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:

MitoNEET-driven alterations in adipocyte mitochondrial activity reveal a crucial adaptive process that preserves insulin sensitivity in obesity

Abstract

We examined mouse models with altered adipocyte expression of mitoNEET, a protein residing in the mitochondrial outer membrane, to probe its impact on mitochondrial function and subsequent cellular responses. We found that overexpression of mitoNEET enhances lipid uptake and storage, leading to an expansion of the mass of adipose tissue. Despite the resulting massive obesity, benign aspects of adipose tissue expansion prevail, and insulin sensitivity is preserved. Mechanistically, we also found that mitoNEET inhibits mitochondrial iron transport into the matrix and, because iron is a rate-limiting component for electron transport, lowers the rate of β-oxidation. This effect is associated with a lower mitochondrial membrane potential and lower levels of reactive oxygen species–induced damage, along with increased production of adiponectin. Conversely, a reduction in mitoNEET expression enhances mitochondrial respiratory capacity through enhanced iron content in the matrix, ultimately corresponding to less weight gain on a high-fat diet. However, this reduction in mitoNEET expression also causes heightened oxidative stress and glucose intolerance. Thus, manipulation of mitochondrial function by varying mitoNEET expression markedly affects the dynamics of cellular and whole-body lipid homeostasis.

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: MitoNEET causes massive adipose tissue expansion and improves hepatic insulin sensitivity.
Figure 2: MitoNEET promotes lipid uptake by stimulating adiponectin production and heightening β-3 adrenergic agonist sensitivity.
Figure 3: MitoNEET-induced alterations in fatty acid metabolism.
Figure 4: MitoNEET compromises mitochondrial dynamics and morphology by modulating mitochondrial iron content.
Figure 5: A lack of mitoNEET enhances mitochondrial oxidative capacity.
Figure 6: Proposed mechanism of mitoNEET action.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Muoio, D.M. & Newgard, C.B. Obesity-related derangements in metabolic regulation. Annu. Rev. Biochem. 75, 367–401 (2006).

    Article  CAS  PubMed  Google Scholar 

  2. Lowell, B.B. & Shulman, G.I. Mitochondrial dysfunction and type 2 diabetes. Science 307, 384–387 (2005).

    Article  CAS  PubMed  Google Scholar 

  3. Mehta, J.L., Rasouli, N., Sinha, A.K. & Molavi, B. Oxidative stress in diabetes: a mechanistic overview of its effects on atherogenesis and myocardial dysfunction. Int. J. Biochem. Cell Biol. 38, 794–803 (2006).

    Article  CAS  PubMed  Google Scholar 

  4. Morino, K., Petersen, K.F. & Shulman, G.I. Molecular mechanisms of insulin resistance in humans and their potential links with mitochondrial dysfunction. Diabetes 55 (suppl. 2), S9–S15 (2006).

    Article  CAS  PubMed  Google Scholar 

  5. 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  PubMed  PubMed Central  Google Scholar 

  6. Scherer, P.E., Williams, S., Fogliano, M., Baldini, G. & Lodish, H.F. A novel serum protein similar to C1q, produced exclusively in adipocytes. J. Biol. Chem. 270, 26746–26749 (1995).

    Article  CAS  PubMed  Google Scholar 

  7. 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  PubMed  Google Scholar 

  8. Kelley, D.E., He, J., Menshikova, E.V. & Ritov, V.B. Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes. Diabetes 51, 2944–2950 (2002).

    Article  CAS  PubMed  Google Scholar 

  9. Ritov, V.B. et al. Deficiency of subsarcolemmal mitochondria in obesity and type 2 diabetes. Diabetes 54, 8–14 (2005).

    Article  CAS  PubMed  Google Scholar 

  10. Simoneau, J.A., Veerkamp, J.H., Turcotte, L.P. & Kelley, D.E. Markers of capacity to utilize fatty acids in human skeletal muscle: relation to insulin resistance and obesity and effects of weight loss. FASEB J. 13, 2051–2060 (1999).

    Article  CAS  PubMed  Google Scholar 

  11. Colca, J.R. et al. Identification of a novel mitochondrial protein (“mitoNEET”) cross-linked specifically by a thiazolidinedione photoprobe. Am. J. Physiol. Endocrinol. Metab. 286, E252–E260 (2004).

