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:

Rapid depot-specific activation of adipocyte precursor cells at the onset of obesity

Abstract

Excessive accumulation of white adipose tissue (WAT) is the defining characteristic of obesity. WAT mass is composed primarily of mature adipocytes, which are generated through the proliferation and differentiation of adipocyte precursors (APs). Although the production of new adipocytes contributes to WAT growth in obesity, little is known about the cellular and molecular mechanisms underlying adipogenesis in vivo. Here, we show that high-fat diet feeding in mice rapidly and transiently induces proliferation of APs within WAT to produce new adipocytes. Importantly, the activation of adipogenesis is specific to the perigonadal visceral depot in male mice, consistent with the patterns of obesogenic WAT growth observed in humans. Furthermore, we find that in multiple models of obesity, the activation of APs is dependent on the phosphoinositide 3-kinase (PI3K)-AKT2 pathway; however, the development of WAT does not require AKT2. These data indicate that developmental and obesogenic adipogenesis are regulated through distinct molecular mechanisms.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: High-fat diet feeding induces depot-specific adipocyte hyperplasia.
Figure 2: High-fat diet feeding induces adipocyte precursor activation.
Figure 3: Activated adipocyte precursors undergo adipogenesis in vivo.
Figure 4: Diet-induced proliferation of adipocyte precursors correlates with cell-intrinsic Akt phosphorylation.
Figure 5: Diet-induced adipocyte precursor activation and adipogenesis requires Akt2 in adipocyte lineage cells.
Figure 6: Akt2 is required for activation of adipocyte precursors in multiple models of obesity.
Figure 7: Akt2 is not required for normal development of white adipose tissue.

Similar content being viewed by others

References

  1. Kelly, T., Yang, W., Chen, C. S., Reynolds, K. & He, J. Global burden of obesity in 2005 and projections to 2030. Int. J. Obes. (Lond). 32, 1431–1437 (2008).

    Article  CAS  Google Scholar 

  2. Peckham, S. C., Entenman, C. & Carroll, H. W. The influence of a hypercaloric diet on gross body and adipose tissue composition in the rat. J. Nutr. 77, 187–197 (1962).

    Article  CAS  PubMed  Google Scholar 

  3. Steinberg, M. D., Zingg, W. & Angel, A. Studies of the number and volume of fat cells in adipose tissue. J. Pediatr. 61, 299–300 (1962).

    Article  CAS  PubMed  Google Scholar 

  4. Enesco, M. & Leblond, C. P. Increase in cell number as a factor in the growth of the organs and tissues of the young male rat. J. Embryol. Exp. Morphol. 10, 530–562 (1962).

    Google Scholar 

  5. Spalding, K. L. et al. Dynamics of fat cell turnover in humans. Nature 453, 783–787 (2008).

    Article  CAS  PubMed  Google Scholar 

  6. Arner, P. et al. Variations in the size of the major omentum are primarily determined by fat cell number. J. Clin. Endocrinol. Metab. 98, E897–E901 (2013).

    Article  PubMed  Google Scholar 

  7. Phillips, L. K. & Prins, J. B. The link between abdominal obesity and the metabolic syndrome. Curr. Hypertens. Rep. 10, 156–164 (2008).

    Article  CAS  PubMed  Google Scholar 

  8. Bjorntorp, P. Metabolic difference between visceral fat and subcutaneous abdominal fat. Diabetes Metab. 26 (Suppl. 3), 10–12 (2000)

  9. Wang, Y., Rimm, E. B., Stampfer, M. J., Willett, W. C. & Hu, F. B. Comparison of abdominal adiposity and overall obesity in predicting risk of type 2 diabetes among men. Am. J. Clin. Nutr. 81, 555–563 (2005).

    Article  CAS  PubMed  Google Scholar 

  10. Hirsch, J. & Batchelor, B. Adipose tissue cellularity in human obesity. Clin. Endocrinol. Metab. 5, 299–311 (1976).

    Article  CAS  PubMed  Google Scholar 

  11. Faust, I. M., Johnson, P. R., Stern, J. S. & Hirsch, J. Diet-induced adipocyte number increase in adult rats: a new model of obesity. Am. J. Physiol. 235, E279–E286 (1978).

