Role of gut microbial metabolites in nonalcoholic fatty liver disease
Ze Hua Zhao
Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
Search for more papers by this authorJonathan King-Lam Lai
Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney and Westmead Hospital, Sydney, New South Wales, Australia
Search for more papers by this authorCorresponding Author
Liang Qiao
Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney and Westmead Hospital, Sydney, New South Wales, Australia
Correspondence
Jian Gao FAN, Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China.
Email: [email protected]
Liang QIAO, Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney and Westmead Hospital, 176 Hawkesbury Road, Westmead, Sydney, NSW 2145, Australia.
Email: [email protected]
Search for more papers by this authorCorresponding Author
Jian Gao Fan
Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
Correspondence
Jian Gao FAN, Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China.
Email: [email protected]
Liang QIAO, Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney and Westmead Hospital, 176 Hawkesbury Road, Westmead, Sydney, NSW 2145, Australia.
Email: [email protected]
Search for more papers by this authorZe Hua Zhao
Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
Search for more papers by this authorJonathan King-Lam Lai
Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney and Westmead Hospital, Sydney, New South Wales, Australia
Search for more papers by this authorCorresponding Author
Liang Qiao
Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney and Westmead Hospital, Sydney, New South Wales, Australia
Correspondence
Jian Gao FAN, Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China.
Email: [email protected]
Liang QIAO, Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney and Westmead Hospital, 176 Hawkesbury Road, Westmead, Sydney, NSW 2145, Australia.
Email: [email protected]
Search for more papers by this authorCorresponding Author
Jian Gao Fan
Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
Correspondence
Jian Gao FAN, Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China.
Email: [email protected]
Liang QIAO, Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney and Westmead Hospital, 176 Hawkesbury Road, Westmead, Sydney, NSW 2145, Australia.
Email: [email protected]
Search for more papers by this authorAbstract
Nonalcoholic fatty liver disease (NAFLD) is a common, multifactorial liver disease that has emerged as a global challenge due to its increasing prevalence and lack of sustainable treatment options. Gut microbiota possess vital functions in fermenting dietary nutrients and synthesizing bioactive molecules. This function is of great importance in maintaining health because these microbial metabolites are essential in regulating energy metabolism, immune response, and other vital physiological processes. Altered gut flora can result in a change in gut microbial metabolites, affecting the onset and progression of multiple diseases. In this review we summarize the metabolites that may have beneficial or harmful effects on the development and progression of NAFLD. This will help us better understand the possible mechanisms underlying the pathogenesis of NAFLD and facilitate the identification of potential therapeutic approaches for NAFLD.
REFERENCES
- 1Hollister EB, Gao C, Versalovic J. Compositional and functional features of the gastrointestinal microbiome and their effects on human health. Gastroenterology. 2014; 146(6): 1449-1458.
- 2Bäckhed F, Manchester JK, Semenkovich CF, Gordon JI. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci U S A. 2007; 104(3): 979-984.
- 3Flint HJ, Scott KP, Duncan SH, Louis P, Forano E. Microbial degradation of complex carbohydrates in the gut. Gut Microbes. 2012; 3(4): 289-306.
- 4Kamada N, Chen GY, Inohara N, Núñez G. Control of pathogens and pathobionts by the gut microbiota. Nat Immunol. 2013; 14(7): 685-690.
- 5Kau AL, Ahern PP, Griffin NW, Goodman AL, Gordon JI. Human nutrition, the gut microbiome and the immune system. Nature. 2011; 474(7351): 327-336.
- 6Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006; 444(7122): 1027-1031.
- 7Yamamoto M, Yamaguchi R, Munakata K, et al. A microarray analysis of gnotobiotic mice indicating that microbial exposure during the neonatal period plays an essential role in immune system development. BMC Genomics. 2012; 13: 335. https://doi.org/10.1186/1471-2164-13-335.
- 8Tilg H, Cani PD, Mayer EA. Gut microbiome and liver diseases. Gut. 2016; 65(12): 2035-2044.
- 9Mouzaki M, Comelli EM, Arendt BM, et al. Intestinal microbiota in patients with nonalcoholic fatty liver disease. Hepatology. 2013; 58(1): 120-127.
