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.

  • Review Article
  • Published:

Stem cell aging: mechanisms, regulators and therapeutic opportunities

Abstract

Aging tissues experience a progressive decline in homeostatic and regenerative capacities, which has been attributed to degenerative changes in tissue-specific stem cells, stem cell niches and systemic cues that regulate stem cell activity. Understanding the molecular pathways involved in this age-dependent deterioration of stem cell function will be critical for developing new therapies for diseases of aging that target the specific causes of age-related functional decline. Here we explore key molecular pathways that are commonly perturbed as tissues and stem cells age and degenerate. We further consider experimental evidence both suppoxrting and refuting the notion that modulation of these pathways per se can reverse aging phenotypes. Finally, we ask whether stem cell aging establishes an epigenetic 'memory' that is indelibly written or one that can be reset.

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: Common pathways contributing to stem cell loss and dysfunction in the aging process.

Debbie Maizels / Nature Publishing Group

Figure 2: The effects of DNA damage in the aging genome that may affect stem cell function.

Debbie Maizels / Nature Publishing Group

Figure 3: Signaling pathways involved in aging of stem cells.

Debbie Maizels / Nature Publishing Group

Similar content being viewed by others

References

  1. Cheung, T.H. & Rando, T.A. Molecular regulation of stem cell quiescence. Nat. Rev. Mol. Cell Biol. 14, 329–340 (2013).

    CAS  PubMed  Google Scholar 

  2. Takubo, K. et al. Regulation of glycolysis by Pdk functions as a metabolic checkpoint for cell cycle quiescence in hematopoietic stem cells. Cell Stem Cell 12, 49–61 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Harris, J.M. et al. Glucose metabolism impacts the spatiotemporal onset and magnitude of HSC induction in vivo. Blood 121, 2483–2493 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Yu, W.-M. et al. Metabolic regulation by the mitochondrial phosphatase PTPMT1 is required for hematopoietic stem cell differentiation. Cell Stem Cell 12, 62–74 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Pervaiz, S., Taneja, R. & Ghaffari, S. Oxidative stress regulation of stem and progenitor cells. Antioxid. Redox Signal. 11, 2777–2789 (2009).

    CAS  PubMed  Google Scholar 

  6. Harman, D. Free radical theory of aging: dietary implications. Am. J. Clin. Nutr. 25, 839–843 (1972).

    CAS  PubMed  Google Scholar 

  7. Clément, M.V. & Stamenkovic, I. Superoxide anion is a natural inhibitor of FAS-mediated cell death. EMBO J. 15, 216–225 (1996).

    PubMed  PubMed Central  Google Scholar 

  8. Ahmad, K.A., Clement, M.-V.V. & Pervaiz, S. Pro-oxidant activity of low doses of resveratrol inhibits hydrogen peroxide-induced apoptosis. Ann. NY Acad. Sci. 1010, 365–373 (2003).

    CAS  PubMed  Google Scholar 

  9. Stolzing, A., Jones, E., McGonagle, D. & Scutt, A. Age-related changes in human bone marrow-derived mesenchymal stem cells: consequences for cell therapies. Mech. Ageing Dev. 129, 163–173 (2008).

    CAS  PubMed  Google Scholar 

  10. Jang, Y.Y. & Sharkis, S. A low level of reactive oxygen species selects for primitive hematopoietic stem cells that may reside in the low-oxygenic niche. Blood 110, 3056–3063 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Ito, K. et al. Regulation of oxidative stress by ATM is required for self-renewal of haematopoietic stem cells. Nature 431, 997–1002 (2004).

    CAS  PubMed  Google Scholar 

  12. Sahin, E. & Depinho, R.A. Linking functional decline of telomeres, mitochondria and stem cells during ageing. Nature 464, 520–528 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. 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 

  14. Paik, J.H. et al. FoxOs cooperatively regulate diverse pathways governing neural stem cell homeostasis. Cell Stem Cell 5, 540–553 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Tothova, Z. et al. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell 128, 325–339 (2007).

    CAS  PubMed  Google Scholar 

  16. Miyamoto, K. et al. Foxo3a is essential for maintenance of the hematopoietic stem cell pool. Cell Stem Cell 1, 101–112 (2007).

    CAS  PubMed  Google Scholar 

  17. Yalcin, S. et al. Foxo3 is essential for the regulation of ataxia telangiectasia mutated and oxidative stress-mediated homeostasis of hematopoietic stem cells. J. Biol. Chem. 283, 25692–25705 (2008).

    CAS  PubMed  Google Scholar 

  18. Renault, V.M. et al. FoxO3 regulates neural stem cell homeostasis. Cell Stem Cell 5, 527–539 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Zhang, J. et al. PTEN maintains haematopoietic stem cells and acts in lineage choice and leukaemia prevention. Nature 441, 518–522 (2006).