    CAS  PubMed  Google Scholar 

  12. Lemonnier, D. Effect of age, sex, and sites on the cellularity of the adipose tissue in mice and rats rendered obese by a high-fat diet. J. Clin. Invest. 51, 2907–2915 (1972).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Wang, Q. A., Tao, C., Gupta, R. K. & Scherer, P. E. Tracking adipogenesis during white adipose tissue development, expansion and regeneration. Nat. Med. 19, 1338–1344 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Cleary, M. P., Brasel, J. A. & Greenwood, M. R. Developmental changes in thymidine kinase, DNA, and fat cellularity in Zucker rats. Am. J. Physiol. 236, E508–E513 (1979).

    CAS  PubMed  Google Scholar 

  15. Cristancho, A. G. & Lazar, M. A. Forming functional fat: a growing understanding of adipocyte differentiation. Nat. Rev. Mol. Cell Biol. 12, 722–734 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Rodeheffer, M. S., Birsoy, K. & Friedman, J. M. Identification of white adipocyte progenitor cells in vivo. Cell 135, 240–249 (2008).

    Article  CAS  PubMed  Google Scholar 

  17. Jeffery, E. et al. Characterization of Cre recombinase models for the study of adipose tissue. Adipocyte 3, 206–211 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Muzumdar, M. D., Tasic, B., Miyamichi, K., Li, L. & Luo, L. A global double-fluorescent Cre reporter mouse. Genesis 45, 593–605 (2007).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Pellettieri, J. & Sanchez Alvarado, A. Cell turnover and adult tissue homeostasis: from humans to planarians. Annu. Rev. Genet. 41, 83–105 (2007).

    Article  CAS  PubMed  Google Scholar 

  21. Joe, A. W., Yi, L., Even, Y., Vogl, A. W. & Rossi, F. M. Depot-specific differences in adipogenic progenitor abundance and proliferative response to high-fat diet. Stem Cells 27, 2563–2570 (2009).

    Article  CAS  PubMed  Google Scholar 

  22. Hudak, C. S. et al. Pref-1 marks very early mesenchymal precursors required for adipose tissue development and expansion. Cell Rep. 8, 678–687 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Church, C. D., Berry, R. & Rodeheffer, M. S. Isolation and study of adipocyte precursors. Methods Enzymol. 537, 31–46 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Berry, R., Jeffery, E. & Rodeheffer, M. S. Weighing in on adipocyte precursors. Cell Metab. 19, 8–20 (2014).

    Article  CAS  PubMed  Google Scholar 

  25. 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  PubMed  PubMed Central  Google Scholar 

  26. Hietakangas, V. & Cohen, S. M. Regulation of tissue growth through nutrient sensing. Annu. Rev. Genet. 43, 389–410 (2009).

    Article  CAS  PubMed  Google Scholar 

  27. Pearce, L. R., Komander, D. & Alessi, D. R. The nuts and bolts of AGC protein kinases. Nat. Rev. Mol. Cell Biol. 11, 9–22 (2010).

    Article  CAS  PubMed  Google Scholar 

  28. Eto, H. et al. Characterization of structure and cellular components of aspirated and excised adipose tissue. Plast. Reconstr. Surg. 124, 1087–1097 (2009).

    Article  CAS  PubMed  Google Scholar 

  29. Gonzalez, E. & McGraw, T. E. The Akt kinases: isoform specificity in metabolism and cancer. Cell Cycle 8, 2502–2508 (2009).

    Article  CAS  PubMed  Google Scholar 

  30. Cho, H., Thorvaldsen, J. L., Chu, Q., Feng, F. & Birnbaum, M. J. Akt1/PKBα is required for normal growth but dispensable for maintenance of glucose homeostasis in mice. J. Biol. Chem. 276, 38349–38352 (2001).

    Article  CAS  PubMed  Google Scholar 

  31. Chen, W. S. et al. Growth retardation and increased apoptosis in mice with homozygous disruption of the Akt1 gene. Genes Dev. 15, 2203–2208 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Cho, H. et al. Insulin resistance and a diabetes mellitus-like syndrome in mice lacking the protein kinase Akt2 (PKB β). Science 292, 1728–1731 (2001).

