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:

Rejuvenation of the muscle stem cell population restores strength to injured aged muscles

Abstract

The elderly often suffer from progressive muscle weakness and regenerative failure. We demonstrate that muscle regeneration is impaired with aging owing in part to a cell-autonomous functional decline in skeletal muscle stem cells (MuSCs). Two-thirds of MuSCs from aged mice are intrinsically defective relative to MuSCs from young mice, with reduced capacity to repair myofibers and repopulate the stem cell reservoir in vivo following transplantation. This deficiency is correlated with a higher incidence of cells that express senescence markers and is due to elevated activity of the p38α and p38β mitogen-activated kinase pathway. We show that these limitations cannot be overcome by transplantation into the microenvironment of young recipient muscles. In contrast, subjecting the MuSC population from aged mice to transient inhibition of p38α and p38β in conjunction with culture on soft hydrogel substrates rapidly expands the residual functional MuSC population from aged mice, rejuvenating its potential for regeneration and serial transplantation as well as strengthening of damaged muscles of aged mice. These findings reveal a synergy between biophysical and biochemical cues that provides a paradigm for a localized autologous muscle stem cell therapy for the elderly.

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: MuSCs from aged mice have diminished regenerative and self-renewal capacity, revealing an inherent stem cell defect.
Figure 2: MuSCs from aged mice are characterized by a senescence phenotype and elevated p38α/β MAPK signaling.
Figure 3: p38α/β MAPK inhibition induces proliferation and augments stem cell gene expression in MuSCs from aged mice grown in soft hydrogel cultures.
Figure 4: p38α/β inhibition and soft hydrogel substrate synergize to increase the total yield of functional aged stem cells.
Figure 5: Ex vivo–expanded progeny of MuSCs from aged mice reconstitute the stem cell reserve in vivo and can be serially transplanted.
Figure 6: Muscle strength restored in injured aged mice by transplantation of ex vivo–expanded progeny of MuSCs from aged mice.

Similar content being viewed by others

References

  1. Ryall, J.G., Schertzer, J.D. & Lynch, G.S. Cellular and molecular mechanisms underlying age-related skeletal muscle wasting and weakness. Biogerontology 9, 213–228 (2008).

    CAS  PubMed  Google Scholar 

  2. 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).

    CAS  PubMed  Google Scholar 

  3. Benny Klimek, M.E. et al. Acute inhibition of myostatin-family proteins preserves skeletal muscle in mouse models of cancer cachexia. Biochem. Biophys. Res. Commun. 391, 1548–1554 (2010).

    CAS  PubMed  Google Scholar 

  4. Di Marco, S. et al. The translation inhibitor pateamine A prevents cachexia-induced muscle wasting in mice. Nat. Commun. 3, 896 (2012).

    PubMed  Google Scholar 

  5. Sandri, M. et al. Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell 117, 399–412 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Stitt, T.N. et al. The IGF-1/PI3K/Akt pathway prevents expression of muscle atrophy-induced ubiquitin ligases by inhibiting FOXO transcription factors. Mol. Cell 14, 395–403 (2004).

    CAS  PubMed  Google Scholar 

  7. Glass, D. & Roubenoff, R. Recent advances in the biology and therapy of muscle wasting. Ann. NY Acad. Sci. 1211, 25–36 (2010).

    PubMed  Google Scholar 

  8. Sakuma, K. & Yamaguchi, A. Molecular mechanisms in aging and current strategies to counteract sarcopenia. Curr. Aging Sci. 3, 90–101 (2010).

    CAS  PubMed  Google Scholar 

  9. Janssen, I., Shepard, D.S., Katzmarzyk, P.T. & Roubenoff, R. The healthcare costs of sarcopenia in the United States. J. Am. Geriatr. Soc. 52, 80–85 (2004).

