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:

GDF15 mediates the effects of metformin on body weight and energy balance

A Publisher Correction to this article was published on 13 February 2020

This article has been updated

Abstract

Metformin, the world’s most prescribed anti-diabetic drug, is also effective in preventing type 2 diabetes in people at high risk1,2. More than 60% of this effect is attributable to the ability of metformin to lower body weight in a sustained manner3. The molecular mechanisms by which metformin lowers body weight are unknown. Here we show—in two independent randomized controlled clinical trials—that metformin increases circulating levels of the peptide hormone growth/differentiation factor 15 (GDF15), which has been shown to reduce food intake and lower body weight through a brain-stem-restricted receptor. In wild-type mice, oral metformin increased circulating GDF15, with GDF15 expression increasing predominantly in the distal intestine and the kidney. Metformin prevented weight gain in response to a high-fat diet in wild-type mice but not in mice lacking GDF15 or its receptor GDNF family receptor α-like (GFRAL). In obese mice on a high-fat diet, the effects of metformin to reduce body weight were reversed by a GFRAL-antagonist antibody. Metformin had effects on both energy intake and energy expenditure that were dependent on GDF15, but retained its ability to lower circulating glucose levels in the absence of GDF15 activity. In summary, metformin elevates circulating levels of GDF15, which is necessary to obtain its beneficial effects on energy balance and body weight, major contributors to its action as a chemopreventive agent.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Effect of Metformin on circulating GDF15 levels in humans and mice.
Fig. 2: GDF15–GFRAL signalling is required for the weight-loss effects of metformin on a high-fat diet.
Fig. 3: Effects of metformin on glucose homeostasis.
Fig. 4: Metformin increases GDF15 expression in the enterocytes of distal intestine and in renal tubular epithelial cells.

Similar content being viewed by others

Data availability

Source Data for Figs. 14 and Extended Data Figs. 16, 810 are provided with the paper. Other data that support the findings of this study are available from the corresponding authors upon request. The CAMERA trial dataset is held at the University of Glasgow and is available on request from the investigators subject to a signed agreement operating within the confines of the original ethics application.

Change history

References

  1. Knowler, W. C. et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N. Engl. J. Med. 346, 393–403 (2002).

    Article  CAS  Google Scholar 

  2. Ramachandran, A. et al. The Indian Diabetes Prevention Programme shows that lifestyle modification and metformin prevent type 2 diabetes in Asian Indian subjects with impaired glucose tolerance (IDPP-1). Diabetologia 49, 289–297 (2006).

    Article  CAS  Google Scholar 

  3. Lachin, J. M. et al. Factors associated with diabetes onset during metformin versus placebo therapy in the diabetes prevention program. Diabetes 56, 1153–1159 (2007).

    Article  CAS  Google Scholar 

  4. Rena, G., Hardie, D. G. & Pearson, E. R. The mechanisms of action of metformin. Diabetologia 60, 1577–1585 (2017).

    Article  CAS  Google Scholar 

  5. Apolzan, J. W. et al. Long-term weight loss with metformin or lifestyle intervention in the Diabetes Prevention Program Outcomes Study. Ann. Intern. Med. 170, 682–690 (2019).

    Article  Google Scholar 

  6. Gerstein, H. C. et al. Growth differentiation factor 15 as a novel biomarker for metformin. Diabetes Care 40, 280–283 (2017).

    Article  CAS  Google Scholar 

  7. Tsai, V. W. W., Husaini, Y., Sainsbury, A., Brown, D. A. & Breit, S. N. The MIC-1/GDF15–GFRAL pathway in energy homeostasis: implications for obesity, cachexia, and other associated diseases. Cell Metab. 28, 353–368 (2018).

    Article  CAS  Google Scholar 

  8. Mullican, S. E. et al. GFRAL is the receptor for GDF15 and the ligand promotes weight loss in mice and nonhuman primates. Nat. Med. 23, 1150–1157 (2017).

    Article  CAS  Google Scholar 

  9. Emmerson, P. J. et al. The metabolic effects of GDF15 are mediated by the orphan receptor GFRAL. Nat. Med. 23, 1215–1219 (2017).

    Article  CAS  Google Scholar 

  10. Yang, L. et al. GFRAL is the receptor for GDF15 and is required for the anti-obesity effects of the ligand. Nat. Med. 23, 1158–1166 (2017).

