Total fecal microbiota transplantation alleviates high-fat diet-induced steatohepatitis in mice via beneficial regulation of gut microbiota

Da Zhou, Qin Pan, Feng Shen, Hai-Xia Cao, Wen-Jin Ding, Yuan-Wen Chen, Jian-Gao Fan, Da Zhou, Qin Pan, Feng Shen, Hai-Xia Cao, Wen-Jin Ding, Yuan-Wen Chen, Jian-Gao Fan

Abstract

Non-alcoholic steatohepatitis (NASH) is an epidemic metabolic disease with limited therapeutic strategies. Cumulative data support the pivotal role of gut microbiota in NASH. Here, we investigated the hypothesis regarding whether fecal microbiota transplantation (FMT) is effective in attenuating high-fat diet (HFD)-induced steatohepatitis in mice. Mice were randomized into control, HFD and HFD + FMT groups. After an 8-week HFD, FMT treatment was initiated and carried out for 8 weeks. The gut microbiota structure, butyrate concentrations of the cecal content, liver pathology and intrahepatic lipid and cytokines were examined. Our results showed that after FMT, the gut microbiota disturbance was corrected in HFD-fed mice with elevated abundances of the beneficial bacteria Christensenellaceae and Lactobacillus. FMT also increased butyrate concentrations of the cecal content and the intestinal tight junction protein ZO-1, resulting in relief of endotoxima in HFD-fed mice. Steatohepatitis was alleviated after FMT, as indicated by a significant decrease in intrahepatic lipid accumulation (reduced Oli-red staining, decreased intrahepatic triglyceride and cholesterol), intrahepatic pro-inflammatory cytokines, and the NAS score. Accordingly, intrahepatic IFN-γ and IL-17 were decreased, but Foxp3, IL-4 and IL-22 were increased after FMT intervention. These data indicate that FMT attenuated HFD-induced steatohepatitis in mice via a beneficial effect on the gut microbiota.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
FMT attenuated HFD-induced obesity, liver injury and metabolic disturbance. (A,B) Body weight changes and body weight at 16th week. (C) Liver index = liver weight/body weight × 100. (D) Epididymal fat index = epididymal fat weight/body weight × 100. (E) Energy intake per mouse per week. (F,I) Fasting serum glucose, fasting serum insulin, HOMA-IR and ISI of the three groups. J,K) Serum ALT and AST. The data represent the mean ± S.E.M. (n = 12 mice per group), *P < 0.05, **P < 0.01 and ***P < 0.001.
Figure 2
Figure 2
Influences of FMT on gut microbiota. (A,B) Scatter plot of the unweighted-unifrac-principal co-ordinates analysis (PCOA) score showing the similarity of the 18 bacterial communities based on the Unifrac distance and Scatter plot of the weighted-PCOA score. (C) Nonmetric multidimensional scaling (NMDS) showing the difference in bacterial communities according to the Bray-Curtis distance. (D) Hierarchical cluster analysis. (E,F) Bacterial composition of the different communities at the genus level (E) and at phylum level (F). Sequences that could not be classified into any known group were assigned as “no rank.” (G,H) Differences were represented in the color of the most abundant group. Key phylotypes of the gut microbiota responding to FMT treatment (G), the histogram (H) showing the lineages with LDA values as determined by LEfSe. (I,K) The acetic acid, propanoic acid and butyrate acid levels of the cecal content. The data represent the mean ± S.E.M. (n = 12 mice per group), ***P < 0.001.
Figure 3
Figure 3
Beneficial effects of FMT on the small intestine and the level of serum endotoxin. (A) HE staining of the small intestine showing that FMT restored HFD-induced mucosal damage. (B) Immunohistochemistry for ZO-1 showing that FMT increased ZO-1 expression. (C) ZO-1 mRNA expression in the small intestine. (D) The level of serum endotoxin. The data represent the mean ± S.E.M. (n = 12 mice per group), *P < 0.05, **P < 0.01 and ***P < 0.001.
Figure 4
Figure 4
FMT improved inflammation and lipid metabolism in liver. (A,B) HE and oil red O staining of liver. (C) NAS score. (D,E) the level of TG and cholesterol in the liver. (F) Lipid metabolism-associated PPAR-α and PPAR-γ gene expression in the liver. (G) Masson’s staining. (H) Endotoxin-associated gene expression of TLR4 and Myd88 and fibrosis-associated gene expression of TGF-β, Smad2, Smad7 and α-SMA. (I,J) IR protein expression level in liver. Gene expression levels are expressed as values relative to the control group. The data represent the mean ± S.E.M. (n = 12 mice per group), *P < 0.05, **P < 0.01 and ***P < 0.001.
Figure 5
Figure 5
Immunoregulation of FMT in the liver. (A) Pro-inflammation-associated gene expression in the liver. (B,C) Foxp3, IFN-γ, IL-4, IL-17 and IL-22 protein expression levels in the liver. (D–I) Immunohistochemistry for Foxp3, IFN-γ, IL-4, IL-17 and IL-22 in the liver showed that Foxp3, IL-4, and IL-22 were increased and IFN-γ and IL-17 were decreased after FMT intervention. The data represent the mean ± S.E.M. *P < 0.05, **P < 0.01 and ***P < 0.001.
Figure 6
Figure 6
FMT improved inflammation and lipid metabolism in epididymal fat tissue. (A) Lipid metabolism-associated PPAR-α and PPAR-γ gene expression in the epididymal fat tissue. (B) Gene expression levels of MCP-1 and TNF-α in the epididymal fat tissue. Gene expression levels are expressed as values relative to the control group. The data represent the mean ± S.E.M. (n = 12 mice per group), *P < 0.05, **P < 0.01 and ***P < 0.001.