    PubMed  Google Scholar 

  10. Mauro, A. Satellite cell of skeletal muscle fibers. J. Biophys. Biochem. Cytol. 9, 493–495 (1961).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Sacco, A., Doyonnas, R., Kraft, P., Vitorovic, S. & Blau, H.M. Self-renewal and expansion of single transplanted muscle stem cells. Nature 456, 502–506 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Brack, A.S. & Rando, T.A. Tissue-specific stem cells: lessons from the skeletal muscle satellite cell. Cell Stem Cell 10, 504–514 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Brack, A.S. & Rando, T.A. Intrinsic changes and extrinsic influences of myogenic stem cell function during aging. Stem Cell Rev. 3, 226–237 (2007).

    CAS  PubMed  Google Scholar 

  14. Gopinath, S.D. & Rando, T.A. Stem cell review series: aging of the skeletal muscle stem cell niche. Aging Cell 7, 590–598 (2008).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  16. 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).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  18. Collins, C.A., Zammit, P.S., Ruiz, A.P., Morgan, J.E. & Partridge, T.A. A population of myogenic stem cells that survives skeletal muscle aging. Stem Cells 25, 885–894 (2007).

    Article  CAS  PubMed  Google Scholar 

  19. Carlson, M.E., Hsu, M. & Conboy, I.M. Imbalance between pSmad3 and Notch induces CDK inhibitors in old muscle stem cells. Nature 454, 528–532 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Chakkalakal, J.V., Jones, K.M., Basson, M.A. & Brack, A.S. The aged niche disrupts muscle stem cell quiescence. Nature 490, 355–360 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Cerletti, M. et al. Highly efficient, functional engraftment of skeletal muscle stem cells in dystrophic muscles. Cell 134, 37–47 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Kuang, S., Kuroda, K., Le Grand, F. & Rudnicki, M.A. Asymmetric self-renewal and commitment of satellite stem cells in muscle. Cell 129, 999–1010 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Montarras, D. et al. Direct isolation of satellite cells for skeletal muscle regeneration. Science 309, 2064–2067 (2005).

    CAS  PubMed  Google Scholar 

  24. Cornelison, D.D. et al. Essential and separable roles for syndecan-3 and syndecan-4 in skeletal muscle development and regeneration. Genes Dev. 18, 2231–2236 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Bosnakovski, D. et al. Prospective isolation of skeletal muscle stem cells with a Pax7 reporter. Stem Cells 26, 3194–3204 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Lefkovits, I. & Waldmann, H. Limiting dilution analysis of cells of the immune system (Oxford University Press, Oxford, 1999).

  27. Rocheteau, P., Gayraud-Morel, B., Siegl-Cachedenier, I., Blasco, M.A. & Tajbakhsh, S. A subpopulation of adult skeletal muscle stem cells retains all template DNA strands after cell division. Cell 148, 112–125 (2012).

    CAS  PubMed  Google Scholar 

  28. Darabi, R. et al. Assessment of the myogenic stem cell compartment following transplantation of Pax3/Pax7-induced embryonic stem cell–derived progenitors. Stem Cells 29, 777–790 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Rossi, D.J., Jamieson, C.H. & Weissman, I.L. Stems cells and the pathways to aging and cancer. Cell 132, 681–696 (2008).

    CAS  PubMed  Google Scholar 

  30. Seale, P. et al. Pax7 is required for the specification of myogenic satellite cells. Cell 102, 777–786 (2000).

    CAS  PubMed  Google Scholar 

  31. Kang, J.S. & Krauss, R.S. Muscle stem cells in developmental and regenerative myogenesis. Curr. Opin. Clin. Nutr. Metab. Care 13, 243–248 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Gilbert, P.M. et al. Substrate elasticity regulates skeletal muscle stem cell self-renewal in culture. Science 329, 1078–1081 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Cosgrove, B.D., Sacco, A., Gilbert, P.M. & Blau, H.M. A home away from home: challenges and opportunities in engineering in vitro muscle satellite cell niches. Differentiation 78, 185–194 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Shefer, G., Van de Mark, D.P., Richardson, J.B. & Yablonka-Reuveni, Z. Satellite-cell pool size does matter: defining the myogenic potency of aging skeletal muscle. Dev. Biol. 294, 50–66 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. 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).

