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:

The microbial metabolite agmatine acts as an FXR agonist to promote polycystic ovary syndrome in female mice

Abstract

Polycystic ovary syndrome (PCOS), an endocrine disorder afflicting 6–20% of women of reproductive age globally, has been linked to alterations in the gut microbiome. We previously showed that in PCOS, elevation of Bacteroides vulgatus in the gut microbiome was associated with altered bile acid metabolism. Here we show that B. vulgatus also induces a PCOS-like phenotype in female mice via an alternate mechanism independent of bile acids. We find that B. vulgatus contributes to PCOS-like symptoms through its metabolite agmatine, which is derived from arginine by arginine decarboxylase. Mechanistically, agmatine activates the farnesoid X receptor (FXR) pathway to subsequently inhibit glucagon-like peptide-1 (GLP-1) secretion by L cells, which leads to insulin resistance and ovarian dysfunction. Critically, the GLP-1 receptor agonist liraglutide and the arginine decarboxylase inhibitor difluoromethylarginine ameliorate ovarian dysfunction in a PCOS-like mouse model. These findings reveal that agmatine–FXR–GLP-1 signalling contributes to ovarian dysfunction, presenting a potential therapeutic target for PCOS management.

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: B. vulgatus induces PCOS-like phenotype through a non-bile acid-dependent pathway.
Fig. 2: B. vulgatus inhibits GLP-1 secretion, and the GLP-1R agonist liraglutide improves B. vulgatus-induced PCOS-like phenotype.
Fig. 3: B. vulgatus extract inhibits GLP-1 secretion in intestinal epithelial L cells by activating FXR.
Fig. 4: FXR signalling pathway mediates B. vulgatus-induced PCOS-like phenotype by regulating GLP-1 secretion.
Fig. 5: The B. vulgatus derivative agmatine activates FXR in intestinal epithelial L cells and inhibits GLP-1 secretion, inducing a PCOS-like phenotype in mice.
Fig. 6: Inhibiting the production of agmatine prevents B. vulgatus-induced PCOS-like phenotype.
Fig. 7: The speA and bsh of B. vulgatus can participate in the development of PCOS-like phenotypes in mice independently.

Similar content being viewed by others

Data availability

All the data relating to the metagenomic sequencing and RNA sequencing of this study have been uploaded to the Sequence Read Archive database and are available for download via accession numbers PRJNA1080049 and PRJNA1080099. Source data are provided with this paper.

Code availability

No custom code was used for this study.

References

  1. Norman, R. J., Dewailly, D., Legro, R. S. & Hickey, T. E. Polycystic ovary syndrome. Lancet 370, 685–697 (2007).

    Article  CAS  PubMed  Google Scholar 

  2. Escobar-Morreale, H. F. Polycystic ovary syndrome: definition, aetiology, diagnosis and treatment. Nat. Rev. Endocrinol. 14, 270–284 (2018).

    Article  PubMed  Google Scholar 

  3. Azziz, R. et al. Polycystic ovary syndrome. Nat. Rev. Dis. Prim. 2, 16057 (2016).

    Article  PubMed  Google Scholar 

  4. Hoeger, K. M., Dokras, A. & Piltonen, T. Update on PCOS: consequences, challenges and guiding treatment. J. Clin. Endocrinol. Metab. 106, e1071–e1083 (2020).

    Article  Google Scholar 

  5. Martel, J. et al. Gut barrier disruption and chronic disease. Trends Endocrinol. Metab. 33, 247–265 (2022).

    Article  CAS  PubMed  Google Scholar 

  6. Postler, T. S. & Ghosh, S. understanding the holobiont: how microbial metabolites affect human health and shape the immune system. Cell Metab. 26, 110–130 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Han, Q., Wang, J., Li, W., Chen, Z.-J. & Du, Y. Androgen-induced gut dysbiosis disrupts glucolipid metabolism and endocrinal functions in polycystic ovary syndrome. Microbiome 9, 101 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Sun, Y., Gao, S., Ye, C. & Zhao, W. Gut microbiota dysbiosis in polycystic ovary syndrome: mechanisms of progression and clinical applications. Front. Cell. Infect. Microbiol. 13, 1142041 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lindheim, L. et al. Alterations in gut microbiome composition and barrier function are associated with reproductive and metabolic defects in women with polycystic ovary syndrome (PCOS): a pilot study. PLoS ONE 12, e0168390 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Suturina, L. et al. Polycystic ovary syndrome and gut microbiota: phenotype matters. Life 13, 7 (2023).

