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:

Inhibition of the IL-17A axis in adipocytes suppresses diet-induced obesity and metabolic disorders in mice

Abstract

Overnutrition causes obesity, a global health problem without any effective therapy. Obesity is characterized by low-grade inflammation, which predisposes individuals to metabolic syndrome via unknown mechanisms. Here, we demonstrate that abolishing the interleukin-17A (IL-17A) axis in mice by inhibition of RORγt-mediated IL-17A production by digoxin, or by ubiquitous deletion of IL-17 receptor A (Il17ra), suppresses diet-induced obesity (DIO) and metabolic disorders, and promotes adipose-tissue browning, thermogenesis and energy expenditure. Genetic ablation of Il17ra specifically in adipocytes is sufficient to completely prevent DIO and metabolic dysfunction in mice. IL-17A produced in response to DIO induces PPARγ phosphorylation at Ser273 in adipocytes in a CDK5-dependent manner, thereby modifying expression of diabetogenic and obesity genes, which correlates with IL-17A signalling in white adipose tissues of individuals with morbid obesity. These findings reveal an unanticipated role for IL-17A in adipocyte biology, in which its direct action pathogenically reprograms adipocytes, promoting DIO and metabolic syndrome. Targeting the IL-17A axis could be an efficient antiobesity strategy.

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: Digoxin prevents DIO and metabolic disorders.
Fig. 2: Digoxin suppresses DIO and metabolic disorders.
Fig. 3: RORγt inhibitors suppress DIO and metabolic disorders.
Fig. 4: Deletion of IL-17RA prevents DIO and metabolic disorders.
Fig. 5: Inhibition of the IL-17A axis promotes browning and thermogenesis.
Fig. 6: Genetic ablation of Il17ra in adipocytes prevents DIO.
Fig. 7: IL-17A phosphorylates PPARγ in adipocytes to promote obesity.

Similar content being viewed by others

Data availability

All data in this study are available in the text and its Supplementary Information files, which include Extended Data Figs. 18 and Source Data Figs. 1 and 2. The data that support the findings of this study are available from the corresponding author upon request. Source data are provided with this paper.

References

  1. Eckel, R. H., Grundy, S. M. & Zimmet, P. Z. The metabolic syndrome. Lancet 365, 1415–1428 (2005).

    Article  CAS  PubMed  Google Scholar 

  2. Abarca-Gómez, L. et al. Worldwide trends in body-mass index, underweight, overweight, and obesity from 1975 to 2016: a pooled analysis of 2416 population-based measurement studies in 128·9 million children, adolescents, and adults. Lancet 390, 2627–2642 (2017).

    Article  Google Scholar 

  3. Finkelstein, E. A. et al. Obesity and severe obesity forecasts through 2030. Am. J. Prev. Med. 42, 563–570 (2012).

    Article  PubMed  Google Scholar 

  4. Yanovski, S. Z. & Yanovski, J. A. Long-term drug treatment for obesity: a systematic and clinical review. JAMA 11, 74–86 (2014).

    Article  CAS  Google Scholar 

  5. Chouchani, E. T., Kazak, L. & Spiegelman, B. M. New advances in adaptive thermogenesis: UCP1 and beyond. Cell Metab. 29, 27–37 (2019).

    Article  CAS  PubMed  Google Scholar 

  6. Seale, P. et al. Prdm16 determines the thermogenic program of subcutaneous white adipose tissue in mice. J. Clin. Invest. 121, 96–105 (2011).

    Article  CAS  PubMed  Google Scholar 

  7. Choi, J. H. et al. Anti-diabetic drugs inhibit obesity-linked phosphorylation of PPARgamma by Cdk5. Nature 466, 451–456 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Wang, H., Liu, L., Lin, J. Z., Aprahamian, T. R. & Farmer, S. R. Browning of white adipose tissue with roscovitine induces a distinct population of UCP1+ adipocytes. Cell Metab. 24, 835–847 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Mathis, D. Immunological goings-on in visceral adipose tissue. Cell Metab. 17, 851–859 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Gomes, A. L. et al. Metabolic inflammation-associated IL-17A causes non-alcoholic steatohepatitis and hepatocellular carcinoma. Cancer Cell 30, 161–175 (2016).

    Article  CAS  PubMed  Google Scholar 

  11. Ivanov, I. I. et al. The orphan nuclear receptor RORγt directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 126, 1121–1133 (2006).

    Article  CAS  PubMed  Google Scholar 

  12. McGeachy, M. J., Cua, D. J. & Gaffen, S. L. The IL-17 family of cytokines in health and disease. Immunity 50, 892–906 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Su, Y. et al. Interleukin-17 receptor D constitutes an alternative receptor for interleukin-17A important in psoriasis-like skin inflammation. Sci. Immunol 4, eaau9657 (2019).

    Article  CAS  PubMed  Google Scholar 

  14. Goepfert, A., Lehmann, S., Wirth, E. & Rondeau, J. M. The human IL-17A/F heterodimer: a two-faced cytokine with unique receptor recognition properties. Sci. Rep. 7, 8906 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Hu, B. et al. γ∆ T cells and adipocyte IL-17RC control fat innervation and thermogenesis. Nature 578, 610–614 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Zapata-Gonzalez, F. et al. Interleukin-17A gene expression in morbidly obese women. Int. J. Mol. Sci. 16, 17469–17481 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Baeten, D. L. & Kuchroo, V. K. How cytokine networks fuel inflammation: interleukin-17 and a tale of two autoimmune diseases. Nat. Med. 19, 824–825 (2013).

    Article  CAS  PubMed  Google Scholar 

  18. Taams, L. S., Steel, K. J. A., Srenathan, U., Burns, L. A. & Kirkham, B. W. IL-17 in the immunopathogenesis of spondyloarthritis. Nat. Rev. Rheumatol. 14, 453–466 (2018).

    Article  CAS  PubMed  Google Scholar 

  19. Olivieri, I., D’Angelo, S., Palazzi, C. & Padula, A. Advances in the management of psoriatic arthritis. Nat. Rev. Rheumatol. 10, 531–542 (2014).

    Article  CAS  PubMed  Google Scholar 

  20. Armstrong, A. W., Harskamp, C. T. & Armstrong, E. J. The association between psoriasis and obesity: a systematic review and meta-analysis of observational studies. Nutr. Diabetes 2, e54 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. McGinley, A. M. et al. Interleukin-17A serves a priming role in autoimmunity by recruiting IL-1β-producing myeloid cells that promote pathogenic T cells. Immunity 52, 342–356 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Xiao, S. et al. Small-molecule RORγt antagonists inhibit T helper 17 cell transcriptional network by divergent mechanisms. Immunity 40, 477–489 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Huh, J. R. et al. Digoxin and its derivatives suppress TH17 cell differentiation by antagonizing RORγt activity. Nature 472, 486–490 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Rathore, S. S., Wang, Y. & Krumholz, H. M. Sex-based differences in the effect of digoxin for the treatment of heart failure. N. Engl. J. Med. 347, 1403–1411 (2002).

    Article  CAS  PubMed  Google Scholar 

  25. Meissburger, B. et al. Adipogenesis and insulin sensitivity in obesity are regulated by retinoid-related orphan receptor gamma. EMBO Mol. Med. 3, 637–651 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Eberl, G. & Littman, D. R. Thymic origin of intestinal αβ T cells revealed by fate mapping of RORγt+ cells. Science 305, 248–251 (2004).

    Article  CAS  PubMed  Google Scholar 

  27. Buch, T. et al. A Cre-inducible diphtheria toxin receptor mediates cell lineage ablation after toxin administration. Nat. Methods 2, 419–426 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Price, E. M. & Lingrel, J. B. Structure-function relationships in the Na,K-ATPase alpha subunit: site-directed mutagenesis of glutamine-111 to arginine and asparagine-122 to aspartic acid generates a ouabain-resistant enzyme. Biochemistry 27, 8400–8408 (1988).

    Article  CAS  PubMed  Google Scholar 

  29. Coskun, T. et al. Fibroblast growth factor 21 corrects obesity in mice. Endocrinology 149, 6018–6027 (2008).

    Article  CAS  PubMed  Google Scholar 

  30. Ortega-Molina, A. et al. Pharmacological inhibition of PI3K reduces adiposity and metabolic syndrome in obese mice and rhesus monkeys. Cell Metab. 21, 558–570 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Crane, J. D. et al. Inhibiting peripheral serotonin synthesis reduces obesity and metabolic dysfunction by promoting brown adipose tissue thermogenesis. Nat. Med. 21, 166–172 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Liu, J., Lee, J., Salazar Hernandez, M. A., Mazitschek, R. & Ozcan, U. Treatment of obesity with celastrol. Cell 161, 999–1011 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Buganova, M. et al. The effects of liraglutide in mice with diet-induced obesity studied by metabolomics. J. Endocrinol. 233, 93–104 (2017).

    Article  CAS  PubMed  Google Scholar 

  34. Bernier, M. et al. Disulfiram treatment normalizes body weight in obese mice. Cell Metab. 32, 203–214 e204 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ruzankina, Y. et al. Deletion of the developmentally essential gene ATR in adult mice leads to age-related phenotypes and stem cell loss. Cell Stem Cell 1, 113–126 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. El Malki, K. et al. An alternative pathway of imiquimod-induced psoriasis-like skin inflammation in the absence of interleukin-17 receptor A signaling. J. Invest Dermatol 133, 441–451 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Ellacott, K. L., Morton, G. J., Woods, S. C., Tso, P. & Schwartz, M. W. Assessment of feeding behavior in laboratory mice. Cell Metab. 12, 10–17 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Tschop, M. H. et al. A guide to analysis of mouse energy metabolism. Nat. Methods 9, 57–63 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Mina, A. I. et al. CalR: a web-based analysis tool for indirect calorimetry experiments. Cell Metab. 28, 656–666(2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Feldmann, H. M., Golozoubova, V., Cannon, B. & Nedergaard, J. UCP1 ablation induces obesity and abolishes diet-induced thermogenesis in mice exempt from thermal stress by living at thermoneutrality. Cell Metab. 9, 203–209 (2009).

