Journal of Gastroenterology & Hepatology Research Category: Medical Type: Review Article

Deregulation of Lipid Homeostasis in Metabolic dysfunction–Associated Fatty Liver Disease (MAFLD)

Neha Bhat1* and Arya Mani2*
1 Janssen Pharmaceutical Companies of Johnson and Johnson, 1400 McKean Road, Springhouse, PA 19002, United states
2 Yale Cardiovascular Research Center, 300 George Street, New Haven, CT 06511, United states

*Corresponding Author(s):
Neha Bhat
Janssen Pharmaceutical Companies Of Johnson And Johnson, 1400 McKean Road, Springhouse, PA 19002, United States
Email:nehabhat22@gmail.com
Arya Mani
Yale Cardiovascular Research Center, 300 George Street, New Haven, CT 06511, United States
Tel:203-737-6118,
Email:arya.mani@yale.edu

Received Date: Oct 27, 2023
Accepted Date: Nov 06, 2023
Published Date: Nov 13, 2023

Metabolic dysfunction–Associated Fatty Liver Disease (MAFLD) affects a quarter of the world’s population, with a substantial impact on the quality of life, healthcare system, and economy. The pressing need to identify the underlying etiology and a cure is motivated by years of observation that MAFLD significantly increases the risk for fatal outcomes such as cardiovascular abnormalities, type II diabetes, liver fibrosis, liver cirrhosis, and hepatocellular carcinoma [1-3]. MAFLD is a collective term for a set of progressive disease conditions that begins with the benign accumulation of fat in the liver called steatosis, followed by inflammation, fibrosis, cirrhosis, and hepatocellular carcinoma, with ~10% of patients advancing to progressively worsening pathology [1-3]. Hepatic steatosis is often preceded by reduced disposal of glucose and glycogen synthesis by skeletal muscle, resulting in elevated plasma glucose [4-6]. The elevated glucose levels lead to increased secretion of insulin levels from the beta cells of the pancreas causing hyperinsulinemia. The elevated glucose levels and the ensuing hyperinsulinemia appears to de-sensitize the insulin receptors by locking it in a fully occupied conformation that is less competent for downstream signaling [7,8]. Reduced signaling through the receptors causes failure of Glut4 (Glucose transporter4) translocation to the plasma membrane, reduced glucose uptake, and reduced glycogen synthesis in the skeletal muscle [4, 5, 9, 10]. In the liver, insulin resistance causes excess glucose production leading to fasting hyperglycemia and in the adipose tissue, insulin-resistant adipocytes are unable to suppress the lipolysis of triglycerides into Non-Esterified Free Fatty Acids (NEFA) [4-6]. This is primarily because insulin-resistant adipocytes lose their ability to regulate ATGL and HSL lipases. The NEFA is immediately esterified to lipids upon entry into the hepatocytes. The influx of NEFA from insulin-resistant adipose tissue contributes to about 65% of liver triglycerides in MAFLD [11-13]. Glycerol, another byproduct of adipose tissue lipolysis undergoes phosphorylation into Glycerol-3-kinase (G-3K) in the liver. The G-3K leads to the generation of more glucose from oxoacetate through the actions of PEPCK, one of the rate-limiting enzymes of gluconeogenesis pathway [12,14,15]. Overall, the functions of liver, skeletal muscles, adipose tissue, and pancreas, get adversely affected and precipitates into MAFLD. 

In the liver, the insulin-responsive pathway of De-Novo Lipogenesis (DNL) continues unabated despite systemic insulin resistance [16-18]. About 25% of hepatic triglycerides are contributed by de-novo lipogenesis in the liver of MAFLD patients [11]. Hyperinsulinemia is strongly associated with increases DNL in the liver [11,16,17,19-27]. Through substrate labeling studies, it was determined that fructose does not directly contribute to DNL but the gluconeogenic precursors such as lactate and alanine lead to synthesis of lipids [28-30]. Furthermore, excess fructose consumption leads to increased acetate production from the gut microbiota which stimulates hepatic DNL [31]. The lipid accumulation in the liver both by De-Novo Lipogenesis (DNL) and NEFA esterification further suppresses the insulin signaling [32] increasing hepatic glucose production by gluconeogenesis and glycogenolysis. Altogether, a self-stimulating catastrophic cycle of hyperglycemia, hyperinsulinemia, and hyperlipidemia is mobilized in MAFLD [18,22,33]. 