    Article  Google Scholar 

  11. Chu, W. et al. Metagenomic analysis identified microbiome alterations and pathological association between intestinal microbiota and polycystic ovary syndrome. Fertil. Steril. 113, 1286 (2020).

    Article  CAS  PubMed  Google Scholar 

  12. Zeng, B. et al. Structural and functional profiles of the gut microbial community in polycystic ovary syndrome with insulin resistance (IR-PCOS): a pilot study. Res. Microbiol. 170, 43–52 (2019).

    Article  CAS  PubMed  Google Scholar 

  13. Liu, R. et al. Dysbiosis of gut microbiota associated with clinical parameters in polycystic ovary syndrome. Front. Microbiol. 8, 324 (2017).

    PubMed  PubMed Central  Google Scholar 

  14. Qi, X. et al. Gut microbiota-bile acid-interleukin-22 axis orchestrates polycystic ovary syndrome. Nat. Med. 25, 1225–1233 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Zheng, L. et al. CRISPR/Cas-based genome editing for human gut commensal bacteroides species. ACS Synth. Biol. 11, 464–472 (2022).

    Article  CAS  PubMed  Google Scholar 

  16. Jensterle, M. et al. The role of glucagon-like peptide-1 in reproduction: from physiology to therapeutic perspective. Hum. Reprod. Update 25, 504–517 (2019).

    Article  CAS  PubMed  Google Scholar 

  17. Gribble, F. M. & Reimann, F. Enteroendocrine cells: chemosensors in the intestinal epithelium. Annu. Rev. Physiol. 78, 277–299 (2016).

    Article  CAS  PubMed  Google Scholar 

  18. Nauck, M. A. & Meier, J. J. Incretin hormones: their role in health and disease. Diabetes Obes. Metab. 20, 5–21 (2018).

    Article  CAS  PubMed  Google Scholar 

  19. Tan, Q. et al. Recent advances in incretin-based pharmacotherapies for the treatment of obesity and diabetes. Front. Endocrinol. 13, 838410 (2022).

    Article  Google Scholar 

  20. Coll, A. P. et al. GDF15 mediates the effects of metformin on body weight and energy balance. Nature 578, 444 (2020).

    Article  CAS  PubMed  Google Scholar 

  21. McLean, B. A. et al. Revisiting the complexity of GLP-1 action from sites of synthesis to receptor activation. Endocr. Rev. 42, 101–132 (2021).

    Article  PubMed  Google Scholar 

  22. Sandoval, D. A. & D’Alessio, D. A. Physiology of proglucagon peptides: role of glucagon and glp-1 in health and disease. Physiol. Rev. 95, 513–548 (2015).

    Article  CAS  PubMed  Google Scholar 

  23. Trabelsi, M.-S. et al. Farnesoid X receptor inhibits glucagon-like peptide-1 production by enteroendocrine L cells. Nat. Commun. 6, 7629 (2015).

    Article  PubMed  Google Scholar 

  24. Han, S. et al. A metabolomics pipeline for the mechanistic interrogation of the gut microbiome. Nature 595, 415 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Sun, X., Song, W. & Liu, L. Enzymatic production of agmatine by recombinant arginine decarboxylase. J. Mol. Catal. B-Enzym. 121, 1–8 (2015).

    Article  CAS  Google Scholar 

  26. Burrell, M., Hanfrey, C. C., Murray, E. J., Stanley-Wall, N. R. & Michael, A. J. Evolution and multiplicity of arginine decarboxylases in polyamine biosynthesis and essential role in Bacillus subtilis biofilm formation. J. Biol. Chem. 285, 39224–39238 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Bitonti, A. J., Casara, P. J., McCann, P. P. & Bey, P. Catalytic irreversible inhibition of bacterial and plant arginine decarboxylase activities by novel substrate and product analogs. Biochem. J. 242, 69–74 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Pathak, P. et al. Intestine farnesoid X receptor agonist and the gut microbiota activate G-protein bile acid receptor-1 signaling to improve metabolism. Hepatology 68, 1574–1588 (2018).

