Flaxseed Powder Attenuates Non-Alcoholic Steatohepatitis via Modulation of Gut Microbiota and Bile Acid Metabolism through Gut–Liver Axis
Abstract
:1. Introduction
2. Results
2.1. The Different Physiological Changes and Biochemical Parameters in the Control, HFD and FLA-Fed Groups
2.2. Disorders of BA Profiles in the HFD Group and a Relative Increase in the Proportion of Non-12αOH BAs in the FLA-Fed Groups
2.3. Gut Microbiome Alteration Induced by FLA-Fed and Its Correlation with BAs
2.4. FLA-Fed Alleviates NASH via Modulating BA Metabolism by FXR/FGF15 and TGR5/NF-κB Signaling
3. Discussion
4. Materials and Methods
4.1. Animal Experiments and Diets
4.2. Serum Sample Collection and Analysis
4.3. Liver Histological Analyzes
4.4. Quantitative Real-Time PCR
4.5. Western Blot Analyses
4.6. Gut Microbiota Assessment
4.7. Bas Analysis
4.8. Statistical Analysis
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Appendix A. Sequencing
Appendix A.1. Extraction of Genome DNA
Appendix A.2. Amplicon Generation
Appendix A.3. PCR Products Quantification and Qualification
Appendix A.4. Library Preparation and Sequencing
Appendix B. Data Analysis
Appendix B.1. Paired-End Reads Assembly and Quality Control
Appendix B.1.1. Data Split
Appendix B.1.2. Sequence Assembly
Appendix B.1.3. Data Filtration
Appendix B.1.4. Chimera Removal
References
- Younossi, Z.; Anstee, Q.M.; Marietti, M.; Hardy, T.; Henry, L.; Eslam, M.; George, J.; Bugianesi, E. Global burden of NAFLD and NASH: Trends, predictions, risk factors and prevention. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 11–20. [Google Scholar] [CrossRef] [PubMed]
- Younossi, Z.M. Non-alcoholic fatty liver disease—A global public health perspective. J. Hepatol. 2019, 70, 531–544. [Google Scholar] [CrossRef] [Green Version]
- Huang, D.Q.; El-Serag, H.B.; Loomba, R. Global epidemiology of NAFLD-related HCC: Trends, predictions, risk factors and prevention. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 223–238. [Google Scholar] [CrossRef]
- Younossi, Z.M.; Henry, L. Epidemiology of non-alcoholic fatty liver disease and hepatocellular carcinoma. JHEP Rep. Innov. Hepatol. 2021, 3, 100305. [Google Scholar] [CrossRef]
- Safari, Z.; Gérard, P. The links between the gut microbiome and Non-Alcoholic Fatty Liver Disease (NAFLD). Cell. Mol. Life Sci. CMLS 2019, 76, 1541–1558. [Google Scholar] [CrossRef] [PubMed]
- Puri, P.; Daita, K.; Joyce, A.; Mirshahi, F.; Santhekadur, P.K.; Cazanave, S.; Luketic, V.A.; Siddiqui, M.S.; Boyett, S.; Min, H.K.; et al. The presence and severity of nonalcoholic steatohepatitis is associated with specific changes in circulating bile acids. Hepatology 2018, 67, 534–548. [Google Scholar] [CrossRef] [PubMed]
- Chow, M.D.; Lee, Y.H.; Guo, G.L. The role of bile acids in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Mol. Asp. Med. 2017, 56, 34–44. [Google Scholar] [CrossRef]
- Albillos, A.; de Gottardi, A.; Rescigno, M. The gut-liver axis in liver disease: Pathophysiological basis for therapy. J. Hepatol. 2020, 72, 558–577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jia, W.; Xie, G.; Jia, W. Bile acid–microbiota crosstalk in gastrointestinal inflammation and carcinogenesis. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 111–128. [Google Scholar] [CrossRef] [Green Version]
