Metagenomic analysis reveals crosstalk between gut microbiota and glucose-lowering drugs targeting the gastrointestinal tract in Chinese patients with type 2 diabetes: a 6 month, two-arm randomised trial

Xiuying Zhang, Huahui Ren, Cuiling Zhao, Zhun Shi, Li Qiu, Fangming Yang, Xianghai Zhou, Xueyao Han, Kui Wu, Huanzi Zhong, Yufeng Li, Junhua Li, Linong Ji, Xiuying Zhang, Huahui Ren, Cuiling Zhao, Zhun Shi, Li Qiu, Fangming Yang, Xianghai Zhou, Xueyao Han, Kui Wu, Huanzi Zhong, Yufeng Li, Junhua Li, Linong Ji

Abstract

Aims/hypothesis: The use of oral glucose-lowering drugs, particularly those designed to target the gut ecosystem, is often observed in association with altered gut microbial composition or functional capacity in individuals with type 2 diabetes. The gut microbiota, in turn, plays crucial roles in the modulation of drug efficacy. We aimed to assess the impacts of acarbose and vildagliptin on human gut microbiota and the relationships between pre-treatment gut microbiota and therapeutic responses.

Methods: This was a randomised, open-labelled, two-arm trial in treatment-naive type 2 diabetes patients conducted in Beijing between December 2016 and December 2017. One hundred participants with overweight/obesity and newly diagnosed type 2 diabetes were recruited from the Pinggu Hospital and randomly assigned to the acarbose (n=50) or vildagliptin (n=50) group using sealed envelopes. The treatment period was 6 months. Blood, faecal samples and visceral fat data from computed tomography images were collected before and after treatments to measure therapeutic outcomes and gut microbiota. Metagenomic datasets from a previous type 2 diabetes cohort receiving acarbose or glipizide for 3 months were downloaded and processed. Statistical analyses were applied to identify the treatment-related changes in clinical variables, gut microbiota and associations.

Results: Ninety-two participants were analysed. After 6 months of acarbose (n=44) or vildagliptin (n=48) monotherapy, both groups achieved significant reductions in HbA1c (from 60 to 46 mmol/mol [from 7.65% to 6.40%] in the acarbose group and from 59 to 44 mmol/mol [from 7.55% to 6.20%] in the vildagliptin group) and visceral fat areas (all adjusted p values for pre-post comparisons <0.05). Both arms showed drug-specific and shared changes in relative abundances of multiple gut microbial species and pathways, especially the common reductions in Bacteroidetes species. Three months and 6 months of acarbose-induced changes in microbial composition were highly similar in type 2 diabetes patients from the two independent studies. Vildagliptin treatment significantly enhanced fasting active glucagon-like peptide-1 (GLP-1) levels. Baseline gut microbiota, rather than baseline GLP-1 levels, were strongly associated with GLP-1 response to vildagliptin, and to a lesser extent with GLP-1 response to acarbose.

Conclusions/interpretation: This study reveals common microbial responses in type 2 diabetes patients treated with two glucose-lowering drugs targeting the gut differently and acceptable performance of baseline gut microbiota in classifying individuals with different GLP-1 responses to vildagliptin. Our findings highlight bidirectional interactions between gut microbiota and glucose-lowering drugs.

Trial registration: ClinicalTrials.gov NCT02999841 FUNDING: National Key Research and Development Project: 2016YFC1304901.

