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:

Obesity and hyperinsulinemia drive adipocytes to activate a cell cycle program and senesce

Abstract

Obesity is considered an important factor for many chronic diseases, including diabetes, cardiovascular disease and cancer. The expansion of adipose tissue in obesity is due to an increase in both adipocyte progenitor differentiation and mature adipocyte cell size. Adipocytes, however, are thought to be unable to divide or enter the cell cycle. We demonstrate that mature human adipocytes unexpectedly display a gene and protein signature indicative of an active cell cycle program. Adipocyte cell cycle progression associates with obesity and hyperinsulinemia, with a concomitant increase in cell size, nuclear size and nuclear DNA content. Chronic hyperinsulinemia in vitro or in humans, however, is associated with subsequent cell cycle exit, leading to a premature senescent transcriptomic and secretory profile in adipocytes. Premature senescence is rapidly becoming recognized as an important mediator of stress-induced tissue dysfunction. By demonstrating that adipocytes can activate a cell cycle program, we define a mechanism whereby mature human adipocytes senesce. We further show that by targeting the adipocyte cell cycle program using metformin, it is possible to influence adipocyte senescence and obesity-associated adipose tissue inflammation.

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: Mature human adipocytes express a cell cycle transcriptional profile.
Fig. 2: Mature human adipocytes are positive for cell cycle markers.
Fig. 3: Adipocyte cell cycle progression is associated with obesity and hyperinsulinemia.
Fig. 4: Insulin induces adipocyte cell cycle reentry and EdU incorporation.
Fig. 5: Adipocyte senescence is induced by obesity and insulin resistance.
Fig. 6: Cell cycle entry or arrest governs adipocyte senescence.

Similar content being viewed by others

Data availability

The datasets generated and analyzed during the current study can be accessed from https://gitlab.inria.fr/knibbe/adipocyte-senescence/ (cyclin A2 and SABG datasets) and https://github.com/PingChen-Angela/Adipocytes_cell_cycle/ (data repository for mRNA-seq gene expression matrix (RPKMs) and sample group annotations). Source data are provided with this paper.

Code availability

Regression analysis scripts (for cyclin A2 and SABG analyses) are available at https://gitlab.inria.fr/knibbe/adipocyte-senescence/ and mRNA-seq codes are available at https://github.com/PingChen-Angela/Adipocytes_cell_cycle/.

References

  1. Arner, P. et al. Adipose lipid turnover and long-term changes in body weight. Nat. Med. 25, 1385–1389 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Ryden, M. & Arner, P. Cardiovascular risk score is linked to subcutaneous adipocyte size and lipid metabolism. J. Intern. Med. 282, 220–228 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  4. van Harmelen, V. et al. Effect of BMI and age on adipose tissue cellularity and differentiation capacity in women. Int. J. Obes. Relat. Metab. Disord. 27, 889–895 (2003).

    Article  PubMed  Google Scholar 

  5. Hoffstedt, J. et al. Regional impact of adipose tissue morphology on the metabolic profile in morbid obesity. Diabetologia 53, 2496–2503 (2010).

    Article  CAS  PubMed  Google Scholar 

  6. Edgar, B. A., Zielke, N. & Gutierrez, C. Endocycles: a recurrent evolutionary innovation for postmitotic cell growth. Nat. Rev. Mol. Cell Biol. 15, 197–210 (2014).

    Article  PubMed  CAS  Google Scholar 

  7. Gandarillas, A., Molinuevo, R. & Sanz-Gomez, N. Mammalian endoreplication emerges to reveal a potential developmental timer. Cell Death Differ. 25, 471–476 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Gentric, G. & Desdouets, C. Polyploidization in liver tissue. Am. J. Pathol. 184, 322–331 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. Skurk, T., Alberti-Huber, C., Herder, C. & Hauner, H. Relationship between adipocyte size and adipokine expression and secretion. J. Clin. Endocrinol. Metab. 92, 1023–1033 (2007).