- 10Miele L, Valenza V, La Torre G, et al. Increased intestinal permeability and tight junction alterations in nonalcoholic fatty liver disease. Hepatology. 2009; 49(6): 1877-1887.
- 11Kapil S, Duseja A, Sharma BK, et al. Small intestinal bacterial overgrowth and toll-like receptor signaling in patients with non-alcoholic fatty liver disease. J Gastroenterol Hepatol. 2016; 31(1): 213-221.
- 12Sharifnia T, Antoun J, Verriere TG, et al. Hepatic TLR4 signaling in obese NAFLD. Am J Physiol Gastrointest Liver Physiol. 2015; 309(4): G270-G278.
- 13Dumas ME, Barton RH, Toye A, et al. Metabolic profiling reveals a contribution of gut microbiota to fatty liver phenotype in insulin-resistant mice. Proc Natl Acad Sci U S A. 2006; 103(33): 12511-12516.
- 14Koeth RA, Lam-Galvez BR, Kirsop J, et al. l-Carnitine in omnivorous diets induces an atherogenic gut microbial pathway in humans. J Clin Invest. 2019; 129(1): 373-387.
- 15Rahman K, Desai C, Iyer SS, et al. Loss of junctional adhesion molecule A promotes severe steatohepatitis in mice on a diet high in saturated fat, fructose, and cholesterol. Gastroenterology. 2016; 151(4): 733-746.e12.
- 16Sellmann C, Priebs J, Landmann M, et al. Diets rich in fructose, fat or fructose and fat alter intestinal barrier function and lead to the development of nonalcoholic fatty liver disease over time. J Nutr Biochem. 2015; 26(11): 1183-1192.
- 17Thuy S, Ladurner R, Volynets V, et al. Nonalcoholic fatty liver disease in humans is associated with increased plasma endotoxin and plasminogen activator inhibitor 1 concentrations and with fructose intake. J Nutr. 2008; 138(8): 1452-1455.
- 18Pang J, Xu W, Zhang X, et al. Significant positive association of endotoxemia with histological severity in 237 patients with non-alcoholic fatty liver disease. Aliment Pharmacol Ther. 2017; 46(2): 175-182.
- 19Seki E, Tsutsui H, Nakano H, et al. Lipopolysaccharide-induced IL-18 secretion from murine Kupffer cells independently of myeloid differentiation factor 88 that is critically involved in induction of production of IL-12 and IL-1β. J Immunol. 2001; 166(4): 2651-2657.
- 20Seki E, De Minicis S, Osterreicher CH, et al. TLR4 enhances TGF-β signaling and hepatic fibrosis. Nat Med. 2007; 13(11): 1324-1332.
- 21Cummings JH, Pomare EW, Branch WJ, Naylor CP, Macfarlane GT. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut. 1987; 28(10): 1221-1227.
- 22de la Cuesta-Zuluaga J, Mueller NT, Álvarez-Quintero R, et al. Higher fecal short-chain fatty acid levels are associated with gut microbiome dysbiosis, obesity, hypertension and cardiometabolic disease risk factors. Nutrients. 2018; 11(1):51. https://doi.org/10.3390/nu11010051.
- 23Zhao L, Zhang F, Ding X, et al. Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes. Science. 2018; 359(6380): 1151-1156.
- 24Frost G, Sleeth ML, Sahuri-Arisoylu M, et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat Commun. 2014; 5: 3611. https://doi.org/10.1038/ncomms4611.
- 25De Vadder F, Kovatcheva-Datchary P, Goncalves D, et al. Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell. 2014; 156(1-2): 84-96.
- 26Cordomí A, Fourmy D, Tikhonova IG. Gut hormone GPCRs: structure, function, drug discovery. Curr Opin Pharmacol. 2016; 31: 63-67. https://doi.org/10.1016/j.coph.2016.09.001.
- 27Theodorakis MJ, Carlson O, Michopoulos S, et al. Human duodenal enteroendocrine cells: source of both incretin peptides, GLP-1 and GIP. Am J Physiol Endocrinol Metab. 2006; 290(3): E550-E559.
- 28Karaki S, Mitsui R, Hayashi H, et al. Short-chain fatty acid receptor, GPR43, is expressed by enteroendocrine cells and mucosal mast cells in rat intestine. Cell Tissue Res. 2006; 324(3): 353-360.
