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:

Preadipocytes from obese humans with type 2 diabetes are epigenetically reprogrammed at genes controlling adipose tissue function

Abstract

Background

Deterioration of the adipogenic potential of preadipocytes may contribute to adipose tissue dysfunction in obesity and type 2 diabetes (T2D). Here, we hypothesized that extracellular factors in obesity epigenetically reprogram adipogenesis potential and metabolic function of preadipocytes.

Methods

The transcriptomic profile of visceral adipose tissue preadipocytes collected from Lean, Obese and Obese with T2D was assessed throughout in vitro differentiation using RNA sequencing. Reduced Representation Bisulfite Sequencing was used to establish the genome-wide DNA methylation profile of human preadipocytes and 3T3-L1 preadipocytes treated by the inflammatory cytokine Tumour Necrosis Factor-α (TNF-α) or palmitate.

Results

While preadipocytes from all obese subjects (Obese+Obese T2D), compared to those of Lean, were transcriptionally different in response to differentiation in culture, preadipocytes from Obese T2D showed impaired insulin signalling and a further transcriptomic shift towards altered adipocyte function. Cultures with a lower expression magnitude of adipogenic genes throughout differentiation (PLIN1, CIDEC, FABP4, ADIPOQ, LPL, PDK4, APOE, LIPE, FABP3, LEP, RBP4 and CD36) were associated with DNA methylation remodelling at genes controlling insulin sensitivity and adipocytokine signalling pathways. Prior incubation of 3T3-L1 preadipocytes with TNF-α or palmitate markedly altered insulin responsiveness and metabolic function in the differentiated adipocytes, and remodelled DNA methylation and gene expression at specific genes, notably related to PPAR signalling.

Conclusions

Our findings that preadipocytes retain the memory of the donor in culture and can be reprogrammed by extracellular factors support a mechanism by which adipocyte precursors are epigenetically reprogrammed in vivo. Epigenetic reprogramming of preadipocytes represents a mechanism by which metabolic function of visceral adipose tissue may be affected in the long term by past exposure to obesity- or T2D-specific factors.

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
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM. Increased adipose tissue expression of tumor necrosis factor-a in human obesity and insulin resistance. J Clin Invest. 1995;95:2409–15.

    Article  CAS  Google Scholar 

  2. Hajer GR, Van Haeften TW, Visseren FLJ. Adipose tissue dysfunction in obesity, diabetes, and vascular diseases. Eur Hear J. 2008;29:2959–71.

    Article  CAS  Google Scholar 

  3. Rønningen T, Shah A, Reiner AH, Collas P, Moskaug JØ. Epigenetic priming of inflammatory response genes by high glucose in adipose progenitor cells. Biochem Biophys Res Commun. 2015;467:979–86.

    Article  Google Scholar 

  4. Isakson P, Hammarstedt A, Gustafson B, Smith U. Impaired preadipocyte differentiation in human abdominal obesity: role of Wnt, tumor necrosis factor-alpha, and inflammation. Diabetes. 2009;58:1550–7.

    Article  CAS  Google Scholar 

  5. Permana PA, Nair S, Lee Y, Luczy-bachman G, De Courten BV, Tataranni PA, et al. Subcutaneous abdominal preadipocyte differentiation in vitro inversely correlates with central obesity. Am J Physiol Endocrinol Metab. 2004;286:958–62.

    Article  Google Scholar 

  6. Lessard J, Laforest S, Pelletier M, Leboeuf M, Blackburn L, Tchernof A. Low abdominal subcutaneous preadipocyte adipogenesis is associated with visceral obesity, visceral adipocyte hypertrophy, and a dysmetabolic state. Adipocyte. 2014;3:197–205.

    Article  CAS  Google Scholar 

  7. Rossmeislová L, Mališová L, Kračmerová J, Tencerová M, Kováčová Z, Koc M, et al. Weight loss improves the adipogenic capacity of human preadipocytes and modulates their secretory profile. Diabetes. 2013;62:1990–5.

    Article  Google Scholar 

  8. Siersbæk MS, Loft A, Aagaard MM, Nielsen R, Schmidt SF, Petrovic N, et al. Genome-wide profiling of peroxisome proliferator-activated receptor γ in primary epididymal, inguinal, and brown adipocytes reveals depot-selective binding correlated with gene expression. Mol Cell Biol. 2012;32:3452–63.

    Article  Google Scholar 

  9. Mikkelsen TS, Xu Z, Zhang X, Wang L, Gimble JM, Lander ES, et al. Comparative epigenomic analysis of murine and human adipogenesis. Cell. 2010;143:156–69.

    Article  CAS  Google Scholar 

  10. Benton MC, Johnstone A, Eccles D, Harmon B, Hayes MT, Lea RA, et al. An analysis of DNA methylation in human adipose tissue reveals differential modification of obesity genes before and after gastric bypass and weight loss. Genome Biol. 2015;16:8:1–21.

