Semin Liver Dis 2021; 41(02): 191-205
DOI: 10.1055/s-0041-1723752
Review Article

Gut–Liver Axis in Nonalcoholic Fatty Liver Disease: the Impact of the Metagenome, End Products, and the Epithelial and Vascular Barriers

Antonio Gil-Gómez
1   SeLiver Group at Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, Seville, Spain
2   Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
,
Paola Brescia
3   Humanitas Clinical and Research Center – IRCCS, Rozzano, Milan, Italy
,
Maria Rescigno
3   Humanitas Clinical and Research Center – IRCCS, Rozzano, Milan, Italy
,
Manuel Romero-Gómez
1   SeLiver Group at Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, Seville, Spain
2   Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
4   UCM Digestive Diseases, Virgen del Rocío University Hospital, Seville, Spain
› Institutsangaben
Funding This project has been partially funded by the “Asociación Española para el Estudio del Hígado” (Beca de Aprendizaje de Nuevas Tecnologías) and the “Spanish Ministry of Economy, Innovation, and Competition, Instituto de Salud Carlos III” (PI19/00589). The funders have not had any role in the design, writing, or interpretation of this project.

Abstract

Nonalcoholic fatty liver disease (NAFLD) is a systemic, dynamic, heterogeneous, and multiaxis entity, the pathogenesis of which is still uncertain. The gut–liver axis is regulated and stabilized by a complex network encompassing a metabolic, immune, and neuroendocrine cross-talk between the gut, the microbiota, and the liver. Changes in the gut–liver axis affect the metabolism of lipids and carbohydrates in the hepatocytes, and they impact the balance of inflammatory mediators and cause metabolic deregulation, promoting NAFLD and its progression to nonalcoholic steatohepatitis. Moreover, the microbiota and its metabolites can play direct and indirect roles in gut barrier function and fibrosis development. In this review, we will highlight findings from the recent literature focusing on the gut–liver axis and its relation to NAFLD. Finally, we will discuss the impact of technical issues, design bias, and other limitations on current knowledge of the gut microbiota in the context of NAFLD.



Publikationsverlauf

Artikel online veröffentlicht:
08. März 2021

© 2021. Thieme. All rights reserved.

Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

 
  • References

  • 1 Anstee QM, Mantovani A, Tilg H, Targher G. Risk of cardiomyopathy and cardiac arrhythmias in patients with nonalcoholic fatty liver disease. Nat Rev Gastroenterol Hepatol 2018; 15 (07) 425-439
  • 2 Weinstein G, Zelber-Sagi S, Preis SR. et al. Association of nonalcoholic fatty liver disease with lower brain volume in healthy middle-aged adults in the Framingham Study. JAMA Neurol 2018; 75 (01) 97-104
  • 3 VanWagner LB, Rinella ME. Extrahepatic manifestations of nonalcoholic fatty liver disease. Curr Hepatol Rep 2016; 15 (02) 75-85
  • 4 Schuppan D, Surabattula R, Wang XY. Determinants of fibrosis progression and regression in NASH. J Hepatol 2018; 68 (02) 238-250
  • 5 McPherson S, Hardy T, Henderson E, Burt AD, Day CP, Anstee QM. Evidence of NAFLD progression from steatosis to fibrosing-steatohepatitis using paired biopsies: implications for prognosis and clinical management. J Hepatol 2015; 62 (05) 1148-1155
  • 6 Czaja MJ. Function of autophagy in nonalcoholic fatty liver disease. Dig Dis Sci 2016; 61 (05) 1304-1313
  • 7 Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism 2016; 65 (08) 1038-1048
  • 8 Caussy C, Tripathi A, Humphrey G. et al. A gut microbiome signature for cirrhosis due to nonalcoholic fatty liver disease. Nat Commun 2019; 10 (01) 1406
  • 9 Le Roy T, Llopis M, Lepage P. et al. Intestinal microbiota determines development of non-alcoholic fatty liver disease in mice. Gut 2013; 62 (12) 1787-1794
  • 10 Arab JP, Karpen SJ, Dawson PA, Arrese M, Trauner M. Bile acids and nonalcoholic fatty liver disease: molecular insights and therapeutic perspectives. Hepatology 2017; 65 (01) 350-362
  • 11 Cho MS, Kim SY, Suk KT, Kim BY. Modulation of gut microbiome in nonalcoholic fatty liver disease: pro-, pre-, syn-, and antibiotics. J Microbiol 2018; 56 (12) 855-867
