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:

White-to-brown metabolic conversion of human adipocytes by JAK inhibition

Abstract

The rising incidence of obesity and related disorders such as diabetes and heart disease has focused considerable attention on the discovery of new therapeutics. One promising approach has been to increase the number or activity of brown-like adipocytes in white adipose depots, as this has been shown to prevent diet-induced obesity and reduce the incidence and severity of type 2 diabetes. Thus, the conversion of fat-storing cells into metabolically active thermogenic cells has become an appealing therapeutic strategy to combat obesity. Here, we report a screening platform for the identification of small molecules capable of promoting a white-to-brown metabolic conversion in human adipocytes. We identified two inhibitors of Janus kinase (JAK) activity with no precedent in adipose tissue biology that stably confer brown-like metabolic activity to white adipocytes. Importantly, these metabolically converted adipocytes exhibit elevated UCP1 expression and increased mitochondrial activity. We further found that repression of interferon signalling and activation of hedgehog signalling in JAK-inactivated adipocytes contributes to the metabolic conversion observed in these cells. Our findings highlight a previously unknown role for the JAK–STAT pathway in the control of adipocyte function and establish a platform to identify compounds for the treatment of obesity.

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

Figure 1: Browning screen in human stem cell-derived adipocytes.
Figure 2: Selected compounds modulate lipid morphology.
Figure 3: Validation of tofacitinib and R406 browning compounds in primary adipocytes.
Figure 4: Inhibition of STAT phosphorylation downstream of tofacitinib and R406.
Figure 5: JAK inhibition stably induces a brown-like profile in adipocytes.
Figure 6: JAK inhibition induces brown-like metabolic properties in adipocytes.
Figure 7: Gene signature and cellular identity of JAK-inactivated adipocytes.
Figure 8: IFN and SHH signalling contribute to adipocyte browning downstream of JAK inhibition.

Similar content being viewed by others

References

  1. Bartelt, A. et al. Brown adipose tissue activity controls triglyceride clearance. Nat. Med. 17, 200–205 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Stanford, K. I. et al. Brown adipose tissue regulates glucose homeostasis and insulin sensitivity. J. Clin. Invest. 123, 215–223 (2013).

    Article  CAS  PubMed  Google Scholar 

  3. Cypess, A. M. et al. Identification and importance of brown adipose tissue in adult humans. N. Engl. J. Med. 360, 1509–1517 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ghorbani, M. & Himms-Hagen, J. Appearance of brown adipocytes in white adipose tissue during CL 316,243-induced reversal of obesity and diabetes in Zucker fa/fa rats. Int. J. Obes. Relat. Metab. Disord. 21, 465–475 (1997).

    Article  CAS  PubMed  Google Scholar 

  5. Granneman, J. G., Li, P., Zhu, Z. & Lu, Y. Metabolic and cellular plasticity in white adipose tissue I: effects of β3-adrenergic receptor activation. Am. J. Physiol. Endocrinol. Metab. 289, E608–E616 (2005).

    Article  CAS  PubMed  Google Scholar 

  6. Himms-Hagen, J. et al. Multilocular fat cells in WAT of CL-316243-treated rats derive directly from white adipocytes. Am. J. Physiol. Cell Physiol. 279, C670–C681 (2000).

    Article  CAS  PubMed  Google Scholar 

  7. 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  PubMed  Google Scholar 

  8. Koh, Y. J. et al. Activation of PPAR γ induces profound multilocularization of adipocytes in adult mouse white adipose tissues. Exp. Mol. Med. 41, 880–895 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Murano, I., Barbatelli, G., Giordano, A. & Cinti, S. Noradrenergic parenchymal nerve fiber branching after cold acclimatisation correlates with brown adipocyte density in mouse adipose organ. J. Anat. 214, 171–178 (2009).