- Wahlström, A.; Sayin, S.I.; Marschall, H.U.; Bäckhed, F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab. 2016, 24, 41–50. [Google Scholar] [CrossRef] [Green Version]
- Xie, G.; Jiang, R.; Wang, X.; Liu, P.; Zhao, A.; Wu, Y.; Huang, F.; Liu, Z.; Rajani, C.; Zheng, X.; et al. Conjugated secondary 12α-hydroxylated bile acids promote liver fibrogenesis. EBioMedicine 2021, 66, 103290. [Google Scholar] [CrossRef]
- Martin, F.P.; Dumas, M.E.; Wang, Y.; Legido-Quigley, C.; Yap, I.K.; Tang, H.; Zirah, S.; Murphy, G.M.; Cloarec, O.; Lindon, J.C.; et al. A top-down systems biology view of microbiome-mammalian metabolic interactions in a mouse model. Mol. Syst. Biol. 2007, 3, 112. [Google Scholar] [CrossRef] [Green Version]
- Pandak, W.M.; Kakiyama, G. The acidic pathway of bile acid synthesis: Not just an alternative pathway. Liver Res. 2019, 3, 88–98. [Google Scholar] [CrossRef]
- Haeusler, R.A.; Pratt-Hyatt, M.; Welch, C.L.; Klaassen, C.D.; Accili, D. Impaired generation of 12-hydroxylated bile acids links hepatic insulin signaling with dyslipidemia. Cell Metab. 2012, 15, 65–74. [Google Scholar] [CrossRef] [Green Version]
- Haeusler, R.A.; Astiarraga, B.; Camastra, S.; Accili, D.; Ferrannini, E. Human insulin resistance is associated with increased plasma levels of 12α-hydroxylated bile acids. Diabetes 2013, 62, 4184–4191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hori, S.; Abe, T.; Lee, D.G.; Fukiya, S.; Yokota, A.; Aso, N.; Shirouchi, B.; Sato, M.; Ishizuka, S. Association between 12α-hydroxylated bile acids and hepatic steatosis in rats fed a high-fat diet. J. Nutr. Biochem. 2020, 83, 108412. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.Y.; Shimizu, H.; Hagio, M.; Fukiya, S.; Watanabe, M.; Tanaka, Y.; Joe, G.H.; Iwaya, H.; Yoshitsugu, R.; Kikuchi, K.; et al. 12α-hydroxylated bile acid induces hepatic steatosis with dysbiosis in rats. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2020, 1865, 158811. [Google Scholar] [CrossRef] [PubMed]
- Neuschwander-Tetri, B.A.; Loomba, R.; Sanyal, A.J.; Lavine, J.E.; Van Natta, M.L.; Abdelmalek, M.F.; Chalasani, N.; Dasarathy, S.; Diehl, A.M.; Hameed, B.; 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, 956–965. [Google Scholar] [CrossRef] [Green Version]
- Younossi, Z.M.; Ratziu, V.; Loomba, R.; Rinella, M.; Anstee, Q.M.; Goodman, Z.; Bedossa, P.; Geier, A.; Beckebaum, S.; Newsome, P.N.; et al. Obeticholic acid for the treatment of non-alcoholic steatohepatitis: Interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial. Lancet 2019, 394, 2184–2196. [Google Scholar] [CrossRef] [Green Version]
- Arora, T.; Bäckhed, F. The gut microbiota and metabolic disease: Current understanding and future perspectives. J. Intern. Med. 2016, 280, 339–349. [Google Scholar] [CrossRef] [PubMed]
- Begley, M.; Hill, C.; Gahan, C.G. Bile salt hydrolase activity in probiotics. Appl. Environ. Microbiol. 2006, 72, 1729–1738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zheng, X.; Huang, F.; Zhao, A.; Lei, S.; Zhang, Y.; Xie, G.; Chen, T.; Qu, C.; Rajani, C.; Dong, B.; et al. Bile acid is a significant host factor shaping the gut microbiome of diet-induced obese mice. BMC Biol. 2017, 15, 120. [Google Scholar] [CrossRef] [Green Version]
- Huang, F.; Zheng, X.; Ma, X.; Jiang, R.; Zhou, W.; Zhou, S.; Zhang, Y.; Lei, S.; Wang, S.; Kuang, J.; et al. Theabrownin from Pu-erh tea attenuates hypercholesterolemia via modulation of gut microbiota and bile acid metabolism. Nat. Commun. 2019, 10, 4971. [Google Scholar] [CrossRef] [Green Version]
