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:

TGF-β2 is an exercise-induced adipokine that regulates glucose and fatty acid metabolism

Abstract

Exercise improves health and well-being across diverse organ systems, and elucidating mechanisms underlying the beneficial effects of exercise can lead to new therapies. Here, we show that transforming growth factor-β2 (TGF-β2) is secreted from adipose tissue in response to exercise and improves glucose tolerance in mice. We identify TGF-β2 as an exercise-induced adipokine in a gene expression analysis of human subcutaneous adipose tissue biopsies after exercise training. In mice, exercise training increases TGF-β2 in subcutaneous white adipose tissue (scWAT) and serum, and its secretion from fat explants. Transplanting scWAT from exercise-trained wild-type mice, but not from adipose-tissue-specific Tgfb2−/− mice, into sedentary mice improves glucose tolerance. TGF-β2 treatment reverses the detrimental metabolic effects of high-fat feeding in mice. Lactate, a metabolite released from muscle during exercise, stimulates TGF-β2 expression in human adipocytes. Administration of the lactate-lowering agent dichloroacetate during exercise training in mice decreases circulating TGF-β2 levels and reduces exercise-stimulated improvements in glucose tolerance. Thus, exercise training improves systemic metabolism through inter-organ communication with fat via a lactate–TGF-β2 signaling cycle.

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: TGF-β2 is an exercise-induced adipokine.
Fig. 2: Recombinant TGF-β2 treatment stimulates glucose uptake and OCR in vitro.
Fig. 3: TGF-β2 infusion via an osmotic pump stimulates tissue glucose uptake and muscle OCR in mice.
Fig. 4: TGF-β2 infusion via an osmotic pump ameliorates the effects of a HFD in mice.
Fig. 5: TGF-β2 treatment attenuates HFD-induced inflammation in adipose tissue.
Fig. 6: Lactate produced by exercise training stimulates TGF-β2.

Similar content being viewed by others

Data availability

All data underlying the findings reported in this manuscript are provided as part of the article. Source data are available online. Mouse and human microarray data are available in the Gene Expression Omnibus database under accession numbers GSE68161 and GSE116801. The raw data that are not already presented in the figures are available from the corresponding author upon reasonable request.

References

  1. Stanford, K. I. & Goodyear, L. J. Exercise and type 2 diabetes: molecular mechanisms regulating glucose uptake in skeletal muscle. Adv. Physiol. Educ. 38, 308–314 (2014).

    Article  Google Scholar 

  2. Fiuza-Luces, C., Garatachea, N., Berger, N. A. & Lucia, A. Exercise is the real polypill. Physiology 28, 330–358 (2013).

    Article  CAS  Google Scholar 

  3. Booth, F. W., Roberts, C. K. & Laye, M. J. Lack of exercise is a major cause of chronic diseases. Compr. Physiol. 2, 1143–1211 (2012).

    PubMed  PubMed Central  Google Scholar 

  4. Colberg, S. R. et al. Exercise and Type 2 Diabetes: The American College of Sports Medicine and the American Diabetes Association: joint position statement executive summary. Diabetes Care 33, e147–67 (2010).

    Article  Google Scholar 

  5. Gollisch, K. S. C. et al. Effects of exercise training on subcutaneous and visceral adipose tissue in normal- and high-fat diet-fed rats. Am. J. Physiol. Endocrinol. Metab. 297, 495–504 (2009).

    Article  Google Scholar 

  6. Stanford, K. I. & Goodyear, L. J. Muscle–adipose tissue cross talk. Cold Spring Harb. Perspect. Med. 4, a029801 (2017).

    Google Scholar 

  7. Craig, B. W., Hammons, G. T., Garthwaite, S. M., Jarett, L. & Holloszy, J. O. Adaptation of fat cells to exercise: response of glucose uptake and oxidation to insulin. J. Appl. Physiol. 51, 1500–1506 (1981).

    Article  CAS  Google Scholar 

  8. You, T., Arsenis, N. C., Disanzo, B. L. & Lamonte, M. J. Effects of exercise training on chronic inflammation in obesity: current evidence and potential mechanisms. Sports Med. 43, 243–256 (2013).