    Article  CAS  PubMed  Google Scholar 

  12. Paddock, M.L. et al. MitoNEET is a uniquely folded 2Fe 2S outer mitochondrial membrane protein stabilized by pioglitazone. Proc. Natl. Acad. Sci. USA 104, 14342–14347 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Wiley, S.E., Murphy, A.N., Ross, S.A., van der Geer, P. & Dixon, J.E. MitoNEET is an iron-containing outer mitochondrial membrane protein that regulates oxidative capacity. Proc. Natl. Acad. Sci. USA 104, 5318–5323 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Wiley, S.E. et al. The outer mitochondrial membrane protein mitoNEET contains a novel redox-active 2Fe-2S cluster. J. Biol. Chem. 282, 23745–23749 (2007).

    Article  CAS  PubMed  Google Scholar 

  15. Lin, J., Zhou, T., Ye, K. & Wang, J. Crystal structure of human mitoNEET reveals distinct groups of iron sulfur proteins. Proc. Natl. Acad. Sci. USA 104, 14640–14645 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Graves, R.A., Tontonoz, P., Platt, K.A., Ross, S.R. & Spiegelman, B.M. Identification of a fat cell enhancer: analysis of requirements for adipose tissue-specific gene expression. J. Cell. Biochem. 49, 219–224 (1992).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  18. Jornayvaz, F.R. & Shulman, G.I. Diacylglycerol activation of protein kinase cepsilon and hepatic insulin resistance. Cell Metab. 15, 574–584 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Milne, G.L., Sanchez, S.C., Musiek, E.S. & Morrow, J.D. Quantification of F2-isoprostanes as a biomarker of oxidative stress. Nat. Protoc. 2, 221–226 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Langin, D. Adipose tissue lipolysis as a metabolic pathway to define pharmacological strategies against obesity and the metabolic syndrome. Pharmacol. Res. 53, 482–491 (2006).

    Article  CAS  PubMed  Google Scholar 

  21. Asterholm, I.W. & Scherer, P.E. Enhanced metabolic flexibility associated with elevated adiponectin levels. Am. J. Pathol. 176, 1364–1376 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Franckhauser, S. et al. Increased fatty acid re-esterification by PEPCK overexpression in adipose tissue leads to obesity without insulin resistance. Diabetes 51, 624–630 (2002).

    Article  CAS  PubMed  Google Scholar 

  23. Hanson, R.W. & Reshef, L. Glyceroneogenesis revisited. Biochimie 85, 1199–1205 (2003).

    Article  CAS  PubMed  Google Scholar 

  24. Sun, K., Kusminski, C.M. & Scherer, P.E. Adipose tissue remodeling and obesity. J. Clin. Invest. 121, 2094–2101 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zuris, J.A. et al. Facile transfer of [2Fe-2S] clusters from the diabetes drug target mitoNEET to an apo-acceptor protein. Proc. Natl. Acad. Sci. USA 108, 13047–13052 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Macdonald, V.W., Charache, S. & Hathaway, P.J. Iron deficiency anemia: mitochondrial α-glycerophosphate dehydrogenase in guinea pig skeletal muscle. J. Lab. Clin. Med. 105, 11–18 (1985).

    CAS  PubMed  Google Scholar 

  27. Crooks, D.R., Ghosh, M.C., Haller, R.G., Tong, W.H. & Rouault, T.A. Posttranslational stability of the heme biosynthetic enzyme ferrochelatase is dependent on iron availability and intact iron-sulfur cluster assembly machinery. Blood 115, 860–869 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Huang, J. et al. Iron overload and diabetes risk: a shift from glucose to fatty acid oxidation and increased hepatic glucose production in a mouse model of hereditary hemochromatosis. Diabetes 60, 80–87 (2011).

    Article  CAS  PubMed  Google Scholar 

  29. Mitchell, P. Keilin's respiratory chain concept and its chemiosmotic consequences. Science 206, 1148–1159 (1979).

    Article  CAS  PubMed  Google Scholar 

  30. Rogers, G.W. et al. High throughput microplate respiratory measurements using minimal quantities of isolated mitochondria. PLoS ONE 6, e21746 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Rigoulet, M., Yoboue, E.D. & Devin, A. Mitochondrial ROS generation and its regulation: mechanisms involved in H2O2 signaling. Antioxid. Redox Signal. 14, 459–468 (2011).