  • 12 Stärkel P, Schnabl B. Bidirectional communication between liver and gut during alcoholic liver disease. Semin Liver Dis 2016; 36 (04) 331-339
  • 13 Milosevic I, Vujovic A, Barac A. et al. Gut-liver axis, gut microbiota, and its modulation in the management of liver diseases: a review of the literature. Int J Mol Sci 2019; 20 (02) E395
  • 14 Grabherr F, Grander C, Effenberger M, Adolph TE, Tilg H. Gut dysfunction and non-alcoholic fatty liver disease. Front Endocrinol (Lausanne) 2019; 10: 611
  • 15 Bäckhed F, Ding H, Wang T. et al. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A 2004; 101 (44) 15718-15723
  • 16 Rabot S, Membrez M, Bruneau A. et al. Germ-free C57BL/6J mice are resistant to high-fat-diet-induced insulin resistance and have altered cholesterol metabolism. FASEB J 2010; 24 (12) 4948-4959
  • 17 Zeng H, Liu J, Jackson MI, Zhao F-Q, Yan L, Combs Jr GF. Fatty liver accompanies an increase in lactobacillus species in the hind gut of C57BL/6 mice fed a high-fat diet. J Nutr 2013; 143 (05) 627-631
  • 18 Spencer MD, Hamp TJ, Reid RW, Fischer LM, Zeisel SH, Fodor AA. Association between composition of the human gastrointestinal microbiome and development of fatty liver with choline deficiency. Gastroenterology 2011; 140 (03) 976-986
  • 19 Loomba R, Seguritan V, Li W. et al. Gut microbiome-based metagenomic signature for non-invasive detection of advanced fibrosis in human nonalcoholic fatty liver disease. Cell Metab 2017; 25 (05) 1054.e5-1062.e5
  • 20 Mouries J, Brescia P, Silvestri A. et al. Microbiota-driven gut vascular barrier disruption is a prerequisite for non-alcoholic steatohepatitis development. J Hepatol 2019; 71 (06) 1216-1228
  • 21 Nier A, Engstler AJ, Maier IB, Bergheim I. Markers of intestinal permeability are already altered in early stages of non-alcoholic fatty liver disease: Studies in children. PLoS One 2017; 12 (09) e0183282
  • 22 Kolodziejczyk AA, Zheng D, Shibolet O, Elinav E. The role of the microbiome in NAFLD and NASH. EMBO Mol Med 2019; 11 (02) e9302
  • 23 Høverstad T, Midtvedt T. Short-chain fatty acids in germfree mice and rats. J Nutr 1986; 116 (09) 1772-1776
  • 24 Maslowski KM, Vieira AT, Ng A. et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 2009; 461 (7268): 1282-1286
  • 25 Zhou D, Fan J-G. Microbial metabolites in non-alcoholic fatty liver disease. World J Gastroenterol 2019; 25 (17) 2019-2028
  • 26 Zhao L, Zhang F, Ding X. et al. Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes. Science 2018; 359 (6380): 1151-1156
  • 27 Zhou D, Chen Y-W, Zhao Z-H. 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) 1-12
  • 28 Kimura I, Ozawa K, Inoue D. et al. The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat Commun 2013; 4: 1829
  • 29 Gao Z, Yin J, Zhang J. et al. Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes 2009; 58 (07) 1509-1517
  • 30 den Besten G, van Eunen K, Groen AK, Venema K, Reijngoud D-J, Bakker BM. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res 2013; 54 (09) 2325-2340
  • 31 Qin J, Li Y, Cai Z. et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012; 490 (7418): 55-60
  • 32 Cortez-Pinto H, Jesus L, Barros H, Lopes C, Moura MC, Camilo ME. How different is the dietary pattern in non-alcoholic steatohepatitis patients?. Clin Nutr 2006; 25 (05) 816-823
  • 33 Rocha R, Cotrim HP, Siqueira AC, Floriano S. Non alcoholic fatty liver disease: treatment with soluble fibres [in Portuguese]. Arq Gastroenterol 2007; 44 (04) 350-352
  • 34 Lu Y, Fan C, Li P, Lu Y, Chang X, Qi K. Short chain fatty acids prevent high-fat-diet-induced obesity in mice by regulating G protein-coupled receptors and gut microbiota. Sci Rep 2016; 6: 37589
  • 35 Chambers ES, Viardot A, Psichas A. et al. Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Gut 2015; 64 (11) 1744-1754
  • 36 Schwiertz A, Taras D, Schäfer K. et al. Microbiota and SCFA in lean and overweight healthy subjects. Obesity (Silver Spring) 2010; 18 (01) 190-195
  • 37 Wahlström A, Sayin SI, Marschall H-U, Bäckhed F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab 2016; 24 (01) 41-50