- Ferrell, J.M.; Pathak, P.; Boehme, S.; Gilliland, T.; Chiang, J.Y.L. Deficiency of both Farnesoid X receptor and Takeda G protein-coupled receptor 5 exacerbated liver fibrosis in mice. Hepatology 2019, 70, 955–970. [Google Scholar] [CrossRef] [PubMed]
- Kuipers, F.; Bloks, V.W.; Groen, A.K. Beyond intestinal soap-bile acids in metabolic control. Nat. Rev. Endocrinol. 2014, 10, 488–498. [Google Scholar] [CrossRef] [PubMed]
- Sayin, S.I.; Wahlstrom, A.; Felin, J.; Jantti, S.; Marschall, H.-U.; Bamberg, K.; Angelin, B.; Hyotylainen, T.; Oresic, M.; Backhed, F. Gut microbiota regulates bile acid metabolism by reducing the levels of tauro-beta-muricholic acid, a naturally occurring FXR antagonist. Cell Metab. 2013, 17, 225–235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, L.; Cai, J.; Gonzalez, F.J. The role of farnesoid X receptor in metabolic diseases, and gastrointestinal and liver cancer. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 335–347. [Google Scholar] [CrossRef]
- Svegliati-Baroni, G.; Patricio, B.; Lioci, G.; Macedo, M.P.; Gastaldelli, A. Gut-pancreas-liver axis as a target for treatment of NAFLD/NASH. Int. J. Mol. Sci. 2020, 21, 5820. [Google Scholar] [CrossRef]
- Yari, Z.; Cheraghpour, M.; Alavian, S.M.; Hedayati, M.; Eini-Zinab, H.; Hekmatdoost, A. The efficacy of flaxseed and hesperidin on non-alcoholic fatty liver disease: An open-labeled randomized controlled trial. Eur. J. Clin. Nutr. 2021, 75, 99–111. [Google Scholar] [CrossRef]
- Yari, Z.; Rahimlou, M.; Eslamparast, T.; Ebrahimi-Daryani, N.; Poustchi, H.; Hekmatdoost, A. Flaxseed supplementation in non-alcoholic fatty liver disease: A pilot randomized, open labeled, controlled study. Int. J. Food Sci. Nutr. 2016, 67, 461–469. [Google Scholar] [CrossRef] [PubMed]
- Luo, J.; Qi, J.; Wang, W.; Luo, Z.; Liu, L.; Zhang, G.; Zhou, Q.; Liu, J.; Peng, X. Antiobesity effect of flaxseed polysaccharide via inducing satiety due to leptin resistance removal and promoting lipid metabolism through the AMP-Activated Protein Kinase (AMPK) signaling pathway. J. Agric. food Chem. 2019, 67, 7040–7049. [Google Scholar] [CrossRef] [PubMed]
- Rezaei, S.; Sasani, M.R.; Akhlaghi, M.; Kohanmoo, A. Flaxseed oil in the context of a weight loss programme ameliorates fatty liver grade in patients with non-alcoholic fatty liver disease: A randomised double-blind controlled trial. Br. J. Nutr. 2020, 123, 994–1002. [Google Scholar] [CrossRef] [PubMed]
- Xu, J.Q.; Rong, S.; Gao, H.; Chen, C.; Yang, W.; Deng, Q.C.; Huang, Q.D.; Xiao, L.Y.; Huang, F.H. A Combination of flaxseed oil and astaxanthin improves hepatic lipid accumulation and reduces oxidative stress in high fat-diet fed rats. Nutrients 2017, 9, 271. [Google Scholar] [CrossRef]
- Nogueira, M.A.; Oliveira, C.P.; Alves, V.A.F.; Stefano, J.T.; Rodrigues, L.S.D.; Torrinhas, R.S.; Cogliati, B.; Barbeiro, H.; Carrilho, F.J.; Waitzberg, D.L. Omega-3 polyunsaturated fatty acids in treating non-alcoholic steatohepatitis: A randomized, double-blind, placebo-controlled trial. Clin. Nutr. 2016, 35, 578–586. [Google Scholar] [CrossRef]
- Han, H.; Ma, H.F.; Rong, S.; Chen, L.; Shan, Z.L.; Xu, J.Q.; Zhang, Y.J.; Liu, L.G. Flaxseed oil containing flaxseed oil ester of plant sterol attenuates high-fat diet-induced hepatic steatosis in apolipoprotein-E knockout mice. J. Funct. Foods 2015, 13, 169–182. [Google Scholar] [CrossRef]