    CAS  PubMed  Google Scholar 

  36. Mouly, V. et al. The mitotic clock in skeletal muscle regeneration, disease and cell mediated gene therapy. Acta Physiol. Scand. 184, 3–15 (2005).

    CAS  PubMed  Google Scholar 

  37. García-Prat, L., Sousa-Victor, P. & Munoz-Canoves, P. Functional dysregulation of stem cells during aging: a focus on skeletal muscle stem cells. FEBS J. 280, 4051–4062 (2013).

    PubMed  Google Scholar 

  38. Beccafico, S. et al. Human muscle satellite cells show age-related differential expression of S100B protein and RAGE. Age (Dordr.) 33, 523–541 (2011).

    CAS  Google Scholar 

  39. Wong, E.S. et al. p38MAPK controls expression of multiple cell cycle inhibitors and islet proliferation with advancing age. Dev. Cell 17, 142–149 (2009).

    CAS  PubMed  Google Scholar 

  40. Ito, K. et al. Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells. Nat. Med. 12, 446–451 (2006).

    CAS  PubMed  Google Scholar 

  41. Iwasa, H., Han, J. & Ishikawa, F. Mitogen-activated protein kinase p38 defines the common senescence-signalling pathway. Genes Cells 8, 131–144 (2003).

    CAS  PubMed  Google Scholar 

  42. Bain, J. et al. The selectivity of protein kinase inhibitors: a further update. Biochem. J. 408, 297–315 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Jones, N.C. et al. The p38α/β MAPK functions as a molecular switch to activate the quiescent satellite cell. J. Cell Biol. 169, 105–116 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Troy, A. et al. Coordination of satellite cell activation and self-renewal by Par-complex-dependent asymmetric activation of p38α/β MAPK. Cell Stem Cell 11, 541–553 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Tajbakhsh, S. et al. Gene targeting the myf-5 locus with nlacZ reveals expression of this myogenic factor in mature skeletal muscle fibres as well as early embryonic muscle. Dev. Dyn. 206, 291–300 (1996).

    CAS  PubMed  Google Scholar 

  46. Sacco, A. et al. Short telomeres and stem cell exhaustion model Duchenne muscular dystrophy in mdx/mTR mice. Cell 143, 1059–1071 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Llewellyn, M.E., Thompson, K.R., Deisseroth, K. & Delp, S.L. Orderly recruitment of motor units under optical control in vivo. Nat. Med. 16, 1161–1165 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Thompson, L.V. Effects of age and training on skeletal muscle physiology and performance. Phys. Ther. 74, 71–81 (1994).

    CAS  PubMed  Google Scholar 

  49. Arpke, R.W. et al. A new immuno-, dystrophin-deficient model, the NSG-mdx4Cv mouse, provides evidence for functional improvement following allogeneic satellite cell transplantation. Stem Cells 31, 1611–1620 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Lluís, F., Perdiguero, E., Nebreda, A.R. & Munoz-Canoves, P. Regulation of skeletal muscle gene expression by p38 MAP kinases. Trends Cell Biol. 16, 36–44 (2006).

    PubMed  Google Scholar 

  51. Palacios, D. et al. TNF/p38α/polycomb signaling to Pax7 locus in satellite cells links inflammation to the epigenetic control of muscle regeneration. Cell Stem Cell 7, 455–469 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Brien, P., Pugazhendhi, D., Woodhouse, S., Oxley, D. & Pell, J.M. p38α MAPK regulates adult muscle stem cell fate by restricting progenitor proliferation during postnatal growth and repair. Stem Cells 31, 1597–1610 (2013).