  • 38 Sayin SI, Wahlström A, Felin J. et al. Gut microbiota regulates bile acid metabolism by reducing the levels of tauro-beta-muricholic acid, a naturally occurring FXR antagonist. Cell Metab 2013; 17 (02) 225-235
  • 39 Kakiyama G, Pandak WM, Gillevet PM. et al. Modulation of the fecal bile acid profile by gut microbiota in cirrhosis. J Hepatol 2013; 58 (05) 949-955
  • 40 Ferslew BC, Xie G, Johnston CK. et al. Altered bile acid metabolome in patients with nonalcoholic steatohepatitis. Dig Dis Sci 2015; 60 (11) 3318-3328
  • 41 Long SL, Gahan CGM, Joyce SA. Interactions between gut bacteria and bile in health and disease. Mol Aspects Med 2017; 56: 54-65
  • 42 Ticho AL, Malhotra P, Dudeja PK, Gill RK, Alrefai WA. Bile acid receptors and gastrointestinal functions. Liver Res 2019; 3 (01) 31-39
  • 43 Gonzalez FJ, Jiang C, Xie C, Patterson AD. Intestinal farnesoid X receptor signaling modulates metabolic disease. Dig Dis 2017; 35 (03) 178-184
  • 44 Gadaleta RM, van Erpecum KJ, Oldenburg B. et al. Farnesoid X receptor activation inhibits inflammation and preserves the intestinal barrier in inflammatory bowel disease. Gut 2011; 60 (04) 463-472
  • 45 Parséus A, Sommer N, Sommer F. et al. Microbiota-induced obesity requires farnesoid X receptor. Gut 2017; 66 (03) 429-437
  • 46 Jiang C, Xie C, Li F. et al. Intestinal farnesoid X receptor signaling promotes nonalcoholic fatty liver disease. J Clin Invest 2015; 125 (01) 386-402
  • 47 Li F, Jiang C, Krausz KW. et al. Microbiome remodelling leads to inhibition of intestinal farnesoid X receptor signalling and decreased obesity. Nat Commun 2013; 4: 2384
  • 48 Lund ML, Sorrentino G, Egerod KL. et al. L-Cell differentiation is induced by bile acids through GPBAR1 and paracrine GLP-1 and serotonin signaling. Diabetes 2020; 69 (04) 614-623
  • 49 Alemi F, Poole DP, Chiu J. et al. The receptor TGR5 mediates the prokinetic actions of intestinal bile acids and is required for normal defecation in mice. Gastroenterology 2013; 144 (01) 145-154
  • 50 Broeders EPM, Nascimento EBM, Havekes B. et al. The bile acid chenodeoxycholic acid increases human brown adipose tissue activity. Cell Metab 2015; 22 (03) 418-426
  • 51 Ockenga J, Valentini L, Schuetz T. et al. Plasma bile acids are associated with energy expenditure and thyroid function in humans. J Clin Endocrinol Metab 2012; 97 (02) 535-542
  • 52 Volynets V, Küper MA, Strahl S. et al. Nutrition, intestinal permeability, and blood ethanol levels are altered in patients with nonalcoholic fatty liver disease (NAFLD). Dig Dis Sci 2012; 57 (07) 1932-1941
  • 53 Zhu L, Baker SS, Gill C. et al. Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: a connection between endogenous alcohol and NASH. Hepatology 2013; 57 (02) 601-609
  • 54 Aragonès G, Colom-Pellicer M, Aguilar C. et al. Circulating microbiota-derived metabolites: a “liquid biopsy?. Int J Obes 2019
  • 55 Cope K, Risby T, Diehl AM. Increased gastrointestinal ethanol production in obese mice: implications for fatty liver disease pathogenesis. Gastroenterology 2000; 119 (05) 1340-1347
  • 56 Baker SS, Baker RD, Liu W, Nowak NJ, Zhu L. Role of alcohol metabolism in non-alcoholic steatohepatitis. PLoS One 2010; 5 (03) e9570
  • 57 Zhu R, Baker SS, Moylan CA. et al. Systematic transcriptome analysis reveals elevated expression of alcohol-metabolizing genes in NAFLD livers. J Pathol 2016; 238 (04) 531-542
  • 58 Engstler AJ, Aumiller T, Degen C. et al. Insulin resistance alters hepatic ethanol metabolism: studies in mice and children with non-alcoholic fatty liver disease. Gut 2016; 65 (09) 1564-1571
  • 59 Yuan J, Chen C, Cui J. et al. Fatty liver disease caused by high-alcohol-producing Klebsiella pneumoniae. Cell Metab 2019; 30 (04) 675.e7-688.e7
  • 60 Chaudhry KK, Shukla PK, Mir H. et al. Glutamine supplementation attenuates ethanol-induced disruption of apical junctional complexes in colonic epithelium and ameliorates gut barrier dysfunction and fatty liver in mice. J Nutr Biochem 2016; 27: 16-26
  • 61 Rao R. Acetaldehyde-induced barrier disruption and paracellular permeability in Caco-2 cell monolayer. Methods Mol Biol 2008; 447: 171-183