    Article  CAS  PubMed  Google Scholar 

  33. Garofalo, R. S. et al. Severe diabetes, age-dependent loss of adipose tissue, and mild growth deficiency in mice lacking Akt2/PKB β. J. Clin. Invest. 112, 197–208 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Gautron, L. & Elmquist, J. K. Sixteen years and counting: an update on leptin in energy balance. J. Clin. Invest. 121, 2087–2093 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Han, J. et al. The spatiotemporal development of adipose tissue. Development 138, 5027–5037 (2011).

    Article  CAS  PubMed  Google Scholar 

  36. Birsoy, K. et al. Analysis of gene networks in white adipose tissue development reveals a role for ETS2 in adipogenesis. Development 138, 4709–4719 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Pollack, A. A.M.A. Recognizes Obesity as a Disease New York Times Online (18 June 2013); http://www.nytimes.com/2013/06/19/business/ama-recognizes-obesity-as-a-disease.html

  38. Leavens, K. F., Easton, R. M., Shulman, G. I., Previs, S. F. & Birnbaum, M. J. Akt2 is required for hepatic lipid accumulation in models of insulin resistance. Cell Metab. 10, 405–418 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Czech, M. P., Tencerova, M., Pedersen, D. J. & Aouadi, M. Insulin signalling mechanisms for triacylglycerol storage. Diabetologia 56, 949–964 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Tontonoz, P. & Spiegelman, B. M. Fat and beyond: the diverse biology of PPARγ. Annu. Rev. Biochem. 77, 289–312 (2008).

    Article  CAS  PubMed  Google Scholar 

  41. Lodhi, I. J. et al. Inhibiting adipose tissue lipogenesis reprograms thermogenesis and PPARγ activation to decrease diet-induced obesity. Cell Metab. 16, 189–201 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. George, S. et al. A family with severe insulin resistance and diabetes due to a mutation in AKT2. Science 304, 1325–1328 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Hussain, K. et al. An activating mutation of AKT2 and human hypoglycemia. Science 334, 474 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Tran, T. T., Yamamoto, Y., Gesta, S. & Kahn, C. R. Beneficial effects of subcutaneous fat transplantation on metabolism. Cell Metab. 7, 410–420 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wajchenberg, B. L. Subcutaneous and visceral adipose tissue: their relation to the metabolic syndrome. Endocr. Rev. 21, 697–738 (2000).

    Article  CAS  PubMed  Google Scholar 

  46. Yamamoto, Y. et al. Adipose depots possess unique developmental gene signatures. Obesity 18, 872–878 (2010).

    Article  CAS  PubMed  Google Scholar 

  47. Grove, K. L., Fried, S. K., Greenberg, A. S., Xiao, X. Q. & Clegg, D. J. A microarray analysis of sexual dimorphism of adipose tissues in high-fat-diet-induced obese mice. Int. J. Obes. (Lond.) 34, 989–1000 (2010).

    Article  CAS  Google Scholar 

  48. Macotela, Y., Boucher, J., Tran, T. T. & Kahn, C. R. Sex and depot differences in adipocyte insulin sensitivity and glucose metabolism. Diabetes 58, 803–812 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Chau, Y. Y. et al. Visceral and subcutaneous fat have different origins and evidence supports a mesothelial source. Nat. Cell Biol. 16, 367–375 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Krueger, K. C., Costa, M. J., Du, H. & Feldman, B. J. Characterization of cre recombinase activity for in vivo targeting of adipocyte precursor cells. Stem Cell Rep. 3, 1147–1158 (2014).

    Article  CAS  Google Scholar 

  51. Rosen, E. D. & Spiegelman, B. M. What we talk about when we talk about fat. Cell 156, 20–44 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Van Herpen, N. A. & Schrauwen-Hinderling, V. B. Lipid accumulation in non-adipose tissue and lipotoxicity. Physiol. Behav. 94, 231–241 (2008).