    Article  CAS  PubMed  Google Scholar 

  29. Yoon, H. S. et al. Akkermansia muciniphila secretes a glucagon-like peptide-1-inducing protein that improves glucose homeostasis and ameliorates metabolic disease in mice. Nat. Microbiol. 6, 563–573 (2021).

    Article  CAS  PubMed  Google Scholar 

  30. Rakipovski, G. et al. The GLP-1 analogs liraglutide and semaglutide reduce atherosclerosis in ApoE(−/−) and LDLr(−/−) mice by a mechanism that includes inflammatory pathways. JACC Basic Transl. Sci. 3, 844–857 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  31. McCartney, C. R. & Marshall, J. C. Polycystic ovary syndrome. N. Engl. J. Med. 375, 54–64 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Wahlstrom, A., Sayin, S. I., Marschall, H.-U. & Backhed, F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab. 24, 41–50 (2016).

    Article  PubMed  Google Scholar 

  33. Teodoro, J. S., Rolo, A. P. & Palmeira, C. M. Hepatic FXR: key regulator of whole-body energy metabolism. Trends Endocrinol. Metab. 22, 458–466 (2011).

    Article  CAS  PubMed  Google Scholar 

  34. Lefebvre, P., Cariou, B., Lien, F., Kuipers, F. & Staels, B. Role of bile acids and bile acid receptors in metabolic regulation. Physiol. Rev. 89, 147–191 (2009).

    Article  CAS  PubMed  Google Scholar 

  35. Jiang, C. et al. Intestine-selective farnesoid X receptor inhibition improves obesity-related metabolic dysfunction. Nat. Commun. 6, 10166 (2015).

    Article  CAS  PubMed  Google Scholar 

  36. Jiang, C. et al. Intestinal farnesoid X receptor signaling promotes nonalcoholic fatty liver disease. J. Clin. Invest. 125, 386–402 (2015).

    Article  PubMed  Google Scholar 

  37. Fiorucci, S., Distrutti, E., Carino, A., Zampella, A. & Biagioli, M. Bile acids and their receptors in metabolic disorders. Prog. Lipid Res. 82, 101094 (2021).

    Article  CAS  PubMed  Google Scholar 

  38. Liu, H., Hu, C., Zhang, X. & Jia, W. Role of gut microbiota, bile acids and their cross-talk in the effects of bariatric surgery on obesity and type 2 diabetes. J. Diabetes Investig. 9, 13–20 (2018).

    Article  PubMed  Google Scholar 

  39. Pyke, C. et al. GLP-1 receptor localization in monkey and human tissue: novel distribution revealed with extensively validated monoclonal antibody. Endocrinology 155, 1280–1290 (2014).

    Article  PubMed  Google Scholar 

  40. Drucker, D. J. The cardiovascular biology of glucagon-like peptide-1. Cell Metab. 24, 15–30 (2016).

    Article  CAS  PubMed  Google Scholar 

  41. Glintborg, D., Mumm, H., Holst, J. J. & Andersen, M. Effect of oral contraceptives and/or metformin on GLP-1 secretion and reactive hypoglycaemia in polycystic ovary syndrome. Endocr. Connect. 6, 267–277 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Vrbikova, J. et al. Incretin levels in polycystic ovary syndrome. Eur. J. Endocrinol. 159, 121–127 (2008).

    Article  CAS  PubMed  Google Scholar 

  43. Gama, R. et al. The entero-insular axis in polycystic ovarian syndrome. Ann. Clin. Biochem. 33, 190–195 (1996).

    Article  PubMed  Google Scholar 

  44. Pontikis, C. et al. The incretin effect and secretion in obese and lean women with polycystic ovary syndrome: a pilot study. J. Women’s Health 20, 971–976 (2011).

    Article  Google Scholar 

  45. Cassar, S. et al. Biomarkers and insulin sensitivity in women with polycystic ovary syndrome: characteristics and predictive capacity. Clin. Endocrinol. 83, 50–58 (2015).