    Article  CAS  PubMed  Google Scholar 

  10. Deng, Y. & Scherer, P. E. Adipokines as novel biomarkers and regulators of the metabolic syndrome. Ann. N. Y. Acad. Sci. 1212, E1–E19 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Kuilman, T., Michaloglou, C., Mooi, W. J. & Peeper, D. S. The essence of senescence. Genes Dev. 24, 2463–2479 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lee, S. & Schmitt, C. A. The dynamic nature of senescence in cancer. Nat. Cell Biol. 21, 94–101 (2019).

    Article  CAS  PubMed  Google Scholar 

  13. Burton, D. G. A. & Faragher, R. G. A. Obesity and type-2 diabetes as inducers of premature cellular senescence and ageing. Biogerontology 19, 447–459 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Minamino, T. et al. A crucial role for adipose tissue p53 in the regulation of insulin resistance. Nat. Med. 15, 1082–1087 (2009).

    Article  CAS  PubMed  Google Scholar 

  15. Palmer, A. K. et al. Targeting senescent cells alleviates obesity-induced metabolic dysfunction. Aging Cell 18, e12950 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Xu, M. et al. Targeting senescent cells enhances adipogenesis and metabolic function in old age. Elife 4, e12997 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Baker, D. J. et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 479, 232–236 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Bian, X. et al. Senescence marker activin A is increased in human diabetic kidney disease: association with kidney function and potential implications for therapy. BMJ Open Diabetes Res. Care 7, e000720 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Gustafson, B., Nerstedt, A. & Smith, U. Reduced subcutaneous adipogenesis in human hypertrophic obesity is linked to senescent precursor cells. Nat. Commun. 10, 2757 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Xu, M. et al. JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Proc. Natl Acad. Sci. USA 112, E6301–E6310 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Matsui-Hirai, H. et al. Dose-dependent modulatory effects of insulin on glucose-induced endothelial senescence in vitro and in vivo: a relationship between telomeres and nitric oxide. J. Pharmacol. Exp. Ther. 337, 591–599 (2011).

    Article  CAS  PubMed  Google Scholar 

  22. Grosse, L. et al. Defined p16high senescent cell types are indispensable for mouse healthspan. Cell Metab. 32, 87–99 (2020).

    Article  CAS  PubMed  Google Scholar 

  23. Chen, Y. W., Harris, R. A., Hatahet, Z. & Chou, K. M. Ablation of XP-V gene causes adipose tissue senescence and metabolic abnormalities. Proc. Natl Acad. Sci. USA 112, E4556–E4564 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Vergoni, B. et al. DNA damage and the activation of the p53 pathway mediate alterations in metabolic and secretory functions of Adipocytes. Diabetes 65, 3062–3074 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Tchkonia, T. et al. Fat tissue, aging and cellular senescence. Aging Cell 9, 667–684 (2010).

    Article  CAS  PubMed  Google Scholar 

  26. Ryden, M. et al. Transplanted bone marrow-derived cells contribute to human adipogenesis. Cell Metab. 22, 408–417 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Lagarrigue, S. et al. CDK4 is an essential insulin effector in adipocytes. J. Clin. Invest. 126, 335–348 (2016).

    Article  PubMed  Google Scholar 

  28. van der Meijden, C. M. et al. Gene profiling of cell cycle progression through S phase reveals sequential expression of genes required for DNA replication and nucleosome assembly. Cancer Res. 62, 3233–3243 (2002).

    PubMed  Google Scholar 

  29. Morgan, D. O. Cell Cycle: Principles of Control (New Science Press, 2007).

  30. Hendzel, M. J. et al. Mitosis-specific phosphorylation of histone H3 initiates primarily within pericentromeric heterochromatin during G2 and spreads in an ordered fashion coincident with mitotic chromosome condensation. Chromosoma 106, 348–360 (1997).

    Article  CAS  PubMed  Google Scholar 

  31. Li, D. W. et al. Dynamic distribution of Ser-10 phosphorylated histone H3 in cytoplasm of MCF-7 and CHO cells during mitosis. Cell Res. 15, 120–126 (2005).

    Article  PubMed  Google Scholar 

  32. Hesse, M. et al. Direct visualization of cell division using high-resolution imaging of M phase of the cell cycle. Nat. Commun. 3, 1076 (2012).

