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.

  • Letter
  • Published:

Activation of calcium signaling through Trpv1 by nNOS and peroxynitrite as a key trigger of skeletal muscle hypertrophy

Abstract

Skeletal muscle atrophy occurs in aging and pathological conditions, including cancer, diabetes and AIDS1. Treatment of atrophy is based on either preventing protein-degradation pathways, which are activated during atrophy, or activating protein-synthesis pathways, which induce muscle hypertrophy2. Here we show that neuronal nitric oxide synthase (nNOS) regulates load-induced hypertrophy by activating transient receptor potential cation channel, subfamily V, member 1 (TRPV1). The overload-induced hypertrophy was prevented in nNOS-null mice. nNOS was transiently activated within 3 min after overload. This activation promoted formation of peroxynitrite, a reaction product of nitric oxide with superoxide3, which was derived from NADPH oxidase 4 (Nox4). Nitric oxide and peroxynitrite then activated Trpv1, resulting in an increase of intracellular Ca2+ concentration ([Ca2+]i) that subsequently triggered activation of mammalian target of rapamycin (mTOR). Notably, administration of the TRPV1 agonist capsaicin induced hypertrophy without overload and alleviated unloading- or denervation-induced atrophy. These findings identify nitric oxide, peroxynitrite and [Ca2+]i as the crucial mediators that convert a mechanical load into an intracellular signaling pathway and lead us to suggest that TRPV1 could be a new therapeutic target for treating muscle atrophy.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Immediate activation of nNOS promotes overload-induced muscle hypertrophy.
Figure 2: Overload-induced muscle hypertrophy is regulated by peroxynitrite.
Figure 3: Nitric oxide and peroxynitrite regulate the immediate activation of mTOR in an [Ca2+]i-dependent manner.
Figure 4: Trpv1-mediated increase in [Ca2+]i induces hypertrophy and alleviates unload- or denervation-induced muscle atrophy.

Similar content being viewed by others

References

  1. Lecker, S.H., Goldberg, A.L. & Mitch, W.E. Protein degradation by the ubiquitin-proteasome pathway in normal and disease states. J. Am. Soc. Nephrol. 17, 1807–1819 (2006).

    Article  CAS  Google Scholar 

  2. Sandri, M. Signaling in muscle atrophy and hypertrophy. Physiology (Bethesda) 23, 160–170 (2008).

    CAS  Google Scholar 

  3. Heck, D.E. *NO, RSNO, ONOO-, NO+, *NOO, NOx–dynamic regulation of oxidant scavenging, nitric oxide stores, and cyclic GMP-independent cell signaling. Antioxid. Redox Signal. 3, 249–260 (2001).

    Article  CAS  Google Scholar 

  4. Hoffman, E.P. & Nader, G.A. Balancing muscle hypertrophy and atrophy. Nat. Med. 10, 584–585 (2004).

    Article  CAS  Google Scholar 

  5. Bodine, S.C. et al. Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nat. Cell Biol. 3, 1014–1019 (2001).

    Article  CAS  Google Scholar 

  6. Goodman, C.A. et al. A phosphatidylinositol 3-kinase/protein kinase B–independent activation of mammalian target of rapamycin signaling is sufficient to induce skeletal muscle hypertrophy. Mol. Biol. Cell 21, 3258–3268 (2010).

    Article  CAS  Google Scholar 

  7. Hornberger, T.A. et al. Mechanical stimuli regulate rapamycin-sensitive signalling by a phosphoinositide 3-kinase–, protein kinase B– and growth factor–independent mechanism. Biochem. J. 380, 795–804 (2004).

    Article  CAS  Google Scholar 

  8. Zoncu, R., Efeyan, A. & Sabatini, D.M. mTOR: from growth signal integration to cancer, diabetes and ageing. Nat. Rev. Mol. Cell Biol. 12, 21–35 (2011).

    Article  CAS  Google Scholar 

  9. Ma, X.M. & Blenis, J. Molecular mechanisms of mTOR-mediated translational control. Nat. Rev. Mol. Cell Biol. 10, 307–318 (2009).

    Article  Google Scholar 

  10. Brenman, J.E. et al. Interaction of nitric oxide synthase with the postsynaptic density protein PSD-95 and α1-syntrophin mediated by PDZ domains. Cell 84, 757–767 (1996).

    Article  CAS  Google Scholar 

  11. Stamler, J.S. & Meissner, G. Physiology of nitric oxide in skeletal muscle. Physiol. Rev. 81, 209–237 (2001).

    Article  CAS  Google Scholar 

  12. Asai, A. et al. Primary role of functional ischemia, quantitative evidence for the two-hit mechanism, and phosphodiesterase-5 inhibitor therapy in mouse muscular dystrophy. PLoS ONE 2, e806 (2007).

    Article  Google Scholar 

  13. Wozniak, A.C. & Anderson, J.E. Nitric oxide–dependence of satellite stem cell activation and quiescence on normal skeletal muscle fibers. Dev. Dyn. 236, 240–250 (2007).

    Article  CAS  Google Scholar 

  14. Yoshida, T. et al. Nitric oxide activates TRP channels by cysteine S-nitrosylation. Nat. Chem. Biol. 2, 596–607 (2006).

    Article  CAS  Google Scholar 

  15. Adachi, T. et al. S-Glutathiolation by peroxynitrite activates SERCA during arterial relaxation by nitric oxide. Nat. Med. 10, 1200–1207 (2004).

    Article  CAS  Google Scholar 

  16. Trebak, M., Ginnan, R., Singer, H.A. & Jourd′heuil, D. Interplay between calcium and reactive oxygen/nitrogen species: an essential paradigm for vascular smooth muscle signaling. Antioxid. Redox Signal. 12, 657–674 (2010).