The dysregulated substrate flux at the cellular level is evident as increased plasma glucose and lipid at the macroscopic organismal level. The substrate flux through various metabolic pathways is regulated by the rate-limiting enzymes, which are effectors of signaling pathways including insulin, and glucagon. The anabolic processes such as triglyceride, cholesterol, glucose, and nucleic acid syntheses are favored more in the MAFLD compared to healthy livers [11,12,34]. Accordingly, downstream effectors of Insulin signaling such as mTORC2 (mammalian target of rapamycin complex2), and PKB (Protein kinase B), increase the expression of full-length Srepb1c and Srebp2 and their downstream targets in MAFLD liver [27,35,36]. Srebp1c activates the expression of a number of rate-limiting enzymes in the triglyceride synthesis pathways such as Fasn, Acc1, Scd1, Dgat1/2, and Gpat1 while Srebp2 increases the expression of HMGCS1 and HMGCR, two rate-limiting enzymes in the cholesterol synthesis pathway [37-39]. The increased lipid accumulation contributes to reduced insulin sensitivity in the liver and skeletal muscle due to diacylglycerol-dependent inactivation of the insulin Receptor beta subunit [40-42] and/or continuous Insulin-mTORC1 signaling which negatively feedbacks to reduce the insulin-signaling output [43,44]. Despite these observations, it seems counter-intuitive that these insulin-dependent pathways remain active in the MAFLD liver despite an insulin-resistant state in the liver. A possible explanation is that a high-energy state (increased glucose, lipids, or both) increases the expression of genes that activates lipogenesis in the hepatocytes but are normally expressed at low levels. One such factor could be Dyrk1b (dual specificity tyrosine phosphorylation regulated kinase 1b), which is increased in the liver of the high calorie fed mice and also in the human liver biopsies from MASH patients [32]. Dyrk1b induces lipogenesis by directly activating mTORC2, the central regulator of lipogenesis, in the fasting liver, when endogenous insulin signaling is minimal [32]. The gain of function mutations in Dyrk1b was previously associated with metabolic syndrome in humans in multiple large families in southwest Iran and in the United States [45]. We showed that Dyrk1b increases lipogenesis in an insulin-resistant liver providing direct relevance to the human disease condition [32]. 

The concurrent existence of type II diabetes and MAFLD indicates shared metabolic pathways that could be targeted therapeutically by drugs that achieve glycemic control. However, glucose-reducing drugs such as metformin, Sglt2 inhibitors, and PPAR agonists have been unsuccessful in reducing hepatic steatosis, inflammation, and fibrosis [46-55]. Several other therapeutic targets that were deduced rationally from metabolic pathways that trigger steatoses such as Acc1, DGAT1, and Scd1 [56, 57], bile acid analogs [58] that prohibit liver steatosis by transcriptional activation of Fgf19 [59], and Fgf21 analogs [60-62] have not been successful either. This is either due to non-specific targeting of unintended pathways, and/or compensatory activation of feedback pathways resulting in diminished efficacy. The field needs a therapeutic modality that can be delivered specifically to the hepatocytes and targets the intended molecule very specifically resulting in reduced hepatic steatosis, and inflammation without any non-specific effects. Further, to treat advanced pathologies such as fibrosis and cirrhosis, novel solutions and a meta-analysis of intercellular and inter-organ signaling networks is imperative. A recent FDA-approved agonist for gut hormone GLP-1 (glucagon-like peptide-1) has been very popular in reducing obesity by up to 22% [63]. Considering that reduction in body weight is an approved first-line recommendation for MAFLD [64], liver steatosis was improved in patients administered with semaglutides [47,51]. However, advanced MAFLD pathologies may necessitate a different therapeutic intervention. Another promising drug that has reached phase III is the Thyroid hormone beta-agonist which is specifically directed to the liver where it stimulates mitochondrial respiration and promotes the breakdown of lipids [65,66]. Altogether, the emerging therapies are promising and offer possibilities of a cure for MAFLD in the near future.