    Article  PubMed  CAS  Google Scholar 

  33. Gillooly, J. F., Hein, A. & Damiani, R. Nuclear DNA content varies with cell size across human cell types. Cold Spring Harb. Perspect. Biol. 7, a019091 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Orr-Weaver, T. L. When bigger is better: the role of polyploidy in organogenesis. Trends Genet. 31, 307–315 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Biran, A. et al. Quantitative identification of senescent cells in aging and disease. Aging Cell 16, 661–671 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Hydbring, P., Malumbres, M. & Sicinski, P. Noncanonical functions of cell cycle cyclins and cyclin-dependent kinases. Nat. Rev. Mol. Cell Biol. 17, 280–292 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Hernandez-Segura, A. et al. Unmasking transcriptional heterogeneity in senescent cells. Curr. Biol. 27, 2652–2660 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Liu, Z. et al. The dysfunctional MDM2–p53 axis in adipocytes contributes to aging-related metabolic complications by induction of lipodystrophy. Diabetes 67, 2397–2409 (2018).

    Article  CAS  PubMed  Google Scholar 

  39. Hickson, L. J. et al. Senolytics decrease senescent cells in humans: preliminary report from a clinical trial of dasatinib plus quercetin in individuals with diabetic kidney disease. EBioMedicine 47, 446–456 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Hernandez-Segura, A., Nehme, J. & Demaria, M. Hallmarks of cellular senescence. Trends Cell Biol. 28, 436–453 (2018).

    Article  CAS  PubMed  Google Scholar 

  41. Kim, D. E. & Davalos, A. R. Alarmin detection in senescent cells. Methods Mol. Biol. 1896, 71–81 (2019).

    Article  CAS  PubMed  Google Scholar 

  42. Neurohr, G. E. et al. Excessive cell growth causes cytoplasm dilution and contributes to senescence. Cell 176, 1083–1097 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Sharpless, N. E. & Sherr, C. J. Forging a signature of in vivo senescence. Nat. Rev. Cancer 15, 397–408 (2015).

    Article  CAS  PubMed  Google Scholar 

  44. Feringa, F. M. et al. Persistent repair intermediates induce senescence. Nat. Commun. 9, 3923 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. te Poele, R. H., Okorokov, A. L., Jardine, L., Cummings, J. & Joel, S. P. DNA damage is able to induce senescence in tumor cells in vitro and in vivo. Cancer Res. 62, 1876–1883 (2002).

    Google Scholar 

  46. Rodier, F. et al. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat. Cell Biol. 11, 973–979 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Tu, W. Z. et al. γH2AX foci formation in the absence of DNA damage: mitotic H2AX phosphorylation is mediated by the DNA–PKcs/CHK2 pathway. FEBS Lett. 587, 3437–3443 (2013).

    Article  CAS  PubMed  Google Scholar 

  48. Shimizu, I., Yoshida, Y., Suda, M. & Minamino, T. DNA damage response and metabolic disease. Cell Metab. 20, 967–977 (2014).

    Article  CAS  PubMed  Google Scholar 

  49. Furukawa, S. et al. Increased oxidative stress in obesity and its impact on metabolic syndrome. J. Clin. Invest. 114, 1752–1761 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Murdolo, G. et al. Oxidative stress and lipid peroxidation by-products at the crossroad between adipose organ dysregulation and obesity-linked insulin resistance. Biochimie 95, 585–594 (2013).

    Article  CAS  PubMed  Google Scholar 

  51. Monickaraj, F. et al. Accelerated fat cell aging links oxidative stress and insulin resistance in adipocytes. J. Biosci. 38, 113–122 (2013).

    Article  CAS  PubMed  Google Scholar 

  52. Terzi, M. Y., Izmirli, M. & Gogebakan, B. The cell fate: senescence or quiescence. Mol. Biol. Rep. 43, 1213–1220 (2016).

    Article  CAS  PubMed  Google Scholar 

  53. Coppe, J. P., Desprez, P. Y., Krtolica, A. & Campisi, J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev. Pathol. 5, 99–118 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. McHugh, D. & Gil, J. Senescence and aging: causes, consequences, and therapeutic avenues. J. Cell Biol. 217, 65–77 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Greten, T. F. & Eggert, T. Cellular senescence associated immune responses in liver cancer. Hepat. Oncol. 4, 123–127 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Aguayo-Mazzucato, C. et al. Acceleration of beta cell aging determines diabetes and senolysis improves disease outcomes. Cell Metab. 30, 129–142 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Gorgoulis, V. et al. Cellular senescence: defining a path forward. Cell 179, 813–827 (2019).