  • 62 Yan AWE, Fouts DE, Brandl J. et al. Enteric dysbiosis associated with a mouse model of alcoholic liver disease. Hepatology 2011; 53 (01) 96-105
  • 63 McVicker BL, Rasineni K, Tuma DJ, McNiven MA, Casey CA. Lipid droplet accumulation and impaired fat efflux in polarized hepatic cells: consequences of ethanol metabolism. Int J Hepatol 2012; 2012: 978136
  • 64 Lieber CS. Hepatic, metabolic and toxic effects of ethanol: 1991 update. Alcohol Clin Exp Res 1991; 15 (04) 573-592
  • 65 Hoang-Yen Tran D, Hoang-Ngoc Tran D, Mattai SA. et al. Cathelicidin suppresses lipid accumulation and hepatic steatosis by inhibition of the CD36 receptor. Int J Obes 2016; 40 (09) 1424-1434
  • 66 Su D, Nie Y, Zhu A. et al. Vitamin D signaling through induction of paneth cell defensins maintains gut microbiota and improves metabolic disorders and hepatic steatosis in animal models. Front Physiol 2016; 7: 498
  • 67 Corbin KD, Zeisel SH. Choline metabolism provides novel insights into nonalcoholic fatty liver disease and its progression. Curr Opin Gastroenterol 2012; 28 (02) 159-165
  • 68 Sherriff JL, O'Sullivan TA, Properzi C, Oddo J-L, Adams LA. Choline, its potential role in nonalcoholic fatty liver disease, and the case for human and bacterial genes. Adv Nutr 2016; 7 (01) 5-13
  • 69 Shih DM, Wang Z, Lee R. et al. Flavin containing monooxygenase 3 exerts broad effects on glucose and lipid metabolism and atherosclerosis. J Lipid Res 2015; 56 (01) 22-37
  • 70 Tang WH, Hazen SL. Microbiome, trimethylamine N-oxide, and cardiometabolic disease. Transl Res 2017; 179: 108-115
  • 71 Koeth RA, Wang Z, Levison BS. et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med 2013; 19 (05) 576-585
  • 72 Chen 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
  • 73 Barrea L, Annunziata G, Muscogiuri G. et al. Trimethylamine-N-oxide (TMAO) as novel potential biomarker of early predictors of metabolic syndrome. Nutrients 2018; 10 (12) E1971
  • 74 Wang Z, Roberts AB, Buffa JA. et al. Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell 2015; 163 (07) 1585-1595
  • 75 Zimmermann E, Anty R, Tordjman J. et al. C-reactive protein levels in relation to various features of non-alcoholic fatty liver disease among obese patients. J Hepatol 2011; 55 (03) 660-665
  • 76 Pihlajamäki J, Kuulasmaa T, Kaminska D. et al. Serum interleukin 1 receptor antagonist as an independent marker of non-alcoholic steatohepatitis in humans. J Hepatol 2012; 56 (03) 663-670
  • 77 Lefere S, Van de Velde F, Devisscher L. et al. Serum vascular cell adhesion molecule-1 predicts significant liver fibrosis in non-alcoholic fatty liver disease. Int J Obes 2017; 41 (08) 1207-1213
  • 78 Liang H, Lum H, Alvarez A, Garduno-Garcia JJ, Daniel BJ, Musi N. A low dose lipid infusion is sufficient to induce insulin resistance and a pro-inflammatory response in human subjects. PLoS One 2018; 13 (04) e0195810
  • 79 Kubes P, Mehal WZ. Sterile inflammation in the liver. Gastroenterology 2012; 143 (05) 1158-1172
  • 80 Ertunc ME, Hotamisligil GS. Lipid signaling and lipotoxicity in metaflammation: indications for metabolic disease pathogenesis and treatment. J Lipid Res 2016; 57 (12) 2099-2114
  • 81 Hosogai N, Fukuhara A, Oshima K. et al. Adipose tissue hypoxia in obesity and its impact on adipocytokine dysregulation. Diabetes 2007; 56 (04) 901-911
  • 82 Rathinam VAK, Zhao Y, Shao F. Innate immunity to intracellular LPS. Nat Immunol 2019; 20 (05) 527-533
  • 83 Vijayan A, Rumbo M, Carnoy C, Sirard JC. Compartmentalized antimicrobial defenses in response to flagellin. Trends Microbiol 2018; 26 (05) 423-435
  • 84 Wolf AJ, Underhill DM. Peptidoglycan recognition by the innate immune system. Nat Rev Immunol 2018; 18 (04) 243-254
  • 85 Yiu JH, Dorweiler B, Woo CW. Interaction between gut microbiota and toll-like receptor: from immunity to metabolism. J Mol Med (Berl) 2017; 95 (01) 13-20
  • 86 Kazankov K, Jørgensen SMD, Thomsen KL. et al. The role of macrophages in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Nat Rev Gastroenterol Hepatol 2019; 16 (03) 145-159
  • 87 Reilly SM, Saltiel AR. Adapting to obesity with adipose tissue inflammation. Nat Rev Endocrinol 2017; 13 (11) 633-643