    Article  CAS  PubMed  Google Scholar 

  41. Cui, H., Lopez, M. & Rahmouni, K. The cellular and molecular bases of leptin and ghrelin resistance in obesity. Nat. Rev. Endocrinol. 13, 338–351 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Ingalls, A. M., Dickie, M. M. & Snell, G. D. Obese, a new mutation in the house mouse. J. Hered. 41, 317–318 (1950).

    Article  CAS  PubMed  Google Scholar 

  43. Chen, H. et al. Evidence that the diabetes gene encodes the leptin receptor: identification of a mutation in the leptin receptor gene in db/db mice. Cell 84, 491–495 (1996).

    Article  CAS  PubMed  Google Scholar 

  44. Howard, J. K. et al. Leptin protects mice from starvation-induced lymphoid atrophy and increases thymic cellularity in ob/ob mice. J. Clin. Invest. 104, 1051–1059 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Reis, B. S. et al. Leptin receptor signaling in T cells is required for TH17 differentiation. J. Immunol. 194, 5253–5260 (2015).

    Article  CAS  PubMed  Google Scholar 

  46. Yu, Y. et al. Cutting edge: leptin-induced RORγt expression in CD4+ T cells promotes TH17 responses in systemic lupus erythematosus. J. Immunol. 190, 3054–3058 (2013).

    Article  CAS  PubMed  Google Scholar 

  47. Nishimura, S. et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat. Med. 15, 914–920 (2009).

    Article  CAS  PubMed  Google Scholar 

  48. Patsouris, D. et al. Ablation of CD11c-positive cells normalizes insulin sensitivity in obese insulin resistant animals. Cell Metab. 8, 301–309 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Clausen, B. E., Burkhardt, C., Reith, W., Renkawitz, R. & Forster, I. Conditional gene targeting in macrophages and granulocytes using LysMcre mice. Transgenic Res 8, 265–277 (1999).

    Article  CAS  PubMed  Google Scholar 

  50. Eguchi, J. et al. Transcriptional control of adipose lipid handling by IRF4. Cell Metab. 13, 249–259 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Pestel, J. et al. IL-17A contributes to propagation of inflammation but does not impair adipogenesis and/or insulin response, in adipose tissue of obese individuals. Cytokine 126, 154865 (2019).

    Article  PubMed  CAS  Google Scholar 

  52. Hu, E., Liang, P. & Spiegelman, B. M. AdipoQ is a novel adipose-specific gene dysregulated in obesity. J. Biol. Chem. 271, 10697–10703 (1996).

    Article  CAS  PubMed  Google Scholar 

  53. Yamauchi, T. et al. The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat. Med. 7, 941–946 (2001).

    Article  CAS  PubMed  Google Scholar 

  54. Lo, J. C. et al. Adipsin is an adipokine that improves beta cell function in diabetes. Cell 158, 41–53 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Rosen, B. S. et al. Adipsin and complement factor D activity: an immune-related defect in obesity. Science 244, 1483–1487 (1989).

    Article  CAS  PubMed  Google Scholar 

  56. Pellicori, P., Kaur, K. & Clark, A. L. Fluid management in patients with chronic heart failure. Card. Fail Rev. 1, 90–95 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Kohlgruber, A. C. et al. γ∆ T cells producing interleukin-17A regulate adipose regulatory T cell homeostasis and thermogenesis. Nat. Immunol. 19, 464–474 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Ussar, S. et al. Interactions between gut microbiota, host genetics and diet modulate the predisposition to obesity and metabolic syndrome. Cell Metab. 22, 516–530 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Nair, A. B. & Jacob, S. A simple practice guide for dose conversion between animals and human. J. Basic Clin. Pharm. 7, 27–31 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Brandt, M. et al. mTORC1 inactivation promotes colitis-Induced colorectal cancer but protects from APC loss-dependent tumorigenesis. Cell Metab. 27, 118–135 (2018).

    Article  CAS  PubMed  Google Scholar 

  61. Chaves-Perez, A., Yilmaz, M., Perna, C., de la Rosa, S. & Djouder, N. URI is required to maintain intestinal architecture during ionizing radiation. Science 364, eaaq1165 (2019).