    Article  CAS  PubMed  Google Scholar 

  10. Petrovic, N. et al. Chronic peroxisome proliferator-activated receptor γ (PPARγ) activation of epididymally derived white adipocyte cultures reveals a population of thermogenically competent, UCP1-containing adipocytes molecularly distinct from classic brown adipocytes. J. Biol. Chem. 285, 7153–7164 (2010).

    Article  CAS  PubMed  Google Scholar 

  11. Wu, J. et al. Beige adipocytes are a distinct type of thermogenic fat cell in mouse and human. Cell 150, 366–376 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Rosenwald, M., Perdikari, A., Rulicke, T. & Wolfrum, C. Bi-directional interconversion of brite and white adipocytes. Nat. Cell Biol. 15, 659–667 (2013).

    Article  CAS  PubMed  Google Scholar 

  13. Van der Lans, A. A. et al. Cold acclimation recruits human brown fat and increases nonshivering thermogenesis. J. Clin. Invest. 123, 3395–3403 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Yoneshiro, T. et al. Recruited brown adipose tissue as an antiobesity agent in humans. J. Clin. Invest. 123, 3404–3408 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ahfeldt, T. et al. Programming human pluripotent stem cells into white and brown adipocytes. Nat. Cell Biol. 14, 209–219 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Bonet, M. L., Oliver, P. & Palou, A. Pharmacological and nutritional agents promoting browning of white adipose tissue. Biochim. Biophys. Acta 1831, 969–985 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Kozak, U. C. et al. An upstream enhancer regulating brown-fat-specific expression of the mitochondrial uncoupling protein gene. Mol. Cell. Biol. 14, 59–67 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Rubio, A., Raasmaja, A., Maia, A. L., Kim, K. R. & Silva, J. E. Effects of thyroid hormone on norepinephrine signaling in brown adipose tissue. I. β 1- and β 2-adrenergic receptors and cyclic adenosine 3’,5’-monophosphate generation. Endocrinology 136, 3267–3276 (1995).

    Article  CAS  PubMed  Google Scholar 

  19. Rubio, A., Raasmaja, A. & Silva, J. E. Thyroid hormone and norepinephrine signaling in brown adipose tissue. II: differential effects of thyroid hormone on β 3-adrenergic receptors in brown and white adipose tissue. Endocrinology 136, 3277–3284 (1995).

    Article  CAS  PubMed  Google Scholar 

  20. Rabelo, R., Schifman, A., Rubio, A., Sheng, X. & Silva, J. E. Delineation of thyroid hormone-responsive sequences within a critical enhancer in the rat uncoupling protein gene. Endocrinology 136, 1003–1013 (1995).

    Article  CAS  PubMed  Google Scholar 

  21. Mocsai, A., Ruland, J. & Tybulewicz, V. L. The SYK tyrosine kinase: A crucial player in diverse biological functions. Nat. Rev. Immunol. 10, 387–402 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Richard, A. J. & Stephens, J. M. The role of JAK-STAT signaling in adipose tissue function. Biochim. Biophys. Acta 1842, 431–439 (2014).

    Article  CAS  PubMed  Google Scholar 

  23. Meyer, D. M. et al. Anti-inflammatory activity and neutrophil reductions mediated by the JAK1/JAK3 inhibitor, CP-690,550, in rat adjuvant-induced arthritis. J. Inflamm. 7, 41–51 (2010).

    Article  Google Scholar 

  24. Flanagan, M. E. et al. Discovery of CP-690,550: a potent and selective Janus kinase (JAK) inhibitor for the treatment of autoimmune diseases and organ transplant rejection. J. Med. Chem. 53, 8468–8484 (2010).

    Article  CAS  PubMed  Google Scholar 

  25. Braselmann, S. et al. R406, an orally available spleen tyrosine kinase inhibitor blocks fc receptor signaling and reduces immune complex-mediated inflammation. J. Pharmacol. Exp. Ther. 319, 998–1008 (2006).