    Google Scholar 

  11. Nylander V, Ingerslev LR, Andersen E, Fabre O, Garde C, Rasmussen M, et al. Ionizing radiation potentiates high-fat diet–induced insulin resistance and reprograms skeletal muscle and adipose progenitor cells. Diabetes. 2016;65:3573–84.

    Article  CAS  Google Scholar 

  12. Nilsson E, Jansson PA, Perfilyev A, Volkov P, Pedersen M, Svensson MK, et al. Altered DNA methylation and differential expression of genes influencing metabolism and inflammation in adipose tissue from subjects with type 2 diabetes. Diabetes. 2014;63:2962–276.

    Article  Google Scholar 

  13. Rönn T, Volkov P, Davegårdh C, Dayeh T, Hall E, Olsson AH, et al. A six months exercise intervention influences the genome-wide DNA methylation pattern in human adipose tissue. PLoS Genet. 2013;9:e1003572.

    Article  Google Scholar 

  14. Mikkelsen TS, Ku M, Jaffe DB, Issac B, Lieberman E, Giannoukos G, et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature. 2007;448:553–60.

    Article  CAS  Google Scholar 

  15. Reik W. Stability and flexibility of epigenetic gene regulation in mammalian development. Nature. 2007;447:425–32.

    Article  CAS  Google Scholar 

  16. Arner P, Arner E, Hammarstedt A, Smith U. Genetic predisposition for type 2 diabetes, but not for overweight/obesity, is associated with a restricted adipogenesis. PLoS ONE. 2011;6:e18284.

    Article  CAS  Google Scholar 

  17. Pattamaprapanont P, Garde C, Fabre O, Barrès R. Muscle contraction induces acute hydroxymethylation of the exercise-responsive gene Nr4a3. Front Endocrinol (Lausanne). 2016;7:1–9.

    Article  Google Scholar 

  18. Yates A, Akanni W, Amode MR, Barrell D, Billis K, Carvalho-Silva D, et al. Ensembl 2016. Nucleic Acids Res. 2016;44:D710–6.

    Article  CAS  Google Scholar 

  19. Bray NL, Pimentel H, Melsted P, Pachter L. Near-optimal probabilistic RNA-seq quantification. Nat Biotechnol. 2016;34:525–7.

    Article  CAS  Google Scholar 

  20. Pimentel H, Bray NL, Puente S, Melsted P, Pachter L. Differential analysis of RNA-seq incorporating quantification uncertainty. Nat Methods. 2017;14:687–90.

    Article  CAS  Google Scholar 

  21. Busser H, Najar M, Raicevic G, Pieters K, Velez Pombo R, Philippart P, et al. Isolation and characterization of human mesenchymal stromal cell subpopulations: comparison of bone marrow and adipose tissue. Stem Cells Dev. 2015;24:2142–57.

    Article  CAS  Google Scholar 

  22. Boyle P, Clement K, Gu H, Smith ZD, Ziller M, Fostel JL, et al. Gel-free multiplexed reduced representation bisulfite sequencing for large-scale DNA methylation profiling. Genome Biol. 2012;13:R92.

    Article  CAS  Google Scholar 

  23. Krueger F, Andrews SR. Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications. Bioinformatics. 2011;27:1571–2.

    Article  CAS  Google Scholar 

  24. Hebestreit K, Dugas M, Klein H-U. Detection of significantly differentially methylated regions in targeted bisulfite sequencing data. Bioinformatics. 2013;29:1647–53.

    Article  CAS  Google Scholar 

  25. Bjornsson HT, Sigurdsson MI, Fallin MD, Irizarry RA, Aspelund T, Cui H, et al. Intra-individual change over time in DNA methylation with familial clustering. JAMA. 2008;299:2877–83.

    Article  CAS  Google Scholar 

  26. Broholm C, Olsson AH, Perfilyev A, Gillberg L, Hansen NS, Ali A, et al. Human adipogenesis is associated with genome-wide DNA methylation and gene-expression changes. Epigenomics. 2016;8:1601–17.

    Article  CAS  Google Scholar 

  27. Van Harmelen V, Röhrig K, Hauner H. Comparison of proliferation and differentiation capacity of human adipocyte precursor cells from the omental and subcutaneous adipose tissue depot of obese subjects. Metabolism. 2004;53:632–7.

    Article  Google Scholar 

  28. Pinnick KE, Nicholson G, Manolopoulos KN, McQuaid SE, Valet P, Frayn KN, et al. Distinct developmental profile of lower-body adipose tissue defines resistance against obesity-associated metabolic complications. Diabetes. 2014;63:3785–97.

    Article  CAS  Google Scholar 

  29. Baglioni S, Cantini G, Poli G, Francalanci M, Squecco R, Franco A, et al. Functional differences in visceral and subcutaneous fat pads originate from differences in the adipose stem cell. PLoS ONE. 2012;7:e36569.