    Article  CAS  Google Scholar 

  46. Abdalla, M. A., Deshmukh, H., Atkin, S. & Sathyapalan, T. The potential role of incretin-based therapies for polycystic ovary syndrome: a narrative review of the current evidence. Ther. Adv. Endocrinol. Metab. 12, 2042018821989238 (2021).

  47. Baranowska-Bik, A. Therapy of obesity in women with PCOS using GLP-1-benefits and limitations. Endokrynol. Pol. 73, 627–643 (2022).

    Article  CAS  PubMed  Google Scholar 

  48. Helvaci, N. & Yildiz, B. O. Current and emerging drug treatment strategies for polycystic ovary syndrome. Expert Opin. Pharmacother. 24, 105–120 (2023).

    Article  CAS  PubMed  Google Scholar 

  49. Morris, S. M. Jr. Arginine metabolism revisited. J. Nutr. 146, 2579–2586 (2016).

    Article  Google Scholar 

  50. Yu, J., Lee, J., Lee, S.-H., Cho, J.-H. & Kim, H.-S. A study on weight loss cause as per the side effect of liraglutide. Cardiovasc. Ther. 2022, 5201684 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Baekdal, T. A. et al. Effect of various dosing conditions on the pharmacokinetics of oral semaglutide, a human glucagon-like peptide-1 analogue in a tablet formulation. Diabetes Ther. 12, 1915–1927 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Saxena, A. R. et al. Efficacy and safety of oral small molecule glucagon-like peptide 1 receptor agonist danuglipron for glycemic control among patients with type 2 diabetes: a randomized clinical trial. JAMA Netw. Open 6, e2314493 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Zheng, X. et al. Hyocholic acid species improve glucose homeostasis through a distinct TGR5 and FXR signaling mechanism. Cell Metab. 33, 791–803.e7 (2020).

    Article  PubMed  Google Scholar 

  54. Liu, C. et al. Gut commensal Christensenella minuta modulates host metabolism via acylated secondary bile acids. Nat. Microbiol 9, 434–450 (2024).

    Article  PubMed  Google Scholar 

  55. Li, T. et al. A gut microbiota-bile acid axis promotes intestinal homeostasis upon aspirin-mediated damage. Cell Host Microbe 32, 191–208.e9 (2024).

    Article  CAS  PubMed  Google Scholar 

  56. Yildiz, M. & Ofori-Mensah, S. The effects of different commercial feeds and seasonal variation on fillet amino acid profile of sea bream (Sparus aurata) and sea bass (Dicentrarchus labrax). Turkish J. Fish. Aquat. Sci. 17, 1297–1307 (2017).

    Google Scholar 

  57. Gorska-Warsewicz, H. et al. Food products as sources of protein and amino acidsthe case of poland. Nutrients 10, 1977 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Guan, F. et al. Amino acids and lipids associated with long-term and short-term red meat consumption in the chinese population: an untargeted metabolomics study. Nutrients 13, 4567 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Rossocha, M., Schultz-Heienbrok, R., von Moeller, H., Coleman, J. P. & Saenger, W. Conjugated bile acid hydrolase is a tetrameric N-terminal thiol hydrolase with specific recognition of its cholyl but not of its tauryl product. Biochemistry 44, 5739–5748 (2005).

    Article  CAS  PubMed  Google Scholar 

  60. Yao, L. et al. A selective gut bacterial bile salt hydrolase alters host metabolism. eLife 7, e37182 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Arifuzzaman, M. et al. Inulin fibre promotes microbiota-derived bile acids and type 2 inflammation. Nature 611, 578–584 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Caldwell, A. S. L. et al. Neuroendocrine androgen action is a key extraovarian mediator in the development of polycystic ovary syndrome. Proc. Natl Acad. Sci. USA 114, E3334–E3343 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–U354 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Mortazavi, A., Williams, B. A., McCue, K., Schaeffer, L. & Wold, B. Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat. Methods 5, 621–628 (2008).

