Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Combining a diet rich in fermentable carbohydrates with metformin improves glycaemic control and reshapes the gut microbiota in people with prediabetes

Abstract

Metformin efficiently lowers blood glucose levels but leads to gastrointestinal side effects. However, whether dietary interventions can improve metformin tolerability and glucose-lowering efficacy remains unknown. Here we investigate the effects of pretreatment with a diet rich in fermentable oligosaccharides, disaccharides, monosaccharides and polyols (FODMAPs) in combination with metformin on postprandial glycaemia and gut microbiota in people with prediabetes. In a double-blind, randomised, crossover trial, 26 individuals with prediabetes received an isocaloric diet with moderate or low FODMAPs for 10 d, concomitantly with metformin for 5 d, separated by a washout period of 2 weeks. The primary endpoint is the difference in postprandial glycaemia assessed by total postprandial incremental area under the curve through continuous glucose monitoring. Secondary endpoints are differences in glucose, insulin and glucagon-like peptide 1 (GLP-1) levels after an oral glucose tolerance test, gut microbiota, gastrointestinal symptoms and body weight. We show that moderate FODMAPs with metformin, as compared with low FODMAPs with metformin, result in lower postprandial glycaemia, higher GLP-1 secretion and higher Butyricimonas virosa abundance. We also show that a higher baseline abundance of Dorea formicigenerans predicts gastrointestinal intolerance to metformin. These findings have implications for personalizing nutritional and pharmacological interventions to prevent diabetes. ClinicalTrials.gov registration: NCT05628584.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Study design and flow.
Fig. 2: Daily total energy, macronutrient, FODMAP intake and physical activity.
Fig. 3: Averaged 24-h sensor glucose profiles captured by CGM.
Fig. 4: PG, late-phase insulin and total GLP-1 response during OGTTs in each intervention period.
Fig. 5: Diversity comparisons of gut microbiome between M-M and L-M.
Fig. 6: Metabolic pathway prediction of gut microbiome between baseline, M-M and L-M.

Similar content being viewed by others

Data availability

The individual de-identified participant microbiota sequencing data can be accessed at https://doi.org/10.6084/m9.figshare.28123295.v1 (ref. 70). Other de-identified participant data are not openly available due to participant confidentiality and will be shared with the corresponding author upon reasonable request, and requests will be responded to within 4 weeks. The study protocol is available in the Supplementary Information. Source data are provided with this paper.

References

  1. American Diabetes Association Professional Practice Committee 3. Prevention or delay of diabetes and associated comorbidities: Standards of Care in Diabetes—2024. Diabetes Care 47, S43–S51 (2024).

  2. Wang, Z. et al. Gut microbiome differences between metformin‐ and liraglutide‐treated T2DM subjects. Endocrinol. Diabetes Metab. 1, e00009 (2018).

    Article  PubMed  Google Scholar 

  3. Tian, R. et al. Metformin ameliorates endotoxemia-induced endothelial pro-inflammatory responses via AMPK-dependent mediation of HDAC5 and KLF2. Biochim. Biophys. Acta 1865, 1701–1712 (2019).

    Article  CAS  Google Scholar 

  4. Qin, J. et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 490, 55–60 (2012).

    Article  CAS  PubMed  Google Scholar 

  5. Mei, Z. et al. Strain-specific gut microbial signatures in type 2 diabetes identified in a cross-cohort analysis of 8,117 metagenomes. Nat. Med. 30, 2265–2276 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Shin, N.-R. et al. An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut 63, 727–735 (2014).

    Article  CAS  PubMed  Google Scholar 

  7. Mueller, N. T. et al. Metformin affects gut microbiome composition and function and circulating short-chain fatty acids: a randomized trial. Diabetes Care 44, 1462–1471 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Brønden, A. et al. Single-dose metformin enhances bile acid-induced glucagon-like peptide-1 secretion in patients with type 2 diabetes. J. Clin. Endocrinol. Metab. 102, 4153–4162 (2017).