    Article  CAS  PubMed  Google Scholar 

  58. Blagosklonny, M. V. Rapamycin, proliferation and geroconversion to senescence. Cell Cycle 17, 2655–2665 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Ben Sahra, I. et al. Metformin, independent of AMPK, induces mTOR inhibition and cell cycle arrest through REDD1. Cancer Res. 71, 4366–4372 (2011).

    Article  PubMed  CAS  Google Scholar 

  60. Andrzejewski, S., Gravel, S. P., Pollak, M. & St-Pierre, J. Metformin directly acts on mitochondria to alter cellular bioenergetics. Cancer Metab. 2, 12 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Goel, S. et al. CDK4/6 inhibition triggers anti-tumour immunity. Nature 548, 471–475 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Ruscetti, M. et al. NK cell-mediated cytotoxicity contributes to tumor control by a cytostatic drug combination. Science 362, 1416–1422 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Ruscetti, M. et al. Senescence-induced vascular remodeling creates therapeutic vulnerabilities in pancreas cancer. Cell 181, 424–441 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Hammarstedt, A., Gogg, S., Hedjazifar, S., Nerstedt, A. & Smith, U. Impaired adipogenesis and dysfunctional adipose tissue in human hypertrophic obesity. Physiol. Rev. 98, 1911–1941 (2018).

    Article  CAS  PubMed  Google Scholar 

  65. Nevzorova, Y. A. et al. Aberrant cell cycle progression and endoreplication in regenerating livers of mice that lack a single E-type cyclin. Gastroenterology 137, 691–703 (2009).

    Article  CAS  PubMed  Google Scholar 

  66. Levine, E. M. Cell cycling through development. Development 131, 2241–2246 (2004).

    Article  CAS  PubMed  Google Scholar 

  67. Sher, N. et al. Fundamental differences in endoreplication in mammals and Drosophila revealed by analysis of endocycling and endomitotic cells. Proc. Natl Acad. Sci. USA 110, 9368–9373 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Fox, D. T. & Duronio, R. J. Endoreplication and polyploidy: insights into development and disease. Development 140, 3–12 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Campisi, J. & d’Adda di Fagagna, F. Cellular senescence: when bad things happen to good cells. Nat. Rev. Mol. Cell Biol. 8, 729–740 (2007).

    Article  CAS  PubMed  Google Scholar 

  70. Wang, M. J., Chen, F., Lau, J. T. Y. & Hu, Y. P. Hepatocyte polyploidization and its association with pathophysiological processes. Cell Death Dis. 8, e2805 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Celton-Morizur, S., Merlen, G., Couton, D., Margall-Ducos, G. & Desdouets, C. The insulin/Akt pathway controls a specific cell division program that leads to generation of binucleated tetraploid liver cells in rodents. J. Clin. Invest. 119, 1880–1887 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Chow, H. M. et al. Age-related hyperinsulinemia leads to insulin resistance in neurons and cell cycle-induced senescence. Nat. Neurosci. 22, 1806–1819 (2019).

    Article  CAS  PubMed  Google Scholar 

  73. Fung, J. & Berger, A. Hyperinsulinemia and insulin resistance: scope of the problem. J. Insul. Resist. 1, a18 (2016).

    Article  Google Scholar 

  74. Xu, M. et al. Senolytics improve physical function and increase lifespan in old age. Nat. Med. 24, 1246–1256 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Hagberg, C. E. et al. Flow cytometry of mouse and human adipocytes for the analysis of browning and cellular heterogeneity. Cell Rep. 24, 2746–2756 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  77. Ramskold, D., Wang, E. T., Burge, C. B. & Sandberg, R. An abundance of ubiquitously expressed genes revealed by tissue transcriptome sequence data. PLoS Comput. Biol. 5, e1000598 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Kamburov, A. et al. ConsensusPathDB: toward a more complete picture of cell biology. Nucleic Acids Res. 39, D712–D717 (2011).