    Article  CAS  Google Scholar 

  11. Hsu, J. Y. et al. Non-homeostatic body weight regulation through a brainstem-restricted receptor for GDF15. Nature 550, 255–259 (2017).

    Article  ADS  Google Scholar 

  12. Konopka, A. R. et al. Hyperglucagonemia mitigates the effect of metformin on glucose production in prediabetes. Cell Rep. 15, 1394–1400 (2016).

    Article  CAS  Google Scholar 

  13. Preiss, D. et al. Metformin for non-diabetic patients with coronary heart disease (the CAMERA study): a randomised controlled trial. Lancet Diabetes Endocrinol. 2, 116–124 (2014).

    Article  CAS  Google Scholar 

  14. McCreight, L. J. et al. Pharmacokinetics of metformin in patients with gastrointestinal intolerance. Diabetes Obes. Metab. 20, 1593–1601 (2018).

    Article  CAS  Google Scholar 

  15. Forouhi, N. G., Luan, J., Hennings, S. & Wareham, N. J. Incidence of Type 2 diabetes in England and its association with baseline impaired fasting glucose: the Ely study 1990-2000. Diabet. Med. 24, 200–207 (2007).

    Article  CAS  Google Scholar 

  16. Chung, H. K. et al. Growth differentiation factor 15 is a myomitokine governing systemic energy homeostasis. J. Cell Biol. 216, 149–165 (2017).

    Article  CAS  Google Scholar 

  17. Li, D., Zhang, H. & Zhong, Y. Hepatic GDF15 is regulated by CHOP of the unfolded protein response and alleviates NAFLD progression in obese mice. Biochem. Biophys. Res. Commun. 498, 388–394 (2018).

    Article  CAS  Google Scholar 

  18. Patel, S. et al. GDF15 provides an endocrine signal of nutritional stress in mice and humans. Cell Metab. 29, 707–718 (2019).

    Article  CAS  Google Scholar 

  19. Shu, Y. et al. Effect of genetic variation in the organic cation transporter 1 (OCT1) on metformin action. J. Clin. Invest. 117, 1422–1431 (2007).

    Article  CAS  Google Scholar 

  20. 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  Google Scholar 

  21. Preiss, D. et al. Sustained influence of metformin therapy on circulating glucagon-like peptide-1 levels in individuals with and without type 2 diabetes. Diabetes Obes. Metab. 19, 356–363 (2017).

    Article  CAS  Google Scholar 

  22. Bahne, E. et al. Metformin-induced glucagon-like peptide-1 secretion contributes to the actions of metformin in type 2 diabetes. JCI Insight 3, 93936 (2018).

    Article  Google Scholar 

  23. Maida, A., Lamont, B. J., Cao, X. & Drucker, D. J. Metformin regulates the incretin receptor axis via a pathway dependent on peroxisome proliferator-activated receptor-α in mice. Diabetologia 54, 339–349 (2011).

    Article  CAS  Google Scholar 

  24. de la Cuesta-Zuluaga, J. et al. Metformin is associated with higher relative abundance of mucin-degrading Akkermansia muciniphila and several short-chain fatty acid-producing microbiota in the gut. Diabetes Care 40, 54–62 (2017).

    Article  Google Scholar 

  25. 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  Google Scholar 

  26. Forslund, K. et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature 528, 262–266 (2015).

    Article  CAS  Google Scholar 

  27. Foretz, M., Guigas, B. & Viollet, B. Understanding the glucoregulatory mechanisms of metformin in type 2 diabetes mellitus. Nat. Rev. Endocrinol. 15, 569–589 (2019).

    Article  CAS  Google Scholar 

  28. Massollo, M. et al. Metformin temporal and localized effects on gut glucose metabolism assessed using 18F-FDG PET in mice. J. Nucl. Med. 54, 259–266 (2013).

    Article  CAS  Google Scholar 

  29. 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  Google Scholar 

  30. Skarnes, W. C. et al. A conditional knockout resource for the genome-wide study of mouse gene function. Nature 474, 337–342 (2011).

    Article  CAS  Google Scholar 

  31. Bradley, A. et al. The mammalian gene function resource: the International Knockout Mouse Consortium. Mamm. Genome 23, 580–586 (2012).

    Article  Google Scholar 

  32. Pettitt, S. J. et al. Agouti C57BL/6N embryonic stem cells for mouse genetic resources. Nat. Methods 6, 493–495 (2009).

    Article  CAS  Google Scholar 

  33. McNeilly, A. D., Williamson, R., Balfour, D. J., Stewart, C. A. & Sutherland, C. A high-fat-diet-induced cognitive deficit in rats that is not prevented by improving insulin sensitivity with metformin. Diabetologia 55, 3061–3070 (2012).

    Article  CAS  Google Scholar 

  34. Ortega-Cava, C. F. et al. Strategic compartmentalization of Toll-like receptor 4 in the mouse gut. J. Immunol. 170, 3977–3985 (2003).

    Article  CAS  Google Scholar 

  35. Goldspink, D. A. et al. Mechanistic insights into the detection of free fatty and bile acids by ileal glucagon-like peptide-1 secreting cells. Mol. Metab. 7, 90–101 (2018).

    Article  CAS  Google Scholar 

  36. Sato, T. et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology 141, 1762–1772 (2011).

    Article  CAS  Google Scholar 

  37. Rashid, S. T. et al. Modeling inherited metabolic disorders of the liver using human induced pluripotent stem cells. J. Clin. Invest. 120, 3127–3136 (2010).

    Article  CAS  Google Scholar 

  38. Yusa, K. et al. Targeted gene correction of α1-antitrypsin deficiency in induced pluripotent stem cells. Nature 478, 391–394 (2011).

    Article  ADS  CAS  Google Scholar 

  39. Chen, G. et al. Chemically defined conditions for human iPSC derivation and culture. Nat. Methods 8, 424–429 (2011).

    Article  CAS  Google Scholar 

  40. Hannan, N. R., Segeritz, C. P., Touboul, T. & Vallier, L. Production of hepatocyte-like cells from human pluripotent stem cells. Nat. Protoc. 8, 430–437 (2013).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The CAMERA trial is funded by a project grant from the Chief Scientist Office, Scotland (CZB/4/613). D.P. is supported by a University of Oxford British Heart Foundation Centre of Research Excellence Senior Transition Fellowship (RE/13/1/30181). N.S. and P.W. acknowledge support from a BHF Centre of Excellence award (COE/RE/18/6/34217). We thank P. Barker, K. Burling and other members of the Cambridge Biochemical Assay Laboratory (CBAL). This project is supported by the National Institute for Health Research (NIHR) Cambridge Biomedical Research Centre. The views expressed are those of the authors and not necessarily those of the NIHR or the Department of Health and Social Care. A.P.C., D. Rimmington, J.A.T., I.C., Y.C.L.T. and G.S.H.Y. are supported by the Medical Research Council (MRC Metabolic Diseases Unit (MC_UU_00014/1)). Mouse studies in Cambridge are supported by S. Grocott and the Disease Model Core, with pathology support from J. Warner and the Histopathology Core (MRC Metabolic Diseases Unit (MC_UU_00014/5) and Wellcome Trust Strategic Award (100574/Z/12/Z). D.B.S. and S.O. are supported by the Wellcome Trust (WT 107064 and WT 095515/Z/11/Z), the MRC Metabolic Disease Unit (MC_UU_00014/1) and The National Institute for Health Research (NIHR) Cambridge Biomedical Research Centre and NIHR Rare Disease Translational Research Collaboration. We thank J. Jones and other members of the Histopathology and ISH Core Facility, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre. D. Ron is supported by a Wellcome Trust Principal Research Fellowship (Wellcome 200848/Z/16/Z) and a Wellcome Trust Strategic Award to the Cambridge Institute for Medical Research (Wellcome 100140). A.V.-P., S.R.-C. and S.V. are supported by the BHF (RG/18/7/33636) and MRC (MC_UU_00014/2). A.M. is supported by a studentship from the Experimental Medicine Training Initiative/AstraZeneca. R.A.T. and L.V. are supported by an ERC advanced grant NewChol and core support from the Wellcome Trust and Medical Research Council to the Wellcome–Medical Research Council Cambridge Stem Cell Institute. M.Y., D.A.G., E.L.M., F.M.G. and F.R. are supported by the MRC (MC_UU_00014/3) and Wellcome Trust (106262/Z/14/Z and 106263/Z/14/Z). M.Y. is supported by a BBSRC-DTP studentship. A.R.K., R.R.E. and K.S.N. are supported by NIH Grants R21 AG60139, UL1 TR000135 and T32DK007352 and acknowledge K. Klaus for technical assistance. N.J.W. is supported by the MRC (MC_UU_12015/1) and is an NIHR Senior Investigator. We acknowledge J. Luan for statistical assistance. CHOP-null mice were a gift from J. Goodall.

Author information

Authors and Affiliations

Authors

Contributions

The overall conceptualization of studies included in this work was done by A.P.C., N.S., D.B.S., B.B.A. and S.O.: these authors contributed equally to this work. A.P.C., M.C., P.T., D. Rimmington, I.C. and Y.C.L.T. designed, managed, performed and analysed data from mouse experiments. S.V. designed experiments and analysed data. A.M. and G.S.H.Y. contributed to conceptualization of experiments and data analysis. J.A.T. performed in situ hybridization experiments. S.P. designed, managed and performed cell based assays along with E.L.M., S.R.-C., R.A.T., H.P.H., A.V-P., L.V. and D. Ron. J.T.-J.H. performed measurement of serum metformin levels. M.Y., D.A.G., F.M.G. and F.R. designed, performed and analysed organoid experiments. A.R.K., R.R.E. and K.S.N. designed and performed short-term metformin studies in humans. N.J.W. analysed the Ely Study Cohort. P.W., D.P. and N.S. designed, analysed and interpreted data arising from the CAMERA study. A.P.C., D.B.S., B.B.A. and S.O. wrote the paper, which was reviewed and edited by all the authors.

Corresponding authors

Correspondence to Anthony P. Coll or Stephen O’Rahilly.

Ethics declarations

Competing interests

P.W. has received grant support from Roche Diagnostics, AstraZeneca and Boehringer Ingelheim. N.S. has consulted for AstraZeneca, Boehringer Ingelheim, Eli Lilly, Napp, Novo Nordisk and Sanofi, and received grant support from Boehringer Ingelheim. M.C., P.T. and B.B.A. are or were employees of NGM Biopharmaceuticals and may hold NGM stock or stock options. F.R. and F.M.G. have received support from AstraZeneca and Eli Lilly. F.M.G. has provided remunerated consultancy services to Kallyope. S.O. has provided remunerated consultancy services to Pfizer, AstraZeneca, Novo-Nordisk and ERX Pharmaceuticals. All other authors declare no competing interests.

Additional information

Peer review information Nature thanks Samuel Breit, Daniel Drucker, Jerrold M. Olefsky, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data figures and tables

Extended Data Fig. 1 Expanded CAMERA dataset.

a, Linear association between change in body weight and change in plasma GDF15 between 0 and 18 months among metformin-treated participants (n = 74, Spearman correlation r = –0.26, two-sided P = 0.024). The red line is the linear regression slope, and grey area is 95% CI for the slope. b, Absolute and relative differences in plasma GDF15 concentration between metformin and placebo groups at each time point (total 625 observations in 173 participants). c, d, Individual measures of plasma GDF15 levels in placebo group (c) and metformin group (d) over time. e, Plasma GDF15 concentration (95% CI) in overweight or obese non-diabetic participants with known cardiovascular disease randomized to metformin or placebo in CAMERA; modelled using a mixed linear model as per Fig. 1 and grouped as ‘all participants’ and ‘all participants not reporting diarrhoea and vomiting’. Model includes all participants.

Source data

Extended Data Fig. 2 Effect of single oral dose of metformin in chow-fed mice.

Serum GDF15 levels in male mice measured 2, 4 or 8 h after a single gavage dose of metformin (300 mg kg−1). a, Mice fed ad libitum overnight before gavage. b, Mice fasted for 12 h before gavage. Data are mean ± s.e.m. (a; n = 6 per group, b; n = 4 per group); P values by two-way ANOVA with Tukey’s correction for multiple comparisons.

Source data

Extended Data Fig. 3 Body weight changes with metformin treatment in mice with disrupted GDF15–GFRAL signalling.

a, Absolute body weight in Gdf15+/+ and Gdf15−/− mice on a high-fat diet treated with metformin (300 mg kg−1 day−1) for 11 days, mice as in Fig. 2a. Data are mean ± s.e.m., P values by two-way ANOVA with Tukey’s correction for multiple comparisons. b, Absolute body weight in high-fat diet-fed Gfral+/+ and Gfral−/− mice given an oral dose of metformin (300 mg kg−1) once daily for 11 days, mice as in Fig. 2c. Data are mean ± s.e.m. c, Absolute body weight of metformin-treated, obese mice dosed with an anti-GFRAL antagonist antibody or with control IgG weekly for five weeks, starting four weeks after initial metformin exposure; mice as in Fig. 2d. Data are mean ± s.e.m. P values by two-way ANOVA with Tukey’s correction for multiple comparisons.

