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:

Sarcolemma-localized nNOS is required to maintain activity after mild exercise

Abstract

Many neuromuscular conditions are characterized by an exaggerated exercise-induced fatigue response that is disproportionate to activity level. This fatigue is not necessarily correlated with greater central or peripheral fatigue in patients1, and some patients experience severe fatigue without any demonstrable somatic disease2. Except in myopathies that are due to specific metabolic defects, the mechanism underlying this type of fatigue remains unknown2. With no treatment available, this form of inactivity is a major determinant of disability3. Here we show, using mouse models, that this exaggerated fatigue response is distinct from a loss in specific force production by muscle, and that sarcolemma-localized signalling by neuronal nitric oxide synthase (nNOS) in skeletal muscle is required to maintain activity after mild exercise. We show that nNOS-null mice do not have muscle pathology and have no loss of muscle-specific force after exercise but do display this exaggerated fatigue response to mild exercise. In mouse models of nNOS mislocalization from the sarcolemma, prolonged inactivity was only relieved by pharmacologically enhancing the cGMP signal that results from muscle nNOS activation during the nitric oxide signalling response to mild exercise. Our findings suggest that the mechanism underlying the exaggerated fatigue response to mild exercise is a lack of contraction-induced signalling from sarcolemma-localized nNOS, which decreases cGMP-mediated vasomodulation in the vessels that supply active muscle after mild exercise. Sarcolemmal nNOS staining was decreased in patient biopsies from a large number of distinct myopathies, suggesting a common mechanism of fatigue. Our results suggest that patients with an exaggerated fatigue response to mild exercise would show clinical improvement in response to treatment strategies aimed at improving exercise-induced signalling.

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: Loss of sarcolemma-localized nNOS leads to skeletal muscle vascular narrowings, decreased capillary perfusion and an exaggerated fatigue response after mild exercise in dystrophic and non-dystrophic mouse models.
Figure 2: Enhancing the cGMP signal resulting from muscle nNOS activation decreases the exaggerated fatigue response to mild exercise.
Figure 3: Treatment with PDE5A inhibitor improves exercised-induced vasomodulation and decreases exercise-induced oedema in mdx mice.
Figure 4: nNOS levels in sarcolemma are decreased in human muscle diseases.

Similar content being viewed by others

References

  1. Schillings, M. L. et al. Experienced and physiological fatigue in neuromuscular disorders. Clin. Neurophysiol. 118, 292–300 (2007)

    Article  CAS  Google Scholar 

  2. Zwarts, M. J., Bleijenberg, G. & van Engelen, B. G. Clinical neurophysiology of fatigue. Clin. Neurophysiol doi: 10.1016/j.clinph.2007.09.126 (2007)

  3. Kalkman, J. S., Schillings, M. L., Zwarts, M. J., van Engelen, B. G. & Bleijenberg, G. The development of a model of fatigue in neuromuscular disorders: A longitudinal study. J. Psychosom. Res. 62, 571–579 (2007)

    Article  Google Scholar 

  4. Radley, H. G., De Luca, A., Lynch, G. S. & Grounds, M. D. Duchenne muscular dystrophy: focus on pharmaceutical and nutritional interventions. Int. J. Biochem. Cell Biol. 39, 469–477 (2007)

    Article  CAS  Google Scholar 

  5. Duclos, F. et al. Progressive muscular dystrophy in α-sarcoglycan-deficient mice. J. Cell Biol. 142, 1461–1471 (1998)

    Article  CAS  Google Scholar 

  6. Ozawa, E., Mizuno, Y., Hagiwara, Y., Sasaoka, T. & Yoshida, M. Molecular and cell biology of the sarcoglycan complex. Muscle Nerve 32, 563–576 (2005)

    Article  CAS  Google Scholar 

  7. Consolino, C. M. et al. Muscles of mice deficient in α-sarcoglycan maintain large masses and near control force values throughout the life span. Physiol. Genomics 22, 244–256 (2005)

    Article  CAS  Google Scholar 

  8. Yokoyama, T., Lisi, T. L., Moore, S. A. & Sluka, K. A. Muscle fatigue increases the probability of developing hyperalgesia in mice. J. Pain 8, 692–699 (2007)

    Article  Google Scholar 

  9. Harper, S. Q. et al. Modular flexibility of dystrophin: implications for gene therapy of Duchenne muscular dystrophy. Nature Med. 8, 253–261 (2002)

    Article  CAS  Google Scholar 

  10. Imamura, M., Mochizuki, Y., Engvall, E. & Takeda, S. I. ε-Sarcoglycan compensates for lack of α-sarcoglycan in a mouse model of limb-girdle muscular dystrophy. Hum. Mol. Genet. 14, 775–783 (2005)

    Article  CAS  Google Scholar 

  11. Phillips, B. A. & Mastaglia, F. L. Exercise therapy in patients with myopathy. Curr. Opin. Neurol. 13, 547–552 (2000)

    Article  CAS  Google Scholar 

  12. Torelli, S. et al. Absence of neuronal nitric oxide synthase (nNOS) as a pathological marker for the diagnosis of Becker muscular dystrophy with rod domain deletions. Neuropathol. Appl. Neurobiol. 30, 540–545 (2004)