    Article  CAS  PubMed  Google Scholar 

  65. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Natural Science Foundation of the Peoples’ Republic of China (grant nos. 31925021, 82130022, 92357305 and 82341226 to C.J.), the National Key Research and Development Program of China (grant nos. 2022YFA0806400 to C.J. and 2022YFC2702500 to R.L.), the National Natural Science Foundation of the Peoples’ Republic of China (grant nos. 82022028 and 82171627 to Y.P., 82288102 to J.Q., 82001506 to X.Q., 92149306 and 81921001 to C.J., 82301841 to S.Y.). C.J. acknowledges the support from the Tencent Foundation through the Xplorer Prize. We thank L. Dai (Shenzhen Institute of Synthetic Biology, Institute of Advanced Technology) for providing the Bacteroides tool plasmid pB041; D. L. Gumucio (University of Michigan) for providing the villin Cre mice; G. L. Guo (Rutgers University) for providing the PGL4-Shp-TK firefly luciferase construct and human FXR expression plasmid; P. A. Dawson (Emory University) for providing the human ASBT expression plasmid and D. Drucker for providing the GLUTag cell line.

Author information

Authors and Affiliations

Authors

Contributions

Y.P., C.J., J.Q. and R.L. designed the study. C. Yun and X.Q. enroled the patients and collected the patient samples. C. Yun, S.Y., B.L. and M.Z. performed the animal experiments and analysed the data. Y.D. and K.W. guided the microbial genetic operations. S.Y. and Q.N. performed the LC–MS analysis. Y.Z., C. Ye, P.X. and M.M. analysed the metagenomic and RNA sequencing data. S.Y., C. Yun, C.J., J.Q. and Y.P. wrote the paper with input from all authors.

Corresponding authors

Correspondence to Rong Li, Changtao Jiang, Jie Qiao or Yanli Pang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Metabolism thanks John Chiang, Wendong Huang and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Yanina-Yasmin Pesch, 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 Bvbsh colonization leads to a PCOS-like phenotype in mice without affecting the bile acid profile.

a, DNA gel electrophoresis experiment to verify the knockout of Bv1032, Bv2699, and Bv3993 in B. vulgatus. b, Growth curve of wild type B. vulgatus (Bv) and Bv2699 (Bvbsh) in BHI medium. c-l, SPF mice were gavaged with wild type B. vulgatus (Bv), BVU2699-knockout B. vulgatus (Bvbsh), or PBS (Vehicle) for three weeks. c, Colonization of Bv and Bvbsh in mice. d, The total bile acid profile of serum. e, The content of UDCA in feces. f, The content of UDCA in serum. g, The levels of IL-22 in serum. h, Quantitative analysis of cystic follicles in the ovaries. i, Area under the curve (AUC) of GTT. j, The levels of fasting glucose in serum. k, The levels of insulin in serum. l, HOMA-IR. All data are presented as the mean ± s.e.m. (b-g, i-l) n = 6/group; h, n = 5/group. In c, the P values were determined by Kruskal–Wallis test followed by Dunn’s post hoc test. The P values were determined by one-way ANOVA with Tukey’s multiple comparison post hoc test in e, i, j, and one-way ANOVA with LSD test in f-h, k, l.

Source data

Extended Data Fig. 2 Cholestyramine cannot reverse B. vulgatus-induced PCOS-like phenotype.

a and b, Cholestyramine reduces the content of bile acids in mouse intestinal epithelium (a) and serum (b). (a) CA, P = 2.32 × 10−7; α-MCA, P = 0.000008; TCA, P = 2.85 × 10−7. (b) TUDCA, P = 0.000016. c-j, Cholestyramine treatment mice were gavaged with wild type B. vulgatus (Bv) or PBS (Vehicle) for three weeks. c, Quantitative analysis of cystic follicles in the ovaries. d, Levels of testosterone in serum. e, Levels of LH in serum. f, Glucose tolerance test (GTT) (15min: ***P = 0.0007; 30min: *P = 0.0249; *P = 0.0183; *compared with the vehicle + Cholestyramine). g, Area under the curve (AUC) of GTT. h, Insulin tolerance test (ITT) (15min: *P = 0.0152; 30min: **P = 0.0087; *compared with the vehicle + Cholestyramine). i, The levels of fasting glucose in serum. j, The level of insulin in serum. All data are presented as the mean ± s.e.m. (a, b, c) n = 5/group. (d-j) n = 6/group. In a, b, d-g, i, j, the P values were determined by two-tailed Student’s t-test. In c, h, the P values were determined by two-tailed Mann–Whitney U-test.

