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:

Knockdown of ANT2 reduces adipocyte hypoxia and improves insulin resistance in obesity

Abstract

Decreased adipose tissue oxygen tension and increased expression of the transcription factor hypoxia-inducible factor–1α (HIF-1α) can trigger adipose tissue inflammation and dysfunction in obesity. Our current understanding of obesity-associated decreased adipose tissue oxygen tension is mainly focused on changes in oxygen supply and angiogenesis. Here, we demonstrate that increased adipocyte oxygen demand, mediated by activity of the mitochondrial protein adenine nucleotide translocase 2 (ANT2), is the dominant cause of adipocyte hypoxia. Deletion of adipocyte Ant2 (also known as Scl25a5) improves obesity-induced intracellular adipocyte hypoxia by decreasing obesity-induced adipocyte oxygen demand, without effects on mitochondrial number or mass, or oligomycin-sensitive respiration. This effect of adipocyte ANT2 knockout led to decreased adipose tissue HIF-1α expression and inflammation with improved glucose tolerance and insulin resistance in both preventative and therapeutic settings. Our results suggest that ANT2 may be a target for the development of insulin-sensitizing drugs and that ANT2 inhibition might have clinical utility.

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: Adipocyte-specific ANT2 knockout mice exhibit normal body weight with increased adipose tissue mass.
Fig. 2: Intracellular oxygen tension and HIF-1α expression are decreased in eWAT of ANT2 AKO mice.
Fig. 3: ANT2 AKO mice are protected from HFD-induced adipose tissue inflammation, glucose intolerance and insulin resistance.
Fig. 4: HFD-induced ATM accumulation and M1-like polarization is reduced in ANT2 AKO mice with improved adipose tissue fibrosis.
Fig. 5: Inducible ANT2 AKO reverses established glucose and insulin intolerance.
Fig. 6: HFD-induced adipocyte apoptosis is decreased in ANT2 AKO mice.

Similar content being viewed by others

Data availability

All data that support the findings of this study are included in the paper or its supplementary information.

References

  1. Olefsky, J. M. & Glass, C. K. Macrophages, inflammation, and insulin resistance. Annu. Rev. Physiol. 72, 219–246 (2010).

    Article  CAS  Google Scholar 

  2. Kusminski, C. M., Bickel, P. E. & Scherer, P. E. Targeting adipose tissue in the treatment of obesity-associated diabetes. Nat. Rev. Drug. Discov. 15, 639–660 (2016).

    Article  CAS  Google Scholar 

  3. Lee, Y. S., Wollam, J. & Olefsky, J. M. An integrated view of immunometabolism. Cell 172, 22–40 (2018).

    Article  CAS  Google Scholar 

  4. Friedman, J. The long road to leptin. J. Clin. Invest. 126, 4727–4734 (2016).

    Article  Google Scholar 

  5. Yore, M. M. et al. Discovery of a class of endogenous mammalian lipids with anti-diabetic and anti-inflammatory effects. Cell 159, 318–332 (2014).

    Article  CAS  Google Scholar 

  6. Hotamisligil, G. S. Inflammation, metaflammation and immunometabolic disorders. Nature 542, 177–185 (2017).

    Article  CAS  Google Scholar 

  7. Cao, H. et al. Identification of a lipokine, a lipid hormone linking adipose tissue to systemic metabolism. Cell 134, 933–944 (2008).

    Article  CAS  Google Scholar 

  8. Ferrante, A. W. Jr. The immune cells in adipose tissue. Diabetes Obes. Metab. 15 Suppl. 3, 34–38 (2013).

    Article  CAS  Google Scholar 

  9. Ying, W. et al. Adipose tissue macrophage-derived exosomal miRNAs can modulate in vivo and in vitro insulin sensitivity. Cell 171, 372–384 e312 (2017).

    Article  CAS  Google Scholar 

  10. Li, P. et al. Hematopoietic-derived galectin-3 causes cellular and systemic insulin resistance. Cell 167, 973–984 e912 (2016).

    Article  CAS  Google Scholar 

  11. Schwartz, D. R. & Lazar, M. A. Human resistin: found in translation from mouse to man. Trends Endocrinol. Metab. 22, 259–265 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Kanda, H. et al. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J. Clin. Invest. 116, 1494–1505 (2006).

    Article  CAS  Google Scholar 

  13. Solinas, G. et al. JNK1 in hematopoietically derived cells contributes to diet-induced inflammation and insulin resistance without affecting obesity. Cell Metab. 6, 386–397 (2007).

    Article  CAS  Google Scholar 

  14. Saberi, M. et al. Hematopoietic cell-specific deletion of toll-like receptor 4 ameliorates hepatic and adipose tissue insulin resistance in high-fat-fed mice. Cell Metab. 10, 419–429 (2009).

    Article  CAS  Google Scholar 

  15. Patsouris, D. et al. Ablation of CD11c-positive cells normalizes insulin sensitivity in obese insulin resistant animals. Cell Metab. 8, 301–309 (2008).