References

  1. Younossi ZM, Golabi P, Avila LD, Paik JM, Srishord M, et al. (2019) The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: A systematic review and meta-analysis. J Hepatol 71: 793-801.
  2. Loomba R, Abraham M, Unalp A, Wilson L, Lavine J, et al. (2012) Association between diabetes, family history of diabetes, and risk of nonalcoholic steatohepatitis and fibrosis. Hepatology 56: 943-951.
  3. Targher G, Corey KE, Byrne CD, Roden M (2021) The complex link between NAFLD and type 2 diabetes mellitus - mechanisms and treatments. Nat Rev Gastroenterol Hepatol 18: 599-612.
  4. DeFronzo RA, Ferrannini E, Groop L, Henry RR, Herman WH, et al. (2015) Type 2 diabetes mellitus. Nat Rev Dis Primers 1: 15019.
  5. DeFronzo RA, Tripathy D (2009) Skeletal muscle insulin resistance is the primary defect in type 2 diabetes. Diabetes Care 32: 157-163.
  6. Shulman GI (2014) Ectopic fat in insulin resistance, dyslipidemia, and cardiometabolic disease. N Engl J Med 371: 1131-1141.
  7. Scapin, G, Dandey VP, Zhang Z, Prosise W, Hruza A, et al. (2018) Structure of the insulin receptor-insulin complex by single-particle cryo-EM analysis. Nature 556: 122-125.
  8. Nielsen J, Brandt J, Boesen T, Hummelshøj T, Slaaby R, et al. (2022) Structural Investigations of Full-Length Insulin Receptor Dynamics and Signalling. J Mol Biol 434: 167458.
  9. Kashyap SR, Belfort R, Berria R, Suraamornku S, Pratipranawatr T, et al. (2004) Discordant effects of a chronic physiological increase in plasma FFA on insulin signaling in healthy subjects with or without a family history of type 2 diabetes. Am J Physiol Endocrinol Metab 287: 537-546.
  10. Shulman GI, Rothman DL, Jue T, Stein P, DeFronzo RA, et al. (1990) Quantitation of muscle glycogen synthesis in normal subjects and subjects with non-insulin-dependent diabetes by 13C nuclear magnetic resonance spectroscopy. N Engl J Med 322: 223-228.
  11. Donnelly KL, Smith CI, Schwarzenberg SJ, Jessurun J, Boldt MD, et al. (2005) Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J Clin Invest 115: 1343-1351.
  12. Perry RJ, Camporez JPGC, Kursawe R, Titchenell PM, Zhang D, et al. (2015) Hepatic acetyl CoA links adipose tissue inflammation to hepatic insulin resistance and type 2 diabetes. Cell 160: 745-758.
  13. Vatner DF, Majumdar SK, Kumashiro N, Petersen MC, Rahimi Y, et al. (2015) Insulin-independent regulation of hepatic triglyceride synthesis by fatty acids. Proc Natl Acad Sci USA 112: 1143-1148.
  14. Kalemba KM, Wang Y, Xu H, Chiles E, McMillin SM, et al. (2019) Glycerol induces G6pc in primary mouse hepatocytes and is the preferred substrate for gluconeogenesis both in vitro and in vivo. J Biol Chem 294: 18017-18028.
  15. Nurjhan N, Consoli A, Gerich J (1992) Increased lipolysis and its consequences on gluconeogenesis in non-insulin-dependent diabetes mellitus. J Clin Invest 89: 169-175.
  16. Parks EJ, Skokan LE, Timlin MT, Dingfelder CS (2008) Dietary sugars stimulate fatty acid synthesis in adults. J Nutr 138: 1039-1046.
  17. Parks EJ, Krauss RM, Christiansen MP, Neese RA, Hellerstein MK (1999) Effects of a low-fat, high-carbohydrate diet on VLDL-triglyceride assembly, production, and clearance. J Clin Invest 104: 1087-1096.
  18. Bhat N, Mani A (2023) Dysregulation of Lipid and Glucose Metabolism in Nonalcoholic Fatty Liver Disease. Nutrients 15: 2323.
