Diabetes Metab J.  2021 Sep;45(5):655-674. 10.4093/dmj.2021.0197.

Rho-Kinase as a Therapeutic Target for Nonalcoholic Fatty Liver Diseases

Affiliations
  • 1CEDOC-Chronic Disease Research Center, NOVA Medical School/ Faculty of Medical Sciences, New University of Lisbon, Lisbon, Portugal
  • 2Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
  • 3Center for Neuroscience and Cell Biology, University of Coimbra, Marquis of Pombal Square, Coimbra, Portugal

Abstract

Nonalcoholic fatty liver disease (NAFLD) is a major public health problem and the most common form of chronic liver disease, affecting 25% of the global population. Although NAFLD is closely linked with obesity, insulin resistance, and type 2 diabetes mellitus, knowledge on its pathogenesis remains incomplete. Emerging data have underscored the importance of Rho-kinase (Rho-associated coiled-coil-containing kinase [ROCK]) action in the maintenance of normal hepatic lipid homeostasis. In particular, pharmacological blockade of ROCK in hepatocytes or hepatic stellate cells prevents the progression of liver diseases such as NAFLD and fibrosis. Moreover, mice lacking hepatic ROCK1 are protected against obesity-induced fatty liver diseases by suppressing hepatic de novo lipogenesis. Here we review the roles of ROCK as an indispensable regulator of obesity-induced fatty liver disease and highlight the key cellular pathway governing hepatic lipid accumulation, with focus on de novo lipogenesis and its impact on therapeutic potential. Consequently, a comprehensive understanding of the metabolic milieu linking to liver dysfunction triggered by ROCK activation may help identify new targets for treating fatty liver diseases such as NAFLD.

Keyword

Rho-kinase; AMP-activated protein kinase; Diet, high-fat; Lipogenesis; Nonalcoholic fatty liver disease

Figure

  • Fig. 1. Schematic of de novo lipogenesis. Entry points for the main nutrient precursors are shown in gray and the sources of hydrogens of the fatty acyl coenzyme A (acyl-CoA) product are represented by different colors: ‘H+’ hydrogens derived from body water, black ‘H’ for the methyl hydrogens of the initial acetyl-CoA that binds to acyl-CoA synthase, red ‘H’ for the hydrogens derived from nicotinamide adenine dinucleotide phosphate (NADPH) via the pentose phosphate pathway (PPP), and ‘H’ for the hydrogens derived from malonyl-CoA. Also represented is the acyl-CoA synthase complex (ACoAS) following the initial acetylCoA and malonyl-CoA additions. Intracellular acetyl-CoA is represented as distinct mitochondrial (Acetyl-CoAMITO) and cytosolic (acetyl-CoACYTO) pools. CoASH, free acetyl CoA; SCoA, CoA moiety; SCFA, short chain fatty acids; OAA, oxaloacetate; CIT, citrate; AoCS, Co moiety

  • Fig. 2. Role of Rho-associated coiled-coil-containing kinase (ROCK1) in the development of nonalcoholic fatty liver disease (NAFLD). High-fat feeding increases endocannabinoids 2-arachidonoylglycerol and anandamide levels produced from hepatic stellate cells of the liver. Endocannabinoids stimulate ROCK1 activity via cannabinoid receptor type 1 (CB1), which inhibits AMP-activated protein kinase (AMPK) activity. Suppressed AMPK activity then drives the sterol regulatory element-binding protein 1c (SREBP1c)-mediated lipogenic program, leading to a rise of lipid accumulation. NAFLD ultimately develops with the progression of obesity. On the other hand, it is hypothesized that activated ROCK1 binds to liver X receptor (LXRα) and may prompt the lipogenic pathway through SREBP1c. As an early event of ROCK1 signaling, CB1-mediated Gα12 (or Gαs) activation could be an initial step for ROCK1-dependent de novo lipogenesis. GTP, guanosine triphosphate; FAS, fatty acid synthase; SCD1, stearoyl-CoA desaturase-1; ACC, acetyl-CoA carboxylase.


Reference

1. Birkenfeld AL, Shulman GI. Nonalcoholic fatty liver disease, hepatic insulin resistance, and type 2 diabetes. Hepatology. 2014; 59:713–23.
Article
2. Kopelman PG. Obesity as a medical problem. Nature. 2000; 404:635–43.
Article
3. Thibaut R, Gage MC, Pineda-Torra I, Chabrier G, Venteclef N, Alzaid F. Liver macrophages and inflammation in physiology and physiopathology of non-alcoholic fatty liver disease. FEBS J. 2021 Apr 15 [Epub]. https://doi.org/10.1111/febs.15877.
Article
4. Ayonrinde OT. Historical narrative from fatty liver in the nineteenth century to contemporary NAFLD: reconciling the present with the past. JHEP Rep. 2021; 3:100261.
