Role of gut microbiota in type 2 diabetes pathophysiology

Manoj Gurung, Zhipeng Li, Hannah You, Richard Rodrigues, Donald B Jump, Andrey Morgun, Natalia Shulzhenko, Manoj Gurung, Zhipeng Li, Hannah You, Richard Rodrigues, Donald B Jump, Andrey Morgun, Natalia Shulzhenko

Abstract

A substantial body of literature has provided evidence for the role of gut microbiota in metabolic diseases including type 2 diabetes. However, reports vary regarding the association of particular taxonomic groups with disease. In this systematic review, we focused on the potential role of different bacterial taxa affecting diabetes. We have summarized evidence from 42 human studies reporting microbial associations with disease, and have identified supporting preclinical studies or clinical trials using treatments with probiotics. Among the commonly reported findings, the genera of Bifidobacterium, Bacteroides, Faecalibacterium, Akkermansia and Roseburia were negatively associated with T2D, while the genera of Ruminococcus, Fusobacterium, and Blautia were positively associated with T2D. We also discussed potential molecular mechanisms of microbiota effects in the onset and progression of T2D.

Keywords: 16S rRNA; Insulin resistance; Metagenomics; Microbiota; Type 2 diabetes.

Copyright © 2019 The Authors. Published by Elsevier B.V. All rights reserved.

Figures

Fig. 1
Fig. 1
Microbial genera most frequently found to be associated with T2D. Number of studies reporting one of the indicated genera in association with T2D (without treatment), and including anti-diabetic therapy (All) in addition to the largest human study by He et al., 2018 .
Fig. 2
Fig. 2
Literature-based network analysis of potential effects on metabolism of bacterial taxa consistently found in association with human T2D (shown in Fig. 1). References corresponding to each edge can be found in the text.

References

    1. He Y. Regional variation limits applications of healthy gut microbiome reference ranges and disease models. Nat Med. 2018;24(10):1532–1535.
    1. Gao R. Dysbiosis signatures of gut microbiota along the sequence from healthy, young patients to those with overweight and obesity. Obesity (Silver Spring) 2018;26(2):351–361.
    1. Candela M. Modulation of gut microbiota dysbioses in type 2 diabetic patients by macrobiotic Ma-Pi 2 diet. Br J Nutr. 2016;116(1):80–93.
    1. Sedighi M. Comparison of gut microbiota in adult patients with type 2 diabetes and healthy individuals. Microb Pathog. 2017;111:362–369.
    1. Wu X. Molecular characterisation of the faecal microbiota in patients with type II diabetes. Curr Microbiol. 2010;61(1):69–78.
    1. Wu H. Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug. Nat Med. 2017;23(7):850–858.
    1. Barengolts E. Gut microbiota varies by opioid use, circulating leptin and oxytocin in African American men with diabetes and high burden of chronic disease. PLoS ONE. 2018;13(3)
    1. Xu J. Structural modulation of gut microbiota during alleviation of type 2 diabetes with a Chinese herbal formula. ISME J. 2015;9(3):552–562.
    1. Pedersen C. Host-microbiome interactions in human type 2 diabetes following prebiotic fibre (galacto-oligosaccharide) intake. Br J Nutr. 2016;116(11):1869–1877.
    1. Sasaki M. Transglucosidase improves the gut microbiota profile of type 2 diabetes mellitus patients: a randomized double-blind, placebo-controlled study. BMC Gastroenterol. 2013;13:81.
    1. Murphy R. Differential changes in gut microbiota after gastric bypass and sleeve gastrectomy bariatric surgery vary according to diabetes remission. Obes Surg. 2017;27(4):917–925.
    1. Le T.K. Bifidobacterium species lower serum glucose, increase expressions of insulin signaling proteins, and improve adipokine profile in diabetic mice. Biomed Res. 2015;36(1):63–70.
    1. Moya-Perez A, Neef A, Sanz Y. Bifidobacterium pseudocatenulatum CECT 7765 reduces obesity-associated inflammation by restoring the lymphocyte-macrophage balance and gut microbiota structure in high-fat diet-fed mice. PLoS ONE. 2015;10(7)
    1. Kikuchi K., Ben Othman M., Sakamoto K. Sterilized bifidobacteria suppressed fat accumulation and blood glucose level. Biochem Biophys Res Commun. 2018;501(4):1041–1047.
    1. Aoki R. A proliferative probiotic bifidobacterium strain in the gut ameliorates progression of metabolic disorders via microbiota modulation and acetate elevation. Sci Rep. 2017;7:43522.
    1. Wang J. Modulation of gut microbiota during probiotic-mediated attenuation of metabolic syndrome in high fat diet-fed mice. ISME J. 2015;9(1):1–15.
    1. Zhang X. Human gut microbiota changes reveal the progression of glucose intolerance. PLoS ONE. 2013;8(8):e71108.
    1. Lippert K. Gut microbiota dysbiosis associated with glucose metabolism disorders and the metabolic syndrome in older adults. Benef Microbes. 2017;8(4):545–556.
    1. Yamaguchi Y. Association of intestinal microbiota with metabolic markers and dietary habits in patients with type 2 diabetes. Digestion. 2016;94(2):66–72.
    1. Munukka E. Women with and without metabolic disorder differ in their gut microbiota composition. Obesity. 2012;20(5):1082–1087.
    1. Sun L. Gut microbiota and intestinal FXR mediate the clinical benefits of metformin. Nat Med. 2018;24(12):1919–1929.
    1. Malik F. Is metformin poised for a second career as an antimicrobial? Diabetes Metab Res Rev. 2018;34(4):e2975.
    1. He Y. Linking gut microbiota, metabolic syndrome and economic status based on a population-level analysis. Microbiome. 2018;6(1):172.
    1. Karlsson FH. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature. 2013;498(7452):99–103.
    1. Yang J.Y. Gut commensal bacteroides acidifaciens prevents obesity and improves insulin sensitivity in mice. Mucosal Immunol. 2017;10(1):104–116.
    1. Cano G. Bacteroides uniformis CECT 7771 ameliorates metabolic and immunological dysfunction in mice with high-fat-diet induced obesity. PLoS ONE. 2012;7(7):e41079.
    1. Larsen N. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS ONE. 2010;5(2):e9085.
    1. Salamon D. Characteristics of gut microbiota in adult patients with type 1 and type 2 diabetes based on nextgeneration sequencing of the 16S rRNA gene fragment. Pol Arch Intern Med. 2018;128(6):336–343.
    1. Forslund K. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature. 2015;528(7581):262–266.
    1. Tong X. Structural alteration of gut microbiota during the amelioration of human type 2 diabetes with hyperlipidemia by metformin and a traditional Chinese herbal formula: a multicenter, randomized, open label clinical trial. MBio. 2018;9(3) pii: e02392-17.
    1. Patrone V. Postoperative changes in fecal bacterial communities and fermentation products in obese patients undergoing bilio-intestinal bypass. Front Microbiol. 2016;7:200.
    1. Remely M. Effects of short chain fatty acid producing bacteria on epigenetic regulation of FFAR3 in type 2 diabetes and obesity. Gene. 2014;537(1):85–92.
    1. Furet JP. Differential adaptation of human gut microbiota to bariatric surgery-induced weight loss: links with metabolic and low-grade inflammation markers. Diabetes. 2010;59(12):3049–3057.
    1. Graessler J. Metagenomic sequencing of the human gut microbiome before and after bariatric surgery in obese patients with type 2 diabetes: correlation with inflammatory and metabolic parameters. Pharmacogenomics J. 2013;13(6):514–522.
    1. Rossi O. Faecalibacterium prausnitzii strain HTF-F and its extracellular polymeric matrix attenuate clinical parameters in DSS-Induced colitis. PLoS ONE. 2015;10(4)
    1. Munukka E. Faecalibacterium prausnitzii treatment improves hepatic health and reduces adipose tissue inflammation in high-fat fed mice. ISME J. 2017;11(7):1667–1679.
    1. Plovier H. A purified membrane protein from Akkermansia muciniphila or the pasteurized bacterium improves metabolism in obese and diabetic mice. Nat Med. 2017;23(1):107–113.
    1. Greer R.L. Akkermansia muciniphila mediates negative effects of IFNgamma on glucose metabolism. Nat Commun. 2016;7:13329.
    1. Everard A. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci U S A. 2013;110(22):9066–9071.
    1. Zhao S. Akkermansia muciniphila improves metabolic profiles by reducing inflammation in chow diet-fed mice. J Mol Endocrinol. 2017;58(1):1–14.
    1. Hanninen A. Akkermansia muciniphila induces gut microbiota remodelling and controls islet autoimmunity in nod mice. Gut. 2018;67(8):1445–1453.
    1. Chelakkot C. Akkermansia muciniphila-derived extracellular vesicles influence gut permeability through the regulation of tight junctions. Exp Mol Med. 2018;50(2):e450.
    1. Ni Y. Characteristics of gut microbiota and its response to a Chinese herbal formula in elder patients with metabolic syndrome. Drug Discov Ther. 2018;12(3):161–169.
    1. Martinic A. Supplementation of lactobacillus plantarum improves markers of metabolic dysfunction induced by a high fat diet. J Proteome Res. 2018;17(8):2790–2802.
    1. Lee E. Lactobacillus plantarum strain Ln4 attenuates diet-induced obesity, insulin resistance, and changes in hepatic mRNA levels associated with glucose and lipid metabolism. Nutrients. 2018;10(5) pii: E643.
    1. Balakumar M. Improvement in glucose tolerance and insulin sensitivity by probiotic strains of Indian gut origin in high-fat diet-fed C57BL/6J mice. Eur J Nutr. 2018;57(1):279–295.
    1. Okubo T. KK/Ta mice administered lactobacillus plantarum strain no. 14 have lower adiposity and higher insulin sensitivity. Biosci Microbiota Food Health. 2013;32(3):93–100.
    1. Fak F, Backhed F. Lactobacillus reuteri prevents diet-induced obesity, but not atherosclerosis, in a strain dependent fashion in Apoe-/- mice. PLoS ONE. 2012;7(10):e46837.
    1. Naito E. Beneficial effect of oral administration of lactobacillus casei strain shirota on insulin resistance in diet-induced obesity mice. J Appl Microbiol. 2011;110(3):650–657.
    1. Park DY. Supplementation of lactobacillus curvatus HY7601 and lactobacillus plantarum KY1032 in diet-induced obese mice is associated with gut microbial changes and reduction in obesity. PLoS ONE. 2013;8(3):e59470.
    1. Yun S.I., Park H.O., Kang J.H. Effect of lactobacillus gasseri BNR17 on blood glucose levels and body weight in a mouse model of type 2 diabetes. J Appl Microbiol. 2009;107(5):1681–1686.
    1. Dang F. Administration of lactobacillus paracasei ameliorates type 2 diabetes in mice. Food Funct. 2018;9(7):3630–3639.
    1. Park K.Y., Kim B., Hyun C.K. Lactobacillus rhamnosus GG improves glucose tolerance through alleviating er stress and suppressing macrophage activation in db/db mice. J Clin Biochem Nutr. 2015;56(3):240–246.
    1. Lim S.M. Lactobacillus sakei OK67 ameliorates high-fat diet-induced blood glucose intolerance and obesity in mice by inhibiting gut microbiota lipopolysaccharide production and inducing colon tight junction protein expression. Nutr Res. 2016;36(4):337–348.
    1. Sabico S. Effects of a multi-strain probiotic supplement for 12 weeks in circulating endotoxin levels and cardiometabolic profiles of medication naive T2DM patients: a randomized clinical trial. J Transl Med. 2017;15(1):249.
    1. Mazloom Z., Yousefinejad A., Dabbaghmanesh M.H. Effect of probiotics on lipid profile, glycemic control, insulin action, oxidative stress, and inflammatory markers in patients with type 2 diabetes: a clinical trial. Iran J Med Sci. 2013;38(1):38–43.
    1. Ivey K.L. The effects of probiotic bacteria on glycaemic control in overweight men and women: a randomised controlled trial. Eur J Clin Nutr. 2014;68(4):447–452.
    1. Ejtahed H.S. Probiotic yogurt improves antioxidant status in type 2 diabetic patients. Nutrition. 2012;28(5):539–543.
    1. Moroti C. Effect of the consumption of a new symbiotic shake on glycemia and cholesterol levels in elderly people with type 2 diabetes mellitus. Lipids Health Dis. 2012;11:29.
    1. Kijmanawat A. Effects of probiotic supplements on insulin resistance in gestational diabetes mellitus: a double-blind randomized controlled trial. J Diabetes Investig. 2019;10(1):163–170.
    1. Asemi Z. Effect of multispecies probiotic supplements on metabolic profiles, hs-CRP, and oxidative stress in patients with type 2 diabetes. Ann Nutr Metab. 2013;63(1–2):1–9.
    1. Tajabadi-Ebrahimi M. A randomized controlled clinical trial investigating the effect of synbiotic administration on markers of insulin metabolism and lipid profiles in overweight type 2 diabetic patients with coronary heart disease. Exp Clin Endocrinol Diabetes. 2017;125(1):21–27.
    1. Hulston C.J., Churnside A.A., Venables M.C. Probiotic supplementation prevents high-fat, overfeeding-induced insulin resistance in human subjects. Br J Nutr. 2015;113(4):596–602.
    1. Mohamadshahi M. Effects of probiotic yogurt consumption on inflammatory biomarkers in patients with type 2 diabetes. Bioimpacts. 2014;4(2):83–88.
    1. Jung SP. Effect of lactobacillus gasseri BNR17 on overweight and obese adults: a randomized, double-blind clinical trial. Korean J Fam Med. 2013;34(2):80–89.
    1. Brahe L.K. Dietary modulation of the gut microbiota–a randomised controlled trial in obese postmenopausal women. Br J Nutr. 2015;114(3):406–417.
    1. Karlsson Videhult F. Probiotics during weaning: a follow-up study on effects on body composition and metabolic markers at school age. Eur J Nutr. 2015;54(3):355–363.
    1. Feizollahzadeh S. Effect of probiotic soy milk on serum levels of adiponectin, inflammatory mediators, lipid profile, and fasting blood glucose among patients with type II diabetes mellitus. Probiotics Antimicrob Proteins. 2017;9(1):41–47.
    1. Sharafedtinov K.K. Hypocaloric diet supplemented with probiotic cheese improves body mass index and blood pressure indices of obese hypertensive patients–a randomized double-blind placebo-controlled pilot study. Nutr J. 2013;12:138.
    1. Naruszewicz M. Effect of lactobacillus plantarum 299v on cardiovascular disease risk factors in smokers. Am J Clin Nutr. 2002;76(6):1249–1255.
    1. Hutt P. Impact of probiotic lactobacillus plantarum TENSIA in different dairy products on anthropometric and blood biochemical indices of healthy adults. Benef Microbes. 2015;6(3):233–243.
    1. Simon MC. Intake of lactobacillus reuteri improves incretin and insulin secretion in glucose-tolerant humans: a proof of concept. Diabetes Care. 2015;38(10):1827–1834.
    1. Jones M.L. Evaluation of clinical safety and tolerance of a lactobacillus reuteri NCIMB 30242 supplement capsule: a randomized control trial. Regul Toxicol Pharmacol. 2012;63(2):313–320.
    1. Hsieh M.C. The beneficial effects of lactobacillus reuteri ADR-1 or ADR-3 consumption on type 2 diabetes mellitus: a randomized, double-blinded, placebo-controlled trial. Sci Rep. 2018;8(1):16791.
    1. Vajro P. Effects of lactobacillus rhamnosus strain gg in pediatric obesity-related liver disease. J Pediatr Gastroenterol Nutr. 2011;52(6):740–743.
    1. Asemi Z. Effects of synbiotic food consumption on metabolic status of diabetic patients: a double-blind randomized cross-over controlled clinical trial. Clin Nutr. 2014;33(2):198–203.
    1. Asemi Z. Effects of beta-carotene fortified synbiotic food on metabolic control of patients with type 2 diabetes mellitus: a double-blind randomized cross-over controlled clinical trial. Clin Nutr. 2016;35(4):819–825.
    1. Shakeri H. Consumption of synbiotic bread decreases triacylglycerol and VLDL levels while increasing HDL levels in serum from patients with type-2 diabetes. Lipids. 2014;49(7):695–701.
    1. Kobyliak N. Effect of alive probiotic on insulin resistance in type 2 diabetes patients: randomized clinical trial. Diabetes Metab Syndr. 2018;12(5):617–624.
    1. Axling U. Green tea powder and lactobacillus plantarum affect gut microbiota, lipid metabolism and inflammation in high-fat fed C57BL/6J mice. Nutr Metab (Lond) 2012;9(1):105.
    1. Allin KH. Aberrant intestinal microbiota in individuals with prediabetes. Diabetologia. 2018;61(4):810–820.
    1. Egshatyan L. Gut microbiota and diet in patients with different glucose tolerance. Endocr Connect. 2016;5(1):1–9.
    1. Inoue R. Prediction of functional profiles of gut microbiota from 16S rRNA metagenomic data provides a more robust evaluation of gut dysbiosis occurring in Japanese type 2 diabetic patients. J Clin Biochem Nutr. 2017;61(3):217–221.
    1. Aw W., Fukuda S. Understanding the role of the gut ecosystem in diabetes mellitus. J Diabetes Investig. 2018;9(1):5–12.
    1. Shen Z. Insights into roseburia intestinalis which alleviates experimental colitis pathology by inducing anti-inflammatory responses. J Gastroenterol Hepatol. 2018;33(10):1751–1760.
    1. Chang YC. TLR2 and interleukin-10 are involved in bacteroides fragilis-mediated prevention of DSS-induced colitis in gnotobiotic mice. PLoS ONE. 2017;12(7)
    1. Li X. Effects of lactobacillus plantarum CCFM0236 on hyperglycaemia and insulin resistance in high-fat and streptozotocin-induced type 2 diabetic mice. J Appl Microbiol. 2016;121(6):1727–1736.
    1. Chen P. Antidiabetic effect of lactobacillus casei CCFM0412 on mice with type 2 diabetes induced by a high-fat diet and streptozotocin. Nutrition. 2014;30(9):1061–1068.
    1. Dagdeviren S. IL-10 prevents aging-associated inflammation and insulin resistance in skeletal muscle. FASEB J. 2017;31(2):701–710.
    1. Hoffmann T.W. Microorganisms linked to inflammatory bowel disease-associated dysbiosis differentially impact host physiology in gnotobiotic mice. ISME J. 2016;10(2):460–477.
    1. Zhu C. Roseburia intestinalis inhibits interleukin17 excretion and promotes regulatory T cells differentiation in colitis. Mol Med Rep. 2018;17(6):7567–7574.
    1. Wang X. Interleukin-22 alleviates metabolic disorders and restores mucosal immunity in diabetes. Nature. 2014;514(7521):237–241.
    1. Liu WC. Lactobacillus plantarum reverse diabetes-induced Fmo3 and ICAM expression in mice through enteric dysbiosis-related c-Jun NH2-terminal kinase pathways. PLoS ONE. 2018;13(5)
    1. Tian P. Antidiabetic (type 2) effects of lactobacillus G15 and Q14 in rats through regulation of intestinal permeability and microbiota. Food Funct. 2016;7(9):3789–3797.
    1. Sun KY. Lactobacillus paracasei modulates LPS-induced inflammatory cytokine release by monocyte-macrophages via the up-regulation of negative regulators of NF-kappaB signaling in a TLR2-dependent manner. Cytokine. 2017;92:1–11.
    1. Zhang L. Akkermansia muciniphila can reduce the damage of gluco/lipotoxicity, oxidative stress and inflammation, and normalize intestine microbiota in streptozotocin-induced diabetic rats. Pathog Dis. 2018;76(4) fty028.
    1. Wang G. Lactobacillus casei CCFM419 attenuates type 2 diabetes via a gut microbiota dependent mechanism. Food Funct. 2017;8(9):3155–3164.
    1. Singh S. Lactobacillus rhamnosus NCDC17 ameliorates type-2 diabetes by improving gut function, oxidative stress and inflammation in high-fat-diet fed and streptozotocintreated rats. Benef Microbes. 2017;8(2):243–255.
    1. Breyner N.M. Microbial anti-inflammatory molecule (MAM) from faecalibacterium prausnitzii shows a protective effect on DNBS and DSS-Induced colitis model in mice through inhibition of NF-kappaB pathway. Front Microbiol. 2017;8:114.
    1. Inan M.S. The luminal short-chain fatty acid butyrate modulates nf-kappab activity in a human colonic epithelial cell line. Gastroenterology. 2000;118(4):724–734.
    1. Kinoshita M., Suzuki Y., Saito Y. Butyrate reduces colonic paracellular permeability by enhancing PPARgamma activation. Biochem Biophys Res Commun. 2002;293(2):827–831.
    1. Matsuzaki T. Prevention of onset in an insulin-dependent diabetes mellitus model, NOD mice, by oral feeding of lactobacillus casei. APMIS. 1997;105(8):643–649.
    1. Yang Y. Fusobacterium nucleatum increases proliferation of colorectal cancer cells and tumor development in mice by activating toll-like receptor 4 signaling to nuclear Factor-kappaB, and up-regulating expression of microRNA-21. Gastroenterology. 2017;152(4):851–866. e24.
    1. Hall A.B. A novel ruminococcus gnavus clade enriched in inflammatory bowel disease patients. Genome Med. 2017;9(1):103.
    1. Cani PD. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 2007;56(7):1761–1772.
    1. Yoshida N. Bacteroides vulgatus and bacteroides dorei reduce gut microbial lipopolysaccharide production and inhibit atherosclerosis. Circulation. 2018;138(22):2486–2498.
    1. Carlsson A.H. Faecalibacterium prausnitzii supernatant improves intestinal barrier function in mice DSS colitis. Scand J Gastroenterol. 2013;48(10):1136–1144.
    1. Kim S.H. The anti-diabetic activity of bifidobacterium lactis HY8101 in vitro and in vivo. J Appl Microbiol. 2014;117(3):834–845.
    1. Kang JH. Anti-obesity effect of lactobacillus gasseri BNR17 in high-sucrose diet-induced obese mice. PLoS ONE. 2013;8(1):e54617.
    1. Miao J. Flavin-containing monooxygenase 3 as a potential player in diabetes-associated atherosclerosis. Nat Commun. 2015;6:6498.
    1. Li X. Effects of lactobacillus casei CCFM419 on insulin resistance and gut microbiota in type 2 diabetic mice. Benef Microbes. 2017;8(3):421–432.
    1. Zhang Y. Lactobacillus casei reduces susceptibility to type 2 diabetes via microbiota-mediated body chloride ion influx. Sci Rep. 2014;4:5654.
    1. Neyazi N. Potential efficacy of lactobacillus casei IBRC_M10711 on expression and activity of insulin degrading enzyme but not insulin degradation. In Vitro Cell Dev Biol Anim. 2017;53(1):12–19.
    1. Allin KH, Nielsen T, Pedersen O. Mechanisms in endocrinology: gut microbiota in patients with type 2 diabetes mellitus. Eur J Endocrinol. 2015;172(4):R167–R177.
    1. Arora T., Backhed F. The gut microbiota and metabolic disease: current understanding and future perspectives. J Intern Med. 2016;280(4):339–349.
    1. Houmard J.A. Intramuscular lipid oxidation and obesity. Am J Physiol Regul Integr Comp Physiol. 2008;294(4):R1111–R1116.
    1. Gao Z. Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes. 2009;58(7):1509–1517.
    1. den Besten G. Short-Chain fatty acids protect against high-fat diet-induced obesity via a PPARgamma-Dependent switch from lipogenesis to fat oxidation. Diabetes. 2015;64(7):2398–2408.
    1. Moens F., Weckx S., De Vuyst L. Bifidobacterial inulin-type fructan degradation capacity determines cross-feeding interactions between bifidobacteria and faecalibacterium prausnitzii. Int J Food Microbiol. 2016;231:76–85.
    1. Rios-Covian D. Enhanced butyrate formation by cross-feeding between faecalibacterium prausnitzii and bifidobacterium adolescentis. FEMS Microbiol Lett. 2015;362(21)
    1. Whang A, Nagpal R, Yadav H. Bi-directional drug-microbiome interactions of anti-diabetics. EBioMedicine. 2019;39:591–602.
    1. Morgun A. Uncovering effects of antibiotics on the host and microbiota using transkingdom gene networks. Gut. 2015;64(11):1732–1743.
    1. Rodrigues R.R. Antibiotic-Induced alterations in gut microbiota are associated with changes in glucose metabolism in healthy mice. Front Microbiol. 2017;8:2306.
    1. Maier L. Extensive impact of non-antibiotic drugs on human gut bacteria. Nature. 2018;555(7698):623–628.
    1. Klaassen CD, Cui JY. Review: mechanisms of how the intestinal microbiota alters the effects of drugs and bile acids. Drug Metab Dispos. 2015;43(10):1505–1521.
    1. Stenman L.K. Probiotic B420 and prebiotic polydextrose improve efficacy of antidiabetic drugs in mice. Diabetol Metab Syndr. 2015;7:75.
    1. Reimer R.A. Combining sitagliptin/metformin with a functional fiber delays diabetes progression in zucker rats. J Endocrinol. 2014;220(3):361–373.
    1. Zheng J. Prebiotic mannan-oligosaccharides augment the hypoglycemic effects of metformin in correlation with modulating gut microbiota. J Agric Food Chem. 2018;66(23):5821–5831.
    1. Sanna S. Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases. Nat Genet. 2019;51(4):600–605.

Source: PubMed

3
Suscribir