  62. Bernard, H. et al. Coxsackievirus B type 4 infection in beta cells downregulates the chaperone prefoldin URI to induce a MODY4-like diabetes via Pdx1 silencing. Cell Rep. Med. 1, 100125 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Grau, M. et al. Trends in cardiovascular risk factor prevalence (1995–2000–2005) in northeastern Spain. Eur. J. Cardiovasc Prev. Rehabil. 14, 653–659 (2007).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We are very thankful to R. Elosua and I. Subirana for collecting and analysing the epidemiological data (Hospital del Mar Medical Research Institute, Barcelona). We particularly thank the biostatistician C. Coscia for discussing the statistical analysis of EE. We are grateful to the CNIO Biobank for helping us to collect WAT from patients and associated clinical data. We particularly acknowledge the patients enroled in this study for their participation and the Aragon Health Sciences Institute in the framework of the Biobank of the Aragon Health System for its collaboration. We are also thankful to M. Malumbres for critical reading of this manuscript, and to the CNIO Mouse Genome Editing Core Unit and Animal Facility for the mouse re-derivation and maintenance, respectively. This work was funded by the European Foundation for the Study of Diabetes (EFSD) award supported by EFSD/JRDF/Lilly programme (EASD 96103), the Pfizer Foundation, and the State Research Agency (AEI, 10.13039/501100011033) from the Spanish Ministry of Science and Innovation (projects SAF2016-76598-R, SAF2017-92733-EXP and RTI2018-094834-B-I00), cofunded by European Regional Development Fund (ERDF). This work was developed at the CNIO funded by the Health Institute Carlos III (ISCIII) and the Spanish Ministry of Science and Innovation. The authors declare no conflict of interest.

Author information

Authors and Affiliations

Authors

Contributions

A.T. and N.D. designed the experiments. A.T. performed most of the experiments and statistical analyses. A.G. helped to process and analyse human WAT samples. A.F. helped with some experiments performed in 3T3-L1 cells. C.P. histopathologically analysed mouse tissues. A.T. and N.D. analysed the data. N.D. conceived, developed and wrote the project and study. N.D. and A.T. wrote the manuscript. N.D. secured all funding.

Corresponding author

Correspondence to Nabil Djouder.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Metabolism thanks Bruno Silva-Santos and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editors: George Caputa; Pooja Jha.

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

Extended data

Extended Data Fig. 1 Digoxin prevents DIO and metabolic disorders.

a, Gating strategy for flow cytometry analysis of IL-17A-producing cells in ND- or HFD-fed-C57BL/6 mice (beginning at 8 weeks) for 2 months, treated with or without digoxin for 1 week. b, qRT-PCR in spleen of 38-week-old C57BL/6 mice fed with HFD (beginning at 8 weeks) treated with or without digoxin (n = 5 mice per group). c, qRT-PCR of RORγt target genes in spleen and RORγ target genes in WAT of mice from b (n = 5 mice per group; P = 0.0008; 0.0358; 0.0028; 0.0033; 0.0683; 0.0115, from left to right). d, Total lean mass in ND- or HFD-fed-24-week-old C57BL/6 mice treated with or without digoxin (beginning at 8 weeks) (n = 5 mice per group; P = 0.0168; <0.0001; 0.0007, from left to right). e, Liver weight in mice from d (n = 5 mice per group; P = 0.0064; 0.0013; 0.0014, from left to right). f, Bone mineral density (BMD) in mice from d (n = 5 mice per group). Unpaired two-tailed Student’s t test was used. *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001. Data are represented as means ± s.e.m.

Extended Data Fig. 2 Digoxin suppresses DIO and metabolic disorders.

a, Weights of HFD-fed-C57BL/6 mice (beginning at 8 weeks) treated with digoxin (beginning at 16 weeks) and injected with rIL-17C, rIL-17E or rIL-17F (beginning at 21 weeks) (n = 4 mice per group). b, Scheme of Rorc-Cre-DTR mouse model in which RORγt-positive cells are depleted with DT. c, Weights of ND- or HFD-fed-C57BL/6 mice (beginning at weeks) treated with vehicle or ouabain (beginning at 8 weeks) (n = 5 mice per group). d, Total lean mass in ND- or HFD-fed-38-week-old C57BL/6 mice (beginning at 8 weeks) treated with or without digoxin (beginning at 20 weeks) (n = 10, 5, 8, and 15 mice; P = 0.0457; 0.0001, from left to right). e, Liver weight in mice from d (n = 9, 5, 10, and 8 mice; p <0.0001). f, BMD in mice from d (n = 10, 5, 8, and 15 mice). g, Scheme representing digoxin treatment over 4 weeks in HFD-fed-C57BL/6 mice. h, ORO in livers of HFD-fed-24-week-old-C57BL/6 (beginning at 8 weeks) treated with or without digoxin (beginning at 20 weeks). i, ORO quantification from mice from h (n =5 mice per group; P = 0.0084). Scale bar, 50 μm. j, Cholesterol in plasma from ND- or HFD-fed-C57BL/6 mice (beginning at 8 weeks) treated with or without digoxin (beginning at 20 weeks) for 1 week, 2 weeks or 4 weeks (n = 4 mice per group; P = 0.0006; 0.0245; 0.023; 0.0015; 0.001; 0.017; 0.0025; 0.0467, from left to right). k, Fasting glucose levels in mice from j (n = 4 mice per group; p = 0.0053; 0.021; 0.05; 0.0012; 0.0115; 0.003; 0.0009, from left to right). l, m, Results from the GTT (l) (p = 0.0354; <0.0001; 0.0016; 0.0067, from left to right) and ITT (m) (P = 0.001; 0.0003; 0.0328; 0.0038, from left to right) in mice from j (n = 4 mice per group). n, H&E in mice from d. 3V: Third ventricle. Scale bars,100μm, 200μm and 500 μm. o, p, Creatinine (o) and NH3 (p) in plasma from mice from d (n = 4 mice per group). q, Success in the tightrope test in mice from h (n = 4, 4, 5, and 4 mice; P = 0.0394; 0.0395, from left to right). r, Rotarod test in mice from h (n = 4, 4, 10, 4 mice; P = 0.0175; 0.0005, from left to right). s, Open-field test in mice from h (n = 4, 4, 5, and 4 mice). t, Object recognition test in in mice from h (n = 4, 4, 5, and 4 mice). Unpaired two-tailed Student’s t test (d-f, i-k, o-t) or two-way ANOVA (a, c, l, m) were used. *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001. Data are represented as means ± s.e.m.