Keywords: Glucose-lowering drugs; Gut microbiota; Type 2 diabetes.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Major clinical outcomes in newly diagnosed type 2 diabetes patients after 6 month treatment with acarbose or vildagliptin. (ar) Bar charts show changes in HbA1c (a); FPG (b); PPG (c); HOMA-IR (d); Pins (e); Fins (f); weight (g); BMI (h); L2-L3 VFA, L2-L3 SFA, L4-L5 VFA and L4-L5 SFA (il); CCK (m); GLP-1 (n); leptin (o); adiponectin (p); ghrelin (q); and PYY (r), in the acarbose (light green) or vildagliptin (light blue) treatment group. Wilcoxon signed-rank test, *BH-adjusted p<0.05. The y-axis indicates the delta (Δ) post-minus pre-treatment value of each variable. Data are presented as mean+SEM (detailed BH-adjusted p values are presented in Table 1). Individual data points are shown on the graph in grey
Fig. 2
Fig. 2
Changes in the gut microbial structure induced by glucose-lowering treatment. (a) Comparisons of alpha diversity (Shannon index at the species level) between four groups. Wilcoxon signed-rank test for comparisons between pre- and post-treatment groups with the same agent; Wilcoxon rank-sum test for comparisons between groups with acarbose or vildagliptin, *p<0.05, ***p<0.001. (b) Non-metric multidimensional scaling (NMDS) plot illustrating the Bray–Curtis dissimilarities of the gut microbial species composition in pre- and post-treatment samples. (c) Bar plots of pseudo F-statistic values showing the magnitudes of microbial dissimilarities for within- and between-treatment groups. PERMANOVA (N=999 permutations), ***p<0.001. (d) Taxonomic cladogram showing significantly altered microbial taxa in patients treated with 6 months of acarbose or vildagliptin (Wilcoxon signed-rank test, BH-adjusted p<0.05; see details in ESM Tables 2, 3). (e) Gut microbial species consistently respond to acarbose and vildagliptin treatment. Wilcoxon signed-rank test, *BH-adjusted p<0.05. Colour bars indicate pre–post treatment effect sizes estimated from Wilcoxon signed-rank tests on the Clr-transformed RAs of species in the two treatment groups. Effect size >0: dark green and dark blue indicate the higher RAs in pre-treatment groups with acarbose (Acar base) and vildagliptin (Vild base), respectively; effect size ≤0: light green (Acar M6) and light blue (Vild M6) indicate the higher RAs in post-treatment groups. The effect size is calculated as the Z statistic divided by the square root of the sample size. The dashed line indicates an absolute value of effect size at 0.3. (f) Heatmap showing significantly altered species in four treatment arms, including 6 month treatment with acarbose (Acar base vs M6; n=42) or vildagliptin (Vild base vs M6; n=41) in the current study, and 3 month treatment with acarbose (Acar base vs M3; n=51) or glipizide (Glip base vs M3; n=43) in a previous study of Chinese type 2 diabetes patients [5]. The colour key indicates pre–post treatment effect sizes. Wilcoxon signed-rank test, * indicates BH-adjusted p<0.05. Acar, acarbose; Vild, vildagliptin
Fig. 3
Fig. 3
Longitudinal associations between clinical variables and microbial abundances. (a, b) Heatmaps resulting from Wald statistics of the longitudinal associations of clinical variables with the 19 species (a) and 25 pathways (b) with consistent responses to 6 month treatment with acarbose or vildagliptin. All the metabolic pathways are ranked in the same order as presented in ESM Fig. 5. Wald statistics are calculated based on multivariate regression models using GEE, adjusting for sex and age. *BH-adjusted p<0.05, **BH-adjusted p<0.01, ***BH-adjusted p<0.001. Blue indicates species/pathways of higher abundances in pre-treatment groups. Red indicates species/pathways of higher abundances in post-treatment groups
Fig. 4
Fig. 4
Links between baseline gut microbiota and post-treatment GLP-1 response. (a) Density curve of the PC% from baseline of fasting active GLP-1 in response to vildagliptin treatment. The dotted line represents a median value of 50.18%. Low response (LR): ≤50.18%; high response (HR): >50.18%. (b, c) Boxplots showing the comparisons of PC% from baseline (b) and baseline values (c) of GLP-1 and six type 2 diabetes-related variables between the two response subgroups. The p values were calculated using ANCOVA with adjustment for age and sex. *p<0.05, **p<0.01, ***p<0.001. (d) sPLS-DA to select baseline microbial features driving the separation of samples between the two subgroups. Individual samples from different subgroups are presented on a scatter plot using different colours (LR: blue; HR: orange) and 95% confidence ellipses. (e) Bar plot representing the contributions of the ten selected microbial features for the first sPLS-DA component. (f) Heatmap resulting from the coefficients of partial Spearman correlations (adjustment for age and sex) between PC%-GLP-1 and baseline abundances of ten selected microbial features in the two groups. *p<0.05. (g) Scatterplots showing the correlation between the PC%-GLP-1 and the sPLS-DA-based predicted probability in the vildagliptin group and the correlation between the PC%-GLP-1 and the predicted probability in the acarbose group. The p values and ρ values were calculated by Spearman’s rank correlation