5. Wong RJ, Aguilar M, Cheung R, Perumpail RB, Harrison SA, Younossi ZM, et al. Nonalcoholic steatohepatitis is the second leading etiology of liver disease among adults awaiting liver transplantation in the United States. Gastroenterology. 2015; 148:547–55.
Article
6. Bellentani S. The epidemiology of non-alcoholic fatty liver disease. Liver Int. 2017; 37 Suppl 1:81–4.
Article
7. Sayiner M, Koenig A, Henry L, Younossi ZM. Epidemiology of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis in the United States and the rest of the world. Clin Liver Dis. 2016; 20:205–14.
Article
8. Samuel VT, Shulman GI. Nonalcoholic fatty liver disease as a nexus of metabolic and hepatic diseases. Cell Metab. 2018; 27:22–41.
Article
9. Loomba R, Friedman SL, Shulman GI. Mechanisms and disease consequences of nonalcoholic fatty liver disease. Cell. 2021; 184:2537–64.
Article
10. Younossi ZM, Golabi P, de Avila L, Paik JM, Srishord M, Fukui N, et al. The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: a systematic review and meta-analysis. J Hepatol. 2019; 71:793–801.
Article
11. Smith BW, Adams LA. Nonalcoholic fatty liver disease and diabetes mellitus: pathogenesis and treatment. Nat Rev Endocrinol. 2011; 7:456–65.
Article
12. Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism. 2016; 65:1038–48.
Article
13. Katsarou A, Moustakas II, Pyrina I, Lembessis P, Koutsilieris M, Chatzigeorgiou A. Metabolic inflammation as an instigator of fibrosis during non-alcoholic fatty liver disease. World J Gastroenterol. 2020; 26:1993–2011.
Article
14. Prasun P. Mitochondrial dysfunction in metabolic syndrome. Biochim Biophys Acta Mol Basis Dis. 2020; 1866:165838.
Article
15. Masarone M, Rosato V, Dallio M, Gravina AG, Aglitti A, Loguercio C, et al. Role of oxidative stress in pathophysiology of nonalcoholic fatty liver disease. Oxid Med Cell Longev. 2018; 2018:9547613.
Article
16. Bourebaba N, Marycz K. Hepatic stellate cells role in the course of metabolic disorders development: a molecular overview. Pharmacol Res. 2021; 170:105739.
17. Blaner WS, O’Byrne SM, Wongsiriroj N, Kluwe J, D’Ambrosio DM, Jiang H, et al. Hepatic stellate cell lipid droplets: a specialized lipid droplet for retinoid storage. Biochim Biophys Acta. 2009; 1791:467–73.
Article
18. Zisser A, Ipsen DH, Tveden-Nyborg P. Hepatic stellate cell activation and inactivation in NASH-fibrosis: roles as putative treatment targets? Biomedicines. 2021; 9:365.
19. Kitto LJ, Henderson NC. Hepatic stellate cell regulation of liver regeneration and repair. Hepatol Commun. 2020; 5:358–70.
Article
20. Berumen J, Baglieri J, Kisseleva T, Mekeel K. Liver fibrosis: pathophysiology and clinical implications. Wiley Interdiscip Rev Syst Biol Med. 2021; 13:e1499.
Article
21. Kisseleva T, Cong M, Paik Y, Scholten D, Jiang C, Benner C, et al. Myofibroblasts revert to an inactive phenotype during regression of liver fibrosis. Proc Natl Acad Sci U S A. 2012; 109:9448–53.
Article
22. Iredale JP, Benyon RC, Pickering J, McCullen M, Northrop M, Pawley S, et al. Mechanisms of spontaneous resolution of rat liver fibrosis: hepatic stellate cell apoptosis and reduced hepatic expression of metalloproteinase inhibitors. J Clin Invest. 1998; 102:538–49.
Article
23. Duarte N, Coelho IC, Patarrao RS, Almeida JI, Penha-Goncalves C, Macedo MP. How inflammation impinges on NAFLD: a role for Kupffer cells. Biomed Res Int. 2015; 2015:984578.
Article
24. Lanthier N, Molendi-Coste O, Cani PD, van Rooijen N, Horsmans Y, Leclercq IA. Kupffer cell depletion prevents but has no therapeutic effect on metabolic and inflammatory changes induced by a high-fat diet. FASEB J. 2011; 25:4301–11.
Article
25. Neyrinck AM, Cani PD, Dewulf EM, De Backer F, Bindels LB, Delzenne NM. Critical role of Kupffer cells in the management of diet-induced diabetes and obesity. Biochem Biophys Res Commun. 2009; 385:351–6.
Article
26. Nguyen-Lefebvre AT, Horuzsko A. Kupffer cell metabolism and function. J Enzymol Metab. 2015; 1:101.
27. Matsui T, Amano M, Yamamoto T, Chihara K, Nakafuku M, Ito M, et al. Rho-associated kinase, a novel serine/threonine kinase, as a putative target for small GTP binding protein Rho. EMBO J. 1996; 15:2208–16.
