A branched-chain amino acid metabolite drives vascular fatty acid transport and causes insulin resistance (original) (raw)

Nature Medicine volume 22, pages 421–426 (2016)Cite this article

Subjects

Abstract

Epidemiological and experimental data implicate branched-chain amino acids (BCAAs) in the development of insulin resistance, but the mechanisms that underlie this link remain unclear1,2,3. Insulin resistance in skeletal muscle stems from the excess accumulation of lipid species4, a process that requires blood-borne lipids to initially traverse the blood vessel wall. How this trans-endothelial transport occurs and how it is regulated are not well understood. Here we leveraged PPARGC1a (also known as PGC-1α; encoded by Ppargc1a), a transcriptional coactivator that regulates broad programs of fatty acid consumption, to identify 3-hydroxyisobutyrate (3-HIB), a catabolic intermediate of the BCAA valine, as a new paracrine regulator of trans-endothelial fatty acid transport. We found that 3-HIB is secreted from muscle cells, activates endothelial fatty acid transport, stimulates muscle fatty acid uptake in vivo and promotes lipid accumulation in muscle, leading to insulin resistance in mice. Conversely, inhibiting the synthesis of 3-HIB in muscle cells blocks the ability of PGC-1α to promote endothelial fatty acid uptake. 3-HIB levels are elevated in muscle from db/db mice with diabetes and from human subjects with diabetes, as compared to those without diabetes. These data unveil a mechanism in which the metabolite 3-HIB, by regulating the trans-endothelial flux of fatty acids, links the regulation of fatty acid flux to BCAA catabolism, providing a mechanistic explanation for how increased BCAA catabolic flux can cause diabetes.

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

Access options

Subscribe to this journal

Receive 12 print issues and online access

$209.00 per year

only $17.42 per issue

Buy this article

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

Additional access options:

Similar content being viewed by others

References

  1. Wang, T.J. et al. Metabolite profiles and the risk of developing diabetes. Nat. Med. 17, 448–453 (2011).
    Article Google Scholar
  2. Newgard, C.B. et al. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab. 9, 311–326 (2009).
    Article CAS Google Scholar
  3. Newgard, C.B. Interplay between lipids and branched-chain amino acids in development of insulin resistance. Cell Metab. 15, 606–614 (2012).
    Article CAS Google Scholar
  4. Shulman, G.I. Ectopic fat in insulin resistance, dyslipidemia, and cardiometabolic disease. N. Engl. J. Med. 371, 1131–1141 (2014).
    Article Google Scholar
  5. Chan, M.C. & Arany, Z. The many roles of PGC-1α in muscle—recent developments. Metabolism 63, 441–451 (2014).
    Article CAS Google Scholar
  6. Handschin, C. & Spiegelman, B.M. Peroxisome proliferator-activated receptor gamma coactivator 1 coactivators, energy homeostasis, and metabolism. Endocr. Rev. 27, 728–735 (2006).
    Article CAS Google Scholar
  7. Arany, Z. et al. HIF-independent regulation of VEGF and angiogenesis by the transcriptional coactivator PGC-1α. Nature 451, 1008–1012 (2008).
    Article CAS Google Scholar
  8. Hagberg, C.E. et al. Vascular endothelial growth factor B controls endothelial fatty acid uptake. Nature 464, 917–921 (2010).
    Article CAS Google Scholar
  9. Roberts, L.D. et al. β-Aminoisobutyric acid induces browning of white fat and hepatic β-oxidation and is inversely correlated with cardiometabolic risk factors. Cell Metab. 19, 96–108 (2014).
    Article CAS Google Scholar
  10. Lin, J. et al. Transcriptional co-activator PGC-1α drives the formation of slow-twitch muscle fibres. Nature 418, 797–801 (2002).
    Article CAS Google Scholar
  11. Arany, Z. et al. The transcriptional coactivator PGC-1β drives the formation of oxidative type IIX fibers in skeletal muscle. Cell Metab. 5, 35–46 (2007).
    Article CAS Google Scholar
  12. Hatazawa, Y. et al. Metabolomic analysis of the skeletal muscle of mice overexpressing PGC-1α. PLoS One 10, e0129084 (2015).
    Article Google Scholar
  13. Henkin, A.H. et al. Real-time noninvasive imaging of fatty acid uptake in vivo. ACS Chem. Biol. 7, 1884–1891 (2012).
    Article CAS Google Scholar
  14. Choi, C.S. et al. Paradoxical effects of increased expression of PGC-1α on muscle mitochondrial function and insulin-stimulated muscle glucose metabolism. Proc. Natl. Acad. Sci. USA 105, 19926–19931 (2008).
    Article CAS Google Scholar
  15. Avogaro, A. & Bier, D.M. Contribution of 3-hydroxyisobutyrate to the measurement of 3-hydroxybutyrate in human plasma: comparison of enzymatic and gas-liquid chromatography-mass spectrometry assays in normal and in diabetic subjects. J. Lipid Res. 30, 1811–1817 (1989).
    CAS PubMed Google Scholar
  16. Giesbertz, P. et al. Metabolite profiling in plasma and tissues of ob/ob and db/db mice identifies novel markers of obesity and type 2 diabetes. Diabetologia 58, 2133–2143 (2015).
    Article CAS Google Scholar
  17. Mullen, E. & Ohlendieck, K. Proteomic profiling of non-obese type 2 diabetic skeletal muscle. Int. J. Mol. Med. 25, 445–458 (2010).
    CAS PubMed Google Scholar
  18. Sasaki, M. et al. A severely brain-damaged case of 3-hydroxyisobutyric aciduria. Brain Dev. 23, 243–245 (2001).
    Article CAS Google Scholar
  19. Abbott, N.J., Hughes, C.C., Revest, P.A. & Greenwood, J. Development and characterisation of a rat brain capillary endothelial culture: towards an in vitro blood-brain barrier. J. Cell Sci. 103, 23–37 (1992).
    CAS PubMed Google Scholar
  20. Xie, Z. et al. Vascular endothelial hyperpermeability induces the clinical symptoms of Clarkson disease (the systemic capillary leak syndrome). Blood 119, 4321–4332 (2012).
    Article CAS Google Scholar
  21. Rowe, G.C. et al. Disconnecting mitochondrial content from respiratory chain capacity in PGC-1-deficient skeletal muscle. Cell Reports 3, 1449–1456 (2013).
    Article CAS Google Scholar
  22. Sawada, N. et al. Endothelial PGC-1α mediates vascular dysfunction in diabetes. Cell Metab. 19, 246–258 (2014).
    Article CAS Google Scholar
  23. Darland, D.C. & D'Amore, P.A. TGFβ is required for the formation of capillary-like structures in three-dimensional cocultures of 10T1/2 and endothelial cells. Angiogenesis 4, 11–20 (2001).
    Article CAS Google Scholar
  24. Titchenell, P.M., Chu, Q., Monks, B.R. & Birnbaum, M.J. Hepatic insulin signalling is dispensable for suppression of glucose output by insulin in vivo. Nat. Commun. 6, 7078 (2015).
    Article CAS Google Scholar
  25. Forman, D.E. et al. Analysis of skeletal muscle gene expression patterns and the impact of functional capacity in patients with systolic heart failure. J. Card. Fail. 20, 422–430 (2014).
    Article CAS Google Scholar

Download references

Acknowledgements

Human endothelial colony forming cells (ECFCs) were kindly provided by J. Bischoff (Boston Children's Hospital). _Fatp4_−/− and _Cd36_−/− mice were kindly provided by J. Miner (Washington University School of Medicine) and J. Lawler (Harvard Medical School), respectively. _Flt1_flox/flox and _Kdr_flox/flox mice were kindly provided by Genentech. C.J. is supported by the Lotte Scholarship and American Heart Association (AHA). S.F.O. is supported by the Crohn's and Colitis Foundation of America (Research Fellowship Award). S.W. is supported by the Toyobo Biotechnology Foundation. G.C.R. is supported by the US National Institute of Arthritis and Musculoskeletal and Skin Diseases (AR062128). J.R. is supported by the US National Institutes of Health (5 T32 GM7592-35). S.M.P. is supported by the US National Heart, Lung, and Blood Institute (NHLBI) (HL093234; HL125275) and the US National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) (DK095072). Q.C. and J.A.B. are supported by the NIDDK (DK098656; DK049210). Z.A. is supported by the NHLBI (HL094499), the AHA and the Geis Realty Group Emerging Initiatives Fund and Dean and Ann Geis.

Author information

Author notes

  1. Glenn C Rowe
    Present address: Present address: Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.,
  2. Cholsoon Jang and Sungwhan F Oh: These authors contributed equally to this work.

Authors and Affiliations

  1. Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
    Cholsoon Jang, Shogo Wada, Atsushi Hoshino, Boa Kim, Ayon Ibrahim, Qingwei Chu, Saikumari Krishnaiah, Aalim M Weljie, Joseph A Baur & Zoltan Arany
  2. Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
    Cholsoon Jang, Glenn C Rowe, Laura Liu, Mun Chun Chan, James Rhee, Luisa G Baca, Esl Kim, Chandra C Ghosh, Samir M Parikh, Aihua Jiang & Stewart H Lecker
  3. Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
    Sungwhan F Oh & Dennis L Kasper
  4. Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
    James Rhee
  5. Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
    Daniel E Forman
  6. Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, USA
    Joshua D Rabinowitz

Authors

  1. Cholsoon Jang
    You can also search for this author inPubMed Google Scholar
  2. Sungwhan F Oh
    You can also search for this author inPubMed Google Scholar
  3. Shogo Wada
    You can also search for this author inPubMed Google Scholar
  4. Glenn C Rowe
    You can also search for this author inPubMed Google Scholar
  5. Laura Liu
    You can also search for this author inPubMed Google Scholar
  6. Mun Chun Chan
    You can also search for this author inPubMed Google Scholar
  7. James Rhee
    You can also search for this author inPubMed Google Scholar
  8. Atsushi Hoshino
    You can also search for this author inPubMed Google Scholar
  9. Boa Kim
    You can also search for this author inPubMed Google Scholar
  10. Ayon Ibrahim
    You can also search for this author inPubMed Google Scholar
  11. Luisa G Baca
    You can also search for this author inPubMed Google Scholar
  12. Esl Kim
    You can also search for this author inPubMed Google Scholar
  13. Chandra C Ghosh
    You can also search for this author inPubMed Google Scholar
  14. Samir M Parikh
    You can also search for this author inPubMed Google Scholar
  15. Aihua Jiang
    You can also search for this author inPubMed Google Scholar
  16. Qingwei Chu
    You can also search for this author inPubMed Google Scholar
  17. Daniel E Forman
    You can also search for this author inPubMed Google Scholar
  18. Stewart H Lecker
    You can also search for this author inPubMed Google Scholar
  19. Saikumari Krishnaiah
    You can also search for this author inPubMed Google Scholar
  20. Joshua D Rabinowitz
    You can also search for this author inPubMed Google Scholar
  21. Aalim M Weljie
    You can also search for this author inPubMed Google Scholar
  22. Joseph A Baur
    You can also search for this author inPubMed Google Scholar
  23. Dennis L Kasper
    You can also search for this author inPubMed Google Scholar
  24. Zoltan Arany
    You can also search for this author inPubMed Google Scholar

Contributions

C.J. led the studies and was directly involved in most experiments. S.F.O. assigned the structure of the paracrine factor as 3-HIB and performed mass spectrometric profiling. S.W., G.C.R., L.L., M.C.C., J.R., A.H., B.K., A.I., L.G.B., E.K. and A.J. assisted with experiments throughout, including qPCR, cell culture and animal studies. Q.C. and J.A.B. performed the mouse clamp studies. S.K. and A.M.W. performed the lipidomic studies. D.E.F. and S.H.L. isolated the human muscle biopsies. C.C.G. and S.M.P. performed the TEER studies. J.D.R. performed the metabolic flux analysis. D.L.K. and Z.A. oversaw the studies. C.J. and Z.A. designed experiments, interpreted results and wrote the paper. All authors discussed the results and commented on the manuscript.

Corresponding author

Correspondence toZoltan Arany.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Rights and permissions

About this article

Cite this article

Jang, C., Oh, S., Wada, S. et al. A branched-chain amino acid metabolite drives vascular fatty acid transport and causes insulin resistance.Nat Med 22, 421–426 (2016). https://doi.org/10.1038/nm.4057

Download citation

This article is cited by