Farnesoid X Receptor Agonism, Acetyl‐Coenzyme A Carboxylase Inhibition, and Back Translation of Clinically Observed Endpoints of De Novo Lipogenesis in a Murine NASH Model

A promising approach for the treatment of nonalcoholic steatohepatitis (NASH) is the inhibition of enhanced hepatic de novo lipogenesis (DNL), which is the synthesis of fatty acids from nonlipid sources. This study assesses three approaches to DNL suppression in a newly developed dietary NASH mouse model: i) dietary intervention (switch from NASH‐inducing diet to normal diet); ii) inhibition of acetyl‐coenzyme A carboxylase (ACC), the enzyme catalyzing the rate‐limiting step in DNL; and iii) activation of farnesoid X receptor (FXR), a major transcriptional regulator of DNL. C57BL/6J mice on a high‐fat diet combined with ad libitum consumption of a fructose–sucrose solution developed several of the liver histologic features seen in human disease, including steatosis, inflammation, and fibrosis, accompanied by elevated fibrosis biomarkers and liver injury enzymes. Obesity and metabolic impairments were associated with increased intestinal permeability and progression to adenoma and hepatocellular carcinoma. All three approaches led to resolution of established NASH with fibrosis in mice; however, some differences were noted, e.g., with respect to the degree of hepatic steatosis attenuation. While ACC inhibition resulted in elevated blood triglycerides and peripheral obesity, FXR activation prevented peripheral obesity in NASH mice. Comparative transcriptome analysis underlined the translatability of the mouse model to human NASH and revealed novel mechanistic insights into differential regulation of lipid, inflammatory, and extracellular matrix pathways by FXR agonism and ACC inhibition. Conclusion: Novel insights are provided on back translation of clinically observed endpoints of DNL inhibition by targeting ACC or FXR, which are promising therapeutic options for the treatment of NASH, in a newly developed diet‐induced NASH mouse model.

[1]  P. McNamara,et al.  Tropifexor‐Mediated Abrogation of Steatohepatitis and Fibrosis Is Associated With the Antioxidative Gene Expression Profile in Rodents , 2019, Hepatology communications.

[2]  C. Sirlin,et al.  Acetyl‐CoA Carboxylase Inhibitor GS‐0976 for 12 Weeks Reduces Hepatic De Novo Lipogenesis and Steatosis in Patients With Nonalcoholic Steatohepatitis , 2018, Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association.

[3]  R. Loomba,et al.  GS-0976 Reduces Hepatic Steatosis and Fibrosis Markers in Patients With Nonalcoholic Fatty Liver Disease. , 2018, Gastroenterology.

[4]  F. Gonzalez,et al.  Intestine farnesoid X receptor agonist and the gut microbiota activate G‐protein bile acid receptor‐1 signaling to improve metabolism , 2018, Hepatology.

[5]  Guihua Liu,et al.  The PI3K/AKT pathway in obesity and type 2 diabetes , 2018, International journal of biological sciences.

[6]  B. Neuschwander‐Tetri,et al.  Mechanisms of NAFLD development and therapeutic strategies , 2018, Nature Medicine.

[7]  Christopher J. N. Kigongo,et al.  Clinical and metabolic effects associated with weight changes and obeticholic acid in non‐alcoholic steatohepatitis , 2018, Alimentary pharmacology & therapeutics.

[8]  A. Sanyal,et al.  Nonalcoholic Steatohepatitis (NASH) and Hepatic Fibrosis: Emerging Therapies. , 2018, Annual review of pharmacology and toxicology.

[9]  P. McNamara,et al.  Discovery of Tropifexor (LJN452), a Highly Potent Non-bile Acid FXR Agonist for the Treatment of Cholestatic Liver Diseases and Nonalcoholic Steatohepatitis (NASH). , 2017, Journal of medicinal chemistry.

[10]  S. Milstein,et al.  Erratum: Acetyl CoA Carboxylase Inhibition Reduces Hepatic Steatosis but Elevates Plasma Triglycerides in Mice and Humans: A Bedside to Bench Investigation (Cell Metabolism (2017) 26 (394–406.e6) (S1550413117304308) (10.1016/j.cmet.2017.07.009)) , 2017 .

[11]  Xuchen Zhang,et al.  Non-alcoholic fatty liver disease: An expanded review , 2017, World journal of hepatology.

[12]  Xinyi Huang,et al.  Acetyl-CoA carboxylase inhibition by ND-630 reduces hepatic steatosis, improves insulin sensitivity, and modulates dyslipidemia in rats , 2016, Proceedings of the National Academy of Sciences.

[13]  Dajiang J. Liu,et al.  Rare variant in scavenger receptor BI raises HDL cholesterol and increases risk of coronary heart disease , 2016, Science.

[14]  Katherine B. LeClair,et al.  Thioesterase superfamily member 1 suppresses cold thermogenesis by limiting the oxidation of lipid droplet-derived fatty acids in brown adipose tissue , 2016, Molecular metabolism.

[15]  M. Trauner,et al.  Nuclear Receptor Modulation for the Treatment of Nonalcoholic Fatty Liver Disease , 2016, Seminars in Liver Disease.

[16]  S. Friedman,et al.  Weight Loss Through Lifestyle Modification Significantly Reduces Features of Nonalcoholic Steatohepatitis. , 2015, Gastroenterology.

[17]  B. Neuschwander‐Tetri,et al.  Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): a multicentre, randomised, placebo-controlled trial , 2015, The Lancet.

[18]  Wei Wei,et al.  Decreased Cezanne expression is associated with the progression and poor prognosis in hepatocellular carcinoma , 2015, Journal of Translational Medicine.

[19]  D. Brenner,et al.  Intestinal FXR agonism promotes adipose tissue browning and reduces obesity and insulin resistance , 2015, Nature Medicine.

[20]  F. Kuipers,et al.  Beyond intestinal soap—bile acids in metabolic control , 2014, Nature Reviews Endocrinology.

[21]  Alberto Mantovani,et al.  Macrophage plasticity and polarization in liver homeostasis and pathology , 2014, Hepatology.

[22]  J. Browning,et al.  Increased de novo lipogenesis is a distinct characteristic of individuals with nonalcoholic fatty liver disease. , 2014, Gastroenterology.

[23]  J. Dyck,et al.  Single phosphorylation sites in Acc1 and Acc2 regulate lipid homeostasis and the insulin–sensitizing effects of metformin , 2013, Nature Medicine.

[24]  S. Bens,et al.  DNA methylation analysis in nonalcoholic fatty liver disease suggests distinct disease-specific and remodeling signatures after bariatric surgery. , 2013, Cell metabolism.

[25]  P. Larsen,et al.  Targeted deletion of thioesterase superfamily member 1 promotes energy expenditure and protects against obesity and insulin resistance , 2012, Proceedings of the National Academy of Sciences.

[26]  B. Knebel,et al.  Liver-Specific Expression of Transcriptionally Active SREBP-1c Is Associated with Fatty Liver and Increased Visceral Fat Mass , 2012, PloS one.

[27]  V. Koteliansky,et al.  The Scap/SREBP pathway is essential for developing diabetic fatty liver and carbohydrate-induced hypertriglyceridemia in animals. , 2012, Cell metabolism.

[28]  Raj Vuppalanchi,et al.  Presence and significance of microvesicular steatosis in nonalcoholic fatty liver disease. , 2011, Journal of hepatology.

[29]  B. Neuschwander‐Tetri Hepatic lipotoxicity and the pathogenesis of nonalcoholic steatohepatitis: The central role of nontriglyceride fatty acid metabolites , 2010, Hepatology.

[30]  G. Shulman,et al.  Reversal of diet-induced hepatic steatosis and hepatic insulin resistance by antisense oligonucleotide inhibitors of acetyl-CoA carboxylases 1 and 2. , 2006, The Journal of clinical investigation.

[31]  J. Jessurun,et al.  Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. , 2005, The Journal of clinical investigation.

[32]  Sander M Houten,et al.  Bile acids lower triglyceride levels via a pathway involving FXR, SHP, and SREBP-1c. , 2004, The Journal of clinical investigation.

[33]  A. Rigotti,et al.  The role of the high-density lipoprotein receptor SR-BI in the lipid metabolism of endocrine and other tissues. , 2003, Endocrine reviews.

[34]  D. Pessayre,et al.  Microvesicular steatosis and steatohepatitis: role of mitochondrial dysfunction and lipid peroxidation. , 1997, Journal of hepatology.