Article
28. Nakagawa O, Fujisawa K, Ishizaki T, Saito Y, Nakao K, Narumiya S. ROCK-I and ROCK-II, two isoforms of Rho-associated coiled-coil forming protein serine/threonine kinase in mice. FEBS Lett. 1996; 392:189–93.
Article
29. Ishizaki T, Maekawa M, Fujisawa K, Okawa K, Iwamatsu A, Fujita A, et al. The small GTP-binding protein Rho binds to and activates a 160 kDa Ser/Thr protein kinase homologous to myotonic dystrophy kinase. EMBO J. 1996; 15:1885–93.
Article
30. Leung T, Manser E, Tan L, Lim L. A novel serine/threonine kinase binding the Ras-related RhoA GTPase which translocates the kinase to peripheral membranes. J Biol Chem. 1995; 270:29051–4.
Article
31. Uehata M, Ishizaki T, Satoh H, Ono T, Kawahara T, Morishita T, et al. Calcium sensitization of smooth muscle mediated by a Rho-associated protein kinase in hypertension. Nature. 1997; 389:990–4.
Article
32. Hirooka Y, Shimokawa H. Therapeutic potential of rho-kinase inhibitors in cardiovascular diseases. Am J Cardiovasc Drugs. 2005; 5:31–9.
Article
33. Hu E, Lee D. Rho kinase as potential therapeutic target for cardiovascular diseases: opportunities and challenges. Expert Opin Ther Targets. 2005; 9:715–36.
Article
34. Shimokawa H, Sunamura S, Satoh K. RhoA/Rho-kinase in the cardiovascular system. Circ Res. 2016; 118:352–66.
Article
35. Kitamura K, Tada S, Nakamoto N, Toda K, Horikawa H, Kurita S, et al. Rho/Rho kinase is a key enzyme system involved in the angiotensin II signaling pathway of liver fibrosis and steatosis. J Gastroenterol Hepatol. 2007; 22:2022–33.
Article
36. Kuroda S, Tashiro H, Igarashi Y, Tanimoto Y, Nambu J, Oshita A, et al. Rho inhibitor prevents ischemia-reperfusion injury in rat steatotic liver. J Hepatol. 2012; 56:146–52.
Article
37. Kuroda S, Tashiro H, Kimura Y, Hirata K, Tsutada M, Mikuriya Y, et al. Rho-kinase inhibitor targeting the liver prevents ischemia/reperfusion injury in the steatotic liver without major systemic adversity in rats. Liver Transpl. 2015; 21:123–31.
Article
38. Klein S, Van Beuge MM, Granzow M, Beljaars L, Schierwagen R, Kilic S, et al. HSC-specific inhibition of Rho-kinase reduces portal pressure in cirrhotic rats without major systemic effects. J Hepatol. 2012; 57:1220–7.
Article
39. van Beuge MM, Prakash J, Lacombe M, Gosens R, Post E, Reker-Smit C, et al. Reduction of fibrogenesis by selective delivery of a Rho kinase inhibitor to hepatic stellate cells in mice. J Pharmacol Exp Ther. 2011; 337:628–35.
Article
40. Wang J, Jiang W. The effects of RKI-1447 in a mouse model of nonalcoholic fatty liver disease induced by a high-fat diet and in HepG2 human hepatocellular carcinoma cells treated with oleic acid. Med Sci Monit. 2020; 26:e919220.
Article
41. Zhou H, Fang C, Zhang L, Deng Y, Wang M, Meng F. Fasudil hydrochloride hydrate, a Rho-kinase inhibitor, ameliorates hepatic fibrosis in rats with type 2 diabetes. Chin Med J (Engl). 2014; 127:225–31.
42. Xie Y, Song T, Huo M, Zhang Y, Zhang YY, Ma ZH, et al. Fasudil alleviates hepatic fibrosis in type 1 diabetic rats: involvement of the inflammation and RhoA/ROCK pathway. Eur Rev Med Pharmacol Sci. 2018; 22:5665–77.
43. Tada S, Iwamoto H, Nakamuta M, Sugimoto R, Enjoji M, Nakashima Y, et al. A selective ROCK inhibitor, Y27632, prevents dimethylnitrosamine-induced hepatic fibrosis in rats. J Hepatol. 2001; 34:529–36.
Article
44. Ding R, Han J, Zhao D, Hu Z, Ma X. Pretreatment with Rhokinase inhibitor ameliorates lethal endotoxemia-induced liver injury by improving mitochondrial function. Int Immuno-pharmacol. 2016; 40:125–30.