References

    1. Rinella ME. Nonalcoholic Fatty Liver Disease. Jama. 2015;313(2263):5370–73.
    1. Gkolfakis P, Dimitriadis G, Triantafyllou K. Gut microbiota and non-alcoholic fatty liver disease. Hepatobiliary & Pancreatic Diseases International. 2015;14:572–581. doi: 10.1016/S1499-3872(15)60026-1.
    1. Marchesi JR, et al. The gut microbiota and host health: a new clinical frontier. Gut. 2016;65:330–339. doi: 10.1136/gutjnl-2015-309990.
    1. Compare D, et al. Gut–liver axis: The impact of gut microbiota on non alcoholic fatty liver disease. Nutrition, Metabolism and Cardiovascular Diseases. 2012;22:471–476. doi: 10.1016/j.numecd.2012.02.007.
    1. Smits LP, Bouter KEC, de Vos WM, Borody TJ, Nieuwdorp M. Therapeutic Potential of Fecal Microbiota Transplantation. Gastroenterology. 2013;145:946–953. doi: 10.1053/j.gastro.2013.08.058.
    1. Gao X, Zhu Y, Wen Y, Liu G, Wan C. Efficacy of probiotics in nonalcoholic fatty liver disease in adult and children: A meta-analysis of randomized controlled trials. Hepatology Research. 2016;46:1226–1233. doi: 10.1111/hepr.12671.
    1. Das SK, Balakrishnan V. Role of Cytokines in the Pathogenesis of Non-Alcoholic Fatty Liver Disease. Indian Journal of Clinical Biochemistry. 2011;26:202–209. doi: 10.1007/s12291-011-0121-7.
    1. Hooper LV, Littman DR, Macpherson AJ. Interactions Between the Microbiota and the Immune System. Science. 2012;336:1268–1273. doi: 10.1126/science.1223490.
    1. Maynard CL, Elson CO, Hatton RD, Weaver CT. Reciprocal interactions of the intestinal microbiota and immune system. Nature. 2012;489:231–241. doi: 10.1038/nature11551.
    1. Wu H-J, Wu E. The role of gut microbiota in immune homeostasis and autoimmunity. Gut Microbes. 2014;3:4–14. doi: 10.4161/gmic.19320.
    1. Suez J, et al. Artificial sweeteners induce glucose intolerance by altering the gut microbiota. Nature. 2014;514:181–186.
    1. Schloss PD, et al. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl Environ Microbiol. 2009;75:7537–7541. doi: 10.1128/AEM.01541-09.
    1. Noval Rivas M, et al. A microbiota signature associated with experimental food allergy promotes allergic sensitization and anaphylaxis. J Allergy Clin Immunol. 2013;131:201–212. doi: 10.1016/j.jaci.2012.10.026.
    1. Jiang XT, et al. Illumina sequencing of 16S rRNA tag revealed spatial variations of bacterial communities in a mangrove wetland. Microb Ecol. 2013;66:96–104. doi: 10.1007/s00248-013-0238-8.
    1. Segata N, et al. Metagenomic biomarker discovery and explanation. Genome Biol. 2011;12:R60. doi: 10.1186/gb-2011-12-6-r60.
    1. Lynch SV, Phimister EG, Pedersen O. The Human Intestinal Microbiome in Health and Disease. New England Journal of Medicine. 2016;375:2369–2379. doi: 10.1056/NEJMra1600266.
    1. Brandt LJ, Aroniadis OC. An overview of fecal microbiota transplantation: techniques, indications, and outcomes. Gastrointestinal Endoscopy. 2013;78:240–249. doi: 10.1016/j.gie.2013.03.1329.
    1. Vrieze A, et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology. 2012;143:913–916 e917. doi: 10.1053/j.gastro.2012.06.031.
    1. Li J, et al. Probiotics modulated gut microbiota suppresses hepatocellular carcinoma growth in mice. Proc Natl Acad Sci USA. 2016;113:E1306–1315. doi: 10.1073/pnas.1518189113.