    CAS  PubMed  Google Scholar 

  53. Tivey, H.S. et al. Small molecule inhibition of p38 MAP kinase extends the replicative life span of human ATR-Seckel syndrome fibroblasts. J. Gerontol. A Biol. Sci. Med. Sci. 68, 1001–1009 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Zou, J. et al. Inhibition of p38 MAPK activity promotes ex vivo expansion of human cord blood hematopoietic stem cells. Ann. Hematol. 91, 813–823 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Parham, D.M. & Ellison, D.A. Rhabdomyosarcomas in adults and children: an update. Arch. Pathol. Lab. Med. 130, 1454–1465 (2006).

    PubMed  Google Scholar 

  56. Page, T.H., Brown, A., Timms, E.M., Foxwell, B.M. & Ray, K.P. Inhibitors of p38 suppress cytokine production in rheumatoid arthritis synovial membranes: does variable inhibition of interleukin-6 production limit effectiveness in vivo? Arthritis Rheum. 62, 3221–3231 (2010).

    CAS  PubMed  Google Scholar 

  57. Engler, A.J., Sen, S., Sweeney, H.L. & Discher, D.E. Matrix elasticity directs stem cell lineage specification. Cell 126, 677–689 (2006).

    CAS  PubMed  Google Scholar 

  58. Pajcini, K.V., Corbel, S.Y., Sage, J., Pomerantz, J.H. & Blau, H.M. Transient inactivation of Rb and ARF yields regenerative cells from postmitotic mammalian muscle. Cell Stem Cell 7, 198–213 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Le Grand, F., Jones, A.E., Seale, V., Scime, A. & Rudnicki, M.A. Wnt7a activates the planar cell polarity pathway to drive the symmetric expansion of satellite stem cells. Cell Stem Cell 4, 535–547 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Westerman, K.A., Ao, Z.J., Cohen, E.A. & Leboulch, P. Design of a trans protease lentiviral packaging system that produces high titer virus. Retrovirology 4, 96 (2007).

    PubMed  PubMed Central  Google Scholar 

  61. Sacks, R.D. & Roy, R.R. Architecture of the hindlimb muscles of cats: functional significance. J. Morphol. 173, 185–195 (1982).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank K. Koleckar, P. Kraft, N. Nguyen, A. Thayer, C. Marceau, K. Magnusson and A. Ho for technical assistance; K. Havenstrite and R. Haynes for polymer synthesis; and the Stanford Center for Innovation in In-Vivo Imaging (SCI3), the Stanford Shared FACS Facility (SSFF) and IBM Almaden for technical support. This work was funded by US National Institutes of Health (NIH) training grant R25CA118681 and grant K99AG042491 (B.D.C.); NIH training grant T32CA009151 and grant K99AR061465 and California Institute for Regenerative Medicine training grant TG2-01159 (P.M.G.); and NIH grants U01HL100397, U01HL099997, R01AG020961, R01HL096113 and R01AG009521, an IBM Faculty Award, California Institute for Regenerative Medicine grants RT1-01001 and TR3-05501 and the Baxter Foundation (H.M.B.).

Author information

Authors and Affiliations

Authors

Contributions

B.D.C., P.M.G. and E.P. designed and performed experiments, analyzed data and wrote the manuscript. F.M., S.P.L., S.Y.C. and M.E.L. developed methods, performed experiments and analyzed data. S.L.D. developed methods and wrote the manuscript. H.M.B. designed experiments, analyzed data and wrote the manuscript.

Corresponding authors

Correspondence to Penney M Gilbert or Helen M Blau.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 (PDF 4343 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Cite this article

Cosgrove, B., Gilbert, P., Porpiglia, E. et al. Rejuvenation of the muscle stem cell population restores strength to injured aged muscles. Nat Med 20, 255–264 (2014). https://doi.org/10.1038/nm.3464

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3464

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