    Article  CAS  PubMed  Google Scholar 

  32. Wu, M. et al. Multiparameter metabolic analysis reveals a close link between attenuated mitochondrial bioenergetic function and enhanced glycolysis dependency in human tumor cells. Am. J. Physiol. Cell Physiol. 292, C125–C136 (2007).

    Article  CAS  PubMed  Google Scholar 

  33. Houstis, N., Rosen, E.D. & Lander, E.S. Reactive oxygen species have a causal role in multiple forms of insulin resistance. Nature 440, 944–948 (2006).

    Article  CAS  PubMed  Google Scholar 

  34. Tarnopolsky, M.A. et al. Influence of endurance exercise training and sex on intramyocellular lipid and mitochondrial ultrastructure, substrate use, and mitochondrial enzyme activity. Am. J. Physiol. Regul. Integr. Comp. Physiol. 292, R1271–R1278 (2007).

    Article  CAS  PubMed  Google Scholar 

  35. Lin, J., Puigserver, P., Donovan, J., Tarr, P. & Spiegelman, B.M. Peroxisome proliferator-activated receptor γ coactivator 1β (PGC-1β), a novel PGC-1–related transcription coactivator associated with host cell factor. J. Biol. Chem. 277, 1645–1648 (2002).

    Article  CAS  PubMed  Google Scholar 

  36. Bonnard, C. et al. Mitochondrial dysfunction results from oxidative stress in the skeletal muscle of diet-induced insulin-resistant mice. J. Clin. Invest. 118, 789–800 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Curtis, J.M. et al. Downregulation of adipose glutathione S-transferase A4 leads to increased protein carbonylation, oxidative stress, and mitochondrial dysfunction. Diabetes 59, 1132–1142 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Karelis, A.D. et al. The metabolically healthy but obese individual presents a favorable inflammation profile. J. Clin. Endocrinol. Metab. 90, 4145–4150 (2005).

    Article  CAS  PubMed  Google Scholar 

  39. McGarry, J.D. Banting lecture 2001: dysregulation of fatty acid metabolism in the etiology of type 2 diabetes. Diabetes 51, 7–18 (2002).

    Article  CAS  PubMed  Google Scholar 

  40. Koh, E.H. et al. Essential role of mitochondrial function in adiponectin synthesis in adipocytes. Diabetes 56, 2973–2981 (2007).

    Article  CAS  PubMed  Google Scholar 

  41. Frizzell, N. et al. Succination of thiol groups in adipose tissue proteins in diabetes: succination inhibits polymerization and secretion of adiponectin. J. Biol. Chem. 284, 25772–25781 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Iwabu, M. et al. Adiponectin and AdipoR1 regulate PGC-1α and mitochondria by Ca2+ and AMPK/SIRT1. Nature 464, 1313–1319 (2010).

    Article  CAS  PubMed  Google Scholar 

  43. Sherratt, H.S. Mitochondria: structure and function. Rev. Neurol. (Paris) 147, 417–430 (1991).

    CAS  Google Scholar 

  44. Dey, R. & Moraes, C.T. Lack of oxidative phosphorylation and low mitochondrial membrane potential decrease susceptibility to apoptosis and do not modulate the protective effect of Bcl-x(L) in osteosarcoma cells. J. Biol. Chem. 275, 7087–7094 (2000).

    Article  CAS  PubMed  Google Scholar 

  45. 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 

  46. Anderson, E.J., Yamazaki, H. & Neufer, P.D. Induction of endogenous uncoupling protein 3 suppresses mitochondrial oxidant emission during fatty acid–supported respiration. J. Biol. Chem. 282, 31257–31266 (2007).

    Article  CAS  PubMed  Google Scholar 

  47. Anderson, E.J. et al. Mitochondrial H2O2 emission and cellular redox state link excess fat intake to insulin resistance in both rodents and humans. J. Clin. Invest. 119, 573–581 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Korshunov, S.S., Skulachev, V.P. & Starkov, A.A. High protonic potential actuates a mechanism of production of reactive oxygen species in mitochondria. FEBS Lett. 416, 15–18 (1997).

    Article  CAS  PubMed  Google Scholar 

  49. Yu, T., Robotham, J.L. & Yoon, Y. Increased production of reactive oxygen species in hyperglycemic conditions requires dynamic change of mitochondrial morphology. Proc. Natl. Acad. Sci. USA 103, 2653–2658 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Finck, B.N. et al. A potential link between muscle peroxisome proliferator- activated receptor-α signaling and obesity-related diabetes. Cell Metab. 1, 133–144 (2005).