    Article  PubMed  Google Scholar 

  9. Cui, J. et al. Butyrate-producing bacteria and insulin homeostasis: the Microbiome and Insulin Longitudinal Evaluation Study (MILES). Diabetes 71, 2438–2446 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bahne, E. et al. Metformin-induced glucagon-like peptide-1 secretion contributes to the actions of metformin in type 2 diabetes. JCI Insight 3, e93936 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Diabetes Prevention Program Research Group Long-term safety, tolerability, and weight loss associated with metformin in the Diabetes Prevention Program Outcomes Study. Diabetes Care 35, 731–737 (2012).

  12. Zhang, L. et al. Safety and effectiveness of metformin plus lifestyle intervention compared with lifestyle intervention alone in preventing progression to diabetes in a Chinese population with impaired glucose regulation: a multicentre, open-label, randomised controlled trial. Lancet Diabetes Endocrinol. 11, 567–577 (2023).

    Article  CAS  PubMed  Google Scholar 

  13. Bryrup, T. et al. Metformin-induced changes of the gut microbiota in healthy young men: results of a non-blinded, one-armed intervention study. Diabetologia 62, 1024–1035 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Díaz-Perdigones, C. M., Muñoz-Garach, A., Álvarez-Bermúdez, M. D., Moreno-Indias, I. & Tinahones, F. J. Gut microbiota of patients with type 2 diabetes and gastrointestinal intolerance to metformin differs in composition and functionality from tolerant patients. Biomed. Pharmacother. 145, 112448 (2022).

    Article  PubMed  Google Scholar 

  15. Gibson, P. R. History of the low FODMAP diet. J. Gastroenterol. Hepatol. 32, 5–7 (2017).

    Article  CAS  PubMed  Google Scholar 

  16. Vandeputte, D. & Joossens, M. Effects of low and high FODMAP diets on human gastrointestinal microbiota composition in adults with intestinal diseases: a systematic review. Microorganisms 8, 1638 (2020).

  17. Halmos, E. P., Power, V. A., Shepherd, S. J., Gibson, P. R. & Muir, J. G. A diet low in FODMAPs reduces symptoms of irritable bowel syndrome. Gastroenterology 146, 67–75 (2014).

    Article  CAS  PubMed  Google Scholar 

  18. Ong, D. K. et al. Manipulation of dietary short chain carbohydrates alters the pattern of gas production and genesis of symptoms in irritable bowel syndrome. J. Gastroenterol. Hepatol. 25, 1366–1373 (2010).

    Article  CAS  PubMed  Google Scholar 

  19. Nybacka, S. et al. A low FODMAP diet plus traditional dietary advice versus a low-carbohydrate diet versus pharmacological treatment in irritable bowel syndrome (CARIBS): a single-centre, single-blind, randomised controlled trial. Lancet Gastroenterol. Hepatol. 9, 507–520 (2024).

    Article  CAS  PubMed  Google Scholar 

  20. Black, C. J., Staudacher, H. M. & Ford, A. C. Efficacy of a low FODMAP diet in irritable bowel syndrome: systematic review and network meta-analysis. Gut 71, 1117–1126 (2022).

    Article  CAS  PubMed  Google Scholar 

  21. McIntosh, K. et al. FODMAPs alter symptoms and the metabolome of patients with IBS: a randomised controlled trial. Gut 66, 1241–1251 (2017).

    Article  CAS  PubMed  Google Scholar 

  22. Chu, N. H. et al. Higher short-chain fermentable carbohydrates are associated with lower body fat and higher insulin sensitivity in people with prediabetes. Nutrients 15, 5070 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Burton, J. H. et al. Addition of a gastrointestinal microbiome modulator to metformin improves metformin tolerance and fasting glucose levels. J. Diabetes Sci. Technol. 9, 808–814 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Chumpitazi, B. P. et al. Randomised clinical trial: gut microbiome biomarkers are associated with clinical response to a low FODMAP diet in children with the irritable bowel syndrome. Aliment. Pharmacol. Ther. 42, 418–427 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. DeFronzo, R. A. et al. Once-daily delayed-release metformin lowers plasma glucose and enhances fasting and postprandial GLP-1 and PYY: results from two randomised trials. Diabetologia 59, 1645–1654 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Buse, J. B. et al. The primary glucose-lowering effect of metformin resides in the gut, not the circulation: results from short-term pharmacokinetic and 12-week dose-ranging studies. Diabetes Care 39, 198–205 (2016).