    Article  CAS  Google Scholar 

  16. Lee, Y. S. et al. Inflammation is necessary for long-term but not short-term high-fat diet-induced insulin resistance. Diabetes 60, 2474–2483 (2011).

    Article  CAS  Google Scholar 

  17. Lee, Y. S. et al. Increased adipocyte O2 consumption triggers HIF-1α, causing inflammation and insulin resistance in obesity. Cell 157, 1339–1352 (2014).

    Article  CAS  Google Scholar 

  18. Halberg, N. et al. Hypoxia-inducible factor 1α induces fibrosis and insulin resistance in white adipose tissue. Mol. Cell. Biol. 29, 4467–4483 (2009).

    Article  CAS  Google Scholar 

  19. Hosogai, N. et al. Adipose tissue hypoxia in obesity and its impact on adipocytokine dysregulation. Diabetes 56, 901–911 (2007).

    Article  CAS  Google Scholar 

  20. Palazon, A., Goldrath, A. W., Nizet, V. & Johnson, R. S. HIF transcription factors, inflammation, and immunity. Immunity 41, 518–528 (2014).

    Article  CAS  Google Scholar 

  21. Jiang, C. et al. Disruption of hypoxia-inducible factor 1 in adipocytes improves insulin sensitivity and decreases adiposity in high-fat diet-fed mice. Diabetes 60, 2484–2495 (2011).

    Article  CAS  Google Scholar 

  22. Lee, K. Y., Gesta, S., Boucher, J., Wang, X. L. & Kahn, C. R. The differential role of Hif1β/Arnt and the hypoxic response in adipose function, fibrosis, and inflammation. Cell Metab. 14, 491–503 (2011).

    Article  CAS  Google Scholar 

  23. Gealekman, O. et al. Depot-specific differences and insufficient subcutaneous adipose tissue angiogenesis in human obesity. Circulation 123, 186–194 (2011).

    Article  Google Scholar 

  24. Cao, Y. Angiogenesis and vascular functions in modulation of obesity, adipose metabolism, and insulin sensitivity. Cell Metab. 18, 478–489 (2013).

    Article  CAS  Google Scholar 

  25. Sun, K., Halberg, N., Khan, M., Magalang, U. J. & Scherer, P. E. Selective inhibition of hypoxia-inducible factor 1α ameliorates adipose tissue dysfunction. Mol. Cell. Biol. 33, 904–917 (2013).

  26. Pasarica, M. et al. Reduced adipose tissue oxygenation in human obesity: evidence for rarefaction, macrophage chemotaxis, and inflammation without an angiogenic response. Diabetes 58, 718–725 (2009).

    Article  CAS  Google Scholar 

  27. Keith, B., Johnson, R. S. & Simon, M. C. HIF1α and HIF2α: sibling rivalry in hypoxic tumour growth and progression. Nat. Rev. Cancer 12, 9–22 (2011).

  28. Lumeng, C. N., Bodzin, J. L. & Saltiel, A. R. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J. Clin. Invest. 117, 175–184 (2007).

    Article  CAS  Google Scholar 

  29. Amano, S. U. et al. Local proliferation of macrophages contributes to obesity-associated adipose tissue inflammation. Cell Metab. 19, 162–171 (2014).

    Article  CAS  Google Scholar 

  30. Haase, J. et al. Local proliferation of macrophages in adipose tissue during obesity-induced inflammation. Diabetologia 57, 562–571 (2014).

    Article  CAS  Google Scholar 

  31. Regula, K. M., Ens, K. & Kirshenbaum, L. A. Inducible expression of BNIP3 provokes mitochondrial defects and hypoxia-mediated cell death of ventricular myocytes. Circ. Res. 91, 226–231 (2002).

    Article  CAS  Google Scholar 

  32. Kubli, D. A., Ycaza, J. E. & Gustafsson, A. B. Bnip3 mediates mitochondrial dysfunction and cell death through Bax and Bak. Biochem. J. 405, 407–415 (2007).

    Article  CAS  Google Scholar 

  33. Vande Velde, C. et al. BNIP3 and genetic control of necrosis-like cell death through the mitochondrial permeability transition pore. Mol. Cell. Biol. 20, 5454–5468 (2000).

    Article  CAS  Google Scholar 

  34. Chavez, J. A. & Summers, S. A. A ceramide-centric view of insulin resistance. Cell Metab. 15, 585–594 (2012).

    Article  CAS  Google Scholar 

  35. Holland, W. L. et al. Receptor-mediated activation of ceramidase activity initiates the pleiotropic actions of adiponectin. Nat. Med. 17, 55–63 (2011).

    Article  CAS  Google Scholar 

  36. Andreyev, A. et al. The ATP/ADP-antiporter is involved in the uncoupling effect of fatty acids on mitochondria. Eur. J. Biochem. 182, 585–592 (1989).

    Article  Google Scholar 

  37. Fabbrini, E. et al. Metabolically normal obese people are protected from adverse effects following weight gain. J. Clin. Invest. 125, 787–795 (2015).

    Article  Google Scholar 

  38. McLaughlin, T. et al. Subcutaneous adipose cell size and distribution: relationship to insulin resistance and body fat. Obes. (Silver Spring) 22, 673–680 (2014).