Source data

Extended Data Fig. 3 GLP-1R agonist liraglutide improves B. vulgatus-induced PCOS-like phenotype.

a, The levels of GIP in patients with PCOS and healthy control serum. b, The levels of GDF15 in patients with PCOS and healthy control serum. c, The correlation between the total GLP-1 level in the serum and LH/FSH ratio. d, The correlation between the total GLP-1 level in the serum and HOMA-IR. e, Quantitative analysis of cystic follicles in the ovaries. f, Area under the curve (AUC) of GTT. g, The levels of fasting glucose in serum. h, The level of insulin in serum. All data are presented as the mean ± s.e.m. (a-d) n = 20 in the control group and n = 20 in the PCOS group. (e) n = 5/group. (f-h) n = 6/group. In a, the P values were determined by two-tailed Mann–Whitney U-test. In b, the P values were determined by two-tailed Student’s t-test. In c, d, correlation analysis was determined by one tailed Pearson r-test. In e, the P values were determined by Kruskal–Wallis test followed by Dunn’s post hoc test. In f-h, the P values were determined by one-way ANOVA with Tukey’s multiple comparison post hoc test.

Source data

Extended Data Fig. 4 B. vulgatus inhibits GLP-1 secretion and induces PCOS-like phenotype depended on intestinal epithelial FXR.

a, The expression levels of Fgf15 and Shp in intestinal epithelium. b, The levels of C4 in serum. c, FXR expression in siFXR STC-1 cells. d, The levels of total GLP-1 in the cellular supernatant. e, The relative expression of Gcg. f, The levels of total GLP-1 in the cellular supernatant. g, Luciferase activity. h, The expression levels of Tgr5 in GLUTag cells. i, The levels of total GLP-1 in the cellular supernatant. j, Relative fold change of total GLP-1 levels to vehicle. k, The expression levels of Fxr and downstream genes in the intestinal epithelium of Fxrfl/fl and FxrΔIE mice. l-q, FxrΔIE or Fxrfl/fl mice were gavaged with Bv or PBS for three weeks. l, Quantitative analysis of corpora lutea. m, Quantitative analysis of cystic follicles. n, AUC of GTT. o, The levels of fasting glucose in serum. p, The level of insulin in serum. q, HOMA-IR. All data are presented as the mean ± s.e.m. (a, k-q) n = 5/group. (b) n = 20 in both control and PCOS group. (c, h) n = 3/group. (d-g, i, j) n = 4/group. In a, the P values were determined by two-tailed Mann–Whitney U-test. In b, d, e, k, the P values were determined by two-tailed Student’s t-test. The P values were determined by one-way ANOVA with Tukey’s multiple comparison post hoc test in c, f, h, m, o-q; one-way ANOVA with Dunnett’s T3 test in g and one-way ANOVA with LSD test in n. In i, j, l, the P values were determined by Kruskal–Wallis test followed by Dunn’s post hoc test.

Source data

Extended Data Fig. 5 GLP-1 receptor inhibitor reversed the improvement of B. vulgatus-induced PCOS-like phenotype in intestinal Fxr knockout mice.

a, Quantitative analysis of corpora lutea in the ovaries. b, Quantitative analysis of cystic follicles in the ovaries. c, Area under the curve (AUC) of GTT. d, The levels of fasting glucose in serum. e, The level of insulin in serum. f, HOMA-IR. All data are presented as the mean ± s.e.m. (a-f) n = 5/group. In a, b, d, the P values were determined by Kruskal–Wallis test followed by Dunn’s post hoc test. In c, e, f, the P values were determined by one-way ANOVA with LSD test.

Source data

Extended Data Fig. 6 Agmatine induces PCOS-like phenotype via a FXR signaling pathway.

a, The content of cytidine 5′-diphosphocholine in the feces of B. vulgatus model mice. b-h, FxrΔIE or Fxrfl/fl mice were gavaged with agmatine or PBS (Vehicle) for three weeks. b, Quantitative analysis of cystic follicles in the ovaries. c, Levels of testosterone in serum. d, Levels of LH in serum. e, Area under the curve (AUC) of GTT. f, The levels of fasting glucose in serum. g, The level of insulin in serum. h, HOMA-IR. All data are presented as the mean ± s.e.m. (a) n = 6/group. (b-h) n = 5/group. In a, the P values were determined by two-tailed Student’s t-test. In b, the P values were determined by Kruskal–Wallis test followed by Dunn’s post hoc test. The P values were determined by one-way ANOVA with Tukey’s multiple comparison post hoc test in c-f, and one-way ANOVA with LSD test in g, h.