  19. Smith GI, Shankaran M, Yoshino M, Schweitzer GG, Chondronikola M, et al. (2020) Insulin resistance drives hepatic de novo lipogenesis in nonalcoholic fatty liver disease. J Clin Invest 130: 1453-1460.
  20. Cook JR, Langlet F, Kido Y, Accili D (2015) Pathogenesis of selective insulin resistance in isolated hepatocytes. J Biol Chem 290: 13972-13980.
  21. Bril F, Lomonaco R, Orsak B, Ortiz-Lopez C, Webb A, et al. (2014) Relationship between disease severity, hyperinsulinemia, and impaired insulin clearance in patients with nonalcoholic steatohepatitis. Hepatology 59: 2178-2187.
  22. Rhee EJ, Lee WY, Cho YK, Kim BI, Sung KC (2011) Hyperinsulinemia and the development of nonalcoholic Fatty liver disease in nondiabetic adults. Am J Med 124: 69-76.
  23. Ota T, Takamura T, Kurita S, Matsuzawa N, Kita Y, et al. (2007) Insulin resistance accelerates a dietary rat model of nonalcoholic steatohepatitis. Gastroenterology 132: 282-293.
  24. George S, Rochford JJ, Wolfrum C, Gray SL, Schinner S, et al. (2004) A family with severe insulin resistance and diabetes due to a mutation in AKT2. Science 304: 1325-1328.
  25. Miyake K, Ogawa W, Matsumoto M, Nakamura T, Sakaue H, et al. (2002) Hyperinsulinemia, glucose intolerance, and dyslipidemia induced by acute inhibition of phosphoinositide 3-kinase signaling in the liver. J Clin Invest 110: 1483-1491.
  26. Chitturi S, Abeygunasekera S, Farrell GC, Holmes-Walker J, Hui JM, et al. (2002) NASH and insulin resistance: Insulin hypersecretion and specific association with the insulin resistance syndrome. Hepatology 35: 373-379.
  27. Shimomura I, Matsuda M, Hammer RE, Bashmakov Y, Brown MS, et al. (2000) Decreased IRS-2 and increased SREBP-1c lead to mixed insulin resistance and sensitivity in livers of lipodystrophic and ob/ob mice. Mol Cell 6: 77-86.
  28. Sun SZ, Empie MW (2012) Fructose metabolism in humans - what isotopic tracer studies tell us. Nutr Metab (Lond) 9: 89.
  29. Palacín M, Lasunción MA, Herrera E (1988) Utilization of glucose, alanine, lactate, and glycerol as lipogenic substrates by periuterine adipose tissue in situ in fed and starved rats. J Lipid Res 29: 26-32.
  30. Domènech M, López-Soriano FJ, Argilés JM (1993) Alanine as a lipogenic precursor in isolated hepatocytes from obese Zucker rats. Cell Mol Biol (Noisy-le-grand) 39: 693-699.
  31. Zhao S, Jang C, Liu J, Uehara K, Gilbert M, et al. (2020) Dietary fructose feeds hepatic lipogenesis via microbiota-derived acetate. Nature 579: 586-591.
  32. Bhat N, Narayanan A, Fathzadeh M, Kahn M, Zang D, et al. (2022) Dyrk1b promotes hepatic lipogenesis by bypassing canonical insulin signaling and directly activating mTORC2 in mice. J Clin Invest 132.
  33. El Ouaamari A, Kawamori D, Dirice E, Liew CW, Shadrach Jl, et al. (2013) Liver-derived systemic factors drive beta cell hyperplasia in insulin-resistant states. Cell Rep 3: 401-410.
  34. Fletcher JA, Deja S, Satapati S, Fu X, Burgess SC, et al. (2019) Impaired ketogenesis and increased acetyl-CoA oxidation promote hyperglycemia in human fatty liver. JCI Insight 5.
  35. Shimomura I, Ikemoto YB, Horton JD, Brown MS, Goldstein JL, et al. (1999) Insulin selectively increases SREBP-1c mRNA in the livers of rats with streptozotocin-induced diabetes. Proc Natl Acad Sci USA 96: 13656-13661.
  36. Yahagi N, Shimano H, Hasty AH, Matsuzaka T, Ide T, et al. (2002) Absence of sterol regulatory element-binding protein-1 (SREBP-1) ameliorates fatty livers but not obesity or insulin resistance in Lep(ob)/Lep(ob) mice. J Biol Chem 277: 19353-19357.