    Article  CAS  PubMed  Google Scholar 

  27. Chu, N., Chan, J. C. & Chow, E. Pharmacomicrobiomics in western medicine and traditional Chinese medicine in type 2 diabetes. Front. Endocrinol. 13, 857090 (2022).

    Article  Google Scholar 

  28. Jia, L. & Zhu, C. Pharmacomicrobiomics and type 2 diabetes mellitus: a novel perspective towards possible treatment. Front. Endocrinol. 14, 1149256 (2023).

    Article  Google Scholar 

  29. Halmos, E. P. et al. Diets that differ in their FODMAP content alter the colonic luminal microenvironment. Gut 64, 93–100 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Staudacher, H. M. et al. A diet low in FODMAPs reduces symptoms in patients with irritable bowel syndrome and a probiotic restores Bifidobacterium species: a randomized controlled trial. Gastroenterology 153, 936–947 (2017).

    Article  CAS  PubMed  Google Scholar 

  31. Chu, N., Chan, J. C. & Chow, E. A diet high in FODMAPs as a novel dietary strategy in diabetes? Clin. Nutr. 41, 2103–2112 (2022).

  32. Lee, H. et al. A novel bacterium, Butyricimonas virosa, preventing HFD-induced diabetes and metabolic disorders in mice via GLP-1 receptor. Front. Microbiol. 13, 858192 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Tsai, C.-Y. et al. Gut microbial signatures for glycemic responses of GLP-1 receptor agonists in type 2 diabetic patients: a pilot study. Front. Endocrinol. 12, 814770 (2022).

    Article  Google Scholar 

  34. Ellingsgaard, H. et al. GLP-1 secretion is regulated by IL-6 signalling: a randomised, placebo-controlled study. Diabetologia 63, 362–373 (2020).

    Article  CAS  PubMed  Google Scholar 

  35. Zhang, L. et al. Gut microbiota and type 2 diabetes mellitus: association, mechanism, and translational applications. Mediators Inflamm. 2021, 5110276 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Sayin, S. I. et al. Gut microbiota regulates bile acid metabolism by reducing the levels of tauro-β-muricholic acid, a naturally occurring FXR antagonist. Cell Metab. 17, 225–235 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Layden, B. T., Angueira, A. R., Brodsky, M., Durai, V. & Lowe, W. L. Jr Short chain fatty acids and their receptors: new metabolic targets. Transl. Res. 161, 131–140 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. Liu, Y. et al. Pediococcus pentosaceus PR-1 modulates high-fat-died-induced alterations in gut microbiota, inflammation, and lipid metabolism in zebrafish. Front. Nutr. 10, 1087703 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Zhao, C., Dong, H., Zhang, Y. & Li, Y. Discovery of potential genes contributing to the biosynthesis of short-chain fatty acids and lactate in gut microbiota from systematic investigation in E. coli. NPJ Biofilms Microbiomes 5, 19 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Wang, Z. et al. Causal effects of gut microbiota on appendicitis: a two-sample Mendelian randomization study. Front. Cell. Infect. Microbiol. 13, 1320992 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Adela, R. & Banerjee, S. K. GDF‐15 as a target and biomarker for diabetes and cardiovascular diseases: a translational prospective. J. Diabetes Res. 2015, 490842 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Xiao, S. et al. Lac-Phe mediates the effects of metformin on food intake and body weight. Nat. Metab. 6, 659–669 (2024).

