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:

Clonal analyses and gene profiling identify genetic biomarkers of the thermogenic potential of human brown and white preadipocytes

Abstract

Targeting brown adipose tissue (BAT) content or activity has therapeutic potential for treating obesity and the metabolic syndrome by increasing energy expenditure. However, both inter- and intra-individual differences contribute to heterogeneity in human BAT and potentially to differential thermogenic capacity in human populations. Here we generated clones of brown and white preadipocytes from human neck fat and characterized their adipogenic and thermogenic differentiation. We combined an uncoupling protein 1 (UCP1) reporter system and expression profiling to define novel sets of gene signatures in human preadipocytes that could predict the thermogenic potential of the cells once they were maturated. Knocking out the positive UCP1 regulators, PREX1 and EDNRB, in brown preadipocytes using CRISPR-Cas9 markedly abolished the high level of UCP1 in brown adipocytes differentiated from the preadipocytes. Finally, we were able to prospectively isolate adipose progenitors with great thermogenic potential using the cell surface marker CD29. These data provide new insights into the cellular heterogeneity in human fat and offer potential biomarkers for identifying thermogenically competent preadipocytes.

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: Generation and characterization of immortalized human brown and white fat progenitors.
Figure 2: Use of a UCP1 reporter system for in vitro and in vivo monitoring of UCP1 expression.
Figure 3: Clonal analysis of human brown and white fat progenitors.
Figure 4: Gene expression profiles in adipose progenitors predict the thermogenic capacity of mature adipocytes.
Figure 5: PREX1 and EDNRB are required for determining thermogenic competency.
Figure 6: Isolation of progenitors possessing thermogenic potential using a cell surface marker.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Cannon, B. & Nedergaard, J. Brown adipose tissue: function and physiological significance. Physiol. Rev. 84, 277–359 (2004).

    Article  CAS  Google Scholar 

  2. Schulz, T.J. & Tseng, Y.H. Brown adipose tissue: development, metabolism and beyond. Biochem. J. 453, 167–178 (2013).

    Article  CAS  Google Scholar 

  3. 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 

  4. Petrovic, N. et al. Chronic peroxisome proliferator-activated receptor gamma (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 

  5. Harms, M. & Seale, P. Brown and beige fat: development, function and therapeutic potential. Nat. Med. 19, 1252–1263 (2013).

    Article  CAS  Google Scholar 

  6. Nedergaard, J. & Cannon, B. The browning of white adipose tissue: some burning issues. Cell Metab. 20, 396–407 (2014).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  9. 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 

  10. 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 

  11. 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 

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

    Article  CAS  Google Scholar 

  13. Saito, M. et al. High incidence of metabolically active brown adipose tissue in healthy adult humans: effects of cold exposure and adiposity. Diabetes 58, 1526–1531 (2009).

    Article  CAS  Google Scholar 

  14. Zingaretti, M.C. et al. The presence of UCP1 demonstrates that metabolically active adipose tissue in the neck of adult humans truly represents brown adipose tissue. FASEB J. 23, 3113–3120 (2009).

    Article  CAS  Google Scholar 

  15. Himms-Hagen, J. et al. Effect of CL-316,243, a thermogenic β3-agonist, on energy balance and brown and white adipose tissues in rats. Am. J. Physiol. 266, R1371–R1382 (1994).

    CAS  PubMed  Google Scholar 

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

    Article  CAS  Google Scholar 

  17. 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  Google Scholar 

  18. Ouellet, V. et al. Brown adipose tissue oxidative metabolism contributes to energy expenditure during acute cold exposure in humans. J. Clin. Invest. 122, 545–552 (2012).

    Article  Google Scholar 

  19. 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 

  20. Cypess, A.M. et al. Anatomical localization, gene expression profiling and functional characterization of adult human neck brown fat. Nat. Med. 19, 635–639 (2013).

    Article  CAS  Google Scholar 

  21. Lidell, M.E. et al. Evidence for two types of brown adipose tissue in humans. Nat. Med. 19, 631–634 (2013).

    Article  CAS  Google Scholar 

  22. Jespersen, N.Z. et al. A classical brown adipose tissue mRNA signature partly overlaps with brite in the supraclavicular region of adult humans. Cell Metab. 17, 798–805 (2013).