  37. Foretz M, Guichard C, Foufelle F (1999) Sterol regulatory element binding protein-1c is a major mediator of insulin action on the hepatic expression of glucokinase and lipogenesis-related genes. Proc Natl Acad Sci U S A 96: 12737-12742.
  38. Horton JD, Goldstein JL, Brown MS (2002) SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J Clin Invest 109: 1125-1131.
  39. Liang G, Yang J, Horton JD, Hammer RE, Goldstein JL, et al. (2002) Diminished hepatic response to fasting/refeeding and liver X receptor agonists in mice with selective deficiency of sterol regulatory element-binding protein-1c. J Biol Chem 277: 9520-9528.
  40. Lyu K, Zhang D, Kahn M, Horst KWT, Rodrigues RC, et al. (2020) A Membrane-Bound Diacylglycerol Species Induces PKC-Mediated Hepatic Insulin Resistance. Cell Metab 32: 654-664.
  41. Petersen MC, Madiraju Ak, Gassaway BM, Marcel M, Nasiri AR (2016) et al. Insulin receptor Thr1160 phosphorylation mediates lipid-induced hepatic insulin resistance. J Clin Invest 126: 4361-4371.
  42. Samuel VT, Liu ZX, Wang A, Beddow SA, Geisler JG, et al. (2007) Inhibition of protein kinase Cepsilon prevents hepatic insulin resistance in nonalcoholic fatty liver disease. J Clin Invest 117: 739-745.
  43. Ardestani A, Lupse B, Kido Y, Leibowitz G, Maedler, et al. (2018) mTORC1 Signaling: A Double-Edged Sword in Diabetic beta Cells. Cell Metab 27: 314-331.
  44. Um SH, Frigerio F, Watanabe M, Picard F, Joaquin M, et al. (2004) Absence of S6K1 protects against age- and diet-induced obesity while enhancing insulin sensitivity. Nature 431: 200-205.
  45. Keramati AR, Fathzadeh M, Go GW, Singh R, Choi M, et al. (2014) A form of the metabolic syndrome associated with mutations in DYRK1B. N Engl J Med 370: 1909-1919.
  46. Francque SM, Bedossa P, Ratziu V, Anstee QM, Bugainesi E, et al. (2021) A Randomized, Controlled Trial of the Pan-PPAR Agonist Lanifibranor in NASH. N Engl J Med 385: 1547-1558.
  47. Flint A, Andersen G, Hockings P, Jonhansson L, Morsing A, et al. (2021) Randomised clinical trial: semaglutide versus placebo reduced liver steatosis but not liver stiffness in subjects with non-alcoholic fatty liver disease assessed by magnetic resonance imaging. Aliment Pharmacol Ther 54: 1150-1161.
  48. Santos RD, Filho RB (2020) Treatment of nonalcoholic fatty liver disease with dapagliflozin in non-diabetic patients. Metabol Open 5: 100028.
  49. Akuta N (2019) Impact of sodium glucose cotransporter 2 inhibitor on histological features and glucose metabolism of non-alcoholic fatty liver disease complicated by diabetes mellitus. Hepatol Res 49: 531-539.
  50. Armstrong MJ, Gaunt P, Aithal GP, Barton D, Hull D, et al. (2016) Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet 387: 679-690.
  51. Newsome PN, Cusi K, Linder M, Okanoue T, Ratziu V, et al. (2021) A Placebo-Controlled Trial of Subcutaneous Semaglutide in Nonalcoholic Steatohepatitis. N Engl J Med 384: 1113-1124.
  52. Shimizu M, Suzuki K, Kato K, Jojima T, Lijima T, et al. (2019) Evaluation of the effects of dapagliflozin, a sodium-glucose co-transporter-2 inhibitor, on hepatic steatosis and fibrosis using transient elastography in patients with type 2 diabetes and non-alcoholic fatty liver disease. Diabetes Obes Metab 21: 285-292.
  53. Harrison SA, Smith WB, Chen CY, Cheng P, Badman MK, et al. (2022) Licogliflozin for nonalcoholic steatohepatitis: a randomized, double-blind, placebo-controlled, phase 2a study. Nat Med 28:1432-1438.