References

    1. Fan Y, Pedersen O. Gut microbiota in human metabolic health and disease. Nat Rev Microbiol. 2021;19(1):55–71. doi: 10.1038/s41579-020-0433-9.
    1. Wu H, Esteve E, Tremaroli V, et al. 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. doi: 10.1038/nm.4345.
    1. Sun L, Xie C, Wang G, et al. Gut microbiota and intestinal FXR mediate the clinical benefits of metformin. Nat Med. 2018;24(12):1919–1929. doi: 10.1038/s41591-018-0222-4.
    1. Mueller NT, Differding MK, Zhang M, et al. Metformin Affects Gut Microbiome Composition and Function and Circulating Short-Chain Fatty Acids: A Randomized Trial. Diabetes Care. 2021;44(7):1462–1471. doi: 10.2337/dc20-2257.
    1. Gu Y, Wang X, Li J, et al. Analyses of gut microbiota and plasma bile acids enable stratification of patients for antidiabetic treatment. Nat Commun. 2017;8:1785. doi: 10.1038/s41467-017-01682-2.
    1. Olivares M, Neyrinck AM, Pötgens SA, et al. The DPP-4 inhibitor vildagliptin impacts the gut microbiota and prevents disruption of intestinal homeostasis induced by a Western diet in mice. Diabetologia. 2018;61(8):1838–1848. doi: 10.1007/s00125-018-4647-6.
    1. Liao X, Song L, Zeng B, et al. Alteration of gut microbiota induced by DPP-4i treatment improves glucose homeostasis. EBioMedicine. 2019;44:665–674. doi: 10.1016/j.ebiom.2019.03.057.
    1. Zhao L, Chen Y, Xia F, et al. A glucagon-like peptide-1 receptor agonist lowers weight by modulating the structure of gut microbiota. Front Endocrinol. 2018;9:233. doi: 10.3389/fendo.2018.00233.
    1. Ashcroft FM, Gribble FM. ATP-sensitive K+ channels and insulin secretion: their role in health and disease. Diabetologia. 1999;42:903–919. doi: 10.1007/s001250051247.
    1. Clissold SP, Edwards C. Acarbose: A Preliminary Review of its Pharmacodynamic and Pharmacokinetic Properties, and Therapeutic Potential. Drugs. 1988;35(3):214–243. doi: 10.2165/00003495-198835030-00003.
    1. Flatt PR, Bailey CJ, Green BD. Dipeptidyl peptidase IV (DPP IV) and related molecules in type 2 diabetes. Front Biosci. 2008;13:3648–3660. doi: 10.2741/2956.
    1. Yang W, Liu J, Shan Z, et al. Acarbose compared with metformin as initial therapy in patients with newly diagnosed type 2 diabetes: An open-label, non-inferiority randomised trial. Lancet Diabetes Endocrinol. 2014;2(1):46–55. doi: 10.1016/S2213-8587(13)70021-4.
    1. Zheng MY, Yang JH, Shan CY, et al. Effects of 24-week treatment with acarbose on glucagon-like peptide 1 in newly diagnosed type 2 diabetic patients: A preliminary report. Cardiovasc Diabetol. 2013;12:73. doi: 10.1186/1475-2840-12-73.
    1. Enç FY, Imeryüz N, Akin L, et al. Inhibition of gastric emptying by acarbose is correlated with GLP-1 response and accompanied by CCK release. Am J Physiol Gastrointest Liver Physiol. 2001;281(3):752–763. doi: 10.1152/ajpgi.2001.281.3.g752.