Source data

Extended Data Fig. 7 The ability of BvspeA to induce PCOS-like phenotype is significantly reduced compared to B. vulgatus.

a, The percentage Bacteroides vulgatus strains with or without speA. b, The relative abundance of speA gene in the metagenomic dataset from PMID: 31332392. n = 25 in Control (BMI < 25) group; n = 25 in PCOS (BMI < 25) group; n = 18 in Control (BMI ≥ 25) group; n = 25 in PCOS (BMI ≥ 25) group. For box plots, the midline represents the median; box represents the interquartile range (IQR) between the first and third quartiles, and whiskers represent the minimum to maximum values. Bacteroides vulgatus: Control (BMI < 25) vs PCOS (BMI < 25) P = 2.56 × 10−7, Control (BMI ≥ 25) vs PCOS (BMI ≥ 25) P = 0.000015; Bifidobacterium longum: Control (BMI ≥ 25) vs PCOS (BMI ≥ 25) P = 0.000025. c, DNA gel electrophoresis experiment to verify the knockout of BvspeA in B. vulgatus. d, Growth curve of wild type B. vulgatus (Bv) and BvspeA in BHI medium. e-m, SPF mice were gavaged with Bv or BvspeA for three weeks. e, Colonization of Bv and BvspeA in mice. f, Quantitative analysis of corpora lutea in the ovaries. g, Quantitative analysis of cystic follicles in the ovaries. h, Levels of testosterone in serum. i, Levels of LH in serum. j, Area under the curve (AUC) of GTT. k, The levels of fasting glucose in serum. l, The level of insulin in serum. m, HOMA-IR. All data are presented as the mean ± s.e.m. (d) n = 3/group. (e, h-m) n = 6/group. (f, g) n = 5/group. In b, the P values were determined by Kruskal–Wallis test followed by Dunn’s post hoc test. In d, i-m, the P values were determined by two-tailed Student’s t-test. In e-h, the P values were determined by two-tailed Mann–Whitney U-test.

Source data

Extended Data Fig. 8 The arginine decarboxylase (ADC) inhibitor DFMA improves the B. vulgatus-induced PCOS-like phenotype.

a, Inhibition rate of agmatine production by adding DFMA to BHI medium. b, Growth curve of Bv and Bv + DFMA in BHI medium. c-l, SPF mice were gavaged with wild type B. vulgatus (Bv), and treated with PBS or arginine decarboxylase inhibitor DFMA for three weeks. c, Quantitative analysis of corpora lutea in the ovaries. d, Quantitative analysis of cystic follicles in the ovaries. e, Levels of testosterone in serum. f, Levels of LH in serum. g, Glucose tolerance test (GTT) (15min: *P = 0.0401; 30min:*P = 0.0203; * compared with the Bv group). h, Area under the curve (AUC) of GTT. i, Insulin tolerance test (ITT) (15min: ***P = 0.000003; 30min: ***P = 0.0006; 60min: *P = 0.0342; 90min: *P =0.0181; * compared with the Bv group). j, The levels of fasting glucose in serum. k, The level of insulin in serum. l, HOMA-IR. All data are presented as the mean ± s.e.m. (a, b) n = 3/group. (c and d) n = 5/group. (e-l) n = 6/group. In b, e-l, the P values were determined by two-tailed Student’s t-test. In c, d, the P values were determined by two-tailed Mann–Whitney U-test.