Source data

Extended Data Fig. 4 Response of high-fat diet-fed Gdf15−/− and Gfral−/− mice to metformin.

a, Circulating GDF15 levels in high-fat diet-fed Gdf15+/+ and Gdf15−/− mice given oral dose of metformin (300 mg kg−1) once daily for 11 days. Data are mean ± s.e.m., mice as in Fig. 2a. All Gdf15−/− samples were below lower limit of the assay (<2 pg ml−1); P values by two-way ANOVA with Tukey’s correction for multiple comparisons. b, Circulating GDF15 levels in high-fat diet-fed Gfral+/+ and Gfral−/− mice given oral dose of metformin (300 mg kg−1) once daily for 11 days. Data are mean ± s.e.m., mice as in Fig. 2c; P values by two-way ANOVA with Tukey’s correction for multiple comparisons. c, Cumulative food intake in high-fat diet fed Gfral+/+ and Gfral−/− mice on a high-fat diet given an oral dose of metformin (300 mg kg−1) once daily for 11 days. Data are mean ± s.e.m., mice as in Fig. 2c; no statistically significant difference in vehicle versus metformin by two-way ANOVA. d, Fat mass (left) and lean mass (right) in metformin-treated obese mice dosed with anti-GFRAL antagonist antibody weekly for five weeks, starting four weeks after initial metformin exposure (mice as in Fig. 2d). Body composition was measured using MRI after 4 weeks of metformin exposure, before receiving anti-GFRAL (week 4), after 6 weeks of metformin exposure and 2 weeks after receiving anti-GFRAL (week 6) and after 9 weeks of metformin exposure and 5 weeks after receiving anti-GFRAL (week 9). Data are mean ± s.e.m. (n = 7, vehicle + control IgG and metformin + anti-GFRAL; n = 8, other groups); P values by two-way ANOVA with Tukey’s correction for multiple comparisons.

Source data

Extended Data Fig. 5 Response of second, independent cohort of high-fat diet fed Gdf15+/+ and Gdf15−/− mice to metformin.

ac, Percentage change in body weight (a), absolute body weight (b) and cumulative food intake (c) of Gdf15+/+ and Gdf15−/− mice on a high-fat diet treated with metformin (300 mg kg−1 day−1) for 11 days. Data are mean ± s.e.m. (n = 6 per group, except Gdf15−/− vehicle, n = 7); P values by two-way ANOVA with Tukey’s correction for multiple comparisons. d, Circulating metformin levels in mice 6 h after final dose of metformin on day 11. Data are mean ± s.e.m. (n = 6 per group, except Gdf15+/+ vehicle, n = 4; Gdf15−/− vehicle, n = 7); P values by two-way ANOVA with Tukey’s correction for multiple comparisons.

Source data

Extended Data Fig. 6 Glucose, insulin and GDF15 response to metformin.

a, Fasting glucose from oral GTT as in Fig. 3e, f. ANOVA; effect of antibody, P = 0.028; effect of metformin, P = 0.271; interaction of antibody and metformin, P = 0.707. b, Circulating GDF15 in mice undergoing intraperitoneal GTT after a single dose of metformin as in Fig. 3 k, l. P values by two-way ANOVA with Tukey’s correction for multiple comparisons. c, d, Fasting glucose (c) and fasting insulin (d) at time 0 of intraperitoneal GTT as in Fig. 3k, l; not statistically significant by two-way ANOVA. e, AUC analysis of glucose levels as in Fig. 3k, l. P values by two-way ANOVA, effect of genotype, P = 0.392; interaction of genotype and metformin, P = 0.883. ae, Data all mean ± s.e.m. f, Circulating GDF15 levels in high-fat-diet-fed Gdf15+/+ mice after single oral dose of metformin (600 mg kg−1). Samples were collected 6 h after dosing, data are mean ± s.e.m., n = 7 per group; P values (95% CI) by two-tailed t-test.

Source data

Extended Data Fig. 7 In situ hybrididation for Gdf15 mRNA expression in gut, liver and kidney.

a, Representative images from the mouse with circulating GDF15 level closest to the group median shown in Fig. 4b, with images from other regions of the gut and from liver. b, In situ hybridization for Gdf15 mRNA expression (red spots) in colon. Tissue collected from high-fat-diet-fed wild-type mice, 6 h after a single dose of oral metformin (600 mg kg−1) (right, red box, M1–M7) or vehicle gavage (left, blue box, V1–V7); n = 7 mice per group, mice as in Fig. 4.