- 29Tolhurst G, Heffron H, Lam YS, et al. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein–coupled receptor FFAR2. Diabetes. 2012; 61(2): 364-371.
- 30Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annu Rev Immunol. 2011; 29: 415-445.
- 31Ilan Y. Review article: novel methods for the treatment of non-alcoholic steatohepatitis – targeting the gut immune system to decrease the systemic inflammatory response without immune suppression. Aliment Pharmacol Ther. 2016; 44(11-12): 1168-1182.
- 32Brenner C, Galluzzi L, Kepp O, Kroemer G. Decoding cell death signals in liver inflammation. J Hepatol. 2013; 59(3): 583-594.
- 33Tedelind S, Westberg F, Kjerrulf M, Vidal A. Anti-inflammatory properties of the short-chain fatty acids acetate and propionate: a study with relevance to inflammatory bowel disease. World J Gastroenterol. 2007; 13(20): 2826-2832.
- 34Liu T, Li J, Liu Y, et al. Short-chain fatty acids suppress lipopolysaccharide-induced production of nitric oxide and proinflammatory cytokines through inhibition of NF-κB pathway in RAW264.7 cells. Inflammation. 2012; 35(5): 1676-1684.
- 35Lynch JR, Wang JY. G protein-coupled receptor signaling in stem cells and cancer. Int J Mol Sci. 2016; 17(5): 707. https://doi.org/10.3390/ijms17050707.
- 36Arslan N. Obesity, fatty liver disease and intestinal microbiota. World J Gastroenterol. 2014; 20(44): 16452-16463.
- 37Tan J, McKenzie C, Potamitis M, Thorburn AN, Mackay CR, Macia L. The role of short-chain fatty acids in health and disease. Adv Immunol. 2014; 121: 91-119.
- 38Xiong Y, Miyamoto N, Shibata K, et al. Short-chain fatty acids stimulate leptin production in adipocytes through the G protein-coupled receptor GPR41. Proc Natl Acad Sci U S A. 2004; 101(4): 1045-1050.
- 39Singh N, Gurav A, Sivaprakasam S, et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity. 2014; 40(1): 128-139.
- 40Macia L, Tan J, Vieira AT, et al. Metabolite-sensing receptors GPR43 and GPR109A facilitate dietary fibre-induced gut homeostasis through regulation of the inflammasome. Nat Commun. 2015; 6: 6734. https://doi.org/10.1038/ncomms7734.
- 41Kendrick SF, O'Boyle G, Mann J, et al. Acetate, the key modulator of inflammatory responses in acute alcoholic hepatitis. Hepatology. 2010; 51(6): 1988-1997.
- 42Usami M, Kishimoto K, Ohata A, et al. Butyrate and trichostatin A attenuate nuclear factor kappaB activation and tumor necrosis factor alpha secretion and increase prostaglandin E2 secretion in human peripheral blood mononuclear cells. Nutr Res. 2008; 28(5): 321-328.
- 43Hayden MS, West AP, Ghosh S. NF-κB and the immune response. Oncogene. 2006; 25(51): 6758-6780.
- 44Vinolo MA, Rodrigues HG, Hatanaka E, Sato FT, Sampaio SC, Curi R. Suppressive effect of short-chain fatty acids on production of proinflammatory mediators by neutrophils. J Nutr Biochem. 2011; 22(9): 849-855.
- 45Schaap FG, Trauner M, Jansen PL. Bile acid receptors as targets for drug development. Nat Rev Gastroenterol Hepatol. 2014; 11(1): 55-67.
- 46Lake AD, Novak P, Shipkova P, et al. Decreased hepatotoxic bile acid composition and altered synthesis in progressive human nonalcoholic fatty liver disease. Toxicol Appl Pharmacol. 2013; 268(2): 132-140.
- 47Jiao N, Baker SS, Chapa-Rodriguez A, et al. Suppressed hepatic bile acid signalling despite elevated production of primary and secondary bile acids in NAFLD. Gut. 2018; 67(10): 1881-1891.