    1. Berni Canani R, et al. Lactobacillus rhamnosus GG-supplemented formula expands butyrate-producing bacterial strains in food allergic infants. ISME J. 2016;10:742–750. doi: 10.1038/ismej.2015.151.
    1. Le Chatelier E, et al. Richness of human gut microbiome correlates with metabolic markers. Nature. 2013;500:541–546. doi: 10.1038/nature12506.
    1. Macfarlane S, Macfarlane GT. Regulation of short-chain fatty acid production. Proceedings of the Nutrition Society. 2007;62:67–72. doi: 10.1079/PNS2002207.
    1. Guilloteau P, et al. From the gut to the peripheral tissues: the multiple effects of butyrate. Nutrition Research Reviews. 2010;23:366–384. doi: 10.1017/S0954422410000247.
    1. Plöger S, et al. Microbial butyrate and its role for barrier function in the gastrointestinal tract. Annals of the New York Academy of Sciences. 2012;1258:52–59. doi: 10.1111/j.1749-6632.2012.06553.x.
    1. Engen, P. A. et al. The Potential Role of Gut-Derived Inflammation in Multiple System Atrophy. Journal of Parkinson’s Disease. 1–16, doi:10.3233/JPD-160991 (2017).
    1. Nova, E., Perez de Heredia, F., Gomez-Martinez, S. & Marcos, A. The Role of Probiotics on the Microbiota: Effect on Obesity. Nutr Clin Pract31, 387–400, doi:10.1177/0884533615620350 (2016).
    1. Mishra AK, Dubey V, Ghosh AR. Obesity: An overview of possible role(s) of gut hormones, lipid sensing and gut microbiota. Metabolism. 2016;65:48–65. doi: 10.1016/j.metabol.2015.10.008.
    1. Cho S-Y, et al. Modulation of gut microbiota and delayed immunosenescence as a result of syringaresinol consumption in middle-aged mice. Scientific Reports. 2016;6:39026. doi: 10.1038/srep39026.
    1. Roberts J, et al. An Exploratory Investigation of Endotoxin Levels in Novice Long Distance Triathletes, and the Effects of a Multi-Strain Probiotic/Prebiotic, Antioxidant Intervention. Nutrients. 2016;8:733. doi: 10.3390/nu8110733.
    1. Choi S-Y, et al. The role of Toll-like receptor 4 in high-glucose-induced inflammatory and fibrosis markers in human peritoneal mesothelial cells. International Urology and Nephrology. 2016;49:171–181. doi: 10.1007/s11255-016-1430-9.
    1. Khan S, Jena G. Sodium butyrate, a HDAC inhibitor ameliorates eNOS, iNOS and TGF-beta1-induced fibrogenesis, apoptosis and DNA damage in the kidney of juvenile diabetic rats. Food Chem Toxicol. 2014;73:127–139. doi: 10.1016/j.fct.2014.08.010.
    1. Bulow R, et al. Antifibrogenic effects of histone deacetylase inhibitors on pancreatic stellate cells. Biochem Pharmacol. 2007;74:1747–1757. doi: 10.1016/j.bcp.2007.08.023.
    1. Stenman, L. K., Burcelin, R. & Lahtinen, S. Establishing a causal link between gut microbes, body weight gain and glucose metabolism in humans – towards treatment with probiotics. Beneficial Microbes, 1–12 (2015).
    1. Goodrich JuliaK, et al. Human Genetics Shape the Gut Microbiome. Cell. 2014;159:789–799. doi: 10.1016/j.cell.2014.09.053.
    1. Jung M-J, et al. Chronic Repression of mTOR Complex 2 Induces Changes in the Gut Microbiota of Diet-induced Obese Mice. Scientific Reports. 2016;6:30887. doi: 10.1038/srep30887.
    1. Wang J, et al. Modulation of gut microbiota during probiotic-mediated attenuation of metabolic syndrome in high fat diet-fed mice. The ISME Journal. 2014;9:1–15. doi: 10.1038/ismej.2014.99.
    1. Sommer F, Bäckhed F. The gut microbiota — masters of host development and physiology. Nature Reviews Microbiology. 2013;11:227–238. doi: 10.1038/nrmicro2974.
    1. Nati M, et al. The role of immune cells in metabolism-related liver inflammation and development of non-alcoholic steatohepatitis (NASH) Rev Endocr Metab Disord. 2016;17:29–39. doi: 10.1007/s11154-016-9339-2.
    1. Park J-S, Lee E-J, Lee J-C, Kim W-K, Kim H-S. Anti-inflammatory effects of short chain fatty acids in IFN-γ-stimulated RAW 264.7 murine macrophage cells: Involvement of NF-κB and ERK signaling pathways. International Immunopharmacology. 2007;7:70–77. doi: 10.1016/j.intimp.2006.08.015.
    1. Smith PM, et al. The Microbial Metabolites, Short-Chain Fatty Acids, Regulate Colonic Treg Cell Homeostasis. Science. 2013;341:569–573. doi: 10.1126/science.1241165.
    1. Jin CJ, Sellmann C, Engstler AJ, Ziegenhardt D, Bergheim I. Supplementation of sodium butyrate protects mice from the development of non-alcoholic steatohepatitis (NASH) British Journal of Nutrition. 2015;114:1745–1755. doi: 10.1017/S0007114515003621.
    1. Olefsky JM, Glass CK. Macrophages, Inflammation, and Insulin Resistance. Annual Review of Physiology. 2010;72:219–246. doi: 10.1146/annurev-physiol-021909-135846.
    1. Kelly D, et al. Commensal anaerobic gut bacteria attenuate inflammation by regulating nuclear-cytoplasmic shuttling of PPAR-gamma and RelA. Nat Immunol. 2004;5:104–112. doi: 10.1038/ni1018.
    1. Liu Y, et al. 3, 3′-Diindolylmethane alleviates steatosis and the progression of NASH partly through shifting the imbalance of Treg/Th17 cells to Treg dominance. Int Immunopharmacol. 2014;23:489–498. doi: 10.1016/j.intimp.2014.09.024.
    1. Rolla S, et al. The balance between IL-17 and IL-22 produced by liver-infiltrating T-helper cells critically controls NASH development in mice. Clin Sci (Lond) 2016;130:193–203. doi: 10.1042/CS20150405.
    1. Sell H, Habich C, Eckel J. Adaptive immunity in obesity and insulin resistance. Nat Rev Endocrinol. 2012;8:709–716. doi: 10.1038/nrendo.2012.114.
    1. Ma X, et al. A high-fat diet and regulatory T cells influence susceptibility to endotoxin-induced liver injury. Hepatology. 2007;46:1519–1529. doi: 10.1002/hep.21823.
    1. Ferreyra Solari NE, et al. The role of innate cells is coupled to a Th1-polarized immune response in pediatric nonalcoholic steatohepatitis. J Clin Immunol. 2012;32:611–621. doi: 10.1007/s10875-011-9635-2.
    1. Tang Y, et al. Interleukin-17 exacerbates hepatic steatosis and inflammation in non-alcoholic fatty liver disease. Clin Exp Immunol. 2011;166:281–290. doi: 10.1111/j.1365-2249.2011.04471.x.
    1. Xu R, Tao A, Zhang S, Zhang M. Neutralization of interleukin-17 attenuates high fat diet-induced non-alcoholic fatty liver disease in mice. Acta Biochim Biophys Sin (Shanghai) 2013;45:726–733. doi: 10.1093/abbs/gmt065.
    1. Saalim M, et al. IL-22: a promising candidate to inhibit viral-induced liver disease progression and hepatocellular carcinoma. Tumour Biol. 2016;37:105–114. doi: 10.1007/s13277-015-4294-1.

Source: PubMed

3
Abonnieren