    Article  CAS  PubMed  Google Scholar 

  53. Berry, R. et al. Imaging of adipose tissue. Methods Enzymol. 537, 47–73 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by American Diabetes Association Award 7-12-JF-46, DERC pilot project grant DK045735 and NIDDK grant DK090489 to M.S.R., Lo Graduate Fellowship for Excellence in Stem Cell Research from the Yale Stem Cell Center to E.J., and EMBO long-term fellowship ALTF 132-2011 to C.D.C.

Author information

Authors and Affiliations

Authors

Contributions

E.J., C.D.C. and M.S.R. designed experiments. E.J., C.D.C., B.H., L.C. and M.S.R. performed experiments. E.J., C.D.C. and B.H. analysed data. E.J., C.D.C., B.H. and M.S.R. interpreted data. E.J. and M.S.R. wrote the manuscript.

Corresponding author

Correspondence to Matthew S. Rodeheffer.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 3 Characterization of HFD-induced adipocyte hyperplasia.

(A) Experimental scheme for BrdU time course in Figure 2B. (B) Representative flow cytometry dot plots indicating sequential gating used to identify AP populations, including CD24+ adipocyte progenitors (CD45, CD31, CD34+, CD29+, Sca1+, CD24+), CD24 preadipocytes (CD45, CD31, CD34+, CD29+, Sca1+, CD24), and total APs (CD45, CD31, CD34+, CD29+, Sca1+). (C) Flow cytometry plots of SVF from VWAT of PdgfRα-H2B-GFP mice showing that the majority of GFP+ cells are CD29+; CD34+, of which 97.1 ± 1.5% are also Sca1+. (n = 3 mice) (D) Quantification of BrdU incorporation into GFP+ cells from VWAT of male PdgfRα-H2B-GFP mice during 1 week of SD or HFD feeding and BrdU treatment. (n = 5 mice for SD and 7 mice for HFD.) Significance between the indicated groups in (D) was calculated using a two-tailed Student’s t-test. Exact p-values are listed in Supplementary Table 1. Error bars or values represent mean ± s.e.m. (p < 0.01). HFD: high-fat diet, SD: standard diet, BrdU: bromodeoxyuridine.

Supplementary Figure 4 Adipocyte precursor proliferation during week 2 of HFD feeding does not lead to adipogenesis.

(A) Schematic and depicting the second week BrdU pulse-chase in Supplementary Figure 2B. (B) BrdU incorporation into adipocyte nuclei after pulse-chase from the second week of HFD feeding. (n = 5 for all groups). Error bars or values represent mean ± s.e.m. HFD: high-fat diet, SD: standard diet, BrdU: bromodeoxyuridine.

Supplementary Figure 5 High-fat diet does not induce AKT phosphorylation in whole fat.

(A) Western blot analysis for phosphorylated AKT at the indicated sites and total AKT, in lysates from whole VWAT on day 3 of diet. Each lane represents one animal. Uncropped blots are shown in Supplementary Figure 7I-L. (B) Quantification of pAKT western blots in (A), normalized to total AKT. (C-D) VWAT was treated with the indicated concentrations of insulin prior to tissue digestion, and APs were isolated in the presence of wortmannin and phosphatase inhibitors to limit further changes in pathway activation (see methods). APs were then fixed and analyzed by flow cytometry for levels of phosphorylated AKT at threonine 308 (C) or serine 473 (D). The same AP cell isolation procedure was used in the experiments in Figure 4B-F and 5B-C. (E) Western blot for phosphorylated AKT1 (S473) with lysates from APs enriched from SVF via Sca-1 bead pull down (Sca1-positive) and remaining unselected cells (Sca1-negative) (see methods) after 3 days of HFD or SD. Each lane represents pooled cells from 2 mice. Uncropped blots are shown in Supplementary Figure 7M-N. Significance between the indicated groups in (B) was calculated using a two-tailed Student’s t-test. Error bars represent mean ± s.e.m. HFD: high-fat diet, SD: standard diet.

Supplementary Figure 6 Deletion of AKT2 in the adipocyte lineage does not affect body weight or food intake.

(A) Quantification of BrdU incorporation into SWAT APs after 1 week of HFD or SD with daily injection of wortmannin (Wort), or vehicle (Veh) (n = 5 mice for each group). (B) PCR analysis of DNA isolated from AP cells from the VWAT and SWAT depots of female mice show successful excision of the floxed exons 4 and 5 from Akt2 gene, while Lin+ cells (CD45+ blood cells and CD31+ endothelial cells) contain the full-length Akt2 gene. Each lane represents cells isolated from the indicated depot of individual mice. (C) Body weight of Akt2fx/fl (n = 9 mice) and PdgfRα-cre; Akt2fl/fl (n = 12 mice) at 6 weeks of age. (D) Quantification of food intake normalized to body weight in the indicated groups of mice on SD or during the first 4 days of HFD feeding. (n = 3 mice for Akt2fl/fl SD and PdgfRα-cre; Akt2fl/flSD, n = 4 mice for PdgfRα-cre HFD, n = 2 mice for Akt2fl/fl HFD, and n = 3 mice for PdgfRα-cre; Akt2fl/fl HFD. (E) BrdU incorporation into SWAT APs of the indicated groups during the first week of HFD feeding compared to SD controls. (n = 7 mice for Akt2fl/fl SD, n = 4 mice for PdgfRα-cre; Akt2fl/fl SD, n = 14 mice for PdgfRα-cre; Akt2fl/fl HFD, n = 8 mice for Akt2fl/fl HFD, n = 5 mice for wild-type and PdgfRα-cre HFD). Significance in (D) was calculated using one-way ANOVA with Tukey’s test for multiple comparisons. Exact p-values are listed in Supplementary Table 1. Statistics source data for 4D can be found in Supplementary Table 2. Error bars represent mean ± s.e.m. (p < 0.01), (P < 0.0001).

Supplementary Figure 7 AKT2 is required for the activation of adipocyte precursors at the onset of obesity in ob/ob mice.

(A-B) Weekly intake of SD in raw kilocalories (kCal) (A) or kCal normalized to body weight (B) for the indicated groups of young mice after weaning at 3 weeks of age. Week “3” denotes food intake between the ages of 3 and 4 weeks. (n = 4 for wild-type, n = 3 for ob/ob, n = 7-8 for ob/ob; Akt2−/−) (C-D) H&E-stained sections of VWAT (C) and corresponding adipocyte size measurements (D) for the indicated groups. (n = 7 for ob/ob; Akt2−/−, n = 4 for ob/ob). Significance between indicated groups in (A-B) was calculated using a two-tailed Student’s t-test. Significance in (D) was calculated using two-way ANOVA with Bonferroni’s test for multiple comparisons. Scale bars in (C) are 50 μm. or a (P < 0.05), or b (p < 0.01), or c (P < 0.001), or d (P < 0.0001). For (A-B), indicates significance of ob/ob over wild-type, † indicates significance of ob/ob; Akt2−/− over wild-type, and ‡ indicates significance of ob/ob; Akt2−/− over ob/ob. Exact p-values are listed in Supplementary Table 1. Statistics source data for 5A-B can be found in Supplementary Table 2. Error bars represent mean ± s.e.m.

Supplementary Figure 8 AKT2 is not required for the normal development of white adipose tissue.

(A-B) Confocal images (A) of developing wild-type and Akt2−/− SWAT stained with LipidTOX (a lipid stain) and Isolectin IB4 (an endothelial cell stain) and (B) corresponding lipid droplet size quantification based on LipidTOX staining (n = 3 mice for all groups except n = 4 mice for Akt2−/−P5). Each group includes mice from 2-3 different litters. (C) Echo MRI quantification of fat mass from the indicated groups of mice at 6 weeks of age. (n = 13 mice for Akt2fl/fl and n = 14 mice for PdgfRα-cre; Akt2fl/fl). Scale bars in (A) are 50 μm. Error bars represent mean ± s.e.m. PX indicates postnatal day X.

Supplementary Table 1 Exact p-values.
Supplementary Table 2 Individual data points.

Supplementary information

Supplementary Information

Supplementary Information (PDF 592 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jeffery, E., Church, C., Holtrup, B. et al. Rapid depot-specific activation of adipocyte precursor cells at the onset of obesity. Nat Cell Biol 17, 376–385 (2015). https://doi.org/10.1038/ncb3122

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3122

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