    Article  CAS  PubMed  Google Scholar 

  79. Harms, M. J. et al. Mature human white adipocytes cultured under membranes maintain identity, function and can transdifferentiate into brown-like adipocytes. Cell Rep. 27, 213–225 (2019).

    Article  CAS  PubMed  Google Scholar 

  80. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  81. Ferrari, S. L. P. & Cribari-Neto, F. Beta regression for modelling rates and proportions. J. Appl. Stat. 31, 799–815 (2004).

    Article  Google Scholar 

  82. Ospina, R. & Ferrari, S. L. P. Inflated beta distributions. Stat. Pap. 51, 111–126 (2010).

    Article  Google Scholar 

Download references

Acknowledgements

We acknowledge the assistance of research nurses A. Bratt and A. -S. Andersson (Ersta Hospital) and laboratory technicians L. Appelsved and A. Olsson. This study was supported by grants from the Swedish Research Council (no. 542-2013-8358 to K.S., no. 350-2012-6538 to C.E.H.), the Strategic Research Program for Diabetes at Karolinska Institutet (no. C5471152 to K.S.), the Novo Nordisk Foundation (Excellence Project Award no. NNF12OC1016064 to K.S.), the Karolinska Institutet/AstraZeneca Integrated Cardiometabolic Centre (no. H725701603 to K.S.), the Erling-Persson Family Foundation (no. 140604 to A.T.), the Swedish Foundation for Strategic Research (I.A.) and the Vallee Foundation Vallee Scholar Award (no. C5471234 to K.S.). C.E.H. was supported by the Swedish Society for Medical Research (no. P13-0057) and Wilhelm och Else Stockmanns Stiftelse.

Author information

Authors and Affiliations

Authors

Contributions

K.L.S., C.E.H. and Q.L. designed the study. A.T. recruited the participants. F.S., C.E.H., P.C. and J.W. prepared and analyzed sequencing samples. Q.L., C.E.H., S.L., H.S.C., E.T. and M.K. developed and performed adipocyte ICC, IHC and senescence stainings. Q.L., S.L., I.A., N.K., E.T. and M.H. designed and performed in vitro experiments and molecular analyses. M.H., I.A., M.A. and J.B. did western blotting. C.H., Q.L., H.S.C., N.K. and C.K. performed statistical analyses and C.K. performed the multiple-regression analyses. C.E.H., Q.L. and K.L.S. wrote the first version of the paper. Q.L. prepared the figures. All authors contributed to and approved the final version of the paper.

Corresponding author

Correspondence to Kirsty L. Spalding.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information: Nature Medicine thanks Suneil Koliwad, Lluis Fajas, Valery Krizhanovsky and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Jennifer Sargent was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Purity of isolated human adipocytes and the expression of a cell cycle transcriptional profile.

a. Representative transmitted light image showing pure fractions of isolated adipocytes without the presence of contaminating stromal vascular cells. Scale bar, 100 µm. The experiment was repeated for each isolation with similar results. b. QPCR analysis of CD45 mRNA expression in isolated mature adipocytes compared to CD45+/CD14+ macrophages sorted by flow cytometry, showing both raw CT values (left) and relative abundance (right, normalized to 18S expression, mean ± SEM). Each dot represents one individual, with the n indicated in the figure. c. RNA-Seq data (mean ± SD) showing the expression of adipocyte-specific (LEP, ADIPOQ, CIDEC, PLIN1, TUSC5) and pre-adipocyte specific (WNT10B, PDGFRA) genes in isolated human adipocytes (n = 13, blue bars) and in FACS sorted and sequenced preadipocyte samples (n = 3, red bars). Note the logarithmic scale. d. Table showing the absence of immune cell markers in the adipocyte mRNA sequencing dataset. e. RNA-Seq data (mean ± SD) showing expression of cell death related transcripts in isolated human adipocytes. N= 3 lean, 6 OB/NI, and 4 OB/HI individuals. f. Correlation between C-peptide and HOMA-IR levels for each individual using two-sided Spearman correlation. g. Enriched pathways for lean individuals based on the differentially expressed genes shown in Extended Data Fig. 2 based on Kruskal-Wallis tests. h. Percentage of S-phase associated transcripts (as listed by Wikipathways and Pathcards, n=123 genes) detected in four or more individuals with obesity out of the 13 analysed adipocyte mRNA sequencing samples. i. Relative mRNA expression of CCNA2 (cyclin A2) in mature adipocytes from WOB, OB/NI and OB/HI individuals, with the n indicated in the plot (mean ± SD). Data are normalized to TATA-box binding protein (TBP) expression and analyzed using Welch ANOVA test, followed by a Dunnett’s T3 multiple comparisons test.