    1. Seifarth C, Bergmann J, Holst JJ, Ritzel R, Schmiegel W, Nauck MA. Prolonged and enhanced secretion of glucagon-like peptide I (7-36 amide) after oral sucrose due to α-glucosidase inhibition (acarbose) in type 2 diabetic patients. Diabet Med. 1998;15(6):485–491. doi: 10.1002/(SICI)1096-9136(199806)15:6<485::AID-DIA610>;2-Y.
    1. DiNicolantonio JJ, Bhutani J, O’Keefe JH (2015) Acarbose: safe and effective for lowering postprandial hyperglycaemia and improving cardiovascular outcomes. Open Heart 2(1):e000327. 10.1136/openhrt-2015-000327
    1. Tolhurst G, Heffron H, Lam YS, et al. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes. 2012;61(2):364–371. doi: 10.2337/db11-1019.
    1. Thomas C, Gioiello A, Noriega L, et al. TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metab. 2009;10(3):167–177. doi: 10.1016/J.CMET.2009.08.001.
    1. Trabelsi MS, Daoudi M, Prawitt J, et al. Farnesoid X receptor inhibits glucagon-like peptide-1 production by enteroendocrine L cells. Nat Commun. 2015;6:7629. doi: 10.1038/NCOMMS8629.
    1. Maier L, Pruteanu M, Kuhn M, et al. Extensive impact of non-antibiotic drugs on human gut bacteria. Nature. 2018;555(7698):623–628. doi: 10.1038/NATURE25979.
    1. Cao TTB, Wu KC, Hsu JL, et al. Effects of Non-insulin Anti-hyperglycemic Agents on Gut Microbiota: A Systematic Review on Human and Animal Studies. Front Endocrinol. 2020;11:573891. doi: 10.3389/FENDO.2020.573891.
    1. World Health Organization (1999) The World health report: 1999: Making a difference. Available from . Accessed 1 May 2000
    1. Truong DT, Franzosa EA, Tickle TL, et al. MetaPhlAn2 for enhanced metagenomic taxonomic profiling. Nat Methods. 2015;12(10):902–903. doi: 10.1038/nmeth.3589.
    1. Franzosa EA, McIver LJ, Rahnavard G, et al. Species-level functional profiling of metagenomes and metatranscriptomes. Nat Methods. 2018;15(11):962–968. doi: 10.1038/s41592-018-0176-y.
    1. Aitchison J. The Statistical Analysis of Compositional Data. J R Stat Soc Ser B. 1982;44(2):139–177. doi: 10.1111/j.2517-6161.1982.tb01195.x.
    1. Chinese Diabetes Society Guidelines for the Prevention and Treatment of Type 2 Diabetes in China (2013) Chin J Endocrinol Metab. 2014;30:893–942. doi: 10.3760/cma.j.issn.1000-6699.2014.10.020.
    1. Müller TD, Nogueiras R, Andermann ML, et al. Ghrelin. Mol Metab. 2015;4:437–460. doi: 10.1016/j.molmet.2015.03.005.
    1. Batterham RL, Bloom SR. The gut hormone peptide YY regulates appetite. Ann N Y Acad Sci. 2003;994(1):162–168. doi: 10.1111/j.1749-6632.2003.tb03176.x.
    1. Sedighi M, Razavi S, Navab-Moghadam F, et al. Comparison of gut microbiota in adult patients with type 2 diabetes and healthy individuals. Microb Pathog. 2017;111:362–369. doi: 10.1016/j.micpath.2017.08.038.
    1. Li Q, Chang Y, Zhang K, Chen H, Tao S, Zhang Z. Implication of the gut microbiome composition of type 2 diabetic patients from northern China. Sci Rep. 2020;10:5450. doi: 10.1038/s41598-020-62224-3.
    1. Karlsson FH, Tremaroli V, Nookaew I, et al. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature. 2013;498(7452):99–103. doi: 10.1038/nature12198.