    Article  CAS  Google Scholar 

  13. Judge, L. M., Haraguchiln, M. & Chamberlain, J. S. Dissecting the signaling and mechanical functions of the dystrophin–glycoprotein complex. J. Cell Sci. 119, 1537–1546 (2006)

    Article  CAS  Google Scholar 

  14. Thomas, G. D., Shaul, P. W., Yuhanna, I. S., Froehner, S. C. & Adams, M. E. Vasomodulation by skeletal muscle-derived nitric oxide requires α-syntrophin-mediated sarcolemmal localization of neuronal nitric oxide synthase. Circ. Res. 92, 554–560 (2003)

    Article  CAS  Google Scholar 

  15. Chao, D. S., Silvagno, F. & Bredt, D. S. Muscular dystrophy in mdx mice despite lack of neuronal nitric oxide synthase. J. Neurochem. 71, 784–789 (1998)

    Article  CAS  Google Scholar 

  16. Crosbie, R. H. et al. mdx muscle pathology is independent of nNOS perturbation. Hum. Mol. Genet. 7, 823–829 (1998)

    Article  CAS  Google Scholar 

  17. 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 

  18. 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  ADS  Google Scholar 

  19. Huang, P. L. et al. Hypertension in mice lacking the gene for endothelial nitric oxide synthase. Nature 377, 239–242 (1995)

    Article  ADS  CAS  Google Scholar 

  20. Thomas, G. D. et al. Impaired metabolic modulation of α-adrenergic vasoconstriction in dystrophin-deficient skeletal muscle. Proc. Natl Acad. Sci. USA 95, 15090–15095 (1998)

    Article  ADS  CAS  Google Scholar 

  21. Bloom, T. J. Age-related alterations in cyclic nucleotide phosphodiesterase activity in dystrophic mouse leg muscle. Can. J. Physiol. Pharmacol. 83, 1055–1060 (2005)

    Article  CAS  Google Scholar 

  22. Bloom, T. J. Cyclic nucleotide phosphodiesterase isozymes expressed in mouse skeletal muscle. Can. J. Physiol. Pharmacol. 80, 1132–1135 (2002)

    Article  CAS  Google Scholar 

  23. Kass, D. A., Champion, H. C. & Beavo, J. A. Phosphodiesterase type 5: expanding roles in cardiovascular regulation. Circ. Res. 101, 1084–1095 (2007)

    Article  CAS  Google Scholar 

  24. Persson, J., Ekelund, U. & Grande, P. O. Endogenous nitric oxide reduces microvascular permeability and tissue oedema during exercise in cat skeletal muscle. J. Vasc. Res. 40, 538–546 (2003)

    Article  Google Scholar 

  25. Marden, F. A., Connolly, A. M., Siegel, M. J. & Rubin, D. A. Compositional analysis of muscle in boys with Duchenne muscular dystrophy using MR imaging. Skeletal Radiol. 34, 140–148 (2005)

    Article  Google Scholar 

Download references

Acknowledgements

We thank M. Anderson and M. Henry for comments, and M. M. Kilburg, K. Uppal, B. J. Steinmann and S. Watkins and members of the Campbell laboratory for scientific contributions. This work was supported in part by a Paul D. Wellstone Muscular Dystrophy Cooperative Research Center Grant. Y.M.K. was supported by grants from the University of Iowa Cardiovascular Interdisciplinary Research/ National Research Service Award (NRSA) Fellowship, from an individual NRSA Fellowship from the National Institute of Arthritis and Musculoskeletal and Skin Diseases, from the National Institutes of Health (NIH), and from a Senator Paul D. Wellstone Fellowship. E.P.R. was supported by a Muscular Dystrophy Association Development Grant. R.M.W. was supported by the NIH. K.P.C. is an investigator of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Kevin P. Campbell.

Supplementary information

Supplementary Information

This file contains Supplementary Additional Methods, Supplementary Figures S1-S9 with Legends, Supplementary Table 1, Supplementary Methods and Supplementary References (PDF 1723 kb)

Movie file S1a

Movie file S1a shows Pre-exercise activity of a C57BL/10 wild-type mouse (MOV 6787 kb)

Movie file S1b

Movie file S1b shows Pre-exercise activity of an mdx mouse (MOV 6763 kb)

Movie file S1c

Movie file S1c shows Pre-exercise activity of a C57BL/6 wild-type mouse (MOV 6729 kb)

Movie file 1d

Movie file 1d shows Pre-exercise activity of an Sgca-null mouse (MOV 6831 kb)

Movie file 2a

Movie file 2a shows Post-exercise activity of a C57BL/10 wild-type mouse. (MOV 6728 kb)

Movie file 2b

Movie file 2b shows Post-exercise activity of an mdx mouse (MOV 6765 kb)

Movie file 2c

Movie file 2c shows Post-exercise activity of a C57BL/6 mouse wild-type mouse (MOV 6777 kb)

Movie file 2d

Movie file 2d shows Post-exercise activity of an Sgca-null mouse (MOV 6947 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kobayashi, Y., Rader, E., Crawford, R. et al. Sarcolemma-localized nNOS is required to maintain activity after mild exercise. Nature 456, 511–515 (2008). https://doi.org/10.1038/nature07414

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature07414

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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