    Article  CAS  Google Scholar 

  39. Kolehmainen, M., Vidal, H., Alhava, E. & Uusitupa, M. I. Sterol regulatory element binding protein 1c (SREBP-1c) expression in human obesity. Obes. Res. 9, 706–712 (2001).

    Article  CAS  Google Scholar 

  40. Pasarica, M. et al. Reduced oxygenation in human obese adipose tissue is associated with impaired insulin suppression of lipolysis. J. Clin. Endocrinol. Metab. 95, 4052–4055 (2010).

    Article  CAS  Google Scholar 

  41. Li, P. et al. Adipocyte NCoR knockout decreases PPARγ phosphorylation and enhances PPARγ activity and insulin sensitivity. Cell 147, 815–826 (2011).

  42. Kusminski, C. M. et al. MitoNEET-driven alterations in adipocyte mitochondrial activity reveal a crucial adaptive process that preserves insulin sensitivity in obesity. Nat. Med. 18, 1539–1549 (2012).

    Article  CAS  Google Scholar 

  43. Kim, J. Y. et al. Obesity-associated improvements in metabolic profile through expansion of adipose tissue. J. Clin. Invest. 117, 2621–2637 (2007).

    Article  CAS  Google Scholar 

  44. Liu, Y. & Chen, X. J. Adenine nucleotide translocase, mitochondrial stress, and degenerative cell death. Oxid. Med. Cell. Longev. 2013, 146860 (2013).

    PubMed  PubMed Central  Google Scholar 

  45. Shabalina, I. G., Kramarova, T. V., Nedergaard, J. & Cannon, B. Carboxyatractyloside effects on brown-fat mitochondria imply that the adenine nucleotide translocator isoforms ANT1 and ANT2 may be responsible for basal and fatty-acid-induced uncoupling respectively. Biochem. J. 399, 405–414 (2006).

    Article  CAS  Google Scholar 

  46. Cho, J. et al. Mitochondrial ATP transporter depletion protects mice against liver steatosis and insulin resistance. Nat. Commun. 8, 14477 (2017).

    Article  CAS  Google Scholar 

  47. Kokoszka, J. E. et al. The ADP/ATP translocator is not essential for the mitochondrial permeability transition pore. Nature 427, 461–465 (2004).

    Article  CAS  Google Scholar 

  48. Seo, J. B. et al. Activated liver X receptors stimulate adipocyte differentiation through induction of peroxisome proliferator-activated receptor gamma expression. Mol. Cell. Biol. 24, 3430–3444 (2004).

    Article  CAS  Google Scholar 

  49. Farrall, A. L. & Whitelaw, M. L. The HIF1α-inducible pro-cell death gene BNIP3 is a novel target of SIM2s repression through cross-talk on the hypoxia response element. Oncogene 28, 3671–3680 (2009).

    Article  CAS  Google Scholar 

  50. Quispe-Tintaya, W. et al. Fast mitochondrial DNA isolation from mammalian cells for next-generation sequencing. Biotechniques 55, 133–136 (2013).

    Article  CAS  Google Scholar 

  51. Don, A.S. & Rosen, H. A fluorescent plate reader assay for ceramide kinase. Anal. Biochem. 375, 265–271 (2008).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank D. Wallace (University of Pennsylvania) for providing Ant2 floxed mice. This study was supported by the US National Institute of Diabetes and Digestive and Kidney Diseases (DK063491 and DK101395), a University of California San Diego/University of California Los Angeles Diabetes Research Center P&F grant, the Basic Science Research Program and the Bio-Synergy Research Project through National Research Foundation of Korea (NRF-2017R1C1B2011125 and NRF-2017M3A9C4065956), the POSCO TJ Park Foundation, and a grant from the Merck, Inc., Janssen Pharmceuticals, Inc., and Pershing Square Foundation. M.R. was supported by a postdoctoral fellowship from the American Heart Association (16POST29990015).

Author information

Authors and Affiliations

Authors

Contributions

Y.S.L. and J.B.S. designed and performed the majority of the experiments. M.R. performed glucose clamp experiments. P.C. measured adipose tissue interstitial oxygen tension and hemodynamics. A.N.M. and A.Y.A supported measuring mitochondrial activity and oxygen consumption. J.Y.H. performed flow cytometry analysis of adipose tissue immune cells. S.C.B., G.I.S. and S.K. performed clinical studies in MNL, MAO and MNO subjects and measured human adipose tissue oxygen tension. Y.S.L. and J.M.O. conceived and supervised the project. J.B.S., Y.S.L. and J.M.O. wrote the manuscript and all authors commented on the paper.

Corresponding authors

Correspondence to Yun Sok Lee or Jerrold M. Olefsky.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Tables 1 and 2

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Seo, J.B., Riopel, M., Cabrales, P. et al. Knockdown of ANT2 reduces adipocyte hypoxia and improves insulin resistance in obesity. Nat Metab 1, 86–97 (2019). https://doi.org/10.1038/s42255-018-0003-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-018-0003-x

Keywords

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