  43. Gillani, S. W., Ghayedi, N., Roosta, P., Seddigh, P. & Nasiri, O. Effect of metformin on lipid profiles of type 2 diabetes mellitus: a meta-analysis of randomized controlled trials. J. Pharm. Bioallied Sci. 13, 76–82 (2021).

    Article  CAS  PubMed  Google Scholar 

  44. Kwon, Y.-J., Lee, H. S., Chang, H.-J., Koh, S.-B. & Lee, J.-W. Association of dietary lipid intake with low-density lipoprotein cholesterol levels: analysis of two independent population-based studies. Eur. J. Nutr. 59, 2557–2567 (2020).

    Article  PubMed  Google Scholar 

  45. Chu, N. H., Chow, E. & Chan, J. C. The therapeutic potential of the specific intestinal microbiome (SIM) diet on metabolic diseases. Biology 13, 498 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Luk, A. O. Y. et al. A proof-of-concept study to evaluate the efficacy and safety of BTI320 on post-prandial hyperglycaemia in Chinese subjects with pre-diabetes. BMC Endocr. Disord. 18, 59 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Zeevi, D. et al. Personalized nutrition by prediction of glycemic responses. Cell 163, 1079–1094 (2015).

    Article  CAS  PubMed  Google Scholar 

  48. Zahalka, S. J., Galindo, R. J., Shah, V. N. & Low Wang, C. C. Continuous glucose monitoring for prediabetes: what are the best metrics? J. Diabetes Sci. Technol. 18, 835–846 (2024).

  49. Chow, E. et al. 1827-P: higher glucagon-like peptide-1 response and lower beta-cell glucose sensitivity are associated with increased glycemic variability among individuals with prediabetes. Diabetes https://doi.org/10.2337/db19-1827-P (2019).

  50. Spartano, N. L. et al. Defining continuous glucose monitor time in range in a large, community-based cohort without diabetes. J. Clin. Endocrinol. Metab. 110, 1128–1134 (2024).

  51. Guerci, B. et al. Important decrease in hospitalizations for acute diabetes events following freestyle libre system initiation in people with type 2 diabetes on basal insulin therapy in France. Diabetes Technol. Ther. 25, 20–30 (2023).

    Article  CAS  PubMed  Google Scholar 

  52. Cox, S. R. et al. Effects of low FODMAP diet on symptoms, fecal microbiome, and markers of inflammation in patients with quiescent inflammatory bowel disease in a randomized trial. Gastroenterology 158, 176–188 (2020).

    Article  CAS  PubMed  Google Scholar 

  53. Gonai, M. et al. Galacto-oligosaccharides ameliorate dysbiotic Bifidobacteriaceae decline in Japanese patients with type 2 diabetes. Benef. Microbes 8, 705–716 (2017).

    Article  CAS  PubMed  Google Scholar 

  54. Canfora, E. E. et al. Supplementation of diet with galacto-oligosaccharides increases bifidobacteria, but not insulin sensitivity, in obese prediabetic individuals. Gastroenterology 153, 87–97 (2017).

    Article  CAS  PubMed  Google Scholar 

  55. Attaluri, A., Donahoe, R., Valestin, J., Brown, K. & Rao, S. S. C. Randomised clinical trial: dried plums (prunes) vs. psyllium for constipation. Aliment. Pharmacol. Ther. 33, 822–828 (2011).

    Article  CAS  PubMed  Google Scholar 

  56. Eady, S. L. et al. The effect of ‘Zesy002’ kiwifruit (Actinidia chinensis var. chinensis) on gut health function: a randomised cross-over clinical trial. J. Nutr. Sci. 8, e18 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Müller, M. et al. Distal colonic transit is linked to gut microbiota diversity and microbial fermentation in humans with slow colonic transit. Am. J. Physiol. 318, G361–G369 (2020).

    Google Scholar 

  58. World Health Organization (ed.) Waist Circumference and Waist–Hip Ratio: Report of a WHO Expert Consultation (World Health Organization, 2008).

  59. Jenkins, D. J. et al. Glycemic index, glycemic load, and cardiovascular disease and mortality. N. Engl. J. Med. 384, 1312–1322 (2021).

    Article  CAS  PubMed  Google Scholar 

  60. Salmerón, J. et al. Dietary fiber, glycemic load, and risk of NIDDM in men. Diabetes Care 20, 545–550 (1997).

    Article  PubMed  Google Scholar 

  61. Muir, J. G. et al. Measurement of short-chain carbohydrates in common Australian vegetables and fruits by high-performance liquid chromatography (HPLC). J. Agric. Food Chem. 57, 554–565 (2009).

    Article  CAS  PubMed  Google Scholar 

  62. Hagströmer, M., Oja, P. & Sjöström, M. The International Physical Activity Questionnaire (IPAQ): a study of concurrent and construct validity. Public Health Nutr. 9, 755–762 (2006).

    Article  PubMed  Google Scholar 

  63. Battelino, T. et al. Continuous glucose monitoring and metrics for clinical trials: an international consensus statement. Lancet Diabetes Endocrinol. 11, 42–57 (2023).

    Article  CAS  PubMed  Google Scholar 

  64. Pagacz, K., Stawiski, K., Szadkowska, A., Mlynarski, W. & Fendler, W. GlyCulator2: an update on a web application for calculation of glycemic variability indices. Acta Diabetol. 55, 877–880 (2018).

    Article  PubMed  Google Scholar 

  65. Lam, E. T. P., Lam, C. L. K., Lo, Y. Y. C. & Gandek, B. Psychometrics and population norm of the Chinese (HK) SF-36 Health Survey_Version 2. Hong Kong Pract. 30, 185–198 (2008).

    Google Scholar 

  66. Matsuda, M. & DeFronzo, R. A. Insulin sensitivity indices obtained from oral glucose tolerance testing: comparison with the euglycemic insulin clamp. Diabetes Care 22, 1462–1470 (1999).

    Article  CAS  PubMed  Google Scholar 

  67. Ferrannini, E. et al. Predominant role of reduced beta-cell sensitivity to glucose over insulin resistance in impaired glucose tolerance. Diabetologia 46, 1211–1219 (2003).

    Article  CAS  PubMed  Google Scholar 

  68. Chow, E. et al. A randomized controlled trial of intermittently-scanned continuous glucose monitoring as an adjunct to lifestyle modification in impaired glucose tolerance. Diabetologia 66, 22 (2023).

  69. R Core Team R: A Language and Environment for Statistical Computing (R Foundation for Statistical Computing, 2013).

  70. Chu, N. FODMAPs and metformin study. figshare https://doi.org/10.6084/m9.figshare.28123295.v1 (2025).

Download references

Acknowledgements

This study was supported by a Merck KGaA investigator-initiated study grant to E.C. (MS200084_0060). The funder approved the final protocol and the manuscript but had no role in the design, collection, analyses or interpretation of data, decision to publish or preparation of the manuscript. Dexcom supported provision of Dexcom G6 CGM devices for the trial but had no role in the study design, data collection, interpretation, final decision to publish or preparation of the manuscript. N.H.S.C. was supported by the Chinese University of Hong Kong Research Fellowship scheme (RFS2223/0621/23) and an Innovation and Technology Commission Grant (MRP 029/21). We thank A. Lai, M. Lee, A. Chan and the 3M Clinical Trial Unit, Prince of Wales Hospital, for their support during the study.

Author information

Authors and Affiliations

Authors

Contributions

N.H.S.C. and E.C. conceived the idea of the study; N.H.S.C., E.W.M.P., Q.S. and J.Y.S.L. were involved in data collection and examination, which Z.Z., J.M. and J.C.N.C. supported; E.C., N.H.S.C. and J.L. analysed the data. N.H.S.C. wrote the first draft of the paper. All authors critically reviewed the paper and agreed to the final published version.

Corresponding author

Correspondence to Elaine Chow.