  • 88 Camargo A, Jimenez-Lucena R, Alcala-Diaz JF. et al. Postprandial endotoxemia may influence the development of type 2 diabetes mellitus: from the CORDIOPREV study. Clin Nutr 2019; 38 (02) 529-538
  • 89 Pussinen PJ, Havulinna AS, Lehto M, Sundvall J, Salomaa V. Endotoxemia is associated with an increased risk of incident diabetes. Diabetes Care 2011; 34 (02) 392-397
  • 90 Mehta NN, McGillicuddy FC, Anderson PD. et al. Experimental endotoxemia induces adipose inflammation and insulin resistance in humans. Diabetes 2010; 59 (01) 172-181
  • 91 Cani PD, Bibiloni R, Knauf C. et al. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 2008; 57 (06) 1470-1481
  • 92 Amar J, Burcelin R, Ruidavets JB. et al. Energy intake is associated with endotoxemia in apparently healthy men. Am J Clin Nutr 2008; 87 (05) 1219-1223
  • 93 Pang 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 (02) 175-182
  • 94 Pendyala S, Walker JM, Holt PR. A high-fat diet is associated with endotoxemia that originates from the gut. Gastroenterology 2012; 142 (05) 1100-1101.e2
  • 95 Cani PD, Neyrinck AM, Fava F. et al. Selective increases of bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia. Diabetologia 2007; 50 (11) 2374-2383
  • 96 Fabbiano S, Suárez-Zamorano N, Chevalier C. et al. Functional gut microbiota remodeling contributes to the caloric restriction-induced metabolic improvements. Cell Metab 2018; 28 (06) 907.e7-921.e7
  • 97 Vatanen T, Kostic AD, d'Hennezel E. et al; DIABIMMUNE Study Group. Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans. Cell 2016; 165 (04) 842-853
  • 98 Fei N, Zhao L. An opportunistic pathogen isolated from the gut of an obese human causes obesity in germfree mice. ISME J 2013; 7 (04) 880-884
  • 99 Clarke TB, Davis KM, Lysenko ES, Zhou AY, Yu Y, Weiser JN. Recognition of peptidoglycan from the microbiota by Nod1 enhances systemic innate immunity. Nat Med 2010; 16 (02) 228-231
  • 100 Chan KL, Tam TH, Boroumand P. et al. Circulating NOD1 activators and hematopoietic NOD1 contribute to metabolic inflammation and insulin resistance. Cell Rep 2017; 18 (10) 2415-2426
  • 101 Zhu W, Gregory JC, Org E. et al. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell 2016; 165 (01) 111-124
  • 102 Brown JM, Hazen SL. Microbial modulation of cardiovascular disease. Nat Rev Microbiol 2018; 16 (03) 171-181
  • 103 Roager HM, Vogt JK, Kristensen M. et al. Whole grain-rich diet reduces body weight and systemic low-grade inflammation without inducing major changes of the gut microbiome: a randomised cross-over trial. Gut 2019; 68 (01) 83-93
  • 104 Furusawa Y, Obata Y, Fukuda S. et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013; 504 (7480): 446-450
  • 105 Rescigno M. The microbiota revolution: excitement and caution. Eur J Immunol 2017; 47 (09) 1406-1413
  • 106 Bergström JH, Birchenough GM, Katona G. et al. Gram-positive bacteria are held at a distance in the colon mucus by the lectin-like protein ZG16. Proc Natl Acad Sci U S A 2016; 113 (48) 13833-13838
  • 107 Okumura R, Kurakawa T, Nakano T. et al. Lypd8 promotes the segregation of flagellated microbiota and colonic epithelia. Nature 2016; 532 (7597): 117-121
  • 108 Jakobsson HE, Rodríguez-Piñeiro AM, Schütte A. et al. The composition of the gut microbiota shapes the colon mucus barrier. EMBO Rep 2015; 16 (02) 164-177
  • 109 Groschwitz KR, Hogan SP. Intestinal barrier function: molecular regulation and disease pathogenesis. J Allergy Clin Immunol 2009; 124 (01) 3-20 , quiz 21–22
  • 110 Schroeder BO, Birchenough GMH, Ståhlman M. et al. Bifidobacteria or fiber protects against diet-induced microbiota-mediated colonic mucus deterioration. Cell Host Microbe 2018; 23 (01) 27.e7-40.e7
  • 111 Desai MS, Seekatz AM, Koropatkin NM. et al. A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell 2016; 167 (05) 1339.e21-1353.e21
  • 112 Wrzosek L, Miquel S, Noordine ML. et al. Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii influence the production of mucus glycans and the development of goblet cells in the colonic epithelium of a gnotobiotic model rodent. BMC Biol 2013; 11: 61
