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.

  • Letter
  • Published:

Brown-fat paucity due to impaired BMP signalling induces compensatory browning of white fat

Abstract

Maintenance of body temperature is essential for the survival of homeotherms. Brown adipose tissue (BAT) is a specialized fat tissue that is dedicated to thermoregulation1. Owing to its remarkable capacity to dissipate stored energy and its demonstrated presence in adult humans2,3,4,5, BAT holds great promise for the treatment of obesity and metabolic syndrome1. Rodent data suggest the existence of two types of brown fat cells: constitutive BAT (cBAT), which is of embryonic origin and anatomically located in the interscapular region of mice; and recruitable BAT (rBAT), which resides within white adipose tissue (WAT)6 and skeletal muscle7, and has alternatively been called beige8, brite9 or inducible BAT10. Bone morphogenetic proteins (BMPs) regulate the formation and thermogenic activity of BAT10,11,12. Here we use mouse models to provide evidence for a systemically active regulatory mechanism that controls whole-body BAT activity for thermoregulation and energy homeostasis. Genetic ablation of the type 1A BMP receptor (Bmpr1a) in brown adipogenic progenitor cells leads to a severe paucity of cBAT. This in turn increases sympathetic input to WAT, thereby promoting the formation of rBAT within white fat depots. This previously unknown compensatory mechanism, aimed at restoring total brown-fat-mediated thermogenic capacity in the body, is sufficient to maintain normal temperature homeostasis and resistance to diet-induced obesity. These data suggest an important physiological cross-talk between constitutive and recruitable brown fat cells. This sophisticated regulatory mechanism of body temperature may participate in the control of energy balance and metabolic disease.

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: Loss of Bmpr1a impairs cBAT formation by decreasing embryonic progenitor proliferation.
Figure 2: Ablation of Bmpr1a in brown pre-adipocytes derived from the Myf5 + lineage inhibits differentiation.
Figure 3: Specific ablation of cBAT results in a compensatory response of increased browning by enhanced sympathetic input to WAT.
Figure 4: Loss of sympathetic innervation causes atrophy of cBAT and compensatory browning of white fat.

Similar content being viewed by others

References

  1. Cannon, B. & Nedergaard, J. Metabolic consequences of the presence or absence of the thermogenic capacity of brown adipose tissue in mice (and probably in humans). Int. J. Obes. Lond. 34 (suppl. 1). S7–S16 (2010)

    Article  CAS  Google Scholar 

  2. 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  Google Scholar 

  3. Nedergaard, J., Bengtsson, T. & Cannon, B. Unexpected evidence for active brown adipose tissue in adult humans. Am. J. Physiol. Endocrinol. Metab. 293, E444–E452 (2007)

    Article  CAS  Google Scholar 

  4. van Marken Lichtenbelt, W. D. et al. Cold-activated brown adipose tissue in healthy men. N. Engl. J. Med. 360, 1500–1508 (2009)

    Article  CAS  Google Scholar 

  5. Virtanen, K. A. et al. Functional brown adipose tissue in healthy adults. N. Engl. J. Med. 360, 1518–1525 (2009)

    Article  CAS  Google Scholar 

  6. Guerra, C., Koza, R. A., Yamashita, H., Walsh, K. & Kozak, L. P. Emergence of brown adipocytes in white fat in mice is under genetic control. Effects on body weight and adiposity. J. Clin. Invest. 102, 412–420 (1998)

    Article  CAS  Google Scholar 

  7. Almind, K., Manieri, M., Sivitz, W. I., Cinti, S. & Kahn, C. R. Ectopic brown adipose tissue in muscle provides a mechanism for differences in risk of metabolic syndrome in mice. Proc. Natl Acad. Sci. USA 104, 2366–2371 (2007)

    Article  ADS  CAS  Google Scholar 

  8. 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  Google Scholar 

  9. 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  Google Scholar 

  10. Schulz, T. J. et al. Identification of inducible brown adipocyte progenitors residing in skeletal muscle and white fat. Proc. Natl Acad. Sci. USA 108, 143–148 (2011)

    Article  ADS  CAS  Google Scholar 

  11. Tseng, Y. H. et al. New role of bone morphogenetic protein 7 in brown adipogenesis and energy expenditure. Nature 454, 1000–1004 (2008)

    Article  ADS  CAS  Google Scholar 

  12. Whittle, A. J. et al. BMP8B increases brown adipose tissue thermogenesis through both central and peripheral actions. Cell 149, 871–885 (2012)

    Article  CAS  Google Scholar 

  13. Seale, P. et al. PRDM16 controls a brown fat/skeletal muscle switch. Nature 454, 961–967 (2008)

    Article  ADS  CAS  Google Scholar 

  14. Timmons, J. A. et al. Myogenic gene expression signature establishes that brown and white adipocytes originate from distinct cell lineages. Proc. Natl Acad. Sci. USA 104, 4401–4406 (2007)

    Article  ADS  CAS  Google Scholar 

  15. Atit, R. et al. β-catenin activation is necessary and sufficient to specify the dorsal dermal fate in the mouse. Dev. Biol. 296, 164–176 (2006)

    Article  CAS  Google Scholar 

  16. Lepper, C. & Fan, C. M. Inducible lineage tracing of Pax7-descendant cells reveals embryonic origin of adult satellite cells. Genesis 48, 424–436 (2010)

    Article  CAS  Google Scholar 

  17. Schmid, P., Lorenz, A., Hameister, H. & Montenarh, M. Expression of p53 during mouse embryogenesis. Development 113, 857–865 (1991)

    CAS  PubMed  Google Scholar 

  18. Sanchez-Gurmaches, J. et al. PTEN loss in the Myf5 lineage redistributes body fat and reveals subsets of white adipocytes that arise from Myf5 precursors. Cell Metab. 16, 348–362 (2012)

    Article  CAS  Google Scholar 

  19. Buckingham, M. E., Lyons, G. E., Ott, M. O. & Sassoon, D. A. Myogenesis in the mouse. Ciba Found. Symp. 165, 111–131 (1992)

    CAS  PubMed  Google Scholar 

  20. Kishigami, S. & Mishina, Y. BMP signaling and early embryonic patterning. Cytokine Growth Factor Rev. 16, 265–278 (2005)

    Article  CAS  Google Scholar 

  21. Gupta, R. K. et al. Transcriptional control of preadipocyte determination by Zfp423. Nature 464, 619–623 (2010)

    Article  ADS  CAS  Google Scholar 

  22. Farmer, S. R. Transcriptional control of adipocyte formation. Cell Metab. 4, 263–273 (2006)

    Article  CAS  Google Scholar 

  23. Cannon, B. & Nedergaard, J. Nonshivering thermogenesis and its adequate measurement in metabolic studies. J. Exp. Biol. 214, 242–253 (2011)

    Article  Google Scholar 

  24. Fisher, F. M. et al. FGF21 regulates PGC-1α and browning of white adipose tissues in adaptive thermogenesis. Genes Dev. 26, 271–281 (2012)

    Article  CAS  Google Scholar 

  25. Boström, P. et al. A PGC1-α-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 481, 463–468 (2012)

    Article  ADS  Google Scholar 

  26. Collins, S. β-Adrenoceptor signaling networks in adipocytes for recruiting stored fat and energy expenditure. Front. Endocrinol. 2, 102 (2011)

    Article  Google Scholar 

  27. Tseng, Y. H., Cypess, A. M. & Kahn, C. R. Cellular bioenergetics as a target for obesity therapy. Nature Rev. Drug Discov. 9, 465–482 (2010)

    Article  CAS  Google Scholar 

  28. 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  Google Scholar 

  29. Rothwell, N. J. & Stock, M. J. Surgical removal of brown fat results in rapid and complete compensation by other depots. Am. J. Physiol. 257, R253–R258 (1989)

    CAS  PubMed  Google Scholar 

  30. Xue, B. et al. Genetic variability affects the development of brown adipocytes in white fat but not in interscapular brown fat. J. Lipid Res. 48, 41–51 (2007)

    Article  CAS  Google Scholar 

  31. Dudas, M., Sridurongrit, S., Nagy, A., Okazaki, K. & Kaartinen, V. Craniofacial defects in mice lacking BMP type I receptor Alk2 in neural crest cells. Mech. Dev. 121, 173–182 (2004)

    Article  CAS  Google Scholar 

  32. Mishina, Y., Hanks, M. C., Miura, S., Tallquist, M. D. & Behringer, R. R. Generation of Bmpr/Alk3 conditional knockout mice. Genesis 32, 69–72 (2002)

    Article  CAS  Google Scholar 

  33. Tallquist, M. D., Weismann, K. E., Hellstrom, M. & Soriano, P. Early myotome specification regulates PDGFA expression and axial skeleton development. Development 127, 5059–5070 (2000)

    CAS  PubMed  Google Scholar 

  34. Abel, E. D. et al. Adipose-selective targeting of the GLUT4 gene impairs insulin action in muscle and liver. Nature 409, 729–733 (2001)

    Article  ADS  CAS  Google Scholar 

  35. Yi, S. E., Daluiski, A., Pederson, R., Rosen, V. & Lyons, K. M. The type I BMP receptor BMPRIB is required for chondrogenesis in the mouse limb. Development 127, 621–630 (2000)

    CAS  PubMed  Google Scholar 

  36. Kaufman, M. H. The Atlas of Mouse Development Vol. 1 (Academic, 1992)

    Google Scholar 

  37. Klein, J., Fasshauer, M., Klein, H. H., Benito, M. & Kahn, C. R. Novel adipocyte lines from brown fat: a model system for the study of differentiation, energy metabolism, and insulin action. Bioessays 24, 382–388 (2002)

    Article  CAS  Google Scholar 

  38. Pulinilkunnil, T. et al. Adrenergic regulation of AMP-activated protein kinase in brown adipose tissue in vivo. J. Biol. Chem. 286, 8798–8809 (2011)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported in part by National Institutes of Health (NIH) grants R01 DK077097 (Y.-H.T.), and Joslin Diabetes Center’s Diabetes Research Center (DRC; P30 DK036836 from the NIDDK), a research grant from the Eli Lilly Research Foundation, and by funding from the Harvard Stem Cell Institute (to Y.-H.T.). T.J.S. was supported by the Mary K. Iacocca Foundation and the German Research Foundation (DFG; SCHU2445/1-1). P.H. was supported by a Scientist Development Grant from the American Heart Association (0730285N). K.L.T. was supported by NIH fellowships (T32 DK007260 and F32 DK091996). R.X. was supported by Project 985III-YFX0302 and NSFC81070680 from the National Natural Science Foundation of China. The authors thank Stryker Regenerative Medicine for the gift of recombinant BMP7. We acknowledge V. Kaartinen, B. Kahn, K. Lyons and P. Soriano for providing floxed Acvr1 mice, FABP4-Cre mice, Bmpr1b heterozygous mice and Myf5-Cre mice, respectively. The authors thank C. R. Kahn, L. J. Goodyear, E. Kokkotou and D. Breault for comments on the manuscript. The authors wish to thank J. LaVecchio, G. Buruzula, A. Wakabayashi, A. Pinkhasov, A. Clermont, M. Mulvey, C. Cahill and G. Sankaranarayanan for technical assistance, and E. Caniano for editorial contributions.

Author information

Authors and Affiliations

Authors

Contributions

T.J.S. and Y.-H.T. planned most of the experiments and wrote the paper. T.J.S. performed the majority of the experiments. P.H., T.L.H., L.E.M., R.X. and K.L.T. performed some of the animal and immunofluorescence experiments and/or provided research assistance. A.M.C. helped with the infrared thermography and provided valuable research materials. Y.M. and E.G. planned some of the experiments and contributed valuable research materials.

Corresponding author

Correspondence to Yu-Hua Tseng.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-13 and Supplementary Tables 1 and 2 which list primer sequences and expression levels of gene measured by quantitative real-time PCR. (PDF 1620 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Schulz, T., Huang, P., Huang, T. et al. Brown-fat paucity due to impaired BMP signalling induces compensatory browning of white fat. Nature 495, 379–383 (2013). https://doi.org/10.1038/nature11943

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature11943

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