- 48Caussy C, Hsu C, Singh S, et al. Serum bile acid patterns are associated with the presence of NAFLD in twins, and dose-dependent changes with increase in fibrosis stage in patients with biopsy-proven NAFLD. Aliment Pharmacol Ther. 2019; 49(2): 183-193.
- 49He X, Ji G, Jia W, Li H. Gut microbiota and nonalcoholic fatty liver disease: insights on mechanism and application of metabolomics. Int J Mol Sci. 2016; 17(3): 300. https://doi.org/10.3390/ijms17030300.
- 50Jiao Y, Lu Y, Li XY. Farnesoid X receptor: a master regulator of hepatic triglyceride and glucose homeostasis. Acta Pharmacol Sin. 2015; 36(1): 44-50.
- 51Schmitt J, Kong B, Stieger B, et al. Protective effects of farnesoid X receptor (FXR) on hepatic lipid accumulation are mediated by hepatic FXR and independent of intestinal FGF15 signal. Liver Int. 2015; 35(4): 1133-1144.
- 52Jiang C, Xie C, Li F, et al. Intestinal farnesoid X receptor signaling promotes nonalcoholic fatty liver disease. J Clin Invest. 2015; 125(1): 386-402.
- 53Neuschwander-Tetri BA, Loomba R, Sanyal AJ, et al. Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): a multicentre, randomised, placebo-controlled trial. Lancet. 2015; 385(9972): 956-965.
- 54Kawamata Y, Fujii R, Hosoya M, et al. A G protein-coupled receptor responsive to bile acids. J Biol Chem. 2003; 278(11): 9435-9440.
- 55Stepanov V, Stankov K, Mikov M. The bile acid membrane receptor TGR5: a novel pharmacological target in metabolic, inflammatory and neoplastic disorders. J Recept Signal Transduct Res. 2013; 33(4): 213-223.
- 56Kumar DP, Rajagopal S, Mahavadi S, et al. Activation of transmembrane bile acid receptor TGR5 stimulates insulin secretion in pancreatic β cells. Biochem Biophys Res Commun. 2012; 427(3): 600-605.
- 57Wang YD, Chen WD, Yu D, Forman BM, Huang W. The G-protein-coupled bile acid receptor, Gpbar1 (TGR5), negatively regulates hepatic inflammatory response through antagonizing nuclear factor κ light-chain enhancer of activated B cells (NF-κB) in mice. Hepatology. 2011; 54(4): 1421-1432.
- 58Nagahashi M, Takabe K, Liu R, et al. Conjugated bile acid-activated S1P receptor 2 is a key regulator of sphingosine kinase 2 and hepatic gene expression. Hepatology. 2015; 61(4): 1216-1226.
- 59Cao R, Cronk ZX, Zha W, et al. Bile acids regulate hepatic gluconeogenic genes and farnesoid X receptor via Gαi-protein-coupled receptors and the AKT pathway. J Lipid Res. 2010; 51(8): 2234-2244.
- 60Studer E, Zhou X, Zhao R, et al. Conjugated bile acids activate the sphingosine-1-phosphate receptor 2 in primary rodent hepatocytes. Hepatology. 2012; 55(1): 267-276.
- 61Velasquez MT, Ramezani A, Manal A, Raj DS. Trimethylamine N-oxide: the good, the bad and the unknown. Toxins (Basel). 2016; 8(11): 326. https://doi.org/10.3390/toxins8110326.
- 62Wang Z, Klipfell E, Bennett BJ, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011; 472(7341): 57-63.
- 63Zhu W, Gregory JC, Org E, et al. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell. 2016; 165(1): 111-124.
- 64Seldin MM, Meng Y, Qi H, et al. Trimethylamine N-oxide promotes vascular inflammation through signaling of mitogen-activated protein kinase and nuclear factor-κB. J Am Heart Assoc. 2016; 5(2): e002767. https://doi.org/10.1161/JAHA.115.002767.
- 65Chen YM, Liu Y, Zhou RF, et al. Associations of gut-flora-dependent metabolite trimethylamine-N-oxide, betaine and choline with non-alcoholic fatty liver disease in adults. Sci Rep. 2016; 6: 19076. https://doi.org/10.1038/srep19076.