    1. Wang L, Yu X, Xu X, et al. The Fecal Microbiota Is Already Altered in Normoglycemic Individuals Who Go on to Have Type 2 Diabetes. Front Cell Infect Microbiol. 2021;11:598672. doi: 10.3389/fcimb.2021.598672.
    1. Zhong H, Ren H, Lu Y, et al. Distinct gut metagenomics and metaproteomics signatures in prediabetics and treatment-naïve type 2 diabetics. EBioMedicine. 2019;47:373–383. doi: 10.1016/j.ebiom.2019.08.048.
    1. Qin J, Li Y, Cai Z, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490(7418):55–60. doi: 10.1038/nature11450.
    1. Louis P, Flint HJ. Diversity, metabolism and microbial ecology of butyrate-producing bacteria from the human large intestine. FEMS Microbiol Lett. 2009;294(1):1–8. doi: 10.1111/j.1574-6968.2009.01514.x.
    1. Pearson ER, Flechtner I, Njølstad PR, et al. Switching from Insulin to Oral Sulfonylureas in Patients with Diabetes Due to Kir6.2 Mutations. N Engl J Med. 2006;355(5):467–477. doi: 10.1056/nejmoa061759.
    1. Christiansen CB, Gabe MBN, Svendsen B, Dragsted LO, Rosenkilde MM, Holst JJ. The impact of short-chain fatty acids on GLP-1 and PYY secretion from the isolated perfused rat colon. Am J Physiol Gastrointest Liver Physiol. 2018;315(1):G53–G65. doi: 10.1152/AJPGI.00346.2017.
    1. Brighton CA, Rievaj J, Kuhre RE, et al. Bile acids trigger GLP-1 release predominantly by accessing basolaterally located G protein–coupled bile acid receptors. Endocrinology. 2015;156(11):3961. doi: 10.1210/EN.2015-1321.
    1. Hanefeld M. Cardiovascular benefits and safety profile of acarbose therapy in prediabetes and established type 2 diabetes. Cardiovasc Diabetol. 2007;6:20. doi: 10.1186/1475-2840-6-20.
    1. Monami M, Lamanna C, Desideri CM, Mannucci E. DPP-4 inhibitors and lipids: Systematic review and meta-analysis. Adv Ther. 2012;29(1):14–25. doi: 10.1007/s12325-011-0088-z.
    1. Zhang Q, Xiao X, Li M, et al. Vildagliptin increases butyrate-producing bacteria in the gut of diabetic rats. PLoS One. 2017;12(10):e0184735. doi: 10.1371/journal.pone.0184735.
    1. Wang L, Li P, Tang Z, Yan X, Feng B. Structural modulation of the gut microbiota and the relationship with body weight: Compared evaluation of liraglutide and saxagliptin treatment. Sci Rep. 2016;6:33251. doi: 10.1038/srep33251.
    1. Kim D, Hofstaedter CE, Zhao C, et al. Optimizing methods and dodging pitfalls in microbiome research. Microbiome. 2017;5:52. doi: 10.1186/s40168-017-0267-5.
    1. Bray GA, Edelstein SL, Crandall JP, et al. Long-term safety, tolerability, and weight loss associated with metformin in the diabetes prevention program outcomes study. Diabetes Care. 2012;35(4):731–737. doi: 10.2337/dc11-1299.
    1. Guo W, Nie L, Wang XR, et al. Comparison of Placebo Effect between Asian and Caucasian Type 2 Diabetic Patients: A Meta-Analysis. Chin Med J. 2018;131(13):1605–1612. doi: 10.4103/0366-6999.235107.
    1. Zhang Y, Gu Y, Ren H, et al. Gut microbiome-related effects of berberine and probiotics on type 2 diabetes (the PREMOTE study) Nat Commun. 2020;11:5015. doi: 10.1038/s41467-020-18414-8.

Source: PubMed

3
订阅