Extended Data Fig. 2 Differentially expressed genes between patient groups.

Heatmap of RNA-seq data showing the differentially expressed genes in Lean, OB/NI, and OB/HI individuals (n = 13 individuals). Each column represents data from one individual. Colour intensity represents row Z-score.

Extended Data Fig. 3 Adipocyte cell cycle re-entry markers correlate with metabolic parameters.

a. Correlation between the percentage of adipocytes positive for cell cycle markers (cyclin D1, cyclin A2, pHH3 and cyclin E1) as determined by ICC, and serum insulin, HbA1c (glycosylated hemoglobin A1c), insulin-like growth factor 1 (IGF-1), triglyceride (TG) and low-denisty liporpotein (LDL) levels. Each dot represents the percentage of positive adipocytes from one individual,with individual number (n) indicated in each plot. Plots highlighted in grey indicate a significant correlation (p < 0.05) using two-sided Spearman correlation analysis. b. QPCR quantification of relative CCND1 (cyclin D1) and CCNA2 (cyclin A2) mRNA expression correlated with C-peptide levels and patient BMI (n = 16 individuals) using two-sided Spearman correlation. Data are normalized to TBP expression.

Extended Data Fig. 4 Statistical analysis of correlations between cell cycle–related markers and patient clinical parameters.

Correlation between the percentage of adipocytes positive for cell cycle markers (cyclin E1, cyclin D1, cyclin A2, pHH3, as determined by ICC), adipocyte cell or nuclear size and clinical parameters. Data was analysed using two-sided Spearman correlation analysis, with individual number (n) indicated in each row. Blocks highlighted in light red indicate a significant correlation (p < 0.05), blocks highlighted in dark red indicate a highly significant correlation (p < 0.0001).

Extended Data Fig. 5 Insulin induces adipocyte cell cycle re-entry and EdU incorporation.

a. Representative flow cytometric analysis of EdU staining in nuclei isolated from adipocytes unstained (left) or stained for EdU (right), demonstrating how the gates were set based on unstained samples. b. Image cytometry-based quantification of nuclear DNA, showing the integrated density (area x median staining intensity) of immobilized Dapi-stained nuclei for two lean individuals (grey, n = 52 nuclei in individual 1, n = 109 nuclei in individual 2) and four individuals with obesity (blue, n = 89 nuclei in individual 1, n = 96 nuclei in individual 2, n = 113 nuclei in individual 3, n = 133 nuclei in individual 4), with the corresponding histograms for each group to the right. In the left graph, each dot represents one nucleus with mean ± SD. c. Nuclear size quantification from adipocyte ICC images showing the fold change in mean nuclear size of cyclin A2 positive adipocytes, compared to cyclin A2 negative adipocytes (set to 1.0) from the same individual, analyzed by two-sided Wilcoxon paired test. Each dot represents the fold change from one individual, with >70 nuclei measured per individual by imaging quantification (n = 21 individuals). Mean ± SEM. d. Correlation between the rate of adipocyte EdU incorporation when cultured in basal medium for 4 days, and clinical parameters. For d-e, each dot represents one individual. Grey plot highlighting indicates a significant correlation (p < 0.05) using two-sided Spearman correlation analysis. e. Correlation between the insulin-mediated response in adipocyte EdU incorporation (by comparing the ratio of EdU positive adipocytes when cultured with exogenous insulin to cultures without) and clinical parameters using two-sided Spearman correlation. f. Quantification of western blot shown in Fig. 4i. g. Quantification of western blots shown in Fig. 4j.