    CAS  PubMed  Google Scholar 

  20. Chen, C. et al. TSC-mTOR maintains quiescence and function of hematopoietic stem cells by repressing mitochondrial biogenesis and reactive oxygen species. J. Exp. Med. 205, 2397–2408 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Juntilla, M.M. et al. AKT1 and AKT2 maintain hematopoietic stem cell function by regulating reactive oxygen species. Blood 115, 4030–4038 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Melov, S. et al. Mitochondrial disease in superoxide dismutase 2 mutant mice. Proc. Natl. Acad. Sci. USA 96, 846–851 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Sakata, H. et al. Interleukin 6-preconditioned neural stem cells reduce ischaemic injury in stroke mice. Brain 135, 3298–3310 (2012).

    PubMed  PubMed Central  Google Scholar 

  24. 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 

  25. Liang, R. & Ghaffari, S. Stem cells, redox signaling, and stem cell aging. Antioxid. Redox Signal. 20, 1902–1916 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Owusu-Ansah, E. & Banerjee, U. Reactive oxygen species prime Drosophila haematopoietic progenitors for differentiation. Nature 461, 537–541 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Yuan, H.-F.F. et al. SIRT1 is required for long-term growth of human mesenchymal stem cells. J. Mol. Med. 90, 389–400 (2012).

    CAS  PubMed  Google Scholar 

  28. Chen, H. et al. Role of SIRT1 and AMPK in mesenchymal stem cells differentiation. Ageing Res. Rev. 13, 55–64 (2014).

    CAS  PubMed  Google Scholar 

  29. Kim, H.-S.S. et al. SIRT3 is a mitochondria-localized tumor suppressor required for maintenance of mitochondrial integrity and metabolism during stress. Cancer Cell 17, 41–52 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Brown, K. et al. SIRT3 reverses aging-associated degeneration. Cell Reports 3, 319–327 (2013).

    CAS  PubMed  Google Scholar 

  31. Sadowska, A.M., Manuel- y-Keenoy, B. & De Backer, W.A. Antioxidant and anti-inflammatory efficacy of NAC in the treatment of COPD: discordant in vitro and in vivo dose-effects: a review. Pulm. Pharmacol. Ther. 20, 9–22 (2007).

    CAS  PubMed  Google Scholar 

  32. Abe, M., Takiguchi, Y., Ichimaru, S., Tsuchiya, K. & Wada, K. Comparison of the protective effect of N-acetylcysteine by different treatments on rat myocardial ischemia-reperfusion injury. J. Pharmacol. Sci. 106, 571–577 (2008).

    CAS  PubMed  Google Scholar 

  33. Kondratov, R.V., Vykhovanets, O., Kondratova, A.A. & Antoch, M.P. Antioxidant N-acetyl-L-cysteine ameliorates symptoms of premature aging associated with the deficiency of the circadian protein BMAL1. Aging 1, 979–987 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Drowley, L. et al. Cellular antioxidant levels influence muscle stem cell therapy. Mol. Ther. 18, 1865–1873 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Kolosova, N.G., Stefanova, N.A Muraleva, N.A. & Skulachev, V.P. The mitochondria-targeted antioxidant SkQ1 but not N-acetylcysteine reverses aging-related biomarkers in rats. Aging 4, 689–694 (2012).

    Google Scholar 

  36. Rossi, D.J. et al. Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age. Nature 447, 725–729 (2007).

    CAS  PubMed  Google Scholar 

  37. Rübe, C.E. et al. Accumulation of DNA damage in hematopoietic stem and progenitor cells during human aging. PLoS ONE 6, e17487 (2011).

    PubMed  PubMed Central  Google Scholar 

  38. Sinha, M. et al. Restoring systemic GDF11 levels reverses age-related dysfunction in mouse skeletal muscle. Science 344, 649–652 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Rogakou, E.P., Pilch, D.R., Orr, A.H., Ivanova, V.S. & Bonner, W.M. DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139. J. Biol. Chem. 273, 5858–5868 (1998).

    CAS  PubMed  Google Scholar 

  40. Beerman, I., Seita, J., Inlay, M.A., Weissman, I.L. & Rossi, D.J. Quiescent hematopoietic stem cells accumulate DNA damage during aging that is repaired upon entry into cell cycle. Cell Stem Cell 15, 37–50 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Flores, I. et al. The longest telomeres: a general signature of adult stem cell compartments. Genes Dev. 22, 654–667 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Larsen, B.D. et al. Caspase 3/caspase-activated DNase promote cell differentiation by inducing DNA strand breaks. Proc. Natl. Acad. Sci. USA 107, 4230–4235 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Charville, G.W. & Rando, T.A. Stem cell ageing and non-random chromosome segregation. Phil. Trans. R. Soc. Lond. B 366, 85–93 (2011).