    Article  CAS  Google Scholar 

  23. 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  CAS  Google Scholar 

  24. Lee, Y.H., Petkova, A.P., Mottillo, E.P. & Granneman, J.G. In vivo identification of bipotential adipocyte progenitors recruited by β3-adrenoceptor activation and high-fat feeding. Cell Metab. 15, 480–491 (2012).

    Article  CAS  Google Scholar 

  25. Berry, R. & Rodeheffer, M.S. Characterization of the adipocyte cellular lineage in vivo. Nat. Cell Biol. 15, 302–308 (2013).

    Article  CAS  Google Scholar 

  26. Wang, W. et al. Ebf2 is a selective marker of brown and beige adipogenic precursor cells. Proc. Natl. Acad. Sci. USA 111, 14466–14471 (2014).

    Article  CAS  Google Scholar 

  27. Tchkonia, T. et al. Fat depot–specific characteristics are retained in strains derived from single human preadipocytes. Diabetes 55, 2571–2578 (2006).

    Article  CAS  Google Scholar 

  28. 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 

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

    Article  CAS  Google Scholar 

  30. Seale, P. et al. Transcriptional control of brown fat determination by PRDM16. Cell Metab. 6, 38–54 (2007).

    Article  CAS  Google Scholar 

  31. 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  CAS  Google Scholar 

  32. Welch, H.C. et al. P-Rex1, a PtdIns(3,4,5)P3- and Gβγ-regulated guanine-nucleotide exchange factor for Rac. Cell 108, 809–821 (2002).

    Article  CAS  Google Scholar 

  33. Cheung, J. et al. Identification of the human cortactin-binding protein-2 gene from the autism candidate region at 7q31. Genomics 78, 7–11 (2001).

    Article  CAS  Google Scholar 

  34. Zhang, S.X. et al. Identification of direct serum-response factor gene targets during Me2SO-induced P19 cardiac cell differentiation. J. Biol. Chem. 280, 19115–19126 (2005).

    Article  CAS  Google Scholar 

  35. Yamada, Y. et al. Cloning and functional characterization of a family of human and mouse somatostatin receptors expressed in brain, gastrointestinal tract and kidney. Proc. Natl. Acad. Sci. USA 89, 251–255 (1992).

    Article  CAS  Google Scholar 

  36. Kikkawa, T. et al. Dmrta1 regulates proneural gene expression downstream of Pax6 in the mammalian telencephalon. Genes Cells 18, 636–649 (2013).

    Article  CAS  Google Scholar 

  37. Garciafigueroa, D.Y., Klei, L.R., Ambrosio, F. & Barchowsky, A. Arsenic-stimulated lipolysis and adipose remodeling is mediated by G-protein–coupled receptors. Toxicol. Sci. 134, 335–344 (2013).

    Article  CAS  Google Scholar 

  38. Chen, T.Y. et al. Endogenous n-3 polyunsaturated fatty acids (PUFAs) mitigate ovariectomy-induced bone loss by attenuating bone marrow adipogenesis in FAT1 transgenic mice. Drug Des. Devel. Ther. 7, 545–552 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Behjati, S. et al. Recurrent PTPRB and PLCG1 mutations in angiosarcoma. Nat. Genet. 46, 376–379 (2014).

    Article  CAS  Google Scholar 

  40. Takada, Y., Ye, X. & Simon, S. The integrins. Genome Biol. 8, 215 (2007).

    Article  Google Scholar 

  41. Margadant, C., Monsuur, H.N., Norman, J.C. & Sonnenberg, A. Mechanisms of integrin activation and trafficking. Curr. Opin. Cell Biol. 23, 607–614 (2011).

    Article  CAS  Google Scholar 

  42. Balamatsias, D. et al. Identification of P-Rex1 as a novel Rac1-guanine nucleotide exchange factor (GEF) that promotes actin remodeling and GLUT4 protein trafficking in adipocytes. J. Biol. Chem. 286, 43229–43240 (2011).

    Article  CAS  Google Scholar 

  43. Lewis, J.P. et al. Analysis of candidate genes on chromosome 20q12–13.1 reveals evidence for BMI-mediated association of PREX1 with type 2 diabetes in European Americans. Genomics 96, 211–219 (2010).

    Article  CAS  Google Scholar 

  44. Wu-Wong, J.R., Berg, C.E. & Dayton, B.D. Endothelin-stimulated glucose uptake: effects of intracellular Ca2+, cAMP and glucosamine. Clin. Sci. 103 (suppl. 48), 418S–423S (2002).

    Article  CAS  Google Scholar 

  45. Juan, C.C. et al. Effect of endothelin-1 on lipolysis in rat adipocytes. Obesity (Silver Spring) 14, 398–404 (2006).

    Article  CAS  Google Scholar 

  46. Shinoda, K. et al. Genetic and functional characterization of clonally derived adult human brown adipocytes. Nat. Med. 21, 389–394 (2015).

    Article  CAS  Google Scholar 

  47. Gierloff, M. et al. Adipogenic differentiation potential of rat adipose tissue–derived subpopulations of stromal cells. J. Plast. Reconstr. Aesthet. Surg. 67, 1427–1435 (2014).

    Article  CAS  Google Scholar 

  48. Farnier, C. et al. The signaling pathway for β1-integrin/ERKs is involved in the adaptation of adipocyte functions to cell size. Ann. NY Acad. Sci. 973, 594–597 (2002).

    Article  CAS  Google Scholar 

  49. Kawaguchi, N. et al. ADAM12 induces actin cytoskeleton and extracellular matrix reorganization during early adipocyte differentiation by regulating β1 integrin function. J. Cell Sci. 116, 3893–3904 (2003).

    Article  CAS  Google Scholar 

  50. Irizarry, R.A. et al. Exploration, normalization, and summaries of high-density oligonucleotide array probe level data. Biostatistics 4, 249–264 (2003).

    Article  Google Scholar 

  51. Benjamin, Y. et al. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Statist. Soc. B 57, 289–300 (1995).

    Google Scholar 

Download references

Acknowledgements

This work was supported in part by US National Institutes of Health (NIH) grants R01DK077097 (to Y.-H.T.), R01DK099511 (to L.J.G.), K23DK081604 (to A.M.C.) and P30DK036836 (to Joslin Diabetes Center's Diabetes Research Center, DRC) from the National Institute of Diabetes and Digestive and Kidney Diseases, a sponsored research grant from Chugai Pharmaceutical Co. (to Y.-H.T. and A.M.C.), a research grant from the American Diabetes Association (ADA 7-12-BS-191, to Y.-H.T.), the Intramural Research Program of the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), and by funding from the Harvard Stem Cell Institute (to Y.-H.T.). M.D.L. was supported by NIH fellowships (T32DK007260 and F32DK102320). We thank M.-E. Patti and K. Hughes of the Advanced Genomics and Genetics Core of Joslin's DRC for advice and expert technical assistance. The authors thank Stryker Regenerative Medicine (Hopkinton, Massachusetts) for the generous gift of recombinant BMP7.

Author information

Authors and Affiliations

Authors

Contributions

The study was designed by Y.-H.T., R.X., M.D.L. and A.M.C. The manuscript was written by Y.-H.T., M.D.L., R.X. and J.M.D. R.X. performed the majority of the experiments. M.D.L. did the time-lapse imaging, IVIS scanning and FACS. J.M.D. analyzed microarray data. F.S. performed bioenergetics analyses in knockout cells. T.J.S. and H.Z. established the method of isolation, immortalization and differentiation of human fat progenitors. T.L.H. did the human cell implantation and gene expression microarrays. K.L.T. provided assistance with the Seahorse bioanalyzer. Y.L. provided research assistance. H.T. and L.J.G. helped with fuel utilization experiments. A.M.C., L.S.W. and A.P.W. collected human fat samples. M.S.L. and L.L.R. helped with the time-lapse imaging. All authors contributed to editing the manuscript.

Corresponding author

Correspondence to Yu-Hua Tseng.

Ethics declarations

Competing interests

A.M.C. and Y.-H.T. are recipients of a sponsored research grant and licensing payments from Chugai Pharmaceutical Co., Ltd through Joslin Diabetes Center.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–9 & Supplementary Tables 1–5 (PDF 1480 kb)

Time lapse imaging of hBAT-SVF differentiation (MOV 31877 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xue, R., Lynes, M., Dreyfuss, J. 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). https://doi.org/10.1038/nm.3881

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3881

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