  54. Han E, Lee YH, Lee BW, Kang ES, Cha BS, et al. (2020) Ipragliflozin Additively Ameliorates Non-Alcoholic Fatty Liver Disease in Patients with Type 2 Diabetes Controlled with Metformin and Pioglitazone: A 24-Week Randomized Controlled Trial. J Clin Med 9: 259.
  55. Inoue M (2019) Effects of canagliflozin on body composition and hepatic fat content in type 2 diabetes patients with non-alcoholic fatty liver disease. J Diabetes Investig 10: 1004-1011.
  56. Calle RA, Amin NB, Gonzalez SC, Ross TT, Bergman A, et al. (2021) ACC inhibitor alone or co-administered with a DGAT2 inhibitor in patients with non-alcoholic fatty liver disease: two parallel, placebo-controlled, randomized phase 2a trials. Nat Med 27: 1836-1848.
  57. Ratziu V, Guevara LD, Poordad F, Fuster F, Arrese M, et al. (2021) Aramchol in patients with nonalcoholic steatohepatitis: a randomized, double-blind, placebo-controlled phase 2b trial. Nat Med 27: 1825-1835.
  58. Younossi ZM, Ratziu V, Loomba R, Rinella M, Anstee QM, et al. (2019) Obeticholic acid for the treatment of non-alcoholic steatohepatitis: interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial. Lancet 394: 2184-2196.
  59. Bhat N, Esteghamat F, Chaube BK, Mani M, Jain D, et al. (2022) TCF7L2 transcriptionally regulates Fgf15 to maintain bile acid and lipid homeostasis through gut-liver crosstalk. FASEB J 36: 22185.
  60. Harrison SA, Ruane PJ, Neff G, Yale K, Fong E, et al. (2021) Efruxifermin in non-alcoholic steatohepatitis: a randomized, double-blind, placebo-controlled, phase 2a trial. Nat Med 27: 1262-1271.
  61. Charles ED, Tetri BA, Frias JP, Kundu S, Luo Y, et al. (2019) Pegbelfermin (BMS-986036), PEGylated FGF21, in Patients with Obesity and Type 2 Diabetes: Results from a Randomized Phase 2 Study. Clincal Trial 27: 41-49.
  62. Sanyal A, Charles ED, Tetri BA, Loomba R, Harrison SA, et al. (2019) Pegbelfermin (BMS-986036), a PEGylated fibroblast growth factor 21 analogue, in patients with non-alcoholic steatohepatitis: a randomised, double-blind, placebo-controlled, phase 2a trial. Lancet 392: 2705-2717.
  63. Nogueiras R, Nauck MA, Tschop MH (2023) Gut hormone co-agonists for the treatment of obesity: from bench to bedside. Nat Metab 5: 933-944.
  64. Andreasen CR, Andersen A, Vilsboll T (2023) The future of incretins in the treatment of obesity and non-alcoholic fatty liver disease. Diabetologia 66: 1846-1858.
  65. Harrison SA, Bashir M, Moussa SE, McCarty K, Frias JP, et al. (2021) Effects of Resmetirom on Noninvasive Endpoints in a 36-Week Phase 2 Active Treatment Extension Study in Patients With NASH. Hepatol Commun 5: 573-588.
  66. Harrison SA, Bashir MR, Guy CD, Zhou R, Frias JP, et al. (2019) Resmetirom (MGL-3196) for the treatment of non-alcoholic steatohepatitis: a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet 394: 2012-2024.

Citation: Bhat N, Mani A (2023) Deregulation of Lipid Homeostasis in Metabolic dysfunction–Associated Fatty Liver Disease (MAFLD). J GastroenterolHepatology Res 7: 045

Copyright: © 2023  Neha Bhat, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.


Herald Scholarly Open Access is a leading, internationally publishing house in the fields of Sciences. Our mission is to provide an access to knowledge globally.



© 2024, Copyrights Herald Scholarly Open Access. All Rights Reserved!