Article
45. Thorlacius K, Slotta JE, Laschke MW, Wang Y, Menger MD, Jeppsson B, et al. Protective effect of fasudil, a Rho-kinase inhibitor, on chemokine expression, leukocyte recruitment, and hepatocellular apoptosis in septic liver injury. J Leukoc Biol. 2006; 79:923–31.
Article
46. Murata T, Arii S, Mori A, Imamura M. Therapeutic significance of Y-27632, a Rho-kinase inhibitor, on the established liver fibrosis. J Surg Res. 2003; 114:64–71.
Article
47. van Beuge MM, Prakash J, Lacombe M, Post E, Reker-Smit C, Beljaars L, et al. Increased liver uptake and reduced hepatic stellate cell activation with a cell-specific conjugate of the Rhokinase inhibitor Y27632. Pharm Res. 2011; 28:2045–54.
Article
48. Klein S, Frohn F, Magdaleno F, Reker-Smit C, Schierwagen R, Schierwagen I, et al. Rho-kinase inhibitor coupled to peptidemodified albumin carrier reduces portal pressure and increases renal perfusion in cirrhotic rats. Sci Rep. 2019; 9:2256.
Article
49. Huang H, Lee SH, Sousa-Lima I, Kim SS, Hwang WM, Dagon Y, et al. Rho-kinase/AMPK axis regulates hepatic lipogenesis during overnutrition. J Clin Invest. 2018; 128:5335–50.
Article
50. Murata T, Arii S, Nakamura T, Mori A, Kaido T, Furuyama H, et al. Inhibitory effect of Y-27632, a ROCK inhibitor, on progression of rat liver fibrosis in association with inactivation of hepatic stellate cells. J Hepatol. 2001; 35:474–81.
Article
51. Ikeda H, Kume Y, Tejima K, Tomiya T, Nishikawa T, Watanabe N, et al. Rho-kinase inhibitor prevents hepatocyte damage in acute liver injury induced by carbon tetrachloride in rats. Am J Physiol Gastrointest Liver Physiol. 2007; 293:G911–7.
Article
52. Laschke MW, Dold S, Jeppsson B, Schilling MK, Menger MD, Thorlacius H. Rho-kinase inhibitor attenuates cholestasis-induced CXC chemokine formation, leukocyte recruitment, and hepatocellular damage in the liver. J Surg Res. 2010; 159:666–73.
Article
53. Ogawa T, Tashiro H, Miyata Y, Ushitora Y, Fudaba Y, Kobayashi T, et al. Rho-associated kinase inhibitor reduces tumor recurrence after liver transplantation in a rat hepatoma model. Am J Transplant. 2007; 7:347–55.
Article
54. Mann DA, Smart DE. Transcriptional regulation of hepatic stellate cell activation. Gut. 2002; 50:891–6.
Article
55. Narumiya S, Ishizaki T, Uehata M. Use and properties of ROCK-specific inhibitor Y-27632. Methods Enzymol. 2000; 325:273–84.
Article
56. Ono-Saito N, Niki I, Hidaka H. H-series protein kinase inhibitors and potential clinical applications. Pharmacol Ther. 1999; 82:123–31.
Article
57. Ali Q, Dalapati S, Prakash N, Narayan P, Paka L, Li J, et al. Novel Rho associated coiled kinase 2 (ROCK2) inhibitor reduces steatosis and fibrosis in mice model of liver disease. FASEB J. 2020; 34(S1):1.
Article
58. Ali Q, Prakash N, Dalapati S, Li J, Goldberg I, Paka L. Novel Rho associated coiled kinase inhibitor improves hepatic fibrosis in mice model of non-alcoholic steatohepatitis (NASH). J Hepatol. 2020; 73:S524.
Article
59. Wingard C, Fulton D, Husain S. Altered penile vascular reactivity and erection in the Zucker obese-diabetic rat. J Sex Med. 2007; 4:348–63.
Article
60. Kikuchi Y, Yamada M, Imakiire T, Kushiyama T, Higashi K, Hyodo N, et al. A Rho-kinase inhibitor, fasudil, prevents development of diabetes and nephropathy in insulin-resistant diabetic rats. J Endocrinol. 2007; 192:595–603.
Article
61. Kolavennu V, Zeng L, Peng H, Wang Y, Danesh FR. Targeting of RhoA/ROCK signaling ameliorates progression of diabetic nephropathy independent of glucose control. Diabetes. 2008; 57:714–23.
Article
62. Mishra RK, Alokam R, Sriram D, Yogeeswari P. Potential role of Rho kinase inhibitors in combating diabetes-related complications including diabetic neuropathy: a review. Curr Diabetes Rev. 2013; 9:249–66.
63. Huang H, Lee SH, Ye C, Lima IS, Oh BC, Lowell BB, et al. ROCK1 in AgRP neurons regulates energy expenditure and locomotor activity in male mice. Endocrinology. 2013; 154:3660–70.
