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:

Imbalance between pSmad3 and Notch induces CDK inhibitors in old muscle stem cells

A Corrigendum to this article was published on 27 July 2016

This article has been updated

Abstract

Adult skeletal muscle robustly regenerates throughout an organism’s life, but as the muscle ages, its ability to repair diminishes and eventually fails1,2. Previous work suggests that the regenerative potential of muscle stem cells (satellite cells) is not triggered in the old muscle because of a decline in Notch activation, and that it can be rejuvenated by forced local activation of Notch3. Here we report that, in addition to the loss of Notch activation, old muscle produces excessive transforming growth factor (TGF)-β (but not myostatin), which induces unusually high levels of TGF-β pSmad3 in resident satellite cells and interferes with their regenerative capacity. Importantly, endogenous Notch and pSmad3 antagonize each other in the control of satellite-cell proliferation, such that activation of Notch blocks the TGF-β-dependent upregulation of the cyclin-dependent kinase (CDK) inhibitors p15, p16, p21 and p27, whereas inhibition of Notch induces them. Furthermore, in muscle stem cells, Notch activity determines the binding of pSmad3 to the promoters of these negative regulators of cell-cycle progression. Attenuation of TGF-β/pSmad3 in old, injured muscle restores regeneration to satellite cells in vivo. Thus a balance between endogenous pSmad3 and active Notch controls the regenerative competence of muscle stem cells, and deregulation of this balance in the old muscle microniche interferes with regeneration.

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: TGF-β/pSmad3, but not myostatin, increases in old skeletal muscle.
Figure 2: Notch removes pSmad3 from the 5′ regulatory regions of CDK inhibitors.
Figure 3: Inhibition of endogenous Notch upregulates CDK inhibitors.
Figure 4: Smad3 shRNA rescues responses of satellite cells in old niche in vivo.

Similar content being viewed by others

Change history

  • 27 July 2016

    Nature 454, 528–532 (2008); doi:10.1038/nature07034 In Fig. 1a of this Letter, the immunofluorescence images for myostatin and follistatin are inaccurate owing to many versions of this figure in multiple revisions of our manuscript. Specifically, the immunofluorescence panels representing anti-myostatin and anti-follistatin staining were duplicated.

References

  1. Grounds, M. D. Age-associated changes in the response of skeletal muscle cells to exercise and regeneration. Ann. NY Acad. Sci. 854, 78–91 (1998)

    Article  ADS  CAS  Google Scholar 

  2. Renault, V., Thornell, L. E., Eriksson, P. O., Butler-Browne, G. & Mouly, V. Regenerative potential of human skeletal muscle during aging. Aging Cell 1, 132–139 (2002)

    Article  CAS  Google Scholar 

  3. Conboy, I. M., Conboy, M. J., Smythe, G. M. & Rando, T. A. Notch-mediated restoration of regenerative potential to aged muscle. Science 302, 1575–1577 (2003)

    Article  ADS  CAS  Google Scholar 

  4. Wagers, A. J. & Conboy, I. M. Cellular and molecular signatures of muscle regeneration: current concepts and controversies in adult myogenesis. Cell 122, 659–667 (2005)

    Article  CAS  Google Scholar 

  5. Collins, C. A. & Partridge, T. A. Self-renewal of the adult skeletal muscle satellite cell. Cell Cycle 4, 1338–1341 (2005)

    Article  CAS  Google Scholar 

  6. Morgan, J. E. et al. Myogenic cell proliferation and generation of a reversible tumorigenic phenotype are triggered by preirradiation of the recipient site. J. Cell Biol. 157, 693–702 (2002)

    Article  CAS  Google Scholar 

  7. Schultz, E. & Lipton, B. H. Skeletal muscle satellite cells: changes in proliferation potential as a function of age. Mech. Ageing Dev. 20, 377–383 (1982)

    Article  CAS  Google Scholar 

  8. Brack, A. S. et al. Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science 317, 807–810 (2007)

    Article  ADS  CAS  Google Scholar 

  9. Conboy, I. M. et al. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature 433, 760–764 (2005)

    Article  ADS  CAS  Google Scholar 

  10. Li, L. & Xie, T. Stem cell niche: structure and function. Annu. Rev. Cell Dev. Biol. 21, 605–631 (2005)

    Article  CAS  Google Scholar 

  11. Carlson, M. E. & Conboy, I. M. Loss of stem cell regenerative capacity within aged niches. Aging Cell (2007)

  12. Massague, J. TGF-beta signal transduction. Annu. Rev. Biochem. 67, 753–791 (1998)

    Article  CAS  Google Scholar 

  13. Massague, J. & Chen, Y. G. Controlling TGF-beta signaling. Genes Dev. 14, 627–644 (2000)

    Article  CAS  Google Scholar 

  14. Derynck, R. & Zhang, Y. E. Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature 425, 577–584 (2003)

    Article  ADS  CAS  Google Scholar 

  15. Feng, X. H. & Derynck, R. Specificity and versatility in tgf-beta signaling through Smads. Annu. Rev. Cell Dev. Biol. 21, 659–693 (2005)