- Kezimana, P.; Dmitriev, A.A.; Kudryavtseva, A.V.; Romanova, E.V.; Melnikova, N.V. Secoisolariciresinol diglucoside of flaxseed and its metabolites: Biosynthesis and potential for nutraceuticals. Front. Genet. 2018, 9, 641. [Google Scholar] [CrossRef] [Green Version]
- Wang, H.; Wei, J.; Li, C.; Jing, Y.; Zhao, T.; Zhang, X.; Zhang, C. Effect of dietary flaxseed powder on intestinal flora structure in healthy adults. Food Sci. 2018, 39, 224–229. [Google Scholar]
- Luo, J.; Li, Y.; Mai, Y.; Gao, L.; Ou, S.; Wang, Y.; Liu, L.; Peng, X. Flaxseed gum reduces body weight by regulating gut microbiota. J. Funct. Foods 2018, 47, 136–142. [Google Scholar] [CrossRef]
- Yang, C.; Xu, Z.; Deng, Q.; Huang, Q.; Wang, X.; Huang, F. Beneficial effects of flaxseed polysaccharides on metabolic syndrome via gut microbiota in high-fat diet fed mice. Food Res. Int. 2020, 131, 108994. [Google Scholar] [CrossRef]
- David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jones, B.V.; Begley, M.; Hill, C.; Gahan, C.G.; Marchesi, J.R. Functional and comparative metagenomic analysis of bile salt hydrolase activity in the human gut microbiome. Proc. Nat. Acad. Sci. USA 2008, 105, 13580–13585. [Google Scholar] [CrossRef] [Green Version]
- Qin, J.; Li, Y.; Cai, Z.; Li, S.; Zhu, J.; Zhang, F.; Liang, S.; Zhang, W.; Guan, Y.; Shen, D.; et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012, 490, 55–60. [Google Scholar] [CrossRef] [PubMed]
- Kim, I.; Ahn, S.-H.; Inagaki, T.; Choi, M.; Ito, S.; Guo, G.L.; Kliewer, S.A.; Gonzalez, F.J. Differential regulation of bile acid homeostasis by the farnesoid X receptor in liver and intestine. J. Lipid Res. 2007, 48, 2664–2672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zaborska, K.E.; Cummings, B.P. Rethinking bile acid metabolism and signaling for type 2 diabetes treatment. Curr. Diabetes Rep. 2018, 18, 108. [Google Scholar]
- Pandak, W.M.; Bohdan, P.; Franklund, C.; Mallonee, D.H.; Eggertsen, G.; Björkhem, I.; Gil, G.; Vlahcevic, Z.R.; Hylemon, P.B. Expression of sterol 12alpha-hydroxylase alters bile acid pool composition in primary rat hepatocytes and in vivo. Gastroenterology 2001, 120, 1801–1809. [Google Scholar] [CrossRef] [PubMed]
- Mouzaki, M.; Comelli, E.M.; Arendt, B.M.; Bonengel, J.; Fung, S.K.; Fischer, S.E.; McGilvray, I.D.; Allard, J.P. Intestinal microbiota in patients with nonalcoholic fatty liver disease. Hepatology 2013, 58, 120–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Koliada, A.; Syzenko, G.; Moseiko, V.; Budovska, L.; Puchkov, K.; Perederiy, V.; Gavalko, Y.; Dorofeyev, A.; Romanenko, M.; Tkach, S.; et al. Association between body mass index and firmicutes/bacteroidetes ratio in an adult Ukrainian population. BMC Microbiol. 2017, 17, s12866–s017. [Google Scholar]
- Marques, F.Z.; Nelson, E.; Chu, P.Y.; Horlock, D.; Fiedler, A.; Ziemann, M.; Tan, J.K.; Kuruppu, S.; Rajapakse, N.W.; El-Osta, A.; et al. High-fiber diet and acetate supplementation change the gut microbiota and prevent the development of hypertension and heart failure in hypertensive mice. Circulation 2017, 135, 964–977. [Google Scholar] [CrossRef]
- Jia, B.; Park, D.; Chun, B.H.; Hahn, Y.; Jeon, C.O. Diet-related alterations of gut bile salt hydrolases determined using a metagenomic analysis of the human microbiome. Int J. Mol. Sci 2021, 22, 3652. [Google Scholar] [CrossRef]