Extended Data Fig. 6 Adipocyte senescence is induced by obesity and insulin resistance.

a. Correlation bewteen adipocyte Ki-67 (left) and PCNA (right) positivity assessed by IHC, and individual C-peptide using two-sided Spearman correlation. For panels a, d, e, gi, each dot represents one individual. b. Top 10 enriched pathways of genes significantly correlated with CCND1 (Cyclin D1) expression using two-sided pairwise Spearman correlation, sorted by descending significance. Senescence-associated pathways are highlighted in bold. c. Representative IHC images of human adipose tissue stained for galactosidase beta 1 (GLB1) (upper panel). Scale bar, 50 µm. Detail of an adipocyte nucleus enlarged for clarity (below, scale bar 10 µm). Experiment was repeated twice independently for 13 individuals with similar results. d. Correlation between the percentage of SABG positive adipocytes and clinical parameters using two-sided Spearman correlation. e. BMI distribution for the three different patient groups analysed in Fig. 5e. Individual number (n), is indicated for each plot. Mean ± SEM analyzed by one-way ANOVA, followed with Holm-Sidak’s multiple comparison test. f. Contribution of each predictor (clinical parameter) to the SABG regression model shown in Fig. 5f. A positive value means that the predictor improves the goodness-of-fit when it is added last to a model containing all the other predictors. g. Age distribution for the three different patient groups analysed in Fig. 5e. Individual number (n) is indicated for each plot. Mean ± SEM analyzed by Kruskai-Wallis, followed with Dunn’s multiple comparison test. h. Representative images of adipocytes labelled by ICC with senescence marker HMGB1. Individual positive and negative adipocytes are indicated by white circles. Scale bar, 50 µm. Correlation between the percentage of HMGB1 negative adipocytes and SABG positivity analyzed by two-sided Spearman is shown to the right. i. Relative mRNA expression of SASP-associated transcripts in mature adipocytes from WOB, OB/NI and OB/HI individuals (n = 4 per group). Mean ± SD, analyzed by one-way ANOVA, followed with Holm-Sidak’s multiple comparison test.

Extended Data Fig. 7 Statistical analysis of the correlations between senescence-related markers and individual clinical parameters.

Correlation between the percentage of adipocytes positive for senescence-related markers (SABG, p16, γΗ2ΑΧ) as determined by ICC and clinical parameters. Data was analysed with two-sided Spearman correlation analysis, with individual number (n) indicated for each block. Blocks highlighted in light red indicate a significant correlation (p < 0.05); blocks highlighted in dark red indicate a highly significant correlation (p < 0.0001).

Extended Data Fig. 8 Cell cycle entry or arrest governs adipocyte senescence.

a. Percentage of EdU positive adipocytes cultured with exogenous insulin for 4 or 7 days. Individual number (n), is indicated for each plot. Mean ± SEM, analyzed using two-sided Mann Whitney test. For panels ac, each dot represents one individual. b. RNA-Seq data showing expression of cell death related transcripts in adipocytes cultured with or without insulin for 7 days. N = 3, mean ± SD analyzed with two-way ANOVA with Dunnett’s multiple comparison test. c. Correlation between individual serum insulin levels and the percentage of SABG positive adipocytes after treatment with insulin and palbociclib for 7 days, analyzed by two-sided Spearman correlation. d. Quantification of secreted SASP-proteins from adipocytes cultured with insulin and palbociclib, normalized to cultures with insulin only. Statistics were performed using paired t-tests with a FDR multiple testing correction set to 10% (N = 8 except for TIMP1 (n=10), MMP19, ADAMTS4, IP10 (n= 7), CTGF and IL-6 (n= 6) due to missing samples, mean ± SD). e, f. Western blots (e) and quantifications (f) showing the effect of palbociclib and metformin on the phosphorylation status of p70S6K and AKT in two different individuals. Actin was used as a loading control. g. Correlation between individual serum insulin levels and the percentage of SABG positive adipocytes after treatment with insulin and metformin using two-sided Spearman correlation. h. Quantification of secreted adiponectin from adipocytes cultured for 7 days as in Fig. 6. Data is shown normalized to INS cultures. N = 10 except for INS + Met and INS + Palbo (n= 9) due to missing samples. Mean ± SD analyzed by ANOVA followed with Holm-Sidak’s multiple comparison test. i. LDH-cytotoxicity test showing no detection of released cytoplasmic content to the culture media after any of the indicated treatments, with the lower limit for cell death detection noted by the red line (no statistical test used). As a positive control, adipocytes were lysed on day 1 (d1) or d3 of culture, and media harvested at d7 along with the other samples (n = 3 for each treatment, mean ± SD).