    Article  CAS  PubMed  Google Scholar 

  26. Cao, W., Medvedev, A. V., Daniel, K. W. & Collins, S. β-Adrenergic activation of p38 MAP kinase in adipocytes: cAMP induction of the uncoupling protein 1 (UCP1) gene requires p38 MAP kinase. J. Biol. Chem. 276, 27077–27082 (2001).

    Article  CAS  PubMed  Google Scholar 

  27. Cao, W. et al. p38 mitogen-activated protein kinase is the central regulator of cyclic AMP-dependent transcription of the brown fat uncoupling protein 1 gene. Mol. Cell. Biol. 24, 3057–3067 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Valladares, A., Roncero, C., Benito, M. & Porras, A. TNF-α inhibits UCP-1 expression in brown adipocytes via ERKs. Opposite effect of p38MAPK. FEBS Lett. 493, 6–11 (2001).

    Article  CAS  PubMed  Google Scholar 

  29. Aaronson, D. S. & Horvath, C. M. A road map for those who don’t know JAK-STAT. Science 296, 1653–1655 (2002).

    Article  CAS  PubMed  Google Scholar 

  30. Todoric, J. et al. Cross-talk between interferon-γ and hedgehog signaling regulates adipogenesis. Diabetes 60, 1668–1676 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Pospisilik, J. A. et al. Drosophila genome-wide obesity screen reveals hedgehog as a determinant of brown versus white adipose cell fate. Cell 140, 148–160 (2010).

    Article  CAS  PubMed  Google Scholar 

  32. Derecka, M. et al. Tyk2 and Stat3 regulate brown adipose tissue differentiation and obesity. Cell Metab. 16, 814–824 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Bray, M. A. & Carpenter, A. et al. in Assay Guidance Manual (ed Sittampalam, G. S.) (Bethesda, 2004).

    Google Scholar 

  34. Prummer, M. Hypothesis testing in high-throughput screening for drug discovery. J. Biomol. Screen. 17, 519–529 (2012).

    Article  CAS  PubMed  Google Scholar 

  35. Storey, J. D. A direct approach to false discovery rates. J. Royal Stat. Soc. 64, 479–498 (2002).

    Article  Google Scholar 

Download references

Acknowledgements

The authors thank I. Clausen, M. Kapps, R. Schmucki and A. Schuler for technical support, K. Christensen and M. Graf for stem cell support, L. Badi for preliminary data analysis, C. Solier, A. Schell-Steven and T. Bergauer for experimental planning and M. Pawlak (Natural and Medical Sciences Institute at the University of Tübingen) for RPPA analyses. A.M. was supported by the Roche Postdoctoral Fellowship (RPF) program (2011–2013). This research was supported in part by F. Hoffmann-La Roche; grant R01DK095384 (C.A.C. and Y.K.L.) and R01DK097768 (C.A.C.) from the United States Institutes of Health (NIH); and Harvard University.

Author information

Authors and Affiliations

Authors

Contributions

A.M. designed and performed experiments, analysed data and wrote the manuscript; Y-K.L. performed experiments, analysed data and edited the manuscript; R.G., C.S.H. and F.X. performed experiments; J.D.Z. and M.E. performed bioinformatics analyses and contributed to the main text related to Fig. 7; H.H.T., S.Z. and M.P. performed high-content imaging analysis; A.K. performed RNA-seq; C.A.M. and R.T.S. supervised stem cell activities; K.E.A. supervised the project and C.A.C. supervised the project and wrote the manuscript. A.M., Y-K.L., R.G., C.S.H., M.P., J.D.Z., H.H.T., S.Z. and A.K. contributed to description of Methods.

Corresponding authors

Correspondence to Annie Moisan or Chad A. Cowan.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 3

(a) UCP1 expression is increased in PSC-WA after treatment with indicated compounds. Rosiglitazone induced expression of both UCP1 and FABP4. Values represent the mean of two biological replicates. (b) Browning screen performance: Plate-wise control distribution and Z’ factors for UCP1 and FABP4. For UCP1, <Z’ >= 0.50, for FABP4, <Z’ >= 0.64. (c) Reproducibility of replicates. The correlation of the intra-plate replicates for UCP1 (left) and FABP4 (right) in log–log representation. Controls are plotted in red (negative) and green (positive). Compounds with large inter-run differences were excluded from further analysis. (d) Hit selection. Left panel: Quantile–quantile plot of the normalized UCP1 reads. The blue line indicates the expected profile for a Gaussian distribution without actives. The blue dashed lines limit the confidence band of a correlation test corroborating the hit selection from a different angle, as all the selected compounds (green dots) lie outside. The P-value distribution drawn in the inset, which is the basis of the hit selection, shows no irregular features. Right panel: Scatter plot of the normalized FABP4 and UCP1 signal on logarithmic axes. Black dots: inactive compounds. Color dots: active compounds, red: rosiglitazone, yellow: rosiglitazone-like compounds in screen, blue: potential browning hits in screen, green: rosiglitazone-like compounds and browning hits confirmed in validation run, grey : non-confirmed in validation run.

Supplementary Figure 4 PSC-WA were differentiated and treated as described in Fig. 1b.

At day 14, adipocytes were fixed, stained and imaged by confocal microscopy. Green: lipids, Red: nuclei. Scale bars: 50 mM.

Supplementary Figure 5 Chemical name and structure of the model browning compounds R406 and tofacitinib.

Supplementary Figure 6

(a) Uncropped bright field images showing that tofacitinib and R406 induce brown-like lipid morphology in human primary adipocytes more prominently than BMP7. ADSC: Adipose tissue-derived stem cells. (b) RT-PCR analysis of UCP1 expression in mouse visceral white adipose tissue explants following 7 days of treatment with tofacitinib (tofa.) or R406. Values are mean ± s.e.m. of n = three biological replicates of pooled tissue from 5 mice.

Supplementary Figure 7 Reverse phase protein analysis (RPPA) of tofacitinib and R406-treated PSC-WA.

Cells were collected 20’ after addition of compounds to PSC-WA and processed for RPPA. Data shown is relative to DMSO. All 51 antibodies recognized the phospho-isoforms of indicated proteins. 3 antibodies did not reach detection threshold and were excluded from graph. Data from one experiment (n = 3 biological replicates) representative of 2 independent experiments. Values are mean ± s.d. of three biological replicates.

Supplementary Figure 8

(a) R406 promoted mRNA expression of PGC1α, PGC1β and PPARG but not of PRDM16. (b) The transcriptional changes downstream of R406 include up-regulation of PPARG, SREBF and BMP target genes. (c) Gene expression regulation by tofacitinib (tofa., JAK3i) versus R406 (SYKi) in PSC-WA at day 7. 54 genes were up-regulated by both compounds, 17 of which were BA-specific, that is, low in PSC-WA and high in PSC-BA (yellow dots). (ac) N = 3 biological replicates. Each independent biological replicate was pooled from two individual wells.

Supplementary Figure 9 The ratio of UCP1/FABP4 mRNA level was down-regulated in PSC-BA upon treatment with cyclopamine.

Values represent the mean of two biological replicates.

Supplementary Figure 10 Original scans of western blot analyses presented in Figs 3, 4 and 6.

Supplementary Table 1 Validated browning hits and PPAR agonists shown in Fig. 1d.
Supplementary Table 2 Panomics QuantiGene2.0 Probes used for branched DNA analysis of gene expression.

Supplementary information

Supplementary Information

Supplementary Information (PDF 2148 kb)

Source Data

Supplementary Information (XLSX 427 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Moisan, A., Lee, YK., Zhang, J. et al. White-to-brown metabolic conversion of human adipocytes by JAK inhibition. Nat Cell Biol 17, 57–67 (2015). https://doi.org/10.1038/ncb3075

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3075

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