Extended Data Fig. 8 Analysis of Gdf15 mRNA expression (normalized to expression levels of Actb) in tissue from high-fat diet-fed Gdf15+/+ mice.

Metformin treatment (300 mg kg−1) once daily for 11 days (see Fig. 2a). Data are mean ± s.e.m., n = 6 metformin, n = 7 vehicle; P values (95% CI) by two-tailed t-test.

Source data

Extended Data Fig. 9 Hepatic GDF15 response to biguanides.

a, b,Gdf15 mRNA expression in primary mouse hepatocytes (a) or human iPS-cell-derived hepatocytes (b) treated with vehicle control (Con) or metformin for 6 h. mRNA expression is presented as fold expression relative to control treatment (set at 1), normalized to Hprt and GAPDH in mouse and human cells, respectively. Data are expressed as mean ± s.e.m. from four (a) or two (b) independent experiments. P values (95% CI) by one-way ANOVA with Tukey’s correction for multiple comparisons. c, d, Circulating levels of GDF15 (c) and hepatic Gdf15 mRNA expression (d) (normalized to β2-microglobulin) in chow-fed, wild-type mice 4 h after a single oral dose of phenformin (300 mg kg−1). Data are mean ± s.e.m., n = 6 per group; P values (95% CI) by two tailed t-test. e, Representative image of in situ hybridization for Gdf15 mRNA expression (red spots) of fixed liver tissue derived from animals treated as described in c and d.

Source data

Extended Data Fig. 10 Role of the ISR in biguanide-induced Gdf15 expression.

a, b, mRNA levels in kidney (a) and colon (b) isolated from obese mice 24 h after a single oral dose of metformin (600 mg kg−1). Data are mean ± s.e.m. (n = 5 per group, except for= 4 for colon metformin Slc22a1). P values (95% CI) by two-tailed t-test. Gdf15 mRNA fold induction 24 h after metformin (600 mg kg−1) is positively correlated with Chop mRNA induction in both kidney (a, right) and colon (b, right). Black line shows linear regression analysis. c–g, Immunoblot analysis of ISR components (c) and Gdf15 mRNA expression (d) in wild-type mouse embryonic fibroblasts (MEF) treated with vehicle control (Con), metformin (Met, 2 mM) or phenformin (Phen, 5 mM) or tunicamycin (Tn, 5 μg ml−1, used as a positive control) for 6 h. eg, Gdf15 mRNA expression in ATF4 knockout (KO) MEFs (e), in control siRNA and CHOP siRNA transfected wild-type MEFs treated with Tn or Phen for 6 h (f), or in wild-type MEFs pre-treated for 1 h with either the PERK inhibitor GSK2606414 (GSK, 200 nM) or eIF2α inhibitor ISRIB (ISR, 100 nM) then co-treated with phenformin for a further 6 h (g). mRNA expression is presented as fold expression relative to its respective control treatment (set at 1) or phenformin-treated samples (set as 100) with normalization to Hprt gene expression. Data are mean ± s.e.m. from two (c, d) or at least three (eg) independent experiments. P values (95% CI) by two tailed t-test relative to phenformin-treated control wild-type and control siRNA-treated samples. h, GDF15 protein in supernatant of mouse derived 2D duodenal organoids treated with metformin in the absence or presence of ISRIB (1 μM). Data are expressed as mean ± s.e.m. from two independent experiments. At least duplicate protein measurements for each sample. P values by two-way ANOVA with Sidak’s correction for multiple comparisons. i, GDF15 protein in supernatants of mouse-derived 2D duodenal organoids from wild-type and Chop-null mice treated with metformin from two independent experiments. At least duplicate protein measurements for each sample. Data are mean ± s.e.m.; P values (95% CI) by two-tailed t-test.

Source data

Supplementary information

Supplementary Figure

This file contains the original gels from Extended Data Fig. 10.

Reporting Summary

Supplementary Table

This table shows baseline characteristics of CAMERA.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Coll, A.P., Chen, M., Taskar, P. et al. GDF15 mediates the effects of metformin on body weight and energy balance. Nature 578, 444–448 (2020). https://doi.org/10.1038/s41586-019-1911-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-019-1911-y

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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