    Article  CAS  PubMed  Google Scholar 

  51. Nawrocki, A.R. et al. Mice lacking adiponectin show decreased hepatic insulin sensitivity and reduced responsiveness to peroxisome proliferator-activated receptor γ agonists. J. Biol. Chem. 281, 2654–2660 (2006).

    Article  CAS  PubMed  Google Scholar 

  52. Berglund, E.D. et al. Direct leptin action on POMC neurons regulates glucose homeostasis and hepatic insulin sensitivity in mice. J. Clin. Invest. 122, 1000–1009 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Livak, K.J. & Schmittgen, T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔC(T) method. Methods 25, 402–408 (2001).

    Article  CAS  PubMed  Google Scholar 

  54. Scherer, P.E., Williams, S., Fogliano, M., Baldini, G. & Lodish, H.F. A novel serum protein similar to C1q, produced exclusively in adipocytes. J. Biol. Chem. 270, 26746–26749 (1995).

    Article  CAS  PubMed  Google Scholar 

  55. Combs, T.P. et al. A transgenic mouse with a deletion in the collagenous domain of adiponectin displays elevated circulating adiponectin and improved insulin sensitivity. Endocrinology 145, 367–383 (2004).

    Article  CAS  PubMed  Google Scholar 

  56. Laplante, M. et al. Tissue-specific postprandial clearance is the major determinant of PPARγ-induced triglyceride lowering in the rat. Am. J. Physiol. Regul. Integr. Comp. Physiol. 296, R57–R66 (2009).

    Article  CAS  PubMed  Google Scholar 

  57. Hultin, M., Carneheim, C., Rosenqvist, K. & Olivecrona, T. Intravenous lipid emulsions: removal mechanisms as compared to chylomicrons. J. Lipid Res. 36, 2174–2184 (1995).

    CAS  PubMed  Google Scholar 

  58. Bligh, E.G. & Dyer, W.J. A rapid method of total lipid extraction and purification. Can. J. Biochem. Physiol. 37, 911–917 (1959).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank P.D. Neufer, G. Schatz and C.B. Newgard for helpful comments and suggestions. We also thank J. Song and J. Xia for technical assistance, in addition to the rest of the Scherer laboratory, R. Unger and D. Clegg for helpful discussions. We also thank P. Blanchard and Y. Deshaies for kindly providing LPL activity measurements, R. Hammer and the University of Texas Southwestern (UTSW) Transgenic Core Facility for the generation of mouse models, as well as the UTSW Metabolic Core Facility for help in the phenotypic characterization of the mice and G. Milne from Vanderbilt University Medical Center for the F2-isoprostane analysis. The authors were supported by US National Institutes of Health grants R01-DK55758, RC1-DK086629 and P01-DK088761 (P.E.S.), K99-DK094973 and an American Heart Association Beginning Grant in Aid 12BGIA8910006 (W.L.H.), R01-DK081842 (D.A.M.), T32-DK091317 (J.A.S.) and Department of Defense Fellowship USAMRMC-BC085909 (J.P.). C.M.K. was supported by a fellowship from the Juvenile Diabetes Foundation (JDRF 3-2008-130).

Author information

Authors and Affiliations

Authors

Contributions

C.M.K. conducted all experiments and wrote the manuscript, except the portions indicated below. W.L.H. helped with the 3H-triolein uptake and β-oxidation experiments and performed their analyses. K.S. generated TRE-mitoNEET mice. J.P. helped plan, perform injections and scan fat pads in control AAV and AAV-mitoNEET experiments. S.B.S. generated the AAV-mitoNEET construct. Y.L. performed the DiOC6 ΔΨm experiment using mitoNEET-transfected 3T3-L1 preadipocytes. G.R.A. and C.L. coordinated the generation of shRNA-MitoN knockdown mice. J.A.S. and D.A.M. measured heme iron and provided high-iron-diet–fed and Hfe−/− liver tissues. P.E.S. was involved in the experimental design, experiments, data analysis and data interpretation, in addition to writing the manuscript.

Corresponding author

Correspondence to Philipp E Scherer.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5, Supplementary Tables 1–4 and Supplementary Methods (PDF 6156 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kusminski, C., Holland, W., Sun, K. 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). https://doi.org/10.1038/nm.2899

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2899

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