Article
64. Noda K, Nakajima S, Godo S, Saito H, Ikeda S, Shimizu T, et al. Rho-kinase inhibition ameliorates metabolic disorders through activation of AMPK pathway in mice. PLoS One. 2014; 9:e110446.
Article
65. Kanda T, Wakino S, Homma K, Yoshioka K, Tatematsu S, Hasegawa K, et al. Rho-kinase as a molecular target for insulin resistance and hypertension. FASEB J. 2006; 20:169–71.
Article
66. Alves-Bezerra M, Cohen DE. Triglyceride metabolism in the liver. Compr Physiol. 2017; 8:1–8.
67. Solinas G, Boren J, Dulloo AG. De novo lipogenesis in metabolic homeostasis: more friend than foe? Mol Metab. 2015; 4:367–77.
Article
68. Moffett JR, Puthillathu N, Vengilote R, Jaworski DM, Namboodiri AM. Acetate revisited: a key biomolecule at the nexus of metabolism, epigenetics and oncogenesis. Part 1: acetyl-CoA, acetogenesis and acyl-CoA short-chain synthetases. Front Physiol. 2020; 11:580167.
Article
69. Belew GD, Silva J, Rito J, Tavares L, Viegas I, Teixeira J, et al. Transfer of glucose hydrogens via acetyl-CoA, malonyl-CoA, and NADPH to fatty acids during de novo lipogenesis. J Lipid Res. 2019; 60:2050–6.
Article
70. Girard J, Perdereau D, Foufelle F, Prip-Buus C, Ferre P. Regulation of lipogenic enzyme gene expression by nutrients and hormones. FASEB J. 1994; 8:36–42.
Article
71. Najjar SM, Yang Y, Fernstrom MA, Lee SJ, Deangelis AM, Rjaily GA, et al. Insulin acutely decreases hepatic fatty acid synthase activity. Cell Metab. 2005; 2:43–53.
Article
72. Eissing L, Scherer T, Todter K, Knippschild U, Greve JW, Buurman WA, et al. De novo lipogenesis in human fat and liver is linked to ChREBP-β and metabolic health. Nat Commun. 2013; 4:1528.
Article
73. Beysen C, Ruddy M, Stoch A, Mixson L, Rosko K, Riiff T, et al. Dose-dependent quantitative effects of acute fructose administration on hepatic de novo lipogenesis in healthy humans. Am J Physiol Endocrinol Metab. 2018; 315:E126–32.
Article
74. McDevitt RM, Bott SJ, Harding M, Coward WA, Bluck LJ, Prentice AM. De novo lipogenesis during controlled overfeeding with sucrose or glucose in lean and obese women. Am J Clin Nutr. 2001; 74:737–46.
Article
75. Ter Horst KW, Vatner DF, Zhang D, Cline GW, Ackermans MT, Nederveen AJ, et al. Hepatic insulin resistance is not pathway selective in humans with nonalcoholic fatty liver disease. Diabetes Care. 2021; 44:489–98.
Article
76. Strawford A, Antelo F, Christiansen M, Hellerstein MK. Adipose tissue triglyceride turnover, de novo lipogenesis, and cell proliferation in humans measured with 2H2O. Am J Physiol Endocrinol Metab. 2004; 286:E577–88.
77. Smith GI, Shankaran M, Yoshino M, Schweitzer GG, Chondronikola M, Beals JW, et al. Insulin resistance drives hepatic de novo lipogenesis in nonalcoholic fatty liver disease. J Clin Invest. 2020; 130:1453–60.
Article
78. Silva JCP, Marques C, Martins FO, Viegas I, Tavares L, Macedo MP, et al. Determining contributions of exogenous glucose and fructose to de novo fatty acid and glycerol synthesis in liver and adipose tissue. Metab Eng. 2019; 56:69–76.
Article
79. Diraison F, Pachiaudi C, Beylot M. In vivo measurement of plasma cholesterol and fatty acid synthesis with deuterated water: determination of the average number of deuterium atoms incorporated. Metabolism. 1996; 45:817–21.
Article
80. Duarte JA, Carvalho F, Pearson M, Horton JD, Browning JD, Jones JG, et al. A high-fat diet suppresses de novo lipogenesis and desaturation but not elongation and triglyceride synthesis in mice. J Lipid Res. 2014; 55:2541–53.
Article
81. Donnelly KL, Smith CI, Schwarzenberg SJ, Jessurun J, Boldt MD, Parks EJ. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J Clin Invest. 2005; 115:1343–51.
Article
82. Bergen WG, Mersmann HJ. Comparative aspects of lipid metabolism: impact on contemporary research and use of animal models. J Nutr. 2005; 135:2499–502.
Article
83. Schmidt NH, Svendsen P, Albarran-Juarez J, Moestrup SK, Bentzon JF. High-fructose feeding does not induce steatosis or non-alcoholic fatty liver disease in pigs. Sci Rep. 2021; 11:2807.