    Article  CAS  Google Scholar 

  30. Böhm A, Halama A, Meile T, Zdichavsky M, Lehmann R, Weigert C, et al. Metabolic signatures of cultured human adipocytes from metabolically healthy versus unhealthy obese individuals. PLoS ONE. 2014;9:e93148.

    Article  Google Scholar 

  31. Naukkarinen J, Heinonen S, Hakkarainen A, Lundbom J, Vuolteenaho K. Characterising metabolically healthy obesity in weight-discordant monozygotic twins. Diabetologia. 2014;57:167–76.

    Article  CAS  Google Scholar 

  32. van Tienen FHJ, van der Kallen CJH, Lindsey PJ, Wanders RJ, van Greevenbroek MM, Smeets HJM. Preadipocytes of type 2 diabetes subjects display an intrinsic gene expression profile of decreased differentiation capacity. Int J Obes. 2011;35:1154–64.

    Article  Google Scholar 

  33. Henninger AMJ, Eliasson B, Jenndahl LE, Hammarstedt A. Adipocyte hypertrophy, inflammation and fibrosis characterize subcutaneous adipose tissue of healthy, non-obese subjects predisposed to type 2 diabetes. PLoS ONE. 2014;9:1–8.

    Article  Google Scholar 

  34. Oliva-olivera W, Gea AL, Lhamyani S, Coín-aragüez L, Torres JA, Bernal-lópez MR, et al. Differences in the osteogenic differentiation capacity of omental adipose-derived stem cells in syndrome. Endocrinology. 2015;156:4492–501.

    Article  CAS  Google Scholar 

  35. Arner P, Sinha I, Thorell A, Rydén M, Dahlman-Wright K, Dahlman I. The epigenetic signature of subcutaneous fat cells is linked to altered expression of genes implicated in lipid metabolism in obese women. Clin Epigenet. 2015;7:1–13.

    Article  Google Scholar 

  36. Broholm C, Olsson AH, Perfilyev A, Hansen NS, Schrölkamp M, Strasko KS, et al. Epigenetic programming of adipose-derived stem cells in low birthweight individuals. Diabetologia. 2016;59:2664–73.

    Article  Google Scholar 

  37. Guénard F, Tchernof A, Deshaies Y, Pérusse L, Biron S, Lescelleur O, et al. Differential methylation in visceral adipose tissue of obese men discordant for metabolic disturbances. Physiol Genom. 2014;46:216–22.

    Article  Google Scholar 

  38. Collas P. Programming differentiation potential in mesenchymal stem cells. Epigenetics. 2010;5:476–82.

    Article  CAS  Google Scholar 

  39. Barrès R, Osler ME, Yan J, Rune A, Fritz T, Caidahl K, et al. Non-CpG Methylation of the PGC-1α promoter through DNMT3B controls mitochondrial density. Cell Metab. 2009;10:189–98.

    Article  Google Scholar 

  40. Sharples AP, Polydorou I, Hughes DC, Owens DJ, Hughes TM, Stewart CE. Skeletal muscle cells possess a ‘memory’ of acute early life TNF-a exposure: role of epigenetic adaptation. Biogerontology. 2016;17:603–17.

    Article  CAS  Google Scholar 

  41. Asterholm IW, Tao C, Morley TS, Wang QA, Delgado-Lopez F, Wang ZV, et al. Adipocyte inflammation is essential for healthy adipose tissue expansion and remodeling. Cell Metab. 2014;20:103–18.

    Article  Google Scholar 

Download references

Acknowledgements

OF was recipient of a research grant from the Danish Diabetes Academy supported by the Novo Nordisk Foundation. We would like to acknowledge The Danish National High-Throughput DNA Sequencing Centre, University of Copenhagen, for sequencing services. The Novo Nordisk Foundation Centre for Basic Metabolic Research is an independent research centre at the University of Copenhagen partially funded by an unrestricted donation from the Novo Nordisk Foundation.

Author contribution:

EA collected human samples, performed experiments, analysed the data and wrote the manuscript; LRI and AA performed bioinformatics analysis and generated figures; OF performed experiments, analysed the data and edited the manuscript; ID, SV, TB and VBK collected human samples, contributed to study design and edited the manuscript; DS provided expert advice and edited the manuscript; RB designed the study, analysed the data and wrote the manuscript. All authors approved the final version of the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Romain Barrès.

Ethics declarations

Conflict of interest:

The authors declare that they have no conflict of interest.

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Andersen, E., Ingerslev, L.R., Fabre, O. et al. Preadipocytes from obese humans with type 2 diabetes are epigenetically reprogrammed at genes controlling adipose tissue function. Int J Obes 43, 306–318 (2019). https://doi.org/10.1038/s41366-018-0031-3

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41366-018-0031-3

This article is cited by

Search

Quick links