- Islam, K.B.; Fukiya, S.; Hagio, M.; Fujii, N.; Ishizuka, S.; Ooka, T.; Ogura, Y.; Hayashi, T.; Yokota, A. Bile acid is a host factor that regulates the composition of the cecal microbiota in rats. Gastroenterology 2011, 141, 1773–1781. [Google Scholar] [CrossRef]
- Joyce, S.A.; MacSharry, J.; Casey, P.G.; Kinsella, M.; Murphy, E.F.; Shanahan, F.; Hill, C.; Gahan, C.G. Regulation of host weight gain and lipid metabolism by bacterial bile acid modification in the gut. Proc. Nat. Acad. Sci. USA 2014, 111, 7421–7426. [Google Scholar] [CrossRef] [Green Version]
- Yang, L.; Li, Y.; Wang, S.; Bian, X.; Jiang, X.; Wu, J.; Wang, K.; Wang, Q.; Xia, J.; Jiang, S.; et al. Western diet aggravated carbon tetrachloride-induced chronic liver injury by disturbing gut microbiota and bile acid metabolism. Mol. Nutr. Food Res. 2021, 65, e2000811. [Google Scholar] [CrossRef] [PubMed]
- Ridlon, J.M.; Kang, D.J.; Hylemon, P.B. Bile salt biotransformations by human intestinal bacteria. J. Lipid Res. 2006, 47, 241–259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sang, C.; Wang, X.; Zhou, K.; Sun, T.; Bian, H.; Gao, X.; Wang, Y.; Zhang, H.; Jia, W.; Liu, P.; et al. Bile acid profiles are distinct among patients with different etiologies of chronic liver disease. J. Proteom. Res. 2021, 20, 2340–2351. [Google Scholar] [CrossRef]
- Duboc, H.; Taché, Y.; Hofmann, A.F. The bile acid TGR5 membrane receptor: From basic research to clinical application. Dig. Liver Dis. Off. J. Ital. Soc. Gastroenterol. Ital. Assoc. Study Liver 2014, 46, 302–312. [Google Scholar] [CrossRef] [Green Version]
- Ye, H.L.; Zhang, J.W.; Chen, X.Z.; Wu, P.B.; Chen, L.; Zhang, G. Ursodeoxycholic acid alleviates experimental liver fibrosis involving inhibition of autophagy. Life Sci. 2020, 242, 117175. [Google Scholar] [CrossRef]
- Ratziu, V.; de Ledinghen, V.; Oberti, F.; Mathurin, P.; Wartelle-Bladou, C.; Renou, C.; Sogni, P.; Maynard, M.; Larrey, D.; Serfaty, L.; et al. A randomized controlled trial of high-dose ursodesoxycholic acid for nonalcoholic steatohepatitis. J. Hepatol. 2011, 54, 1011–1019. [Google Scholar] [CrossRef]
- Wei, M.; Huang, F.; Zhao, L.; Zhang, Y.; Yang, W.; Wang, S.; Li, M.; Han, X.; Ge, K.; Qu, C.; et al. A dysregulated bile acid-gut microbiota axis contributes to obesity susceptibility. EBioMedicine 2020, 55, 102766. [Google Scholar] [CrossRef] [PubMed]
- Wang, K.; Liao, M.; Zhou, N.; Bao, L.; Ma, K.; Zheng, Z.; Wang, Y.; Liu, C.; Wang, W.; Wang, J.; et al. Parabacteroides distasonis alleviates obesity and metabolic dysfunctions via production of succinate and secondary bile acids. Cell Rep. 2019, 26, 222–235.e225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hylemon, P.B.; Zhou, H.; Pandak, W.M.; Ren, S.; Gil, G.; Dent, P. Bile acids as regulatory molecules. J. Lipid Res. 2009, 50, 1509–1520. [Google Scholar] [CrossRef] [Green Version]
- Thomas, C.; Pellicciari, R.; Pruzanski, M.; Auwerx, J.; Schoonjans, K. Targeting bile-acid signalling for metabolic diseases. Nat. Rev. Drug Discov. 2008, 7, 678–693. [Google Scholar] [CrossRef] [PubMed]
- Hartmann, P.; Hochrath, K.; Horvath, A.; Chen, P.; Seebauer, C.T.; Llorente, C.; Wang, L.; Alnouti, Y.; Fouts, D.E.; Stärkel, P.; et al. Modulation of the intestinal bile acid/farnesoid X receptor/fibroblast growth factor 15 axis improves alcoholic liver disease in mice. Hepatology 2018, 67, 2150–2166. [Google Scholar] [CrossRef] [PubMed]