  • 113 Martínez-López M, Iborra S, Conde-Garrosa R. et al. Microbiota sensing by Mincle-Syk axis in dendritic cells regulates interleukin-17 and -22 production and promotes intestinal barrier integrity. Immunity 2019; 50 (02) 446.e9-461.e9
  • 114 Sonnenberg GF, Monticelli LA, Alenghat T. et al. Innate lymphoid cells promote anatomical containment of lymphoid-resident commensal bacteria. Science 2012; 336 (6086): 1321-1325
  • 115 Stockinger B, Omenetti S. The dichotomous nature of T helper 17 cells. Nat Rev Immunol 2017; 17 (09) 535-544
  • 116 Hartmann P, Seebauer CT, Mazagova M. et al. Deficiency of intestinal mucin-2 protects mice from diet-induced fatty liver disease and obesity. Am J Physiol Gastrointest Liver Physiol 2016; 310 (05) G310-G322
  • 117 Kurashima Y, Kiyono H. Mucosal ecological network of epithelium and immune cells for gut homeostasis and tissue healing. Annu Rev Immunol 2017; 35: 119-147
  • 118 Brown EM, Sadarangani M, Finlay BB. The role of the immune system in governing host-microbe interactions in the intestine. Nat Immunol 2013; 14 (07) 660-667
  • 119 Tilg H, Zmora N, Adolph TE, Elinav E. The intestinal microbiota fuelling metabolic inflammation. Nat Rev Immunol 2020; 20 (01) 40-54
  • 120 Giorgio V, Miele L, Principessa L. et al. Intestinal permeability is increased in children with non-alcoholic fatty liver disease, and correlates with liver disease severity. Dig Liver Dis 2014; 46 (06) 556-560
  • 121 Miele L, Valenza V, La Torre G. et al. Increased intestinal permeability and tight junction alterations in nonalcoholic fatty liver disease. Hepatology 2009; 49 (06) 1877-1887
  • 122 Thaiss CA, Levy M, Grosheva I. et al. Hyperglycemia drives intestinal barrier dysfunction and risk for enteric infection. Science 2018; 359 (6382): 1376-1383
  • 123 Johnson AM, Costanzo A, Gareau MG. et al. High fat diet causes depletion of intestinal eosinophils associated with intestinal permeability. PLoS One 2015; 10 (04) e0122195
  • 124 Genser L, Aguanno D, Soula HA. et al. Increased jejunal permeability in human obesity is revealed by a lipid challenge and is linked to inflammation and type 2 diabetes. J Pathol 2018; 246 (02) 217-230
  • 125 Monteiro-Sepulveda M, Touch S, Mendes-Sá C. et al. Jejunal T cell inflammation in human obesity correlates with decreased enterocyte insulin signaling. Cell Metab 2015; 22 (01) 113-124
  • 126 Garidou L, Pomié C, Klopp P. et al. The gut microbiota regulates intestinal CD4 T cells expressing RORγt and controls metabolic disease. Cell Metab 2015; 22 (01) 100-112
  • 127 Ghoshal S, Witta J, Zhong J, de Villiers W, Eckhardt E. Chylomicrons promote intestinal absorption of lipopolysaccharides. J Lipid Res 2009; 50 (01) 90-97
  • 128 Laugerette F, Vors C, Géloën A. et al. Emulsified lipids increase endotoxemia: possible role in early postprandial low-grade inflammation. J Nutr Biochem 2011; 22 (01) 53-59
  • 129 Guo S, Al-Sadi R, Said HM, Ma TY. Lipopolysaccharide causes an increase in intestinal tight junction permeability in vitro and in vivo by inducing enterocyte membrane expression and localization of TLR-4 and CD14. Am J Pathol 2013; 182 (02) 375-387
  • 130 Rahman 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 (04) 733.e12-746.e12
  • 131 Zhao Z-H, Xin F-Z, Xue Y. et al. Indole-3-propionic acid inhibits gut dysbiosis and endotoxin leakage to attenuate steatohepatitis in rats. Exp Mol Med 2019; 51 (09) 1-14
  • 132 Spadoni I, Zagato E, Bertocchi A. et al. A gut-vascular barrier controls the systemic dissemination of bacteria. Science 2015; 350 (6262): 830-834
  • 133 Spadoni I, Pietrelli A, Pesole G, Rescigno M. Gene expression profile of endothelial cells during perturbation of the gut vascular barrier. Gut Microbes 2016; 7 (06) 540-548
  • 134 Spadoni I, Fornasa G, Rescigno M. Organ-specific protection mediated by cooperation between vascular and epithelial barriers. Nat Rev Immunol 2017; 17 (12) 761-773
  • 135 Cheng C, Tan J, Qian W, Zhang L, Hou X. Gut inflammation exacerbates hepatic injury in the high-fat diet induced NAFLD mouse: attention to the gut-vascular barrier dysfunction. Life Sci 2018; 209: 157-166
  • 136 Sorribas M, Jakob MO, Yilmaz B. et al. FXR modulates the gut-vascular barrier by regulating the entry sites for bacterial translocation in experimental cirrhosis. J Hepatol 2019; 71 (06) 1126-1140