Article
84. Lambert JE, Ramos-Roman MA, Browning JD, Parks EJ. Increased de novo lipogenesis is a distinct characteristic of individuals with nonalcoholic fatty liver disease. Gastroenterology. 2014; 146:726–35.
Article
85. Postic C, Girard J. The role of the lipogenic pathway in the development of hepatic steatosis. Diabetes Metab. 2008; 34(6 Pt 2):643–8.
Article
86. Diraison F, Moulin P, Beylot M. Contribution of hepatic de novo lipogenesis and reesterification of plasma non esterified fatty acids to plasma triglyceride synthesis during non-alcoholic fatty liver disease. Diabetes Metab. 2003; 29:478–85.
Article
87. Flannery C, Dufour S, Rabol R, Shulman GI, Petersen KF. Skeletal muscle insulin resistance promotes increased hepatic de novo lipogenesis, hyperlipidemia, and hepatic steatosis in the elderly. Diabetes. 2012; 61:2711–7.
Article
88. Beysen C, Schroeder P, Wu E, Brevard J, Ribadeneira M, Lu W, et al. Inhibition of fatty acid synthase with FT-4101 safely reduces hepatic de novo lipogenesis and steatosis in obese subjects with non-alcoholic fatty liver disease: results from two early-phase randomized trials. Diabetes Obes Metab. 2021; 23:700–10.
89. Mallat A, Teixeira-Clerc F, Deveaux V, Manin S, Lotersztajn S. The endocannabinoid system as a key mediator during liver diseases: new insights and therapeutic openings. Br J Pharmacol. 2011; 163:1432–40.
Article
90. Baldassarre M, Giannone FA, Napoli L, Tovoli A, Ricci CS, Tufoni M, et al. The endocannabinoid system in advanced liver cirrhosis: pathophysiological implication and future perspectives. Liver Int. 2013; 33:1298–308.
Article
91. Gotfried J, Naftali T, Schey R. Role of cannabis and its derivatives in gastrointestinal and hepatic disease. Gastroenterology. 2020; 159:62–80.
Article
92. Silvestri C, Di Marzo V. The endocannabinoid system in energy homeostasis and the etiopathology of metabolic disorders. Cell Metab. 2013; 17:475–90.
Article
93. Batkai S, Osei-Hyiaman D, Pan H, El-Assal O, Rajesh M, Mukhopadhyay P, et al. Cannabinoid-2 receptor mediates protection against hepatic ischemia/reperfusion injury. FASEB J. 2007; 21:1788–800.
Article
94. Auguet T, Berlanga A, Guiu-Jurado E, Terra X, Martinez S, Aguilar C, et al. Endocannabinoid receptors gene expression in morbidly obese women with nonalcoholic fatty liver disease. Biomed Res Int. 2014; 2014:502542.
Article
95. Mendez-Sanchez N, Zamora-Valdes D, Pichardo-Bahena R, Barredo-Prieto B, Ponciano-Rodriguez G, Bermejo-Martinez L, et al. Endocannabinoid receptor CB2 in nonalcoholic fatty liver disease. Liver Int. 2007; 27:215–9.
Article
96. Devane WA, Hanus L, Breuer A, Pertwee RG, Stevenson LA, Griffin G, et al. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science. 1992; 258:1946–9.
Article
97. Mechoulam R, Ben-Shabat S, Hanus L, Ligumsky M, Kaminski NE, Schatz AR, et al. Identification of an endogenous 2-monoglyceride, present in canine gut, that binds to cannabinoid receptors. Biochem Pharmacol. 1995; 50:83–90.
Article
98. Sugiura T, Kondo S, Sukagawa A, Nakane S, Shinoda A, Itoh K, et al. 2-Arachidonoylglycerol: a possible endogenous cannabinoid receptor ligand in brain. Biochem Biophys Res Commun. 1995; 215:89–97.
99. Rahman SM, Uyama T, Hussain Z, Ueda N. Roles of endocannabinoids and endocannabinoid-like molecules in energy homeostasis and metabolic regulation: a nutritional perspective. Annu Rev Nutr. 2021 Jun 11 [Epub]. https://doi.org/10.1146/annurev-nutr-043020-090216.
Article
100. Osei-Hyiaman D, DePetrillo M, Pacher P, Liu J, Radaeva S, Batkai S, et al. Endocannabinoid activation at hepatic CB1 receptors stimulates fatty acid synthesis and contributes to dietinduced obesity. J Clin Invest. 2005; 115:1298–305.
Article
101. Bermudez-Silva FJ, Suarez J, Baixeras E, Cobo N, Bautista D, Cuesta-Munoz AL, et al. Presence of functional cannabinoid receptors in human endocrine pancreas. Diabetologia. 2008; 51:476–87.
Article
102. Juan-Pico P, Fuentes E, Bermudez-Silva FJ, Javier Diaz-Molina F, Ripoll C, Rodriguez de Fonseca F, et al. Cannabinoid receptors regulate Ca(2+) signals and insulin secretion in pancreatic beta-cell. Cell Calcium. 2006; 39:155–62.
103. Silvestri C, Ligresti A, Di Marzo V. Peripheral effects of the endocannabinoid system in energy homeostasis: adipose tissue, liver and skeletal muscle. Rev Endocr Metab Disord. 2011; 12:153–62.
Article
104. Eckardt K, Sell H, Taube A, Koenen M, Platzbecker B, Cramer A, et al. Cannabinoid type 1 receptors in human skeletal muscle cells participate in the negative crosstalk between fat and muscle. Diabetologia. 2009; 52:664–74.
Article
105. Bazwinsky-Wutschke I, Zipprich A, Dehghani F. Endocannabinoid system in hepatic glucose metabolism, fatty liver disease, and cirrhosis. Int J Mol Sci. 2019; 20:2516.
Article
106. Gary-Bobo M, Elachouri G, Gallas JF, Janiak P, Marini P, Ravinet-Trillou C, et al. Rimonabant reduces obesity-associated hepatic steatosis and features of metabolic syndrome in obese Zucker fa/fa rats. Hepatology. 2007; 46:122–9.
Article
107. Chang E, Kim DH, Yang H, Lee DH, Bae SH, Park CY. CB1 receptor blockade ameliorates hepatic fat infiltration and inflammation and increases Nrf2-AMPK pathway in a rat model of severely uncontrolled diabetes. PLoS One. 2018; 13:e0206152.
Article
108. Jorgacevic B, Vucevic D, Duricic I, Sobajic S, Mladenovic D, Veskovic M, et al. The effect of cannabinoid receptor 1 blockade on hepatic free fatty acid profile in mice with nonalcoholic fatty liver disease. Chem Phys Lipids. 2017; 204:85–93.
109. Hussien NI, El-Kerdasy HI, Ibrahim ME. Protective effect of rimonabant, a canabinoid receptor 1 antagonist, on nonalcoholic fatty liver disease in a rat model through modulation of the hepatic expression of activin A and follistatin. Can J Physiol Pharmacol. 2017; 95:1433–41.
Article
110. Jorgacevic B, Vucevic D, Veskovic M, Mladenovic D, Vukicevic D, Vukicevic RJ, et al. The effect of cannabinoid receptor 1 blockade on adipokine and proinflammatory cytokine concentration in adipose and hepatic tissue in mice with nonalcoholic fatty liver disease. Can J Physiol Pharmacol. 2019; 97:120–9.
111. Khan N, Laudermilk L, Ware J, Rosa T, Mathews K, Gay E, et al. Peripherally selective CB1 receptor antagonist improves symptoms of metabolic syndrome in mice. ACS Pharmacol Transl Sci. 2021; 4:757–64.
Article
112. Tam J, Cinar R, Liu J, Godlewski G, Wesley D, Jourdan T, et al. Peripheral cannabinoid-1 receptor inverse agonism reduces obesity by reversing leptin resistance. Cell Metab. 2012; 16:167–79.
Article
113. Cinar R, Godlewski G, Liu J, Tam J, Jourdan T, Mukhopadhyay B, et al. Hepatic cannabinoid-1 receptors mediate diet-induced insulin resistance by increasing de novo synthesis of long-chain ceramides. Hepatology. 2014; 59:143–53.
114. Gruden G, Barutta F, Kunos G, Pacher P. Role of the endocannabinoid system in diabetes and diabetic complications. Br J Pharmacol. 2016; 173:1116–27.
Article
115. Tan S, Liu H, Ke B, Jiang J, Wu B. The peripheral CB1 receptor antagonist JD5037 attenuates liver fibrosis via a CB1 receptor/β-arrestin1/Akt pathway. Br J Pharmacol. 2020; 177:2830–47.
116. Yang Q, Sun S, Liu W, Liu Q, Wang J. Hypoxia training improves hepatic steatosis partly by downregulation of CB1 receptor in obese mice. Biochem Biophys Res Commun. 2020; 525:639–45.
Article
117. Killilea M, Kerr DM, Mallard BM, Roche M, Wheatley AM. Exacerbated LPS/GalN-induced liver injury in the stress-sensitive Wistar Kyoto rat is associated with changes in the endocannabinoid system. Molecules. 2020; 25:3834.
Article
118. Assa-Glazer T, Gorelick J, Sela N, Nyska A, Bernstein N, Madar Z. Cannabis extracts affected metabolic syndrome parameters in mice fed high-fat/cholesterol diet. Cannabis Cannabinoid Res. 2020; 5:202–14.
Article
119. Drori A, Gammal A, Azar S, Hinden L, Hadar R, Wesley D, et al. CB1R regulates soluble leptin receptor levels via CHOP, contributing to hepatic leptin resistance. Elife. 2020; 9:e60771.