- Song, M.; Ye, J.; Zhang, F.; Su, H.; Yang, X.; He, H.; Liu, F.; Zhu, X.; Wang, L.; Gao, P.; et al. Chenodeoxycholic acid (CDCA) protects against the lipopolysaccharide-induced impairment of the intestinal epithelial barrier function via the FXR-MLCK pathway. J. Agric. Food Chem. 2019, 67, 8868–8874. [Google Scholar] [CrossRef] [PubMed]
- Sorribas, M.; Jakob, M.O.; Yilmaz, B.; Li, H.; Stutz, D.; Noser, Y.; de Gottardi, A.; Moghadamrad, S.; Hassan, M.; Albillos, A.; et al. FXR modulates the gut-vascular barrier by regulating the entry sites for bacterial translocation in experimental cirrhosis. J. Hepatol. 2019, 71, 1126–1140. [Google Scholar] [CrossRef] [PubMed]
- Wang, C.; Tao, Q.; Wang, X.; Wang, X.; Zhang, X. Impact of high-fat diet on liver genes expression profiles in mice model of nonalcoholic fatty liver disease. Environ. Toxicol. Pharmacol. 2016, 45, 52–62. [Google Scholar] [CrossRef]
- Jiang, C.; Xie, C.; Li, F.; Zhang, L.; Nichols, R.G.; Krausz, K.W.; Cai, J.; Qi, Y.; Fang, Z.Z.; Takahashi, S.; et al. Intestinal farnesoid X receptor signaling promotes nonalcoholic fatty liver disease. J. Clin. Investing. 2015, 125, 386–402. [Google Scholar] [CrossRef]
- Li, F.; Jiang, C.; Krausz, K.W.; Li, Y.; Albert, I.; Hao, H.; Fabre, K.M.; Mitchell, J.B.; Patterson, A.D.; Gonzalez, F.J. Microbiome remodelling leads to inhibition of intestinal farnesoid X receptor signalling and decreased obesity. Nat. Commun. 2013, 4, 2384. [Google Scholar] [CrossRef]
- Bertaggia, E.; Jensen, K.K.; Castro-Perez, J.; Xu, Y.; Di Paolo, G.; Chan, R.B.; Wang, L.; Haeusler, R.A. Cyp8b1 ablation prevents western diet-induced weight gain and hepatic steatosis because of impaired fat absorption. Am. J. Physiol. Endocrinol Metab. 2017, 313, E121–E133. [Google Scholar] [CrossRef] [Green Version]
- Cui, G.; Martin, R.C.; Jin, H.; Liu, X.; Pandit, H.; Zhao, H.; Cai, L.; Zhang, P.; Li, W.; Li, Y. Up-regulation of FGF15/19 signaling promotes hepatocellular carcinoma in the background of fatty liver. J. Exp. Clin. Cancer Res. CR 2018, 37, 136. [Google Scholar] [CrossRef]
- Li, Y.; Zhang, W.; Doughtie, A.; Cui, G.; Li, X.; Pandit, H.; Yang, Y.; Li, S.; Martin, R. Up-regulation of fibroblast growth factor 19 and its receptor associates with progression from fatty liver to hepatocellular carcinoma. Oncotarget 2016, 7, 52339. [Google Scholar] [CrossRef] [Green Version]
- Ocker, M. Fibroblast growth factor signaling in non-alcoholic fatty liver disease and non-alcoholic steatohepatitis: Paving the way to hepatocellular carcinoma. World J. Gastroenterol. 2020, 26, 279–290. [Google Scholar] [CrossRef]
- Chen, Q.; Jiang, Y.; An, Y.; Zhao, N.; Zhao, Y.; Yu, C. Soluble FGFR4 extracellular domain inhibits FGF19-induced activation of FGFR4 signaling and prevents nonalcoholic fatty liver disease. Biochem. Biophys. Res. Commun. 2011, 409, 651–656. [Google Scholar] [CrossRef]
- Ji, C.G.; Xie, X.L.; Yin, J.; Qi, W.; Chen, L.; Bai, Y.; Wang, N.; Zhao, D.Q.; Jiang, X.Y.; Jiang, H.Q. Bile acid receptor TGR5 overexpression is associated with decreased intestinal mucosal injury and epithelial cell proliferation in obstructive jaundice. Transl. Res. J. Lab. Clin. Med. 2017, 182, 88–102. [Google Scholar] [CrossRef]
- Li, Y.; Jadhav, K.; Zhang, Y. Bile acid receptors in non-alcoholic fatty liver disease. Biochem. Pharmacol. 2013, 86, 1517–1524. [Google Scholar] [CrossRef]
- Perino, A.; Schoonjans, K. TGR5 and immunometabolism: Insights from physiology and pharmacology. Trends Pharmacol. Sci. 2015, 36, 847–857. [Google Scholar] [CrossRef] [PubMed]
- Kleiner, D.E.; Brunt, E.M.; Van Natta, M.; Behling, C.; Contos, M.J.; Cummings, O.W.; Ferrell, L.D.; Liu, Y.C.; Torbenson, M.S.; Unalp-Arida, A.; et al. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology 2005, 41, 1313–1321. [Google Scholar] [CrossRef] [PubMed]
- Liu, H.; Zhu, H.; Xia, H.; Yang, X.; Yang, L.; Wang, S.; Wen, J.; Sun, G. Different effects of high-fat diets rich in different oils on lipids metabolism, oxidative stress and gut microbiota. Food Res. Int. 2021, 141, 110078. [Google Scholar] [CrossRef]
- Livak, K.J.; Schmittgen, T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods 2001, 25, 402–408. [Google Scholar] [CrossRef]
- Magoč, T.; Salzberg, S.L. FLASH: Fast length adjustment of short reads to improve genome assemblies. Bioinformatics. 2011, 27, 2957–2963. [Google Scholar] [CrossRef]
- Bokulich, N.A.; Subramanian, S.; Faith, J.J.; Gevers, D.; Gordon, J.I.; Knight, R.; Mills, D.A.; Caporaso, J.G. Quality-filtering vastly improves diversity estimates from Illumina amplicon sequencing. Nat. Methods 2013, 10, 57–59. [Google Scholar] [CrossRef]
- Caporaso, J.G.; Kuczynski, J.; Stombaugh, J.; Bittinger, K.; Bushman, F.; Costello, E.K.; Fierer, N.; Peña, A.G.; Goodrich, J.K.; Gordon, J.I.; et al. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods 2010, 7, 335–336. [Google Scholar] [CrossRef] [Green Version]
- Edgar, R.C.; Haas, B.J.; Clemente, J.C.; Quince, C.; Knight, R. UCHIME improves sensitivity and speed of chimera detection. Bioinformatics 2011, 27, 2194–2200. [Google Scholar] [CrossRef] [Green Version]
- Haas, B.J.; Gevers, D.; Earl, A.M.; Feldgarden, M.; Ward, D.V.; Giannoukos, G.; Ciulla, D.; Tabbaa, D.; Highlander, S.K.; Sodergren, E.; et al. Chimeric 16S rRNA sequence formation and detection in Sanger and 454-pyrosequenced PCR amplicons. Genome Res. 2011, 21, 494–504. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Yang, C.; Wan, M.; Xu, D.; Pan, D.; Xia, H.; Yang, L.; Sun, G. Flaxseed Powder Attenuates Non-Alcoholic Steatohepatitis via Modulation of Gut Microbiota and Bile Acid Metabolism through Gut–Liver Axis. Int. J. Mol. Sci. 2021, 22, 10858. https://doi.org/10.3390/ijms221910858
Yang C, Wan M, Xu D, Pan D, Xia H, Yang L, Sun G. Flaxseed Powder Attenuates Non-Alcoholic Steatohepatitis via Modulation of Gut Microbiota and Bile Acid Metabolism through Gut–Liver Axis. International Journal of Molecular Sciences. 2021; 22(19):10858. https://doi.org/10.3390/ijms221910858
Chicago/Turabian StyleYang, Chao, Min Wan, Dengfeng Xu, Da Pan, Hui Xia, Ligang Yang, and Guiju Sun. 2021. "Flaxseed Powder Attenuates Non-Alcoholic Steatohepatitis via Modulation of Gut Microbiota and Bile Acid Metabolism through Gut–Liver Axis" International Journal of Molecular Sciences 22, no. 19: 10858. https://doi.org/10.3390/ijms221910858
APA StyleYang, C., Wan, M., Xu, D., Pan, D., Xia, H., Yang, L., & Sun, G. (2021). Flaxseed Powder Attenuates Non-Alcoholic Steatohepatitis via Modulation of Gut Microbiota and Bile Acid Metabolism through Gut–Liver Axis. International Journal of Molecular Sciences, 22(19), 10858. https://doi.org/10.3390/ijms221910858