Ethics declarations

Competing interests

E.C. has received speaker honoraria from AstraZeneca, Procter & Gamble and ZP Therapeutics and institutional research support from Merck KGaA, Hua Medicine, Medtronic Diabetes, Powder Pharmaceuticals and Sanofi. J.C.N.C. has received research grants and/or honoraria for consultancy or giving lectures from AstraZeneca, Bayer, Boehringer Ingelheim, Eli Lilly, Hua Medicine, Lee Powder, Merck Serono, Merck Sharp & Dohme, Pfizer, Servier, Sanofi and Viatris Pharmaceutical. Monash University financially benefits from the sales of digital applications and booklets associated with the low-FODMAP diet. The other authors declare no competing interests.

Peer review

Peer review information

Nature Metabolism thanks Holly Willis and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editors: Jean Nakhle and Ashley Castellanos-Jankiewicz, in collaboration with the Nature Metabolism team.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Changes in Gastrointestinal symptoms (a-j) between moderate M-M) and low FODMAP (L-M) interventions.a) Abdominal pain/discomfort, b) Abdominal distension, c) Belching, d) Bloating, e)Flatuence, f) Dissatidfaction of bowel habits, g) Epigastric pain/discomfort, H) Epigastric distension, i) Early satiety, j)Fatigue.

Abbreviation: VAS: visual analogue scale. Differences between interventions are compared using paired t-tests. Sample size n= moderate FODMAP intervention: 25 and low FODMAP intervention: 26 participants.

Source data

Extended Data Fig. 2 Comparisons of species. Volcano diagram in comparing a) the baseline and M-M intervention, b) L-M intervention in the species levels (T-test, adjusted p-value) and (c) Bar plot comparing the M-M and L-M interventions in the species levels.

Bar plot comparing the M-M and L-M interventions in the species levels. A volcano plot is a presentation of a scatter plot, which usually consists of several parts, including significantly up-regulated different species and significantly down-regulated different species. The horizontal axis is the fold difference of the different species in the comparison group. In contrast, the vertical axis is the p-value of the significant between-group difference test for the different species. Statistical significance was assessed using two-sided two-sample t-tests. Each point in the graph represents a differential species. (Total fecal sample size n= baseline:26, M-M:23 and L-M:24 samples.) Up (red points) represents the higher abundance of that differential species in the M-M than the L-M intervention; grey points indicate no significant change, while Down (blue points) is the opposite. Abbreviation: B: baseline, L-M: low FODMAP diet with metformin, M-M: moderate FODMAP diet with metformin.

Source data

Extended Data Fig. 3 Beta-Diversity comparisons of gut microbiome. Principal Coordinates Analysis (PCoA) between a) the baseline and interventions from Jaccard, b) unweighted_unifrac and c) weighted_unifrac.

Each point represents a sample, plotted by a principal component on the X-axis and another principal component on the Y-axis, which was colored by group. (Total fecal sample size n= baseline:26, M-M:23 and L-M:24 samples.) The percentage on each axis indicates the contribution value to discrepancy among samples.

Source data

Extended Data Table 1 Interventional diets provided to subjects
Extended Data Table 2 Comparison in CGM metrics between interventions
Extended Data Table 3 Inflammatory and lipid profiles at the baseline and end of each intervention period
Extended Data Table 4 Gastrointestinal symptoms during metformin only or in combination with moderate or low FODMAP diets
Extended Data Table 5 SCFA /Bile Acids following low or moderate FODMAPs and metformin

Supplementary information

Supplementary Information

Supplementary Tables 1–4, Fig. 1 and Note, including the consort checklist, study protocol and statistical analysis plan.

Reporting Summary

Source data

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chu, N.H.S., Ling, J., Poon, E.W.M. et al. Combining a diet rich in fermentable carbohydrates with metformin improves glycaemic control and reshapes the gut microbiota in people with prediabetes. Nat Metab 7, 1614–1629 (2025). https://doi.org/10.1038/s42255-025-01336-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s42255-025-01336-4

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research