Extended Data Fig. 3 RORγt inhibitors suppress DIO and metabolic disorders.

a-d, gWAT (a) (P = 0.0001), iWAT (b) (P = 0.0206), BAT (c) (P = 0.0062), and liver (d) (pP = 0.0434) weights in HFD-fed-20-week-old C57BL/6 mice, treated with vehicle or GSK805 (beginning at 8 weeks) (n = 5 (HFD + vehicle) and 3 (HFD + GSK805) mice). e, Cholesterol in plasma from mice from a-d (n = 5 (HFD + vehicle) and 3 (HFD + GSK805) mice; P = 0.001). f, Percentage of total fat in HFD-fed-26-week-old C57BL/6 mice (beginning at 8 weeks) treated with vehicle or GSK805 (beginning at 20 weeks) (n = 5 (HFD + vehicle) and 3 (HFD + GSK805) mice; P = 0.0234). g, Total fat mass in mice from f (n = 5 (HFD + vehicle) and 3 (HFD + GSK805) mice; P = 0.0257). h-j, gWAT (h) (P = 0.0443), iWAT (i) (P = 0.0122), BAT (j) (P = 0.0475) weights in mice from f (n = 5 (HFD + vehicle) and 3 (HFD + GSK805) mice). k, Total lean mass in mice from f (n = 5 (HFD + vehicle) and 3 (HFD + GSK805) mice; P = 0.0321). l, Liver weight in mice from f (n = 5 (HFD + vehicle) and 3 (HFD + GSK805) mice; P = 0.0136). m, BMD in mice from f (n = 5 (HFD + vehicle) and 3 (HFD + GSK805) mice). n, Cholesterol in plasma from mice from f (n = 5 (HFD + vehicle) and 3 (HFD + GSK805) mice; P = 0.0325). o-r, gWAT (o) (P = 0.0338), iWAT (p) (P = 0.0061), BAT (q) (P = 0.0192), and liver (r) (P = 0.0065) weights in HFD-fed-18-week-old C57BL/6 mice, treated with vehicle or TMP778 (beginning at 6 weeks) (n = 4 (HFD + vehicle) and 5 (HFD + TMP778) mice). s, Cholesterol in plasma from mice from o-r (n = 4 (HFD + vehicle) and 5 (HFD + TMP778) mice; P = 0.0018). t, Table summarizing the effects of anti-obesity compounds in preclinical mouse models. 1Time: administration time needed for weight stabilization described. 2Length: Long-term administration described. ND: Not determined. IP: intraperitoneal administration. SC: subcutaneous administration. Weight loss was adjusted to ND- or HFD-fed mice as indicated in Methods. Unpaired two-tailed Student’s t test was used. *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001.Data are represented as means ± s.e.m.

Extended Data Fig. 4 Deletion of IL-17RA prevents DIO and metabolic disorders.

a, Scheme of Il17raΔ/Δ mouse model in which Il17ra deletion is controlled by hUBC-CreERT2. b, qRT-PCR of tissues from ND- or HFD-fed-Il17ra+/+ and -Il17raΔ/Δ mice, after deletion of Il17ra (n = 3 mice per group; P = 0.05; 0.0018; 0.0383; 0.047, from left to right). c, Total lean mass in ND- or HFD-fed-38-week-old Il17ra+/+ and Il17raΔ/Δ mice (n = 4, 4, 6, and 5 mice; P = 0.002; 0.017, from left to right). d, Liver weight in mice from c (n = 5, 6, 9, and 6 mice; P = 0.001; 0.0003, from left to right). e, BMD in mice from c (n = 4, 4, 6, and 5 mice). Unpaired two-tailed Student’s t test was used. *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001. Data are represented as means ± s.e.m.