  • 137 Angulo P, Kleiner DE, Dam-Larsen S. et al. Liver fibrosis, but no other histologic features, is associated with long-term outcomes of patients with nonalcoholic fatty liver disease. Gastroenterology 2015; 149 (02) 389.e10-397.e10
  • 138 Gangarapu V, Ince AT, Baysal B. et al. Efficacy of rifaximin on circulating endotoxins and cytokines in patients with nonalcoholic fatty liver disease. Eur J Gastroenterol Hepatol 2015; 27 (07) 840-845
  • 139 Boursier J, Mueller O, Barret M. et al. The severity of nonalcoholic fatty liver disease is associated with gut dysbiosis and shift in the metabolic function of the gut microbiota. Hepatology 2016; 63 (03) 764-775
  • 140 Tripathi A, Debelius J, Brenner DA. et al. The gut-liver axis and the intersection with the microbiome. Nat Rev Gastroenterol Hepatol 2018; 15 (07) 397-411
  • 141 Douhara A, Moriya K, Yoshiji H. et al. Reduction of endotoxin attenuates liver fibrosis through suppression of hepatic stellate cell activation and remission of intestinal permeability in a rat non-alcoholic steatohepatitis model. Mol Med Rep 2015; 11 (03) 1693-1700
  • 142 Abdelmalek MF, Suzuki A, Guy C. et al; Nonalcoholic Steatohepatitis Clinical Research Network. Increased fructose consumption is associated with fibrosis severity in patients with nonalcoholic fatty liver disease. Hepatology 2010; 51 (06) 1961-1971
  • 143 Jang C, Hui S, Lu W. et al. The small intestine converts dietary fructose into glucose and organic acids. Cell Metab 2018; 27 (02) 351.e3-361.e3
  • 144 Csak T, Ganz M, Pespisa J, Kodys K, Dolganiuc A, Szabo G. Fatty acid and endotoxin activate inflammasomes in mouse hepatocytes that release danger signals to stimulate immune cells. Hepatology 2011; 54 (01) 133-144
  • 145 Seki E, Schnabl B. Role of innate immunity and the microbiota in liver fibrosis: crosstalk between the liver and gut. J Physiol 2012; 590 (03) 447-458
  • 146 Wree A, McGeough MD, Peña CA. et al. NLRP3 inflammasome activation is required for fibrosis development in NAFLD. J Mol Med (Berl) 2014; 92 (10) 1069-1082
  • 147 Fouts DE, Torralba M, Nelson KE, Brenner DA, Schnabl B. Bacterial translocation and changes in the intestinal microbiome in mouse models of liver disease. J Hepatol 2012; 56 (06) 1283-1292
  • 148 Seki E, De Minicis S, Osterreicher CH. et al. TLR4 enhances TGF-beta signaling and hepatic fibrosis. Nat Med 2007; 13 (11) 1324-1332
  • 149 Seki E, De Minicis S, Gwak GY. et al. CCR1 and CCR5 promote hepatic fibrosis in mice. J Clin Invest 2009; 119 (07) 1858-1870
  • 150 Luo X, Li H, Ma L. et al. Expression of STING is increased in liver tissues from patients with NAFLD and promotes macrophage-mediated hepatic inflammation and fibrosis in mice. Gastroenterology 2018; 155 (06) 1971.e4-1984.e4
  • 151 Seki E, Schwabe RF. Hepatic inflammation and fibrosis: functional links and key pathways. Hepatology 2015; 61 (03) 1066-1079
  • 152 Zong H, Armoni M, Harel C, Karnieli E, Pessin JE. Cytochrome P-450 CYP2E1 knockout mice are protected against high-fat diet-induced obesity and insulin resistance. Am J Physiol Endocrinol Metab 2012; 302 (05) E532-E539
  • 153 Sinha R, Abnet CC, White O, Knight R, Huttenhower C. The microbiome quality control project: baseline study design and future directions. Genome Biol 2015; 16: 276
  • 154 Cui J, Chen C-H, Lo M-T. et al; For The Genetics Of Nafld In Twins Consortium. Shared genetic effects between hepatic steatosis and fibrosis: a prospective twin study. Hepatology 2016; 64 (05) 1547-1558
  • 155 Goodrich JK, Davenport ER, Beaumont M. et al. Genetic determinants of the gut microbiome in UK twins. Cell Host Microbe 2016; 19 (05) 731-743
  • 156 Hartmann P, Chu H, Duan Y, Schnabl B. Gut microbiota in liver disease: too much is harmful, nothing at all is not helpful either. Am J Physiol Gastrointest Liver Physiol 2019; 316 (05) G563-G573
  • 157 Oh J-H, Schueler KL, Stapleton DS. et al. Secretion of recombinant interleukin-22 by engineered Lactobacillus reuteri reduces fatty liver disease in a mouse model of diet-induced obesity. MSphere 2020; 5 (03) e00183-e20