- 66Romano KA, Vivas EI, Amador-Noguez D, Rey FE. Intestinal microbiota composition modulates choline bioavailability from diet and accumulation of the proatherogenic metabolite trimethylamine-N-oxide. MBio. 2015; 6(2): e02481. https://doi.org/10.1128/mBio.02481-14.
- 67Gao X, Liu X, Xu J, Xue C, Xue Y, Wang Y. Dietary trimethylamine N-oxide exacerbates impaired glucose tolerance in mice fed a high fat diet. J Biosci Bioeng. 2014; 118(4): 476-481.
- 68Dumas ME, Rothwell AR, Hoyles L, et al. Microbial-host co-metabolites are prodromal markers predicting phenotypic heterogeneity in behavior, obesity, and impaired glucose tolerance. Cell Rep. 2017; 20(1): 136-148.
- 69Shimada Y, Kinoshita M, Harada K, et al. Commensal bacteria-dependent indole production enhances epithelial barrier function in the colon. PLoS One. 2013; 8(11): e80604. https://doi.org/10.1371/journal.pone.0080604.
- 70Denison MS, Nagy SR. Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu Rev Pharmacol Toxicol. 2003; 43: 309-334.
- 71Zelante T, Iannitti RG, Cunha C, et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity. 2013; 39(2): 372-385.
- 72Chimerel C, Emery E, Summers DK, Keyser U, Gribble FM, Reimann F. Bacterial metabolite indole modulates incretin secretion from intestinal enteroendocrine L cells. Cell Rep. 2014; 9(4): 1202-1208.
- 73Venkatesh M, Mukherjee S, Wang H, et al. Symbiotic bacterial metabolites regulate gastrointestinal barrier function via the xenobiotic sensor PXR and Toll-like receptor 4. Immunity. 2014; 41(2): 296-310.
- 74Chyan YJ, Poeggeler B, Omar RA, et al. Potent neuroprotective properties against the Alzheimer β-amyloid by an endogenous melatonin-related indole structure, indole-3-propionic acid. J Biol Chem. 1999; 274(31): 21937-21942.
- 75Zhang LS, Davies SS. Microbial metabolism of dietary components to bioactive metabolites: opportunities for new therapeutic interventions. Genome Med. 2016; 8(1): 46. https://doi.org/10.1186/s13073-016-0296-x.
- 76Krishnan S, Ding Y, Saedi N, et al. Gut microbiota-derived tryptophan metabolites modulate inflammatory response in hepatocytes and macrophages. Cell Rep. 2018; 23(4): 1099-1111.
- 77Choi HH, Cho YS. Fecal microbiota transplantation: current applications, effectiveness, and future perspectives. Clin Endosc. 2016; 49(3): 257-265.
- 78Suez J, Elinav E. The path towards microbiome-based metabolite treatment. Nat Microbiol. 2017; 2: 17075. https://doi.org/10.1038/nmicrobiol.2017.75.
- 79Wang Z, Roberts AB, Buffa JA, et al. Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell. 2015; 163(7): 1585-1595.
- 80Endo H, Niioka M, Kobayashi N, Tanaka M, Watanabe T. Butyrate-producing probiotics reduce nonalcoholic fatty liver disease progression in rats: new insight into the probiotics for the gut-liver axis. PLoS One. 2013; 8(5): e63388. https://doi.org/10.1371/journal.pone.0063388.
- 81Zhou D, Pan Q, Liu XL, et al. Clostridium butyricum B1 alleviates high-fat diet-induced steatohepatitis in mice via enterohepatic immunoregulation. J Gastroenterol Hepatol. 2017; 32(9): 1640-1648.
- 82Mattace Raso G, Simeoli R, Russo R, et al. Effects of sodium butyrate and its synthetic amide derivative on liver inflammation and glucose tolerance in an animal model of steatosis induced by high fat diet. PLoS One. 2013; 8(7): e68626. https://doi.org/10.1371/journal.pone.0068626.
- 83Zhou D, Chen YW, Zhao ZH, et al. Sodium butyrate reduces high-fat diet-induced non-alcoholic steatohepatitis through upregulation of hepatic GLP-1R expression. Exp Mol Med. 2018; 50(12): 157. https://doi.org/10.1038/s12276-018-0183-1.