Extended Data Fig. 5 Inhibition of IL-17A axis promotes browning and thermogenesis.

a, b, Cumulative food intake of ND- or HFD-fed-C57BL/6 mice (beginning at 8 weeks), treated with or without digoxin (beginning at 20 weeks) for 1 month (a) (n = 4 mice per group; P = 0.0245) and of ND- or HFD-fed-Il17ra+/+ and -Il17raΔ/Δ mice (beginning at 8 weeks) for 1 month (b) (n = 3, 4, 6, and 4 mice; P = 0.0397). c, d, Cumulative food intake of ND- or HFD-fed-38-week-old C57BL/6 mice (beginning at 8 weeks) treated with or without digoxin (beginning at 20 weeks) (c) (n = 10, 5, 4 and 8 mice; P = 0.0415; 0.0037, from left to right) (c), and of ND- or HFD-fed-38-week-old Il17ra+/+ and Il17raΔ/Δ mice (beginning at 8 weeks) (d) (n = 6, 6, 6 and 4 mice; P = 0.0043). e, Cumulative drink intake of mice from a (n = 4 mice per group). f, Cumulative drink intake of mice from b (n = 3, 4, 6 and 4 mice). g, Hematocrit (HCT), hemoglobin (HGB) and red blood cells (RBC) measured by haemogram in mice from c (n = 9, 5, 10, and 5 mice). h, Weights of ND- or HFD-fed-C57BL/6 mice, treated with or without digoxin in drinking water or intraperitoneally (i.p) (beginning at 8 weeks) (n = 5 mice per group; p <0.0001). i, Locomotor activity in mice from c (n = 10, 9, 8, and 12 mice). j, Locomotor activity in mice from d (n = 7, 6, 7, and 4 mice). k, Respiratory exchange ratio (RER) in mice from c (n = 10, 9, 8, and 12 mice; p <0.0001). l, RER in mice from d (n = 7, 6, 7, and 4 mice; P = 0.0002; <0.0001; 0.0022, from left to right). m, n, EE normalized to body weight and monitored for 3 days (m) and total EE normalized to body weight (n) (P = 0.0004; 0.0009, from left to right) in ND- or HFD-fed-13-week-old C57BL/6 mice (beginning at 8 weeks) treated with or without digoxin (beginning at 12 weeks) for 1 week (n = 10, 8, 9, 8 mice). Grey squares denote night periods. o, p, EE normalized to body weight and monitored for 3 days (o) and total EE normalized to body weight (p) (P = 0.004; 0.0043; 0.0002, from left to right) in mice from c (n = 10, 9, 8, and 12 mice). q, r, EE normalized to body weight and monitored for 3 days (q) and total EE normalized to body weight (r) (P = 0.0117; 0.0002; 0.0035, from left to right) in mice from d (n = 7, 6, 7, and 6 mice). s, t, EE monitored for 3 days (s) and total EE (t) in mice from c (n = 10, 9, 8, and 12 mice). u, Regression plot of total EE from t and body weight from each mouse from c (n = 10, 9, 8, and 12 mice). v, WB of gWAT from mice from c (n = 1, 3, 4, and 4 samples). w, qRT-PCR of β oxidation target genes in BAT of mice from c (n = 6, 3, 6, and 5 mice; P = 0.0253; 0.0091; 0.0003; 0.0109; 0.0329; <0.0001; 0.0074; 0.0364; 0.0029; 0.0055, from left to right). x, H&E and IHC in gWAT, iWAT, and BAT of ND- or HFD-fed-12-week-old C57BL/6 mice, treated with or without digoxin (beginning at 8 weeks) for 1 month after 24 hours of cold exposure. Scale bars, 100 μm in WAT and 50 μm in BAT. Unpaired two-tailed Student’s t test (g, i-l, n, p, r, t, w), two-way ANOVA with interaction (u), “conventional” two-way ANCOVA (t) and two-tailed Tukey’s post-hoc test (t), or two-way ANOVA (a-f, h) were used. Each dot in (u) represents one mouse. *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001. Data are represented as means ± s.e.m.