  • 158 Duan Y, Llorente C, Lang S. et al. Bacteriophage targeting of gut bacterium attenuates alcoholic liver disease. Nature 2019; 575 (7783): 505-511
  • 159 Honda T, Ishigami M, Luo F. et al. Branched-chain amino acids alleviate hepatic steatosis and liver injury in choline-deficient high-fat diet induced NASH mice. Metabolism 2017; 69: 177-187
  • 160 Kaikkonen JE, Würtz P, Suomela E. et al. Metabolic profiling of fatty liver in young and middle-aged adults: cross-sectional and prospective analyses of the Young Finns Study. Hepatology 2017; 65 (02) 491-500
  • 161 Krishnan S, Ding Y, Saedi N. et al. Gut microbiota-derived tryptophan metabolites modulate inflammatory response in hepatocytes and macrophages. Cell Rep 2018; 23 (04) 1099-1111
  • 162 Natividad JM, Agus A, Planchais J. et al. Impaired aryl hydrocarbon receptor ligand production by the gut microbiota is a key factor in metabolic syndrome. Cell Metab 2018; 28 (05) 737.e4-749.e4
  • 163 El Hadi H, Vettor R, Rossato M. Vitamin E as a treatment for nonalcoholic fatty liver disease: reality or myth?. Antioxidants 2018; 7 (01) 12
  • 164 Christensen K, Lawler T, Mares J. Dietary carotenoids and non-alcoholic fatty liver disease among US adults, NHANES 2003–2014. Nutrients 2019; 11 (05) 1101
  • 165 Anhê FF, Nachbar RT, Varin TV. et al. A polyphenol-rich cranberry extract reverses insulin resistance and hepatic steatosis independently of body weight loss. Mol Metab 2017; 6 (12) 1563-1573
  • 166 Kalantar-Zadeh K, Berean KJ, Burgell RE, Muir JG, Gibson PR. Intestinal gases: influence on gut disorders and the role of dietary manipulations. Nat Rev Gastroenterol Hepatol 2019; 16 (12) 733-747
  • 167 Strandwitz P. Neurotransmitter modulation by the gut microbiota. Brain Res 2018; 1693 (Pt B): 128-133
  • 168 Michail S, Lin M, Frey MR. et al. Altered gut microbial energy and metabolism in children with non-alcoholic fatty liver disease. FEMS Microbiol Ecol 2015; 91 (02) 1-9
  • 169 Raman M, Ahmed I, Gillevet PM. et al. Fecal microbiome and volatile organic compound metabolome in obese humans with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2013; 11 (07) 868-75.e1 , 3
  • 170 Wong VW-S, Tse C-H, Lam TT-Y. et al. Molecular characterization of the fecal microbiota in patients with nonalcoholic steatohepatitis--a longitudinal study. PLoS One 2013; 8 (04) e62885
  • 171 Hoyles L, Fernández-Real JM, Federici M. et al. Molecular phenomics and metagenomics of hepatic steatosis in non-diabetic obese women. Nat Med 2018; 24 (07) 1070-1080
  • 172 Shen F, Zheng R-D, Sun X-Q, Ding W-J, Wang X-Y, Fan J-G. Gut microbiota dysbiosis in patients with non-alcoholic fatty liver disease. Hepatobiliary Pancreat Dis Int 2017; 16 (04) 375-381
  • 173 Del Chierico F, Nobili V, Vernocchi P. et al. Gut microbiota profiling of pediatric nonalcoholic fatty liver disease and obese patients unveiled by an integrated meta-omics-based approach. Hepatology 2017; 65 (02) 451-464
  • 174 Mouzaki M, Comelli EM, Arendt BM. et al. Intestinal microbiota in patients with nonalcoholic fatty liver disease. Hepatology 2013; 58 (01) 120-127
  • 175 Bajaj JS, Heuman DM, Hylemon PB. et al. Altered profile of human gut microbiome is associated with cirrhosis and its complications. J Hepatol 2014; 60 (05) 940-947
  • 176 Wang B, Jiang X, Cao M. et al. Altered fecal microbiota correlates with liver biochemistry in nonobese patients with non-alcoholic fatty liver disease. Sci Rep 2016; 6 (01) 32002
  • 177 Li F, Sun G, Wang Z. et al. Characteristics of fecal microbiota in non-alcoholic fatty liver disease patients. Sci China Life Sci 2018; 61 (07) 770-778
  • 178 Jiang W, Wu N, Wang X. et al. Dysbiosis gut microbiota associated with inflammation and impaired mucosal immune function in intestine of humans with non-alcoholic fatty liver disease. Sci Rep 2015; 5: 8096
  • 179 Da Silva HE, Teterina A, Comelli EM. et al. Nonalcoholic fatty liver disease is associated with dysbiosis independent of body mass index and insulin resistance. Sci Rep 2018; 8 (01) 1466
  • 180 Demir M, Lang S, Martin A. et al. Phenotyping non-alcoholic fatty liver disease by the gut microbiota: ready for prime time?. J Gastroenterol Hepatol 2020