    Article  Google Scholar 

  9. Porter, J. W. et al. Anti-inflammatory effects of exercise training in adipose tissue do not require FGF21. J. Endocrinol. 235, 97–109 (2017).

    Article  CAS  Google Scholar 

  10. Kawanishi, N., Yano, H., Yokogawa, Y. & Suzuki, K. Exercise training inhibits inflammation in adipose tissue via both suppression of macrophage infiltration and acceleration of phenotypic switching from M1 to M2 macrophages in high-fat-diet-induced obese mice. Exerc. Immunol. Rev. 16, 105–118 (2010).

    PubMed  Google Scholar 

  11. Rao, R. R. et al. Meteorin-like is a hormone that regulates immune–adipose interactions to increase beige fat thermogenesis. Cell 157, 1279–1291 (2014).

    Article  CAS  Google Scholar 

  12. Bostrom, P. et al. A PGC1 alpha dependent myokine that drives brown fat like development of white fat and thermogenesis. Nature 481, 463–468 (2012).

    Article  Google Scholar 

  13. Kajimura, S., Spiegelman, B. M. & Seale, P. Brown and beige fat: physiological roles beyond heat generation. Cell Metab. 22, 546–559 (2015).

    Article  CAS  Google Scholar 

  14. Stallknecht, B., Vinten, J., Ploug, T. & Galbo, H. Increased activities of mitochondrial enzymes in white adipose tissue in trained rats. Am. J. Physiol. 261, E410–E414 (1991).

    CAS  PubMed  Google Scholar 

  15. Trevellin, E. et al. Exercise training induces mitochondrial biogenesis and glucose uptake in subcutaneous adipose tissue through eNOS-dependent mechanisms. Diabetes 63, 2800–2811 (2014).

    Article  CAS  Google Scholar 

  16. Stanford, K. I. et al. A novel role for subcutaneous adipose tissue in exercise-induced improvements in glucose homeostasis. Diabetes 64, 2002–2014 (2015).

    Article  CAS  Google Scholar 

  17. Massague, J. TGF-β signal transduction. Annu. Rev. Biochem. 67, 753–791 (1998).

    Article  CAS  Google Scholar 

  18. LEASK, A. TGF-β signaling and the fibrotic response. FASEB J. 18, 816–827 (2004).

    Article  CAS  Google Scholar 

  19. Li, M. O., Wan, Y. Y. & Flavell, R. A. T. Cell-produced transforming growth factor-β1 controls T cell tolerance and regulates Th1- and Th17-cell differentiation. Immunity 26, 579–591 (2007).

    Article  CAS  Google Scholar 

  20. Sanford, L. P. et al. TGFβ2 knockout mice have multiple developmental defects that are non-overlapping with other TGFβ knockout phenotypes. Development 124, 2659–2670 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Doetschman, T. et al. Generation of mice with a conditional allele for the transforming growth factor β3 gene. Genesis 50, 59–66 (2012).

    Article  CAS  Google Scholar 

  22. Ishtiaq Ahmed, A. S., Bose, G. C., Huang, L. & Azhar, M. Generation of mice carrying a knockout-first and conditional-ready allele of transforming growth factor β2 gene. Genesis 52, 817–826 (2014).

    Article  CAS  Google Scholar 

  23. Azhar, M. et al. Generation of mice with a conditional allele for transforming growth factor β1 gene. Genesis 47, 423–431 (2009).

    Article  CAS  Google Scholar 

  24. de Martin, R. et al. Complementary DNA for human glioblastoma-derived T cell suppressor factor, a novel member of the transforming growth factor-β gene family. EMBO J. 6, 3673–3677 (1987).

    Article  Google Scholar 

  25. Zhang, H., Yang, P., Zhou, H., Meng, Q. & Huang, X. Involvement of Foxp3-expressing CD4+ CD25+ regulatory T cells in the development of tolerance induced by transforming growth factor-β2-treated antigen-presenting cells. Immunology 124, 304–314 (2008).

    Article  CAS  Google Scholar 

  26. Maheshwari, A. et al. TGF-β2 suppresses macrophage cytokine production and mucosal inflammatory responses in the developing intestine. Gastroenterology 140, 242–253 (2011).

    Article  CAS  Google Scholar 

  27. Shimizu, C. et al. Transforming growth factor-β signaling pathway in patients with Kawasaki disease. Circ. Cardiovasc. Genet. 4, 16–25 (2011).

    Article  CAS  Google Scholar 

  28. Yfanti, C. et al. Effect of antioxidant supplementation on insulin sensitivity in response to endurance exercise training. Am. J. Physiol. Endocrinol. Metab. 300, E761–E770 (2011).

    Article  CAS  Google Scholar 

  29. Yfanti, C. et al. Antioxidant supplementation does not alter endurance training adaptation. Med. Sci. Sports Exerc. 42, 1388–1395 (2010).

    Article  CAS  Google Scholar 

  30. Camon, E. The Gene Ontology Annotation (GOA) Database: sharing knowledge in Uniprot with Gene Ontology. Nucleic Acids Res. 32, 262D–266D (2004).

    Article  Google Scholar 

  31. Motiani, P. et al. Decreased insulin-stimulated brown adipose tissue glucose uptake after short-term exercise training in healthy middle aged men. Diabetes Obes. Metab. 19, 1379–1388 (2017).

    Article  CAS  Google Scholar 

  32. Schulz, T. J. et al. Brown-fat paucity due to impaired BMP signalling induces compensatory browning of white fat. Nature 495, 379–383 (2013).

    Article  CAS  Google Scholar 

  33. Rasbach, Ka et al. PGC-1α regulates a HIF2α-dependent switch in skeletal muscle fiber types. Proc. Natl Acad. Sci. USA 107, 21866–21871 (2010).

    Article  CAS  Google Scholar 

  34. Wu, Z. et al. Mechanisms controlling mitochondrial biogenesis and respiration through the thermogenic coactivator PGC-1. Cell 98, 115–124 (1999).

    Article  CAS  Google Scholar 

  35. Cohen, P. et al. Ablation of PRDM16 and beige adipose causes metabolic dysfunction and a subcutaneous to visceral fat switch. Cell 156, 304–316 (2014).

    Article  CAS  Google Scholar 

  36. Tsunoda, T. & Takagi, T. Estimating transcription factor bindability on DNA. Bioinformatics 15, 622–630 (1999).

    Article  CAS  Google Scholar 

  37. Benatti, F. B. & Pedersen, B. K. Exercise as an anti-inflammatory therapy for rheumatic diseases–myokine regulation. Nat. Rev. Rheumatol. 11, 86–97 (2014).

    Article  Google Scholar 

  38. Stacpoole, P. W., Nagaraja, N. V. & Hutson, A. D. Efficacy of dichloroacetate as a lactate-lowering drug. J. Clin. Pharmacol. 43, 683–691 (2003).

    Article  CAS  Google Scholar 

  39. Goodwin, M. L., Harris, J. E., Hernández, A. & Gladden, L. B. Blood lactate measurements and analysis during exercise: a guide for clinicians. J. Diabetes Sci. Technol. 1, 558–569 (2007).

    Article  Google Scholar 

  40. Hashimoto, T., Hussien, R., Oommen, S., Gohil, K. & Brooks, G. Lactate sensitive transcription factor network in L6 cells: activation of MCT1 and mitochondrial biogenesis. FASEB J. 21, 2602–2612 (2007).

    Article  CAS  Google Scholar 

  41. Carrière, A. et al. Browning of white adipose cells by intermediate metabolites: an adaptive mechanism to alleviate redox pressure. Diabetes 63, 3253–3265 (2014).

    Article  Google Scholar 

  42. Gulick, T., Cresci, S., Caira, T., Moore, D. D. & Kelly, D. P. The peroxisome proliferator-activated receptor regulates mitochondrial fatty acid oxidative enzyme gene expression. Proc. Natl Acad. Sci. USA 91, 11012–11016 (1994).

    Article  CAS  Google Scholar 

  43. Ahmadian, M. et al. PPARγ signaling and metabolism: the good, the bad and the future. Nat. Med. 99, 557–566 (2013).

    Article  Google Scholar 

  44. Li, P., Zhu, Z., Lu, Y. & Granneman, J. G. Metabolic and cellular plasticity in white adipose tissue II: role of peroxisome proliferator-activated receptor-α. Am. J. Physiol. Endocrinol. Metab. 289, E617–E626 (2005).

    Article  CAS  Google Scholar 

  45. Schenk, S., Saberi, M. & Olefsky, J. M. Insulin sensitivity: modulation by nutrients and inflammation. J. Clin. Investig. 118, 2992–3002 (2008).

    Article  CAS  Google Scholar 

  46. Greenberg, A. S. & Obin, M. S. Obesity and the role of adipose tissue in inflammation and metabolism. Am. J. Clin. Nutr. 83, 461–465 (2006).

    Article  Google Scholar 

  47. Xu, H. et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J. Clin. Invest. 112, 1821–1830 (2003).

    Article  CAS  Google Scholar 

  48. Gleeson, M. et al. The anti-inflammatory effects of exercise: mechanisms and implications for the prevention and treatment of disease. Nat. Rev. Immunol. 11, 607–315 (2011).

    Article  CAS  Google Scholar 

  49. Bradley, R. L., Jeon, J. Y., Liu, F. & Maratos-Flier, E. Voluntary exercise improves insulin sensitivity and adipose tissue inflammation in diet-induced obese mice. Am. J. Physiol. Endocrinol. Metab. 295, E586–E594 (2008).

    Article  CAS  Google Scholar 

  50. de Martin, R. et al. Complementary DNA for human glioblastoma-derived T cell suppressor factor, a novel member of the transforming growth factor-β gene family. EMBO J. 6, 3673–3677 (1987).

    Article  Google Scholar 

  51. Xue, R. et al. Clonal analyses and gene profiling identify genetic biomarkers of the thermogenic potential of human brown and white preadipocytes. Nat. Med. 21, 760–768 (2015).

    Article  CAS  Google Scholar 

  52. Shamsi, F. & Tseng, Y. H. Protocols for generation of immortalized human brown and white preadipocyte cell lines. Methods Mol. Biol. 1566, 77–85 (2017).

    Article  CAS  Google Scholar 

  53. Hoque, R., Farooq, A., Ghani, A., Gorelick, F. & Mehal, W. Z. Lactate reduces liver and pancreatic injury in toll-like receptor- and inflammasome-mediated inflammation via gpr81-mediated suppression of innate immunity. Gastroenterology 146, 1763–1774 (2014).

    Article  CAS  Google Scholar 

  54. Ferré, P., Leturque, A., Burnol, A. F., Penicaud, L. & Girard, J .A. A method to quantify glucose utilization in vivo in skeletal muscle and white adipose tissue of the anaesthetized rat. Biochem. J. 228, 103–110 (1985).

    Article  Google Scholar 

  55. Kramer, H. F. et al. AS160 regulates insulin- and contraction-stimulated glucose uptake in mouse skeletal muscle. J. Biol. Chem. 281, 31478–31485 (2006).

    Article  CAS  Google Scholar 

  56. Ho, R. C., Alcazar, O., Fujii, N., Hirshman, M. F. & Goodyear, L. J. p38γ MAPK regulation of glucose transporter expression and glucose uptake in L6 myotubes and mouse skeletal muscle. Am. J. Physiol. Regul. Integr. Comp. Physiol. 286, R342–R349 (2004).

    Article  CAS  Google Scholar 

  57. Lynes, M. D. et al. The cold-induced lipokine 12,13-diHOME promotes fatty acid transport into brown adipose tissue. Nat. Med. 23, 631–637 (2017).

    Article  CAS  Google Scholar 

  58. Townsend, K. L. et al. Increased mitochondrial activity in BMP7-treated brown adipocytes, due to increased CPT1- and CD36-mediated fatty acid uptake. Antioxid. Redox Signal. 19, 243–257 (2013).

    Article  CAS  Google Scholar 

  59. De Keijzer, M. H., Brandts, R. W. & Brans, P. G. W. Evaluation of a biosensor for the measurement of lactate in whole blood. Clin. Biochem. 32, 109–112 (1999).

    Article  Google Scholar 

  60. Li, C. Automating dChip: toward reproducible sharing of microarray data analysis. BMC Bioinformatics 9, 231 (2008).

    Article  Google Scholar 

  61. Ritchie, M. E. et al. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  Google Scholar 

  62. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  Google Scholar 

  63. Tian, L. et al. Discovering statistically significant pathways in expression profiling studies. Proc. Natl Acad. Sci. USA 102, 13544–13549 (2005).

    Article  CAS  Google Scholar 

  64. Gentleman, R. et al. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 5, R80 (2004).

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported by NIH grants R01DK099511 and R01DK112283 (to L.J.G.), K23DK114550 (to R.J.W.M.) and the Joslin Diabetes Center DRC (P30 DK36836). H.T. was supported by individual research fellowships from the Uehara Memorial Foundation and Sumitomo Life Welfare Foundation. Y.-H.T. was supported by NIH grant grants R01DK077097 and R01DK102898. K.I.S. was supported by R01-HL138738. M.D.L. was supported by NIH grants T32DK007260, F32DK102320 and K01DK111714. M.A. was supported by NIH grants R01HL126705 and R01HL145064, and American Heart Association Grant-in-Aid grant 17GRNT33650018. B.K.P. and the Centre for Physical Activity Research were supported by a grant from TrygFonden. We thank K. Longval and A. Clermont from the Joslin Diabetes Center Animal Physiology Core, and L. Kannan from Joslin Special Assay Core. We thank L. Rowland, S. Lessard and A. Queiroz for helpful scientific discussions, and N. Prince and C. Doherty for technical support.

Author information

Authors and Affiliations

Authors

Contributions

H.T. and C.R.R.A. designed research, carried out experiments, analysed data and wrote the paper. K.I.S. performed experiments with trained mice. R.J.W.M. performed and analysed human data. P. Nigro carried out all experiments of adipocyte incubation. R.E.R. carried out experiments and analysed data with Tgfb2-knockout mice and TGF-β2-treated mice. R.X. designed and performed Seahorse assays and provided human white preadipocytes. M.S. carried out experiments and analysed data of cell sorting. M.D.L. carried out in vivo imaging studies for fatty acid uptake. K.S. and J.D.M. performed genotyping of Tgfb2-knockout mice and cell experiments. J.M.D. carried out correlation analysis of microarray data and analysed bioinformatic data. M.-Y.L. carried out gene expression analysis of human adipose tissue. E.B. carried out fatty acid uptake in vitro and Seahorse assays. H.P. and J.M.D. performed bioinformatics analysis. M.F.H. performed in vivo experiments and supervised all experiments. M.A. established and provided Tgfb2-knockout mice. J.C.H., P. Nuutila, K.K.K., B.K.P. and S.N. carried out and provided human samples. C.R.K. supervised in vivo and in vitro experiments with adipocytes or adipose tissue. Y.-H.T. supervised experiments with human preadipocytes and provided immortalized brown preadipocytes. L.J.G. directed the research project, designed experiments and wrote the paper. All authors participated in the manuscript review. All authors approved the final manuscript.

Corresponding author

Correspondence to Laurie J. Goodyear.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Supplementary information

Supplementary Information

Supplementary Figures 1–14 and Supplementary Tables 2 and 3

Reporting Summary

Supplementary Table 1

Human scWAT gene expression data (human_gene_stats_unweighted)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Takahashi, H., Alves, C.R.R., Stanford, K.I. et al. TGF-β2 is an exercise-induced adipokine that regulates glucose and fatty acid metabolism. Nat Metab 1, 291–303 (2019). https://doi.org/10.1038/s42255-018-0030-7

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-018-0030-7

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