Source data

Extended Data Fig. 6 IL-17A mediates obesity independently of the leptin axis or myeloid cells’ activation.

a, Weights of Lep+/+(ob) and Lep-/-(ob) mice treated with or without digoxin (beginning at12 weeks) (n = 7, 11 and 12 mice; p <0.0001). b, Pictures of 24-week-old Lep+/+(ob) and Lep-/-(ob) mice treated with or without digoxin (beginning at 12 weeks). Scale bar, 1 cm. c, Weights of mice from b relative to the weight of Lep+/+(ob) mice (n = 7, 11, and 12 mice; p <0.0001). d, Weights of Lepr+/-(db) and Lepr-/-(db) mice treated with or without digoxin (beginning at 12 weeks) (n = 13, 8, and 7 mice; p <0.0001). e, Scheme of tamoxifen treatment for 2 weeks at 10 weeks of age in Lep+/+(ob);Il17ra+/+, Lep-/-(ob);Il17ra+/+ and Lep-/-(ob);Il17raΔ/Δ mice (upper panel), and weights of Lep+/+(ob);Il17ra+/+, Lep-/-(ob);Il17ra+/+ and Lep-/-(ob);Il17raΔ/Δ mice (lower panel) (n = 8, 5, and 6 mice; p <0.0001). f, Percentage of weight change after 1 month of rLeptin in ND- or HFD-fed-28-week-old C57BL/6 mice (beginning at 8 weeks), treated with or without digoxin (beginning at 20 weeks), relative to 24-week-old weight (n = 4, 5, 4, and 4 mice; P = 0.0135). g, Percentage of weight change after 1 month of rLeptin in 20-week-old Lep-/-(ob) mice treated with or without digoxin (beginning at 12 weeks), relative to 16-week-old weight. (n = 4, 5, 5, and 5 mice; P = 0.0002; 0.0001, from left to right). h, Weights of ND- or HFD-fed-Lep+/+(ob) and Lep-/-(ob) mice (beginning at 6 week)s, treated with or without digoxin (beginning at 12 weeks) (n = 4, 11, 9, and 6 mice; P = 0.0421). i, IHC in gWAT of ND- or HFD-fed-38-week-old C57BL/6 mice (beginning at 8 weeks), treated with or without digoxin (beginning at 20 weeks). Scale bars, 20 μm and 100 μm. j-m B220 (j) (n = 4, 5, 6, and 7 mice; P = 0.0381; 0.0072, from left to right), CD3 (k) (n = 4, 4, 5, and 5 mice; P = 0.044; 0.0115, from left to right), F4/80 (l) (n = 4, 5, 8, and 8 mice; P = 0.0321; 0.0006, from left to right) and MPO (m) (n = 4, 4, 8, and 7 mice; P = 0.0314; 0.0211, from left to right) quantification from gWAT of mice from i. n, Scheme of Il17raΔ/Δ(Myeloid) mouse model in which Il17ra deletion is controlled by LysM-Cre. o, Gating strategy and representative flow cytometry from Il17ra+/+(Myeloid) and Il17raΔ/Δ(Myeloid) mice. p, Quantification of IL-17RA+ CD11b+ cells in Il17ra+/+(Myeloid) and Il17raΔ/Δ(Myeloid) mice (n = 4 and 8 mice; p <0.0001). q, Weights of ND- or HFD-fed-Il17ra+/+(Myeloid) and -Il17raΔ/Δ(Myeloid) mice, treated with or without digoxin (beginning at 8 weeks) (n = 4, 5, 8, and 4 mice; P = 0.0003). Unpaired two-tailed Student’s t test (c, f, g, j-m, p) or two-way ANOVA (a, d, e, h, q) were used. *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001. Data are represented as means ± s.e.m.

Extended Data Fig. 7 IL-17A phosphorylates PPARγ in adipocytes to promote obesity.

a, FITC-dextran in plasma from ND- or HFD-fed-38-week-old C57BL/6 mice (beginning at 8 weeks), treated wit or /without digoxin (beginning at 20 weeks) (n = 5 mice per group; P = 0.0019). b, Daily faecal mass in mice from a (n = 3, 4, 5, and 6 mice; p <0.0001). c, Faecal caloric density measured by bomb calorimetry in mice from a (n = 4 mice per group). d, Faecal trycerides (TGs) (n = 6, 5, 6, and 5 mice per group), proteins (n = 5 mice per group) and carbohydrates (n = 5, 4, 4, and 5 mice) in mice from a. e, Plasma free fatty acids (FFA) in mice from a (n = 5, 6, 5, and 7 mice). f, qRT-PCR of target genes for hepatic β oxidation in livers of mice from a (n = 5, 4, 5, and 7 mice). g, H&E in gWAT of mice from a (upper panel) and of ND- or HFD-fed-38-week-old Il17ra+/+ and -Il17raΔ/Δ mice (lower panel). Scale bar, 100 μm. h, Adipocyte size of mice from a (n = 7, 4, 8, and 7 mice; P = 0.0005; 0.0178; 0.0238, from left to right). i, Adipocyte size of ND- or HFD-fed-38-week-old Il17ra+/+ and Il17raΔ/Δ mice (n = 4, 4, 5, and 3 mice; P = 0.0463; 0.0043; 0.0029, from left to right). j, Scheme of fasting-refeeding and sample collection in ND- or HFD-fed-24-week-old C57BL/6 mice (beginning at 8 weeks, treated with or without digoxin (beginning at 20 weeks) for 1 month. k, l, Pc j after 24 hours of fasting and refeeding represented as ng/ml (k) (p <0.0001; 0.0053) or normalized to fasted levels (T0) (l) (P = 0.0083; 0.0181; 0.0047; 0.0475; 0.0176; 0.0376; 0.0478, from left to right) (n = 5 mice per group). m, Scheme of Il17raΔ/Δ(Adipoq) mouse model in which IL17ra deletion is controlled by Adipoq-Cre. n, Il17ra mRNA levels in WAT from HFD-fed-20-week-old Il17ra+/+(Adipoq) and Il17raΔ/Δ(Adipoq) mice (beginning at 8 weeks) (n = 5 mice per group; P = 0.05; 0.0218, from left to right). o, Weights of HFD-fed-20-week-old Il17ra+/+, Il17raΔ/Δ, Il17ra+/+(Adipoq) and Il17raΔ/Δ(Adipoq) mice (beginning at 8 weeks) (n = 8, 7, 8, and 6 mice; p <0.0001; 0.0026, from left to right). p, Total lean mass in mice from n (n = 6, 4, 8, and 4 mice; P = 0.0092; 0.0244, from left to right). q, Liver weight in mice from n (n = 6, 4, 8, and 6 mice; P = 0.028; 0.0007, from left to right). r, BMD in mice from n (n = 6, 4, 8, and 6 mice). s, qRT-PCR in gWAT from mice from n (n = 4 mice per group). t, u, qRT-PCR of target genes for lipogenesis (t) (P = 0.008; 0.0408; 0.0128; 0.0064, from left to right) and differentiation (u) (P = 0.0228; 0.0462, from left to right) in gWAT of mice from a (n = 6, 4, 7, and 7 mice). v, qRT-PCR of genes modulated by the CDK5-mediated phosphorylation of PPARγ on Ser273 in differentiated 3T3-L1 adipocytes (n = 3 independent experiments; P = 0.0442; 0.0129; 0.0033, from left to right). w, qRT-PCR of genes modulated by the CDK5-mediated phosphorylation of PPARγ on Ser273 in gWAT from mice from a (n = 6, 4, 7, and 7 mice; p <0.0001; 0.0094; 0.0002; 0.0007; <0.0001; 0.0086; 0.0004; 0.0284; 0.0002; 0.0489; <0.0001; 0.0216; 0.0001; 0.0038; 0.0002; 0.0386, from left to right). Unpaired two-tailed Student’s t test (a-f, h, i, l, n-w) or two-way ANOVA (k) were used. *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001. Data are represented as means ± s.e.m.

Extended Data Fig. 8 IL-17A signalling correlates with obese genes in human WAT.

a, b, Linear regression analysis of ADIPOQ (a) and CFD (b) with BMI in visceral WAT from patients with morbid obesity (n = 75). c, Linear regression analysis of IL17A with LCN2 in visceral WAT from patients with morbid obesity (n = 75). d, e, Linear regression analysis of IL17A (d) and LCN2 (e) with BMI in visceral WAT from patients with morbid obesity (n = 75). f, g, Linear regression analysis of ADIPOQ (f) and CFD (g) with IL17A in visceral WAT from patients with morbid obesity (n = 75). h, i, Linear regression analysis of ADIPOQ (h) and CFD (i) with LCN2 in visceral WAT from patients with morbid obesity (n = 75). j, k, Linear regression analysis of ADIPOQ (j) and CFD (k) with BMI in visceral WAT from women with morbid obesity (n = 41). l, Linear regression analysis of IL17A with LCN2 in visceral WAT from women with morbid obesity (n = 41). m, n, Linear regression analysis of IL17A (m) and LCN2 (n) with BMI in visceral WAT from women with morbid obesity (n = 41). o, p, Linear regression analysis of ADIPOQ (o) and CFD (p) with IL17A in visceral WAT from women with morbid obesity (n = 41). q, r, Linear regression analysis of ADIPOQ (q) and CFD (r) with LCN2 in visceral WAT from women with morbid obesity (n = 41). s-u, Linear regression analysis of IL17C (s), IL17E (t), and IL17F (u) with BMI in visceral WAT from patients with morbid obesity (n = 75). v, w, Association of digoxin use with BMI, cholesterol and LDL levels in a cross-sectional population-based survey. Each dot in (a-u) represents one patient. Red dots denote women patients. Linear regression analyses were used.

Supplementary information

Reporting Summary

Supplementary Table

Supplementary Tables 1–8

Source data

Source Data Fig. 7

Unprocessed Western Blots from Fig. 7

Source Data Extended Data Fig. 5

Unprocessed Western Blots from Extended Data Fig. 5

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Teijeiro, A., Garrido, A., Ferre, A. et al. Inhibition of the IL-17A axis in adipocytes suppresses diet-induced obesity and metabolic disorders in mice. Nat Metab 3, 496–512 (2021). https://doi.org/10.1038/s42255-021-00371-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-021-00371-1

This article is cited by

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