Source data

Extended Data Fig. 9 Schematic and omental adipocytes display a lower incidence of cell cycle and senescence markers.

a. Schematic representation of the effects of modulating adipocyte cell cycle entry and block on senescence. (1) During short term culture (4 days) insulin acts as a mitogen promoting cell cycle activity. Endogenous cellular stressors, such as lipotoxicity or oxidative stress, activate a DDR, inducing (amongst others) p16/p21 and blocking cell cycle progression. Continued mitogenic stimulation (insulin, ‘accelerator’), countered by continued cell cycle block (‘brake’) can result in adipocytes becoming senescent (blue adipocytes) and exiting cell cycle. (2) In response to prolonged hyperinsulinemia (adipocytes cultured for 7 days with insulin), an increased number of adipocytes progress through cell cycle, resulting in an increase in the number of internal cell cycle:cell block conflicts, which in turn increases adipocyte senescence. (3) Mimicking endogenous p16/p21 activity by culturing with the CDK4/6 inhibitor, palbociclib, in combination with mitogenic stimulation (insulin) accentuates the internal cell cycle:cell block conflict, resulting in increased adipocyte senescence. (4) Blocking cell cycle entry with metformin significantly reduces adipocyte cell cycle progression and adipocyte senescence. b. Relative mRNA expression of PCNA, CCNA2 (cyclin A2) and CDC20 in paired samples of mature adipocytes isolated from subcutaneous (SC) and omental (OM) fat from WOB (n = 4), OB/NI (n = 4) and OB/HI (n = 4) individuals. Data are normalized to TBP expression. For panels b-h, each dot represents one individual, paired samples are indicated with a line and statistics analyzed using two-sided paired Student’s t tests, except in panel f where a two-sided Wilcoxon paired test was used. c. Percentage of cyclin A2 positive adipocytes in paired SC and OM fat samples from 1 lean individual and 5 individuals with obesity. d. Percentage of EdU positive omental adipocytes following 4 days of floating culture with or without exogenous insulin. e. Representative images of paired SC and OM samples of packed isolated mature human adipocytes stained using the SABG assay. Each tube contains isolated adipocytes from the respective depots of one individual. Green/blue staining indicates increased β-galactosidase activity. f. Percentage of SABG positive adipocytes in paired SC and OM samples. g. Relative mRNA expression of CCND1 (cyclin D1), CDKNA1 (p21) and CCL2 (MCP-1) in the same paired SC and OM samples as in a. Data are normalized to TBP expression. h. Percentage of p16 positive adipocytes in paired SC and OM fat samples from 5 individuals with obesity.

Extended Data Fig. 10 Inguinal hernia repair and gall bladder surgical biopsies display no significant differences in all studied cellular parameters.

Subgroup analysis of the cellular parameters measured for individuals using biopsies obtained from two types of surgeries: inguinal hernia repair (Hernia) and gallstone operations (Gallstone), show no significant difference in cell and nuclear size, the expression of cyclins E1, D1, A2, pHH3 (determined by ICC), the percentage of SABG positive adipocytes, or the level of EdU incorpation after suspension culture (cultured samples). Individual number (n), is indicated for each plot. Data is shown as mean ± SD. Statistics were performed using a two-sided Student’s t test, ns stands for non-significant difference.

Supplementary information

Supplementary Information

Supplementary Tables 1–13. A description of patient cohorts and reagents used in the study.

Reporting Summary

Source data

Source Data Fig. 4

Images from unprocessed blots shown in Fig. 4i,j.

Source Data Extended Data Fig. 8

Images from unprocessed blots shown in Extended Data Fig. 8e.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, Q., Hagberg, C.E., Silva Cascales, H. et al. Obesity and hyperinsulinemia drive adipocytes to activate a cell cycle program and senesce. Nat Med 27, 1941–1953 (2021). https://doi.org/10.1038/s41591-021-01501-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-021-01501-8

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