    Article  CAS  Google Scholar 

  16. Natarajan, E. et al. A keratinocyte hypermotility/growth-arrest response involving laminin 5 and p16INK4A activated in wound healing and senescence. Am. J. Pathol. 168, 1821–1837 (2006)

    Article  CAS  Google Scholar 

  17. Ito, T., Sawada, R., Fujiwara, Y., Seyama, Y. & Tsuchiya, T. FGF-2 suppresses cellular senescence of human mesenchymal stem cells by down-regulation of TGF-beta2. Biochem. Biophys. Res. Commun. 359, 108–114 (2007)

    Article  CAS  Google Scholar 

  18. Untergasser, G. et al. Profiling molecular targets of TGF-beta1 in prostate fibroblast-to-myofibroblast transdifferentiation. Mech. Ageing Dev. 126, 59–69 (2005)

    Article  CAS  Google Scholar 

  19. Olson, N. E., Kozlowski, J. & Reidy, M. A. Proliferation of intimal smooth muscle cells. Attenuation of basic fibroblast growth factor 2-stimulated proliferation is associated with increased expression of cell cycle inhibitors. J. Biol. Chem. 275, 11270–11277 (2000)

    Article  CAS  Google Scholar 

  20. Joulia, D. et al. Mechanisms involved in the inhibition of myoblast proliferation and differentiation by myostatin. Exp. Cell Res. 286, 263–275 (2003)

    Article  CAS  Google Scholar 

  21. Lin, J. et al. P27 knockout mice: reduced myostatin in muscle and altered adipogenesis. Biochem. Biophys. Res. Commun. 300, 938–942 (2003)

    Article  CAS  Google Scholar 

  22. Rao, P. & Kadesch, T. The intracellular form of notch blocks transforming growth factor beta-mediated growth arrest in Mv1Lu epithelial cells. Mol. Cell. Biol. 23, 6694–6701 (2003)

    Article  CAS  Google Scholar 

  23. Janzen, V. et al. Stem-cell ageing modified by the cyclin-dependent kinase inhibitor p16INK4a. Nature 443, 421–426 (2006)

    Article  ADS  CAS  Google Scholar 

  24. Molofsky, A. V. et al. Increasing p16INK4a expression decreases forebrain progenitors and neurogenesis during ageing. Nature 443, 448–452 (2006)

    Article  ADS  CAS  Google Scholar 

  25. Krishnamurthy, J. et al. p16INK4a induces an age-dependent decline in islet regenerative potential. Nature 443, 453–457 (2006)

    Article  ADS  CAS  Google Scholar 

  26. Stepanova, L. & Sorrentino, B. P. A limited role for p16Ink4a and p19Arf in the loss of hematopoietic stem cells during proliferative stress. Blood 106, 827–832 (2005)

    Article  CAS  Google Scholar 

  27. Husmann, I., Soulet, L., Gautron, J., Martelly, I. & Barritault, D. Growth factors in skeletal muscle regeneration. Cytokine Growth Factor Rev. 7, 249–258 (1996)

    Article  CAS  Google Scholar 

  28. McPherron, A. C., Lawler, A. M. & Lee, S. J. Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. Nature 387, 83–90 (1997)

    Article  ADS  CAS  Google Scholar 

  29. Barcellos-Hoff, M. H. Latency and activation in the control of TGF-beta. J. Mammary Gland Biol. Neoplasia 1, 353–363 (1996)

    Article  CAS  Google Scholar 

  30. Svoboda, P. Off-targeting and other non-specific effects of RNAi experiments in mammalian cells. Curr. Opin. Mol. Ther. 3, 248–257 (2007)

    Google Scholar 

Download references

Acknowledgements

We thank R. Derynck and M. Conboy for discussions. This work was supported by National Institutes of Health (NIH) R01 (AG027252), NIH R21 (AG27892) and Ellison’s Medical Foundation grants to I.M.C., and a Pre-doctoral Training Fellowship from the California Institute for Regenerative Medicine training grant to M.E.C.

Author Contributions M.E.C. performed all experiments, analysed the data and contributed to the writing of the manuscript; M.H. performed preliminary experiments for Fig. 1a, b, d and Supplementary Figs 4 and 11; and I.M.C. designed the study, participated in experiments, interpreted the data and wrote the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Irina M. Conboy.

Supplementary information

Supplementary Information

The file contains Supplementary Notes and Supplementary Figures 1-12 with Legends. (PDF 1221 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Carlson, M., Hsu, M. & Conboy, I. Imbalance between pSmad3 and Notch induces CDK inhibitors in old muscle stem cells. Nature 454, 528–532 (2008). https://doi.org/10.1038/nature07034

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature07034

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