Article
120. Ali AM, El-Tawil OS, Al-Mokaddem AK, Abd El-Rahman SS. Promoted inhibition of TLR4/miR-155/NFkB p65 signaling by cannabinoid receptor 2 agonist (AM1241), aborts inflammation and progress of hepatic fibrosis induced by thioacetamide. Chem Biol Interact. 2021; 336:109398.
121. van Eenige R, Ying Z, Tambyrajah L, Pronk AC, Blomberg N, Giera M, et al. Cannabinoid type 1 receptor inverse agonism attenuates dyslipidemia and atherosclerosis in APOE*3-Leiden.CETP mice. J Lipid Res. 2021; 62:100070.
Article
122. Demizieux L, Piscitelli F, Troy-Fioramonti S, Iannotti FA, Borrino S, Gresti J, et al. Early low-fat diet enriched with linolenic acid reduces liver endocannabinoid tone and improves late glycemic control after a high-fat diet challenge in mice. Diabetes. 2016; 65:1824–37.
Article
123. Teixeira-Clerc F, Julien B, Grenard P, Tran Van Nhieu J, Deveaux V, Li L, et al. CB1 cannabinoid receptor antagonism: a new strategy for the treatment of liver fibrosis. Nat Med. 2006; 12:671–6.
Article
124. Irungbam K, Churin Y, Matono T, Weglage J, Ocker M, Glebe D, et al. Cannabinoid receptor 1 knockout alleviates hepatic steatosis by downregulating perilipin 2. Lab Invest. 2020; 100:454–65.
Article
125. Kahn BB, Alquier T, Carling D, Hardie DG. AMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab. 2005; 1:15–25.
Article
126. Li Y, Xu S, Mihaylova MM, Zheng B, Hou X, Jiang B, et al. AMPK phosphorylates and inhibits SREBP activity to attenuate hepatic steatosis and atherosclerosis in diet-induced insulin-resistant mice. Cell Metab. 2011; 13:376–88.
Article
127. Minokoshi Y, Kim YB, Peroni OD, Fryer LG, Muller C, Carling D, et al. Leptin stimulates fatty-acid oxidation by activating AMP-activated protein kinase. Nature. 2002; 415:339–43.
Article
128. Weikel KA, Ruderman NB, Cacicedo JM. Unraveling the actions of AMP-activated protein kinase in metabolic diseases: systemic to molecular insights. Metabolism. 2016; 65:634–45.
Article
129. Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, et al. Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest. 2001; 108:1167–74.
Article
130. Kim SS, Hwang WM, Yang WM, Lee H, Park KS, Dagon Y, et al. Rho-kinase mediates the anorexigenic action of melanocortin by suppressing AMPK. bioRxiv. 2019 Jun 21. https://doi.org/10.1101/677880.
Article
131. Hong C, Tontonoz P. Liver X receptors in lipid metabolism: opportunities for drug discovery. Nat Rev Drug Discov. 2014; 13:433–44.
Article
132. Steffensen KR, Gustafsson JA. Putative metabolic effects of the liver X receptor (LXR). Diabetes. 2004; 53 Suppl 1:S36–42.
Article
133. Bradley MN, Hong C, Chen M, Joseph SB, Wilpitz DC, Wang X, et al. Ligand activation of LXR beta reverses atherosclerosis and cellular cholesterol overload in mice lacking LXR alpha and apoE. J Clin Invest. 2007; 117:2337–46.
134. Joseph SB, Laffitte BA, Patel PH, Watson MA, Matsukuma KE, Walczak R, et al. Direct and indirect mechanisms for regulation of fatty acid synthase gene expression by liver X receptors. J Biol Chem. 2002; 277:11019–25.
Article
135. Chu K, Miyazaki M, Man WC, Ntambi JM. Stearoyl-coenzyme A desaturase 1 deficiency protects against hypertriglyceridemia and increases plasma high-density lipoprotein cholesterol induced by liver X receptor activation. Mol Cell Biol. 2006; 26:6786–98.
Article
136. Beaven SW, Matveyenko A, Wroblewski K, Chao L, Wilpitz D, Hsu TW, et al. Reciprocal regulation of hepatic and adipose lipogenesis by liver X receptors in obesity and insulin resistance. Cell Metab. 2013; 18:106–17.
Article
137. Laffitte BA, Chao LC, Li J, Walczak R, Hummasti S, Joseph SB, et al. Activation of liver X receptor improves glucose tolerance through coordinate regulation of glucose metabolism in liver and adipose tissue. Proc Natl Acad Sci U S A. 2003; 100:5419–24.
Article
138. Chisholm JW, Hong J, Mills SA, Lawn RM. The LXR ligand T0901317 induces severe lipogenesis in the db/db diabetic mouse. J Lipid Res. 2003; 44:2039–48.
Article
Full Text Links
  • DMJ
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