Source data

Extended Data Fig. 9 The arginine decarboxylase (ADC) inhibitor DFMA improves the DHEA-induced hyperandrogenic PCOS-like phenotype.

a, The relative abundance of B. vulgatus in the DHEA model. b, The concentration of agmatine in feces. c, The concentration of agmatine in intestinal epithelium. d, Expression levels of Fgf15 and Shp in intestinal epithelium. e, Levels of total GLP-1 in serum. f, Quantitative analysis of estrous cycles. g, Hematoxylin and eosin staining of representative ovaries. The corpora lutea are indicated by * and the cystic follicle is indicated by #. Scale bar, 200 μm. h, Quantitative analysis of corpora lutea. i, Quantitative analysis of cystic follicles. j, Levels of testosterone in serum. k, Levels of LH in serum. l, Glucose tolerance test (GTT) (15min: ***P = 0.000026, ##P = 0.0034; 30min: ***P = 0.0002, #P = 0.0103; 60min: *P = 0.0297; * compared with the vehicle group, # compared with the DHEA group.). m, Area under the curve (AUC) of GTT. n, Insulin tolerance test (ITT) (15min: **P = 0.0048; 30min: *P = 0.0214; * compared with the vehicle group). o, The levels of fasting glucose in serum. p, The level of insulin in serum. q, HOMA-IR. r, Expression levels of Ucp1, Pgc1a, Cited1, Cox8b, Nr2f6, Prdm16 in the subcutaneous fat. s, Expression levels of Ucp1, Pgc1a, Cited1, Cox8b in the brown adipose tissue. t, Expression levels of Il-8, Il-6, Il-1β, Il-18, Ifng, Ccl20, Ccl2, in the ovaries. All data are presented as the mean ± s.e.m. (a-f, j-t) n = 6/group. (h, i) n = 5/group. In a, the P values were determined by two-tailed Student’s t-test. The P values were determined by one-way ANOVA with Tukey’s multiple comparison post hoc test in c, h-j, l, m, o, q, one-way ANOVA with Dunnett’s T3 test in b and one-way ANOVA with LSD test in k, n. In d-f, p, r-t the P values were determined by Kruskal–Wallis test followed by Dunn’s post hoc test.

Source data

Extended Data Fig. 10 The speA and bsh of B. vulgatus can participate in the development of PCOS-like phenotypes in mice independently.

a, DNA gel electrophoresis experiment to verify the knockout of BvspeAΔbsh in B. vulgatus. b, Growth curve of wild type B. vulgatus (Bv) and Bv-ΔspeAΔbsh in BHI medium. c, The content of agmatine after 24 h cultured with B. vulgatus or Bv-ΔspeAΔbsh in BHI medium. d, The ability of hydrolyzing TUDCA (12 h) after knocking out both the speA and bsh in B. vulgatus. e, Colonization of Bv and BvspeA, Bvbsh and Bv-ΔspeAΔbsh in cecum of mice. f, Quantitative analysis of cystic follicles in the ovaries. g, Area under the curve (AUC) of GTT. h, The levels of fasting glucose in serum. i, The level of insulin in serum. j, Expression levels of Ucp1, Pgc1a, Cited1, Cox8b, Nr2f6, Prdm16 in the subcutaneous fat. Ucp1: Vehicle versus Bv, P = 8.06 × 10−7; Bv versus Bv-ΔspeAΔbsh, P = 8.09 × 10−7. k, Expression levels of Ucp1, Pgc1a, Cited1, Cox8b in the brown adipose tissue. Ucp1: Vehicle versus Bv, P = 0.000019. l, Expression levels of Il-8, Il-6, Il-1β, Il-18, Ifng, Ccl20, Ccl2, in the ovaries. All data are presented as the mean ± s.e.m. (b, e, g-l) n = 6/group. (c, d) n = 3/group. (f) n = 5/group. The P values were determined by one-way ANOVA with Dunnett’s T3 test in c, d, one-way ANOVA with Tukey’s multiple comparison post hoc test in g, k and one-way ANOVA with LSD test in f, h-j. In e, l, the P values were determined by Kruskal–Wallis test followed by Dunn’s post hoc test.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–4 and Tables 1 and 2.

Reporting Summary

Source data

Source Data Fig. 1

Statistical 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 Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed gels.

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.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed gels.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Source Data Extended Data Fig. 10

Unprocessed gels.

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

Yun, C., Yan, S., Liao, B. et al. The microbial metabolite agmatine acts as an FXR agonist to promote polycystic ovary syndrome in female mice. Nat Metab 6, 947–962 (2024). https://doi.org/10.1038/s42255-024-01041-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-024-01041-8

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing