience. 2014; 279:65–76.
Article
70. Wyler SC, Lord CC, Lee S, Elmquist JK, Liu C. Serotonergic control of metabolic homeostasis. Front Cell Neurosci. 2017; 11:277.
Article
71. Choi W, Moon JH, Kim H. Serotonergic regulation of energy metabolism in peripheral tissues. J Endocrinol. 2020; 245:R1–10.
Article
72. Wang L, Fan X, Han J, Cai M, Wang X, Wang Y, et al. Gut-derived serotonin contributes to the progression of non-alcoholic steatohepatitis via the liver HTR2A/PPARγ2 pathway. Front Pharmacol. 2020; 11:553.
Article
Full Text Links
  • ENM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr