当前位置:高等教育资讯网  >  中国高校课件下载中心  >  大学文库  >  浏览文档

《微生物学》课程教学资源(前沿进展)“神药”二甲双胍降糖之谜,肠道微生物或能解答(英文原文)Metformin Is Associated With Higher Relative Abundance of Mucin-Degrading Akkermansia muciniphila and Several Short-Chain Fatty Acid–Producing Microbiota in the Gut

资源类别:文库,文档格式:PDF,文档页数:9,文件大小:1.12MB,团购合买
点击下载完整版文档(PDF)

DiabetesCare Metformin Is Associated With acobo de lo Cuesta-Zuluaga, Higher Relative abundance of tes-Agudelo, Eliang p velasquez-Meiig Mucin-Degrading akkermansia 从d muciniphila and Several Short- Chain Fatty Acid-Producing Microbiota in the Gut D0t:10.2337/dc16-1324 OBJECTIVE t the beneficial effects of metformin on glu memicroballymeddWemnd tection of type2dabetes metformin,and gut microbiota in community-dwelling Colombian adults.On the basis of previous research,we hypoth obiogaofshortehn sized that metformin is associated with fatty acid(SCFA)-producing and m cin-degrading RESEARCH DESIGN AND METHODS Participants were selected from a larger cohort of 459 participants.The present analyses focus on the 28 participants diagnosed with diabetes-14 taking metformin- and the 84 participants without diabetes (3-to-1)to P pants with di betes by s age.am We measured de analyze the composition and structure of the gut microbiota. RESULTS We found an association between diabetes and gut microbiota that was modified by metformin use nts with participants known for m rin de adation and s biota known for production of SCFAs,including Butyrivib Bifid acterium bifidum,Megasph and an ope rati n out diabetes pa 20 June 2016 and accepted 27 Septem 02dos and a distinct pperational taxonomis unit of Prevptell anda lowe r206 abundance of Enterococcus casseliflavus. CONCLUSIONS 2016 by the A Our results support the hypothesis that metformin shifts gut microbiota compo 北as ng as the wo n through the enr n-degrading A.n ld as w for profit.。 shifts mediate met min's glyc emic and anti-infammatory p le ot http://www. Diabetes Care Publish Ahead of Print,published online November 14,2016

Metformin Is Associated With Higher Relative Abundance of Mucin-Degrading Akkermansia muciniphila and Several Short￾Chain Fatty Acid–Producing Microbiota in the Gut DOI: 10.2337/dc16-1324 OBJECTIVE Recent studies suggest the beneficial effects of metformin on glucose metabolism may be microbially mediated. We examined the association of type 2 diabetes, metformin, and gut microbiota in community-dwelling Colombian adults. On the basis of previous research, we hypothesized that metformin is associated with higher levels of short-chain fatty acid (SCFA)–producing and mucin-degrading microbiota. RESEARCH DESIGN AND METHODS Participants were selected from a larger cohort of 459 participants. The present analyses focus on the 28 participants diagnosed with diabetesd14 taking metformind and the 84 participants without diabetes who were matched (3-to-1) to participants with diabetes by sex, age, and BMI. We measured de￾mographic information, anthropometry, and blood biochemical parameters and collected fecal samples from which we performed 16S rRNA gene sequencing to analyze the composition and structure of the gut microbiota. RESULTS We found an association between diabetes and gut microbiota that was modified by metformin use. Compared with participants without diabetes, participants with diabetes taking metformin had higher relative abundance of Akkermansia muciniphila, a microbiota known for mucin degradation, and several gut micro￾biota known for production of SCFAs, including Butyrivibrio, Bifidobacterium bifidum, Megasphaera, and an operational taxonomic unit of Prevotella. In contrast, compared with participants without diabetes, participants with diabetes not taking metformin had higher relative abundance of Clostridiaceae 02d06 and a distinct operational taxonomic unit of Prevotella and a lower abundance of Enterococcus casseliflavus. CONCLUSIONS Our results support the hypothesis that metformin shifts gut microbiota compo￾sition through the enrichment of mucin-degrading A. muciniphila as well as sev￾eral SCFA-producing microbiota. Future studies are needed to determine if these shifts mediate metformin’s glycemic and anti-inflammatory properties. 1 VidariumdNutrition, Health and Wellness Re￾search Center, Grupo Empresarial Nutresa, Me￾dellin, Colombia 2 Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 3 Dinamica I.P.S. ´ dEspecialista en Ayudas Diag￾nosticas, Medellin, Colombia ´ 4 EPS y Medicina Prepagada Suramericana S.A., Medellin, Colombia Corresponding author: Juan S. Escobar, jsescobar@ serviciosnutresa.com. Received 20 June 2016 and accepted 27 Septem￾ber 2016. This article contains Supplementary Data online at http://care.diabetesjournals.org/lookup/ suppl/doi:10.2337/dc16-1324/-/DC1. © 2016 by the American Diabetes Association. Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered.More infor￾mation is available at http://www.diabetesjournals .org/content/license. Jacobo de la Cuesta-Zuluaga,1 Noel T. Mueller,2 Vanessa Corrales-Agudelo,1 Eliana P. Velasquez-Mej ´ ´ıa,1 Jenny A. Carmona,3 Jose M. Abad, ´ 4 and Juan S. Escobar1 Diabetes Care 1 EMERGING TECHNOLOGIES AND THERAPEUTICS Diabetes Care Publish Ahead of Print, published online November 14, 2016

2 Type 2 Diabetes,Metformin,and Gut Microbiota Diabetes Car (11-13).a mucin-degrading bacteria nt.and individuals diae their associated genes(microbiome) with Alzheimer disease,Parkinson dis ute the T our env In numans,parti r any other ne rative d ved to deterministic role in had similar abundance of Subdoliaran and gastrointestinal diseases (crohn dis lum and Akkerm ansi ease.ulcerative colitis short bo wel syn of type .or diah globe,has been linked in nonhuman (1) may help ameliorate a type 2 diabetes dance with the principles of the Dec ation of Helsink the nost rerent hu takir elative abundance of Adlercreutzia(17). and 008430 of 1993).All of the partici don was bn a poten metage ants were th nt (6) and pron e assured of anonymity and conf Metformi 1.1 imethylbigua dia ontrast tiality nfor wa th ith with had Th (7).and findings from recent studi dance of fubacte nmittee of Sede de cio t ma o pre 53 17 min has nleiot ic effe vet the Subdoli anulum and a and an ved the r s de echa nist cluster of butyrate- oducing Clostr 588 ings pro alters hepatic glucose production via to the antidiabe effects of metformin metric,Clinical,and Dietary Evalu through 03 that mucin we BM (kg)/heigh esi the gut (11 n this lean (18.5 BMI ke/m 509% eralizability of ght B/m ng t inte inal an -30 21 ation of metformin is 300 times that of of HDI IDI VIDI total choles dysbiosis in a colombian adult popula iglycerides,apolipoprotein B asting the plasma (16),mal on.Given the conside ariation in etes and that the obtained (collection and m administration of metformin lombians is differen to tha of othe nt explained in Supplemen y Data a14 518 hyp n the 24-h 。e throug ns a Data) al me ormin a rogates in s ferent to those obs and 2 ction and enc effect of metfo nin enrichment of mucin-degrading and scFA Each participant collected their own fe- ela producing micro ealed,ste the RESEARCH DESIGN AND METHODS receptacle ffect than imm r ext Study Design erated in h sehold freezers and brough e July and N ember 2 ty in ea xposure than the other metformin for ld with BMI =18 5 kg/m2 living in sively in the morning (6 AM-12 PM) mulations(15 the Colom of M n,bt ted the anima 11 2,1 16171 nlled in the dav ay that metformin may partially restore sured by the health insu rance prov ide samples were stored on dry ice and ith type next-da formin treatment increased the relativ individual who consu anti evaluated by trained lab abundance of Akkermansia muciniphilo biotics or antiparasitics <3 months prior oratory technicians

Microbial communities (microbiota) and their associated genes (microbiome) constitute the interface of our environs and our cells, and their composition is believed to play a deterministic role in human health and disease. In particular, the development of type 2 diabetes, a disease rising in prevalence around the globe, has been linked in nonhuman (1) and human (2–5) studies to imbalances in microbiota of the intestinal tract (gut). However, the most recent human study on this topic found that the asso￾ciation was modulated in a potent￾ially beneficial manner by metformin treatment (6). Metformin (1,1-dimethylbiguanide hydrochloride) is the most frequent medication used to treat type 2 diabetes (7), and findings from recent studies suggest it may also prevent cancer (7) and cardiovascular events (8). Metfor￾min has pleiotropic effects, yet the majority of mechanistic studies have focused on changes in liver function (7,9,10). Although metformin certainly alters hepatic glucose production via effects on AMPK, there is growing evi￾dence that the genesis of its action is in the gut (11–15). Metformin is ;50% bioavailable, al￾lowing for near-equal intestinal and plasma exposure, but intestinal accumu￾lation of metformin is 300 times that of the plasma (16), making the gut the pri￾mary reservoir for metformin in humans. Unlike oral administration, intravenous administration of metformin in humans does not improve glycemia (14). More￾over, in mice, oral administration of a broad-spectrum antibiotic cocktail with oral metformin abrogates metformin’s glucose-lowering effect (12). Providing yet further evidence that the glucose￾lowering effect of metformin may originate in the lower bowel, a delayed-release oral metformin, which targets the ileum, had a similar or greater glucose-lowering effect than immediate-release or extended￾release metformin, despite the delayed￾release metformin having lower systemic exposure than the other metformin for￾mulations (15). Recent studies in animals (11,12,13) and humans (6,17) provide evidence that metformin may partially restore gut dysbiosis associated with type 2 di￾abetes. In mice fed a high-fat diet, met￾formin treatment increased the relative abundance of Akkermansia muciniphila (11–13), a mucin-degrading bacteria that has been shown to reverse meta￾bolic disorders (1,12). In humans, partic￾ipants with diabetes taking metformin had similar abundance of Subdoligranu￾lum and, to some extent, Akkermansia compared with control subjects without diabetes, suggesting that metformin may help ameliorate a type 2 diabetes– associated gut microbiome (6). It has also been shown that people with diabetes taking metformin had a higher relative abundance of Adlercreutzia (17), and metagenomic functional analyses dem￾onstrated significantly enhanced butyrate and propionate production in people with diabetes using metformin (6). In contrast, people with diabetes who were not treat￾ed with metformin had a higher abun￾dance of Eubacterium and Clostridiaceae SMB53 (17) and lower levels of short￾chain fatty-acid (SCFA)–producers, such as Roseburia, Subdoligranulum, and a cluster of butyrate-producing Clostri￾diales (6). These findings provide evi￾dence that gut microbes may contribute to the antidiabetes effects of metformin through pathways that include mucin degradation and SCFA production. In this study we aimed to test the gen￾eralizability of previous observations con￾cerning the influence of metformin on the association of type 2 diabetes and gut dysbiosis in a Colombian adult popula￾tion. Given the considerable variation in the microbiota associated with type 2 di￾abetes and that the gut microbiota of Co￾lombians is different to that of other populations (18), we hypothesized that the microbial taxa involved in the type 2 diabetes dysbiosis of Colombians are dif￾ferent to those observed in Chinese and European populations (2,3) but that the effect of metformin is similar, i.e., through enrichment of mucin-degrading and SCFA￾producing microbiota. RESEARCH DESIGN AND METHODS Study Design Between July and November 2014, we en￾rolled 459 men and women 18–62 years old, with BMI $18.5 kg/m2 , living in the Colombian cities of Medellin, Bogota, Barranquilla, Bucaramanga, and Cali. All participants enrolled in the study were in￾sured by the health insurance provider EPS y Medicina Prepagada Suramericana S.A. (EPS SURA). We excluded pregnant women, individuals who consumed anti￾biotics or antiparasitics ,3 months prior to enrollment, and individuals diagnosed with Alzheimer disease, Parkinson dis￾ease, or any other neurodegenerative dis￾eases; current or recent cancer (,1 year); and gastrointestinal diseases (Crohn dis￾ease, ulcerative colitis, short bowel syn￾drome, diverticulosis, or celiac disease). This study was conducted in accor￾dance with the principles of the Decla￾ration of Helsinki, as revised in 2008, and had minimal risk according to the Colombian Ministry of Health (Resolution 008430 of 1993). All of the partici￾pants were thoroughly informed about the study and procedures. Participants were assured of anonymity and confi- dentiality. Written informed consent was obtained from all the participants before beginning the study. The Bioethics Committee of Sede de Investigacion´ UniversitariadUniversity of Antioquia re￾viewed the protocol and the consent forms and approved the procedures de￾scribed here (approbation act 14–24–588 dated 28 May 2014). Anthropometric, Clinical, and Dietary Evaluations We calculated BMI as weight (kg)/height squared (m2 ) to classify participants as lean (18.5 # BMI , 25.0 kg/m2 ), over￾weight (25.0 # BMI , 30.0 kg/m2 ), or obese (BMI $30 kg/m2 ). In addition, val￾ues of HDL, LDL, VLDL, total cholesterol, triglycerides, apolipoprotein B, fasting glucose, glycated hemoglobin (HbA1c), fasting insulin, adiponectin, and hs-CRP were obtained (collection and measure￾ment explained in Supplementary Data). Dietary intake was evaluated through 24-h dietary recalls (see Supplementary Data). DNA Extraction and Sequence Analysis Each participant collected their own fe￾cal sample in a hermetically sealed, ster￾ile receptacle provided by the research team. Samples were immediately refrig￾erated in household freezers and brought to an EPS SURA facility in each city within 12 h; receipt of samples occurred exclu￾sively in the morning (6 A.M.–12 P.M.). As such, stools were collected between the evening of the day before and the morn￾ing of the day of sample receipt. Fecal samples were stored on dry ice and sent to a central laboratory via next-day delivery. Before DNA extraction, stool consistency was evaluated by trained lab￾oratory technicians. 2 Type 2 Diabetes, Metformin, and Gut Microbiota Diabetes Care

care.diabete de la Cuesta-Zuluaga and Associates sing 14 with ty (Qiagen,Hilden,Germany)following metformin (T2D-met"),14 with type 2 di- plemented in the Vegan package of R(22). the es (ND (T2-met) We ine be ingstep with the lysis buffer (0at agnostically identify microbial bic 15 Hz using a stainless stee with a stical An markers.LEfS e uses the nonparam d clinical variables ified DNA e compared across study groups us e noDrop spectrophotometer (Ny ing ANOVA and t tests after check ing fo differential abundance amon ote ns,Fra nce)and sent it to th par then p of Michigan Medical School (Ann Art ants using the unpaired wil MI).The V4 hypervariable region of the lity tests sum test,and final lly uses LD est transformed (natural log for unco and R806 (S'-GGACTACHVGGGTWTC ined variables or ar quare roo of lEfse com with sta statis -3)p rs and equenced sing for p rati tica approaches is that, y and the Statistical analyses were formed ing strategy(19).n addition t with Rv..3.2.2(21】 een each OTU and the grou g cat we sequ the QIamp DNA S 's EB el median 28,699).To limit the effects o nicrobial biomarkers in ou tio an hhe da etained f they had a A 0.05 and 0g10 D M e 12D-me na ment's run. quences were curated ugh Finally,across groups,we tested for dif foll ving th eg sta a erences in e the likeli 13620 of detecting false positives.especially maior butyrate. microbia seq tedat the NCBIand mic units a,including Butyrivibni can be th he a nd com as assessed by quantifying and ssified in each of thes ion of Type 2 iab and preting (4 for A.muciniphil 11 for that had typedi;6self-reported and the erence using ANOVA nd t te te t n We compared the nine whet (fasting bloc 2126 mg/d ne analysis。 bunda of the 28 only ir formin treatment,14 not (1 was Diversity using Results from lEfSe and from the pooled treat with insutin alo with gli d g mu of ph otype e corrected fo logical treat nent for =0 5)calcr including the 2 participants unaware of ated with the GUniF ge ofR(23 age of R(25).Tests were considered betes status)(supplementary a P value. We matched each participant with di nce per OTU.then obtained a dis with thr matrix from pairwis (to th finally eda rela cf20nerP0iemtheohg ndrNSteristiG difference between case and control joining phylogenetic tree using Mothur pants.There were no statistically signif mong en es (a ght or ohese)This left us with atotal sing the adonis function (ANOVA using cal narameters hetw en T2D-met*and

Total microbial DNA was extracted using the QIAamp DNA Stool Mini Kit (Qiagen, Hilden, Germany) following the manufacturer’s instructions, with a slight modification consisting in a bead￾beating step with the lysis buffer (20 s at 15 Hz using a stainless steel bead with a 5-mm diameter). After extraction, we quantified DNA concentration using a NanoDrop spectrophotometer (Nyxor Biotech, Paris, France) and sent it to the Microbial Systems Laboratory, University of Michigan Medical School (Ann Arbor, MI). The V4 hypervariable region of the 16S rRNA gene was amplified using the F515 (59-CACGGTCGKCGGCGCCATT-39) and R806 (59-GGACTACHVGGGTWTC TAAT-39) primers and sequenced using the Illumina MiSeq sequencing platform with V2 chemistry and the dual-index sequencing strategy (19). In addition to DNA from fecal samples, we sequenced negative controls (ultrapure water and the QIAamp DNA Stool Mini Kit’s EB elu￾tion buffer), a DNA extraction blank, and a mock community (HM-782D, BEI Resources, Manassas, VA) in each instru￾ment’s run. Sequences were curated following the MiSeq standard operating procedure implemented by Mothur v.1.36 (20) (see Supplementary Data). Raw sequences were deposited at the NCBI and can be accessed through the BioProject (accession number PRJNA325931). Definition of Type 2 Diabetes and Selection of Control Subjects We identified 28 participants in our study that had type 2 diabetes; 26 self-reported physician-diagnosed diabetes prior to the beginning of the study and 2 were diagnosed through laboratory testing (fasting blood glucose $126 mg/dL and HbA1c$6.5%). Of the 28 participants with type 2 diabetes, 14 were under met￾formin treatment, 14 were not (1 was treated with insulin alone, 2 with gliben￾clamide, and 11 were under no pharmaco￾logical treatment for type 2 diabetes, including the 2 participants unaware of their diabetes status) (Supplementary Table 1). We matched each participant with di￾abetes with three participants without diabetes in our study based on sex, age (to the closest possible age; maximum difference between case and control subjects 6 years; mean 1.5 years; median 1 year), and BMI category (lean, over￾weight, or obese). This left us with a total analytic sample of 112 study participants comprising 14 with type 2 diabetes using metformin (T2D-met+ ), 14 with type 2 di￾abetes not using metformin (T2D-met2), and 84 without diabetes (ND). Statistical Analyses Anthropometric and clinical variables were compared across study groups us￾ing ANOVA and t tests after checking for homoscedasticity and normal distribu￾tion of residuals (using Fligner-Killeen tests of homogeneity of variances and Shapiro-Wilk normality tests). When necessary, variables were appropriately transformed (natural log for uncon￾strained variables or arcsin square root for proportions). Sex ratio and stool con￾sistency were compared using x2 tests. Statistical analyses were performed with R v.3.2.2 (21). Curated DNA sequences ranged from 69 to 102,660 sequences per sample (median 28,699). To limit the effects of uneven sampling, we rarefied the data set to 4,091 sequences per sample, result￾ing in the exclusion of one T2D-met2 par￾ticipant with 69 reads. Although rarefaction may lead to missing low-abundance data, it is a powerful way to reduce the likelihood of detecting false positives, especially among those operational taxonomic units (OTUs) with very low abundance. The gut microbiota structure and com￾position was assessed by quantifying and interpreting similarities based on intra￾and intergroup diversity analyses (a and b diversity, respectively). For a diversity, we calculated Good’s coverage and the number of OTUs of each sample using Mothur and constructed rarefaction curves.We compared these indices among groups of participants using analysis of similarity (ANOSIM) with 1,000 permuta￾tions using the Vegan package of R (22). b Diversity was assessed using phylogeny-based generalized UniFrac dis￾tances (with the a parameter controlling weight on abundant lineages = 0.5) calcu￾lated with the GUniFrac package of R (23). For this, we first reduced the alignment and the OTU table to one representative sequence per OTU, then obtained a dis￾tance matrix from uncorrected pairwise distances between aligned sequences, and finally constructed a relaxed neighbor￾joining phylogenetic tree using Mothur and Clearcut. Comparisons among groups of participants were performed using the adonis function (ANOVA using distance matrices) of the permutational multivariate ANOVA (PERMANOVA) im￾plemented in the Vegan package of R (22). We next used linear discriminant analysis (LDA) effect size (LEfSe) (24) to agnostically identify microbial bio￾markers. LEfSe uses the nonparametric factorial Kruskal-Wallis sum rank test to detect individual OTUs with significant differential abundance among groups of participants, then performs a set of pairwise tests among groups of partici￾pants using the unpaired Wilcoxon rank sum test, and finally uses LDA to esti￾mate the effect size of each differen￾tially abundant OTU (24). The strength of LEfSe compared with standard statis￾tical approaches is that, in addition to providing P values, it provides an estima￾tion of the magnitude of the association between each OTU and the grouping cat￾egories (e.g., metformin, type 2 diabetes) through the LDA score. For stringency, microbial biomarkers in our study were retained if they had a P , 0.05 and a (log10) LDA score $3, i.e., one order of magnitude greater than LEfSe’s default. Finally, across groups, we tested for dif￾ferences in relative abundance of the mucin-degrading A. muciniphila and major butyrate-producing microbial genera, including Butyrivibrio, Roseburia, Subdoligranulum, and Faecalibacterium. For this analysis, we pooled all OTUs classified in each of these phylotypes (4 for A. muciniphila, 11 for Butyrivibrio, 4 for Roseburia, 10 for Subdoligranulum, and 5 for Faecalibacterium) and tested for differences using ANOVA and t tests on arcsin square root transformed relative abundances. This pooling served to examine whether differences in relative abundance of these groups of bacteria occurred across all OTUs or only in specific OTUs. Results from LEfSe and from the pooled analysis of phylotypes were corrected for multiple testing using the Bayesian ap￾proach implemented in the qvalue pack￾age of R (25). Tests were considered significant if they had a P value # 0.05 and a q value #0.1. RESULTS In Table 1 we present the characteristics of T2D-met+ , T2D-met2, and ND partic￾ipants. There were no statistically signif￾icant differences (all P values . 0.10) in demographic, anthropometric, or clini￾cal parameters between T2D-met+ and care.diabetesjournals.org de la Cuesta-Zuluaga and Associates 3

4 Type 2 Diabetes,Metformin,and Gut Microbiota Diabetes Care 18g8 8昌888838昌888 3日888B言昌君目8昌日 西始器然路1111 用Eea 888器111 日g 合器器阳9 woy sanjeA d llV.'OS ueaw se pal

Table 1 —General characteristics of T2D-met+, T2D-met2, and ND participants among community-dwelling Colombian adults Group P* T2D-met+ T2D-met2 ND T2D-met+ vs. T2D-met2 T2D-met+ vs. ND T2D-met2 vs. ND n 14 14 84 d dd Age (years) 50 6 10 44 6 9 47 6 9 0.11 0.33 0.24 Sex (F/M) 0.36 0.50 0.43 0.70 0.83 0.84 Anthropometry BMI (kg/m2 ) 31.88 6 4.63 32.15 6 6.36 31.11 6 4.53 0.98 0.56 0.62 Body fat (%) 0.41 6 0.05 0.41 6 0.03 0.40 6 0.04 0.96 0.68 0.58 Waist circumference (cm) 104.6 6 9.8 102.8 6 14.2 102.0 6 11.3 0.70 0.39 0.85 Clinical parameters Total cholesterol (mg/dL) 178 6 51 208 6 44 187 6 30 0.11 0.54 0.10 HDL (mg/dL) 40 6 11 38 6 6 44 6 12 0.77 0.21 0.0136 LDL (mg/dL) 105 6 35 128 6 39 115 6 28 0.11 0.31 0.26 VLDL (mg/dL) 38 6 40 52 6 52 31 6 15 0.19 0.64 0.0471 Apolipoprotein B (mg/dL) 102 6 30 108 6 30 97 6 26 0.56 0.55 0.18 Triglycerides (mg/dL) 176 6 202 244 6 253 154 6 75 0.15 0.97 0.07 Fasting glucose (mg/dL) 127 6 47 145 6 76 90 6 10 0.57 0.0047 0.0060 HbA1c [% (mmol/mol)] 6.9 6 1.4 (52.0 6 15.3) 7.1 6 1.7 (54.0 6 18.6) 5.6 6 0.3 (38.0 6 3.3) 0.78 0.0026 0.0047 Fasting insulin (mU/mL) 22.24 6 12.58 24.24 6 12.86 15.20 6 8.79 0.49 0.11 0.0029 Insulin resistance index 2.9 6 1.5 3.7 6 3.0 1.9 6 1.1 0.39 0.0420 0.0018 Leptin (ng/mL) 7.39 6 6.09 8.34 6 5.40 7.89 6 6.14 0.38 0.91 0.25 Adiponectin (mg/mL) 4.59 6 1.97 4.96 6 3.22 6.79 6 3.90 0.91 0.0195 0.07 hs-CRP (mg/L) 2.70 6 2.53 3.35 6 2.76 4.04 6 5.02 0.61 0.21 0.62 Dietary intake Energy intake (calories) 1,843 6 268 1,865 6 584 1,945 6 572 0.75 0.61 0.57 Carbohydrate (g) 250 6 37 260 6 86 267 6 88 0.92 0.52 0.68 Protein (g) 74.7 6 9.1 69.7 6 13.1 74.1 6 13.8 0.26 0.85 0.26 Fat (g) 60.7 6 13.0 60.2 6 21.3 62.9 6 17.1 0.64 0.76 0.51 Cholesterol (mg) 336 6 31 330 6 41 347 6 38 0.67 0.28 0.18 Dietary fiber (g) 17.9 6 4.8 18.6 6 6.4 17.5 6 4.8 0.88 0.73 0.64 Stool consistency [n (%)] 0.38 0.58 0.50 Hard 4 (28) 2 (14) 13 (15) d d d Normal 8 (57) 9 (64) 54 (64) d dd Mushy 2 (14) 1 (7) 13 (15) d dd Diarrheic 0 (0) 2 (14) 4 (5) d dd Values presented as mean 6 SD. *All P values from t tests except in sex and stool consistency (x2 tests). 4 Type 2 Diabetes, Metformin, and Gut Microbiota Diabetes Care

care.diabetesjo nals.org de la Cuesta-Zuluaga and Associates 5 T2D R2=0019 had higher fasting glucose,HbAand in- p=0.34 rent T20 16S rRNA Gene Sequencing e ND 009 g (mean Good's cov e±5D=09901 PCo1(10.34% 0.001);9%of OTUs were detected at ast bych biota.We next tested for ween participants with 20 diabetes and ND participants(A OSIM 2519 ipants (ANOSIM statistic R=-0.018,P= 0.557 The numbe of obse ved O met"and ND than bet n T2D-met (Supplementary Fig ce- PCo1(10.34% ANOSIM statistic R=0.018,P=0.348; :ANOSIM statistic R- We c s (PE 2D-me A) with dia nd ND partic =0.009,P=0.416)(Fg18) ever,the com significance (R2=0.013,P=0.036)(Fig. C).the PCo1(10.34%) De0036 0.01 T2D-met ND Participants (R 0.008,P=0.9 43).These results suggested and partici s not takir D the -me (T t-and VA.(A high-quality color represe e in the onine issue.) We next use to examine microbiota at the OTU level.Note that (represented by OTUs with [og10]LDA relevant.OTUs (19 displayed in Fig.2 and

T2D-met2 participants. Compared with ND participants, T2D-met+ participants had higher fasting glucose, HbA1c, and in￾sulin resistance than ND participants and lower levels of the insulin-sensitizing hor￾mone adiponectin (P , 0.05). No other demographic, anthropometric, or clinical parameters were statistically different. 16S rRNA Gene Sequencing Gut microbiota communities were spe￾cific to each participant with marked in￾tersubject differences (Fig. 1A) (overall interindividual generalized UniFrac dis￾tance = 0.720 6 0.009). We found high coverage across all groups of participants (mean Good’s coverage 6 SD = 0.990 6 0.001); 99% of OTUs were detected at least by two DNA reads, demonstrating thorough sampling of the gut micro￾biota. We next tested for differences in the number of observed OTUs across the groups of participants. We found no differences between participants with diabetes and ND participants (ANOSIM statistic R = 0.005, P value = 0.425) or between T2D-met+ and T2D-met2 partic￾ipants (ANOSIM statistic R = 20.018, P = 0.557). The number of observed OTUs tended to be more similar between T2D￾met+ and ND than between T2D-met2 and ND participants (Supplementary Fig. 1); however, these differences were not statistically significant (T2D-met+ vs. ND: ANOSIM statistic R = 0.018, P = 0.348; T2D-met2 vs. ND: ANOSIM statistic R = 0.009, P = 0.409). We observed no significant differences in b diversity estimates among the three groups of participants (PERMANOVA: R2 = 0.019, P = 0.335) (Fig. 1A) or between participants with diabetes and ND partic￾ipants (R2 = 0.009, P = 0.416) (Fig. 1B). However, the comparison between met￾formin and nonmetformin users reached significance (R2 = 0.013, P = 0.036) (Fig. 1C), demonstrating differences in the bacterial community structure associated with metformin use. The difference was also significant when comparing T2D￾met+ and ND participants (R2 = 0.015, P = 0.036) but not when comparing T2D-met2 and ND participants (R2 = 0.008, P = 0.943). These results suggested the microbial communities of T2D-met+ versus T2D-met2 were modestly phylo￾genetically dissimilar. We next used LEfSe to examine differ￾ences in the relative abundance of gut microbiota at the OTU level. Note that we were only interested in OTUs dis￾playing strong associations in the LDA (represented by OTUs with [log10] LDA scores $3); such stringency resulted in fewer retained, but more likely biologically relevant, OTUs (19 displayed in Fig. 2 and PCo2 (6.03%) PCo1 (10.34%) ND R2 = 0.019 P = 0.34 T2D-met+ T2D-met￾PCo2 (6.03%) R2 = 0.009 P = 0.42 ND T2D PCo1 (10.34%) PCo2 (6.03%) met￾R2 = 0.013 P = 0.036 T2D-met+ PCo1 (10.34%) A B C Figure 1—Principal coordinates analysis based on generalized UniFrac. A: Comparison among the three groups of participants. B: Comparison between participants with diabetes and ND partici￾pants. C: Comparison between T2D-met+ and participants not taking metformin (T2D-met2 and ND). Ellipses encompass 75% of data distribution in each group of participants. R2 and P values from PERMANOVA. (A high-quality color representation of this figure is available in the online issue.) care.diabetesjournals.org de la Cuesta-Zuluaga and Associates 5

6 Type 2 Diabetes,Metformin,and Gut Microbiota Diabetes Care A ND T2D-met OTUO2).Baresiellaceae(Bacteroidetes Enterococcus casselifiavus(OTU048 Prevotella (OTU236) OTU080)were enriched in T2D-met compared with T2D-met*participants 3 2 0 1 2 3 and F ND T2D-met' obe we ound that A. (3.4 and 44 times.respectively)in T2D. met*than in T2D-met participants tridium celatum (OTU14 Bulleidia p-1630-c5(OTUO7 Mollicutes RF39(OTUO6 00o,0si0eoh0ofbunotur Mollicutes RF39 (OTU284 2 Bifidobacterium bifidum (OTU176 tdiffer Megasphaera(OTU069 otbutyraiteproducesbeteenRC Prevotella (OTU028) n and nonme Butyrivibrio (OTU062) =0001.p -3 -2 0 3 C T2D-met T2D-met+ any of these groups of bacteria betweer Clostridia tes and ND p nd q values>0.2 Me CONCLUSIONS Prevotel a(0u028 mmnity-ba ple of Colom .3 .1 0 3 tent with previous literature (6.11-13.17 LDA score(log10) that the a association between gut micro of -(C)par higher relative abundance of purportedly netcidlmucin.degradingandSCfA 3 out of 273 statistically significant if two OTUs of Molli metndND participants matchedon age,sex,and BMI. hen co that OTand ND eve es hav o Clostridiac 02d06 (Firmicutes T2D-met'than i ND participants.In tMCobiaacompcsion,buiimd lostridiaceae OTU074)and Prev contras fou OTUs idiales in ngs on the taxa involved have been in e cipants,whereas Enterococcus c 556 SMB53(Firmicutes|Clostridiaceael plained by confounding factors,includ- Ent 0TU026 body weight,and ints (Figs.2A and 3A and B).W After matching on age sex and articipa ts t Ruminoc aelOTU025)were more s on we s(E OTU062).a different OTU of Pre Prevotel a(Bacteroide t microbiota composition.One OTU 02 8)and M was higher among OTU)bifidum (Acti in T2D-met*compared with T2D- other otu related to f casseliflayus was nobacterial BifidobacteriaceaelOTU176). participants,whereas OTUs from lower among participants with diabetes

Fig. 3 out of 273 statistically significant if not taking LDA scores into account). When comparing T2D-met2 and ND par￾ticipants, we found that OTUs belonging to Clostridiaceae 02d06 (Firmicutes| Clostridiaceae|OTU074) and Prevotella (Bacteroidetes|Prevotellaceae|OTU236) were overrepresented in T2D-met2 par￾ticipants, whereas Enterococcus casse￾liflavus (Firmicutes|Enterococcaceae| OTU048) was more abundant in ND par￾ticipants (Figs. 2A and 3A and B). When comparing T2D-met+ participants to ND participants, we found that OTUs of Butyrivibrio (Firmicutes|Lachnospiraceae| OTU062), a different OTU of Prevotella (Bacteroidetes|Prevotellaceae|OTU028), Megasphaera (Firmicutes|Veillonellaceae| OTU069), Bifidobacterium bifidum (Acti￾nobacteria|Bifidobacteriaceae|OTU176), two OTUs ofMollicutes RF39 (Tenericutes| OTU284 and OTU067), and Bulleidia p-1630-c5 (Firmicutes|Erysipelotricha￾ceae|OTU077) were more abundant in T2D-met+ than in ND participants. In contrast, four OTUs of Clostridiales in￾cluding Clostridium celatum (Firmicutes| Clostridiaceae|OTU014), Clostridiaceae SMB53 (Firmicutes|Clostridiaceae| OTU026), Oscillospira (Firmicutes| Ruminococcaceae|OTU024), and Cellu￾losibacter alkalithermophilus (Firmicutes| Ruminococcaceae|OTU025) were more abundant in ND than in T2D-met+ partic￾ipants (Figs. 2B and 3C and D). OTUs from Prevotella (Bacteroidetes|Prevotellaceae| OTU028) and Megasphaera (Firmicutes| Veillonellaceae|OTU069) were enriched in T2D-met+ compared with T2D-met2 participants, whereas OTUs from Oscillospira (Firmicutes|Ruminococcaceae| OTU024), Barnesiellaceae (Bacteroidetes| OTU102), and a different OTU of Clostri￾diaceae 02d06 (Firmicutes|Clostridiaceae| OTU080) were enriched in T2D-met2 compared with T2D-met+ participants (Figs. 2C and 3E and F). Finally, when we pooled mucin￾degrading and butyrate-producing mi￾crobes, we found that A. muciniphila and Butyrivibrio were more abundant (3.4 and 4.4 times, respectively) in T2D￾met+ than in T2D-met2 participants; differences were statistically significant for A. muciniphila (F1, 109 = 9.46, P = 0.003, q value = 0.01) but not for Butyr￾ivibrio (F1, 109 = 3.03, P = 0.08, q value = 0.21) (Fig. 3G and H). There were no sig￾nificant differences in the other groups of butyrate producers between metfor￾min and nonmetformin users (Roseburia: F1, 109 = 1.44, P = 0.23, q value = 0.39; Subdoligranulum: F1, 109 = 0.001, P = 0.97, q value = 0.97; Faecalibacterium: F1, 109 = 0.53, P = 0.47, q value = 0.59). There were no significant differences in any of these groups of bacteria between participants with diabetes and ND partic￾ipants (all P . 0.1 and q values .0.2). CONCLUSIONS In our community-based sample of Colom￾bian adults, we provide evidence consis￾tent with previous literature (6,11–13,17) that the association between gut micro￾biota and type 2 diabetes is modified by metformin use. T2D-met+ participants had higher relative abundance of purportedly beneficial mucin-degrading and SCFA￾producing bacteria compared with T2D￾met2 and ND participants matched on age, sex, and BMI. Several studies have demonstrated that type 2 diabetes is associated with gut microbiota composition, but find￾ings on the taxa involved have been in￾consistent (2–5). Some of the variance in previous study findings may be ex￾plained by confounding factors, includ￾ing demographics, body weight, and treatment with drugs, such as metfor￾min. After matching on age, sex, and BMI and stratifying comparisons on metformin, we found only modest asso￾ciations between type 2 diabetes and gut microbiota composition. One OTU related to Prevotella was higher among participants with diabetes, whereas an￾other OTU related to E. casseliflavus was lower among participants with diabetes. -4 -3 -2 -1 0 1 2 3 4 Butyrivibrio (OTU062) Prevotella (OTU028) Megasphaera (OTU069) Bifidobacterium bifidum (OTU176) Mollicutes RF39 (OTU284) Mollicutes RF39 (OTU067) Bulleidia p-1630-c5 (OTU077) Clostridium celatum (OTU014) Clostridiaceae SMB53 (OTU026) Oscillospira (OTU024) Cellulosibacter alkalithermophilus (OTU025) ND T2D-met+ -4 -3 -2 -1 0 1 2 3 4 Clostridiaceae 02d06 (OTU074) Prevotella (OTU236) Enterococcus casseliflavus (OTU048) ND T2D-met- -4 -3 -2 -1 0 1 2 3 4 Prevotella (OTU028) Megasphaera (OTU069) Oscillospira (OTU024) Barnesiellaceae (OTU102) Clostridiaceae 02d06 (OTU080) T2D-met- T2D-met+ LDA score (log10) B A C Figure 2—LDA scores (log10) of the OTUs displaying differences between pairs of groups of participants. ND vs. T2D-met2 (A), ND vs. T2D-met+ (B), T2D-met+ vs. T2D-met2 (C) participants. 6 Type 2 Diabetes, Metformin, and Gut Microbiota Diabetes Care

care diabetesio als.org de la Cuesta-Zuluaga and Associates 7 0.04 A G H f0.30 0.2 0.1 0.0 -Re of th ng diff ts (logLDA >3).Data p nted n±sE.Ope with ND.D 20- 2-met and NE Prevotello has been associated with A.muciniphila alone may explain the derive from the strengthening of the in carbohydrate degradation beneficial effects of m rmin. estinal mucosal barrie terit of the mucin laver.ther gen that may cause se tions in diabetes demonstrated that stopping ing translocation of proinfammatory 527 ormin treat da ipopolys formin use and eut microbiota com glucag on-like peptide 1(17).A three gucose homeostasis (112).Likewise clmtoCioieiectoalimctagenoic r to T2D s p with SCEA t this bad may bacteria(6).Metformin has also been potentially contributing nce al health in the same way T2D-metparticipants in our study mic effect of metfo min stem from it increases SCFA production through also had higher relative abundance o sition on of t e gut microb ing bac na,but not impacts the metformin alters glucose metabolism metabolism f the microbl ota hosted by through effec on bile aci ( studies have shown that met aforeme uman (11-13)and ment in mice on high-fat diet shifts the human (6,17)studies,metformin use in ticipants),Megasphoera (a bac com rd that o b dy was n g ater re nd Butyri LEfSe biom overy,we also 50n5. mi robiota ave beer fat diets ed with aith rate A.muciniphila or metformin had simila (26,33-35).SCFAs may be beneficial for mprovements in mu dance of the mucin-eading bacteri In-pre and Butyrate,in particu one and glucose tolerance,suggesting that that metformin's health henefits mav lonic epithelium (35).Recent studies in

Prevotella has been associated with carbohydrate-based diets and degradation of complex polysaccharides (26), whereas E. casseliflavus is an opportunistic patho￾gen that may cause serious infections in immunosuppressed individuals (27). We found associations between met￾formin use and gut microbiota composi￾tion that were largely consistent whether we compared T2D-met+ participants to ND control subjects or to T2D-met2 par￾ticipants, suggesting that metformin may have direct microbial effects. Our findings are congruent with multiple lines of evi￾dence indicating the gut-mediated glyce￾mic effect of metformin stem from alterations in the gut microbiota compo￾sition. Cabreiro et al. (28) first demon￾strated that metformin impacts the metabolism of the microbiota hosted by Caenorhabditis elegans. Three mouse studies have shown that metformin treat￾ment in mice on high-fat diet shifts the microbiota composition toward that of mice fed normal chow by increasing abundance of Akkermansia spp. (11–13). Moreover, a follow-up experiment by Shin et al. (12) found that mice fed high￾fat diets treated with either cultured A. muciniphila or metformin had similar improvements in mucin-producing gob￾let cells, proinflammatory interleukin-6, and glucose tolerance, suggesting that A. muciniphila alone may explain the beneficial effects of metformin. In human studies, a small nonrandom￾ized clinical trial of 12 patients with type 2 diabetes demonstrated that stopping metformin treatment for 7 days led to alterations in the gut microbiota and glucagon-like peptide 1 (17). A three￾country cross-sectional metagenomic study found that metformin use was pos￾itively associated with SCFA-producing bacteria (6). Metformin has also been shown to enhance active and total glucagon-like peptide 1 (17,29,30), which is consistent with the hypothesis that it increases SCFA production through modification of the gut microbiota com￾position. There is also the possibility that metformin alters glucose metabolism through effects on bile acid secretion (31). As we hypothesized, on the basis of the aforementioned nonhuman (11–13) and human (6,17) studies, metformin use in our study was associated with greater rel￾ative abundance of the mucin-degrading A. muciniphila. Through LEfSe biomarker discovery, we also found metformin was positively associated with the mucolytic bacterium B. bifidum. The higher abun￾dance of the mucin-degrading bacteria A. muciniphila and B. bifidum in the gut microbiota of metformin users suggests that metformin’s health benefits may derive from the strengthening of the in￾testinal mucosal barrier. A. muciniphila plays a crucial role in maintaining the in￾tegrity of the mucin layer, thereby reduc￾ing translocation of proinflammatory lipopolysaccharides and controlling fat storage, adipose tissue metabolism, and glucose homeostasis (1,12). Likewise, B. bifidum can grow on gastric mucin as a sole carbon source, and genome anal￾ysis revealed that this bacterium can use host mucins (32), potentially contributing to gastrointestinal health in the same way as A. muciniphila. T2D-met+ participants in our study also had higher relative abundance of some SCFA-producing bacteria, but not others (e.g., Roseburia, Subdoligranulum, Faecalibacterium). Those positively as￾sociated with metformin use included B. bifidum, Prevotella (an OTU distinct from the OTU enriched in T2D-met2 par￾ticipants), Megasphaera (a bacterium re￾lated to Megamonas), and Butyrivibrio (although this taxa was no longer signifi- cant after adjusting for multiple compar￾isons). These microbiota have been associated with production of SCFAs, in￾cluding butyrate, propionate, and acetate (26,33–35). SCFAs may be beneficial for health. Butyrate, in particular, is one of the preferred energy sources of the co￾lonic epithelium (36). Recent studies in 0 0.01 0.02 0.03 0.04 Relave abundance 0 0.10 0.20 0.30 AB C D E F GH *** *** ** * ** ** ** ** ** * ** ** ** ** + *** * * * * * Figure 3—Relative abundance of the groups of bacteria displaying differences among participants (logLDA .3). Data presented as mean 6 SE. Open bars = T2D-met+ ; gray bars = T2D-met2; black bars = ND. A: OTUs enriched in T2D-met2 compared with ND. B: OTUs enriched in ND compared with T2D-met2. C: OTUs enriched in T2D-met+ compared with ND. D: OTUs enriched in ND compared with T2D-met+ . E: OTUs enriched in T2D-met+ compared with T2D-met2. F: OTUs enriched in T2D-met2 compared with T2D-met+ . G: Eleven pooled OTUs classified as Butyrivibrio enriched in T2D￾met+ compared with T2D-met2 and ND. H: Four pooled OTUs classified as A. muciniphila enriched in T2D-met+ compared with T2D-met2 and ND (note the change in scale). +P , 0.1; *P , 0.05; **P , 0.05 and q value ,0.1; ***P , 0.05 and q value ,0.05. care.diabetesjournals.org de la Cuesta-Zuluaga and Associates 7

8 Type 2 Diabetes,Metformin,and Gut Microbiota Diabetes Care mice showed that an increase in the co n conclusion.our study of Colombian Prior Presentation.Parts of this study wer onic production of SCFAs,especially bu- adults provides eviden sented at the yrate and pro ery homeostasisand he nentation of mucin-dera glucose production Reference appetite body A.muciniphila as well as several SCFA Acetat ng b lo a mediated by epithelia celland antidiabetes and anti-inflammatory effects hehost agains ,U¥Cai01310906 34 5tud85w90556 ota in type pithelial metabolism and dep intra n of the agenc et a ranscri ng ment Forem nN,Vogens FK,van den Berg FW w levels of butyrate-producing ba ut mic e De n assodated with col on of SCFAs in dis tance (39,40) ta cha study is not without limit ata that cannot nrovide causal infer ng by m Korbonits M.Metfo od microbiota associations were Inst cancer.Nat Rev 010:14 argely consistent wheth event not due to confo t by ind funded by Grupo Em we ca other limitation to our study was the lack 307-32 of information on dose and duration of nl,analysis,orinterpretationotthed 201522 K.Molecula ud hav submit the of action hts? 2013:56:189 21ggrnen or ciation(att ation to ee H s of G En rd the P E 。Future studies are No othe nted to 6 of the tributions.Jd.l.C.Z. essed atealceaPobe revious obs on this an N.T.M (Forslund et a alyzed 4 gu I.Lack type 2diabetes)and thus may have be erp ples t af gon and groed assoctions of microbial et a that were st er in mag rec ou abili ults from udies with our modest samplesi and uscript. thors read 6:3919 20 a different populatio 552 intestine is robust and replicable across diverse populations. ut-based oh

mice showed that an increase in the co￾lonic production of SCFAs, especially bu￾tyrate and propionate, triggers intestinal gluconeogenesis, benefiting glucose and energy homeostasis and reducing hepatic glucose production, appetite, and body weight (37). Acetate produced by bifido￾bacteria improves the intestinal defense mediated by epithelial cells and protects the host against lethal infection (34). Also, SCFAs, particularly butyrate, stimulate epithelial metabolism and deplete intra￾cellular O2, resulting in stabilization of the transcription factor HIF-1 and increasing epithelial barrier function (38). In humans, low levels of butyrate-producing bacteria have been associated with colonic disease (e.g., inflammatory bowel disease), high￾lighting the role of SCFAs in disease resis￾tance (39,40). Our study is not without limitations. Our findings are based on observational data that cannot provide causal infer￾ence. We were able to reduce the po￾tential for confounding by matching on age, sex, and BMI. Because our metformin– gut microbiota associations were largely consistent whether we used as a reference group the T2D-met2 or ND participants, we believe our findings were not due to confounding by indi￾cation. However, we cannot rule out unmeasured or residual confounding. An￾other limitation to our study was the lack of information on dose and duration of metformin treatment. This limitation could have resulted in a weaker, more conservative, association (attenuation to￾ward the null) between metformin and gut microbiota composition and struc￾ture. Future studies are warranted to determine the dose-response of the metformin-microbiota relationship. We also had a small sample size relative to a previous observational study on this topic (Forslund et al. [6] analyzed 784 gut metagenomes of Danish, Swedish, and Chinese participants, of which, 199 had type 2 diabetes) and thus may have been underpowered to detect statistical sig￾nificance for measures of a diversity and associations of microbial composi￾tion that were smaller in magnitude. Nev￾ertheless, our ability to largely confirm hypotheses generated from previous studies with our modest sample size and in a different population suggests that the effect of metformin on the gut microbiota is robust and replicable across diverse populations. In conclusion, our study of Colombian adults provides evidence congruent with the hypothesis that metformin has direct effects on gut microbiota composition through augmentation of mucin-degrading A. muciniphila as well as several SCFA￾producing bacteria. Randomized controlled trials are needed to determine whether the antidiabetes and anti-inflammatory effects of metformin are mediated by the changes to gut microbiota composition. Acknowledgments. Foremost, the authors are indebted to all the participants who agreed to take part in this study. The authors also thank Natalia Zuluaga (Vidarium) for preselection of potential participants; Luz G. Betancur and Natalia E. Guarin (bothofVidarium)forhelpinparticipantrecruitment; ErikaM. Loaiza, Natalia Pareja, D. Tatiana Garcia, and Yuliana M. Franco (of Vidarium) for their invaluable help during field work; Amalia Toro, Connie J. Arboleda, and Ana C. Ochoa (of EPS SURA) for their help authorizing and coordinating activities within EPS SURA; and the administrative and laboratory staff of EPS SURA and Dinamica I.P.S. ´ The authors also acknowledge Paola A. Rios (student) and Giovanni Torres (employee) from the Instituto Colombiano de Medicina Tropical for their help with sample handling and pretreat￾ment and APOLO Scientific Computing Center of EAFIT University for hosting the bioinformatics resources for the study. Funding. This work was funded by Grupo Em￾presarial Nutresa, EPS SURA, and Dinamica I.P.S. ´ The funders have not had any role in designing or conducting the study; in the collection, man￾agement, analysis, or interpretation of the data; in the preparation, review, or approval of the manuscript; or in the decision to submit the manuscript for publication. Duality of Interest. J.d.l.C.-Z., V.C.-A., E.P.V.-M., and J.S.E. are employees of Grupo Empresarial Nutresa. J.A.C. is an employee of Dinamica I.P.S. ´ J.M.A. is an employee of EPS SURA. No other potential conflicts of interest relevant to this article were reported. Author Contributions. J.d.l.C.-Z. processed fecal samples and DNA sequences, performed analyses, and wrote the manuscript. N.T.M. conceived the study, put forward hypotheses to be tested, and wrote the manuscript. V.C.-A. designed the cohort study, recruited partici￾pants, coordinatedfield activities, collected fecal and blood samples, and measured anthropo￾metric variables. E.P.V.-M. processed fecal sam￾ples and DNA sequences. J.A.C. coordinated field activities and transport and treatment of sam￾ples. J.M.A. coordinated participant recruitment and field activities. J.S.E. designed the cohort study, coordinated participant recruitment, su￾pervised field activities and transport and treat￾ment of samples, performed analyses, and wrote the manuscript. All authors read and approved the final version of the manuscript. J.S.E. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis. Prior Presentation. Parts of this study were presented at the XXIII Latin American Congress of Microbiology and Food Hygiene, Rosario, Argentina, 26–30 September 2016. References 1. Everard A, Belzer C, Geurts L, et al. Cross-talk between Akkermansia muciniphila and intesti￾nal epithelium controls diet-induced obesity. Proc Natl Acad Sci U S A 2013;110:9066–9071 2. Qin J, Li Y, Cai Z, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012;490:55–60 3. Karlsson FH, Tremaroli V, Nookaew I, et al. Gut metagenome in European women with nor￾mal, impaired and diabetic glucose control. Na￾ture 2013;498:99–103 4. Larsen N, Vogensen FK, van den Berg FWJ, et al. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS One 2010;5:e9085 5. Zhang X, Shen D, Fang Z, et al. Human gut microbiota changes reveal the progression of glucose intolerance. PLoS One 2013;8:e71108 6. Forslund K, Hildebrand F, Nielsen T, et al; MetaHIT consortium. Disentangling type 2 dia￾betes and metformin treatment signatures in the human gut microbiota. Nature 2015;528: 262–266 7. Pernicova I, Korbonits M. Metformin–mode of action and clinical implications for diabetes and cancer. Nat Rev Endocrinol 2014;10:143– 156 8. Lamanna C, Monami M, Marchionni N, Mannucci E. Effect of metformin on cardiovas￾cular events and mortality: a meta-analysis of randomized clinical trials. Diabetes Obes Metab 2011;13:221–228 9. Pryor R, Cabreiro F. Repurposing metformin: an old drug with new tricks in its binding pock￾ets. Biochem J 2015;471:307–322 10. Rena G, Pearson ER, Sakamoto K. Molecular mechanism of action of metformin: old or new insights? Diabetologia 2013;56:1898–1906 11. Lee H, Ko G. Effect of metformin on meta￾bolic improvement and gut microbiota. Appl En￾viron Microbiol 2014;80:5935–5943 12. Shin N-RR, Lee J-CC, Lee H-YY, et al. An in￾crease in the Akkermansia spp. population in￾duced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut 2014;63:727–735 13. Zhang X, Zhao Y, Xu J, et al. Modulation of gut microbiota by berberine and metformin during the treatment of high-fat diet-induced obesity in rats. Sci Rep 2015;5:14405 14. Bonora E, Cigolini M, Bosello O, et al. Lack of effect of intravenous metformin on plasma con￾centrations of glucose, insulin, C-peptide, gluca￾gon and growth hormone in non-diabetic subjects. Curr Med Res Opin 1984;9:47–51 15. Buse JB, DeFronzo RA, Rosenstock J, et al. The primary glucose-lowering effect of metfor￾min resides in the gut, not the circulation: Re￾sults from short-term pharmacokinetic and 12-week dose-ranging studies. Diabetes Care 2016;39:198–205 16. Bailey CJ, Wilcock C, Scarpello JH. Metfor￾min and the intestine. Diabetologia 2008;51: 1552–1553 17. Napolitano A, Miller S, Nicholls AW, et al. Novel gut-based pharmacology of metformin in 8 Type 2 Diabetes, Metformin, and Gut Microbiota Diabetes Care

care.diabetesio als.org de la Cuesta-Zuluaga and Associates s orey 0. eans and Asians.BMC /jdstorey/qvalue et al. ce2011334:103-108 e data o the M infections.N Engl J Med 2000:342:710-721 al ba fecal butyrate in heath rm.Appl Env Appl Environ Microbiol 2016:82:5850 biol201379-5112-5120 Introducing mothur:open urce,pla 29.Maida A Lan B,Cao X,Drucker D ities.Appl Environ Microbiol 2009::537 via a pat dependent on per some 37.De Vadder Kovatcheva-Datchary P 21 2011:54:339-349 abolites Vienna,Aus ib,RFoundationforStatiC3l on glu n-like p 31.Wahlstr ,Marschall H-u ng,201 .26 9 ossens M,Sabino J,et al. ciat urro buria h Fo 2115 et al.M or ho 201010 1951d Gut 20 63 .Low counts Genome Biol 2011:12:60 Sutterell parvirubr sp.nov.and Megomonas biota.Inflamm Bowel Dis 200915:1831189

patients with type 2 diabetes mellitus. PLoS One 2014;9:e100778 18. Escobar JS, Klotz B, Valdes BE, Agudelo GM. The gut microbiota of Colombians differs from that of Americans, Europeans and Asians. BMC Microbiol 2014;14:311 19. Kozich JJ,WestcottSL, BaxterNT,Highlander SK, Schloss PD. Development of a dual-index se￾quencing strategy and curation pipeline for an￾alyzing amplicon sequence data on the MiSeq Illumina sequencing platform. Appl Environ Mi￾crobiol 2013;79:5112–5120 20. Schloss PD, Westcott SL, Ryabin T, et al. Introducing mothur: open-source, platform￾independent, community-supported software for describing and comparing microbial com￾munities. Appl Environ Microbiol 2009;75:7537– 7541 21. R Development Core Team. R: A Language and Environment for Statistical Computing. Vienna, Austria, R Foundation for Statistical Computing, 2012 22. Oksanen J, Blanchet FG, Kindt R, et al. Vegan: Community Ecology Package. R package version 2.3-1, R Foundation for Statistical Com￾puting, 2015. p. 264. 23. Chen J, Bittinger K, Charlson ES, et al. Asso￾ciating microbiome composition with environ￾mental covariates using generalized UniFrac distances. Bioinformatics 2012;28:2106–2113 24. Segata N, Izard J, Waldron L, et al. Metage￾nomic biomarker discovery and explanation. Genome Biol 2011;12:R60 25. Storey JD, Bass AJ, Dabney A, Robinson D. qvalue: Q-value estimation for false discovery rate control. R package version 2.2.2. Available from http://github.com/jdstorey/qvalue. Accessed 19 January 2016 26. Wu GD, Chen J, Hoffmann C, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011;334:105–108 27. Murray BE. Vancomycin-resistant enterococ￾cal infections. N Engl J Med 2000;342:710–721 28. Cabreiro F, Au C, Leung KY, et al. Metformin retards aging in C. elegans by altering microbial folate and methionine metabolism. Cell 2013; 153:228–239 29. Maida A, Lamont BJ, Cao X, Drucker DJ. Metformin regulates the incretin receptor axis via a pathway dependent on peroxisome prolif￾erator-activated receptor-a in mice. Diabetolo￾gia 2011;54:339–349 30. Mannucci E, Ognibene A, Cremasco F, et al. Effect of metformin on glucagon-like peptide 1 (GLP-1) and leptin levels in obese nondiabetic subjects. Diabetes Care 2001;24:489–494 31. Wahlstrom A, Sayin SI, Marschall H-U, ¨ Backhed F. Intestinal crosstalk between bile ¨ acids and microbiota and its impact on host me￾tabolism. Cell Metab 2016;24:41–50 32. Turroni F, Bottacini F, Foroni E, et al. Genome analysis of Bifidobacterium bifidumPRL2010 reveals metabolic pathways for host-derived glycan forag￾ing. Proc Natl Acad Sci U S A 2010;107:19514–19519 33. Sakon H, Nagai F, Morotomi M, Tanaka R. Sutterella parvirubra sp. nov. and Megamonas funiformissp. nov., isolated from human faeces. Int J Syst Evol Microbiol 2008;58:970–975 34. Fukuda S, Toh H, Hase K, et al. Bifidobacte￾ria can protect from enteropathogenic infection through production of acetate. Nature 2011; 469:543–547 35. Gargari G, Taverniti V, Balzaretti S, et al. Consumption of a Bifidobacterium bifidum strain for 4 weeks modulates dominant intesti￾nal bacterial taxa and fecal butyrate in healthy adults. Appl Environ Microbiol 2016;82:5850– 5859 36. Hamer HM, Jonkers D, Venema K, Vanhoutvin S, Troost FJ, Brummer RJ. Review article: the role of butyrate on colonic function. Aliment Pharma￾col Ther 2008;27:104–119 37. De Vadder F, Kovatcheva-Datchary P, Goncalves D, et al. Microbiota-generated me￾tabolites promote metabolic benefits via gut￾brain neural circuits. Cell 2014;156:84–96 38. Kelly CJ, Zheng L, Campbell EL, et al. Cross￾talk between microbiota-derived short-chain fatty acids and intestinal epithelial HIF aug￾ments tissue barrier function. Cell Host Microbe 2015;17:662–671 39. Machiels K, Joossens M, Sabino J, et al. A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium praus￾nitzii defines dysbiosis in patients with ulcera￾tive colitis. Gut 2014;63:1275–1283 40. Sokol H, Seksik P, Furet JP, et al. Low counts of Faecalibacterium prausnitzii in colitis micro￾biota. Inflamm Bowel Dis 2009;15:1183–1189 care.diabetesjournals.org de la Cuesta-Zuluaga and Associates 9

点击下载完整版文档(PDF)VIP每日下载上限内不扣除下载券和下载次数;
按次数下载不扣除下载券;
24小时内重复下载只扣除一次;
顺序:VIP每日次数-->可用次数-->下载券;
已到末页,全文结束
相关文档

关于我们|帮助中心|下载说明|相关软件|意见反馈|联系我们

Copyright © 2008-现在 cucdc.com 高等教育资讯网 版权所有