    Article  CAS  Google Scholar 

  17. Sellman, J.E. et al. In vivo inhibition of nitric oxide synthase impairs upregulation of contractile protein mRNA in overloaded plantaris muscle. J. Appl. Physiol. 100, 258–265 (2006).

    Article  CAS  Google Scholar 

  18. Soltow, Q.A. et al. Ibuprofen inhibits skeletal muscle hypertrophy in rats. Med. Sci. Sports Exerc. 38, 840–846 (2006).

    Article  CAS  Google Scholar 

  19. Kemp-Harper, B. & Feil, R. Meeting report: cGMP matters. Sci. Signal. 1, pe12 (2008).

    Article  Google Scholar 

  20. Carnesecchi, S. et al. A key role for NOX4 in epithelial cell death during development of lung fibrosis. Antioxid. Redox Signal. 15, 607–619 (2011).

    Article  CAS  Google Scholar 

  21. Bedard, K. & Krause, K.H. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol. Rev. 87, 245–313 (2007).

    Article  CAS  Google Scholar 

  22. Laleu, B. et al. First in class, potent, and orally bioavailable NADPH oxidase isoform 4 (Nox4) inhibitors for the treatment of idiopathic pulmonary fibrosis. J. Med. Chem. 53, 7715–7730 (2010).

    Article  CAS  Google Scholar 

  23. Tallini, Y.N. et al. Imaging cellular signals in the heart in vivo: cardiac expression of the high-signal Ca2+ indicator GCaMP2. Proc. Natl. Acad. Sci. USA 103, 4753–4758 (2006).

    Article  CAS  Google Scholar 

  24. Piétri-Rouxel, F. et al. DHPR α1S subunit controls skeletal muscle mass and morphogenesis. EMBO J. 29, 643–654 (2010).

    Article  Google Scholar 

  25. Luo, Z. et al. TRPV1 activation improves exercise endurance and energy metabolism through PGC-1α upregulation in mice. Cell Res. 22, 551–564 (2012).

    Article  CAS  Google Scholar 

  26. Xin, H. et al. Vanilloid receptor expressed in the sarcoplasmic reticulum of rat skeletal muscle. Biochem. Biophys. Res. Commun. 332, 756–762 (2005).

    Article  CAS  Google Scholar 

  27. Suzuki, N. et al. NO production results in suspension-induced muscle atrophy through dislocation of neuronal NOS. J. Clin. Invest. 117, 2468–2476 (2007).

    Article  CAS  Google Scholar 

  28. Gulati, P. et al. Amino acids activate mTOR complex 1 via Ca2+/CaM signaling to hVps34. Cell Metab. 7, 456–465 (2008).

    Article  CAS  Google Scholar 

  29. Cunningham, J.T. et al. mTOR controls mitochondrial oxidative function through a YY1–PGC-1α transcriptional complex. Nature 450, 736–740 (2007).

    Article  CAS  Google Scholar 

  30. Sandri, M. et al. PGC-1α protects skeletal muscle from atrophy by suppressing FoxO3 action and atrophy-specific gene transcription. Proc. Natl. Acad. Sci. USA 103, 16260–16265 (2006).

    Article  CAS  Google Scholar 

  31. Aoki, Y. et al. In-frame dystrophin following exon 51–skipping improves muscle pathology and function in the exon 52–deficient mdx mouse. Mol. Ther. 18, 1995–2005 (2010).

    Article  CAS  Google Scholar 

  32. Kuroda, J. et al. NADPH oxidase 4 (Nox4) is a major source of oxidative stress in the failing heart. Proc. Natl. Acad. Sci. USA 107, 15565–15570 (2010).

    Article  CAS  Google Scholar 

  33. Whitehead, N.P., Yeung, E.W., Froehner, S.C. & Allen, D.G. Skeletal muscle NADPH oxidase is increased and triggers stretch-induced damage in the mdx mouse. PLoS ONE 5, e15354 (2010).

    Article  CAS  Google Scholar 

  34. Ono, Y., Boldrin, L., Knopp, P., Morgan, J.E. & Zammit, P.S. Muscle satellite cells are a functionally heterogeneous population in both somite-derived and branchiomeric muscles. Dev. Biol. 337, 29–41 (2010).

    Article  CAS  Google Scholar 

  35. Bakondi, E. et al. Role of intracellular calcium mobilization and cell-density–dependent signaling in oxidative-stress–induced cytotoxicity in HaCaT keratinocytes. J. Invest. Dermatol. 121, 88–95 (2003).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank M. Kotlikoff (Department of Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA) for providing pCAGGS-GCaMP2 mice, K.-H. Krause (Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland) for Nox4-null mice, Y. Ono for the technical support to isolate satellite cells and GenKyoTex (Geneva, Switzerland) for supplying GKT136901. We also thank D. Glass, I. Kii, V. Ullrich, B. Gähwiler and Y. Aoki for reading the manuscript and contributing to valuable discussions. This work was supported by a Grant-in-Aid for Scientific Research (B), a Grant-in-Aid for Japan Society for the Promotion of Science Fellows and a grant from the Association Française contre les Myopathies (AFM, France).

Author information

Authors and Affiliations

Authors

Contributions

N.I. conceived and performed the experiments. A.K., Y.M.-S., U.T.R. and S.T. conducted and supervised the experiments. N.I. and U.T.R. designed the experiments and wrote the manuscript, and all authors discussed the results and commented on the manuscript.

Corresponding author

Correspondence to Shin'ichi Takeda.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 (PDF 2441 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Ito, N., Ruegg, U., Kudo, A. et al. Activation of calcium signaling through Trpv1 by nNOS and peroxynitrite as a key trigger of skeletal muscle hypertrophy. Nat Med 19, 101–106 (2013). https://doi.org/10.1038/nm.3019

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3019

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