    CAS  Google Scholar 

  44. Ciccia, A. & Elledge, S. The DNA damage response: making it safe to play with knives. Mol. Cell 40, 179–204 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Fortini, P., Ferretti, C. & Dogliotti, E. The response to DNA damage during differentiation: pathways and consequences. Mutat. Res. 743–744, 160–168 (2013).

    PubMed  Google Scholar 

  46. Behrens, A., van Deursen, J.M., Rudolph, K.L. & Schumacher, B. Impact of genomic damage and ageing on stem cell function. Nat. Cell Biol. 16, 201–207 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Moskalev, A.A. et al. The role of DNA damage and repair in aging through the prism of Koch-like criteria. Ageing Res. Rev. 12, 661–684 (2013).

    CAS  PubMed  Google Scholar 

  48. Bernardes de Jesus, B. & Blasco, M.A. Telomerase at the intersection of cancer and aging. Trends Genet. 29, 513–520 (2013).

    CAS  PubMed  Google Scholar 

  49. Dupressoir, A., Puech, A. & Heidmann, T. IAP retrotransposons in the mouse liver as reporters of ageing. Biochim. Biophys. Acta 1264, 397–402 (1995).

    PubMed  Google Scholar 

  50. Li, W. et al. Activation of transposable elements during aging and neuronal decline in Drosophila. Nat. Neurosci. 16, 529–531 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Maxwell, P.H., Burhans, W.C. & Curcio, M.J. Retrotransposition is associated with genome instability during chronological aging. Proc. Natl. Acad. Sci. USA 108, 20376–20381 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Wong, K.-K. et al. Telomere dysfunction and Atm deficiency compromises organ homeostasis and accelerates ageing. Nature 421, 643–648 (2003).

    CAS  PubMed  Google Scholar 

  53. Nijnik, A. et al. DNA repair is limiting for haematopoietic stem cells during ageing. Nature 447, 686–690 (2007).

    CAS  PubMed  Google Scholar 

  54. Wilson, A. et al. Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair. Cell 135, 1118–1129 (2008).

    CAS  PubMed  Google Scholar 

  55. Foudi, A. et al. Analysis of histone 2B-GFP retention reveals slowly cycling hematopoietic stem cells. Nat. Biotechnol. 27, 84–90 (2009).

    CAS  PubMed  Google Scholar 

  56. Mohrin, M. et al. Hematopoietic stem cell quiescence promotes error-prone DNA repair and mutagenesis. Cell Stem Cell 7, 174–185 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Fumagalli, M. et al. Telomeric DNA damage is irreparable and causes persistent DNA-damage-response activation. Nat. Cell Biol. 14, 355–365 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Hewitt, G. et al. Telomeres are favoured targets of a persistent DNA damage response in ageing and stress-induced senescence. Nat. Commun. 3, 708 (2012).

    PubMed  Google Scholar 

  59. Salama, R., Sadaie, M., Hoare, M. & Narita, M. Cellular senescence and its effector programs. Genes Dev. 28, 99–114 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Erol, A. Deciphering the intricate regulatory mechanisms for the cellular choice between cell repair, apoptosis or senescence in response to damaging signals. Cell. Signal. 23, 1076–1081 (2011).

    CAS  PubMed  Google Scholar 

  61. Mandal, P.K., Blanpain, C. & Rossi, D.J. DNA damage response in adult stem cells: pathways and consequences. Nat. Rev. Mol. Cell Biol. 12, 198–202 (2011).

    CAS  PubMed  Google Scholar 

  62. Kirkwood, T.B. Understanding the odd science of aging. Cell 120, 437–447 (2005).

    CAS  PubMed  Google Scholar 

  63. Jaskelioff, M. et al. Telomerase reactivation reverses tissue degeneration in aged telomerase-deficient mice. Nature 469, 102–106 (2011).

    CAS  PubMed  Google Scholar 

  64. Canela, A., Martín-Caballero, J., Flores, J.M. & Blasco, M.A. Constitutive expression of tert in thymocytes leads to increased incidence and dissemination of T-cell lymphoma in Lck-Tert mice. Mol. Cell. Biol. 24, 4275–4293 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Artandi, S.E. et al. Constitutive telomerase expression promotes mammary carcinomas in aging mice. Proc. Natl. Acad. Sci. USA 99, 8191–8196 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. González-Suárez, E. et al. Increased epidermal tumors and increased skin wound healing in transgenic mice overexpressing the catalytic subunit of telomerase, mTERT, in basal keratinocytes. EMBO J. 20, 2619–2630 (2001).

    PubMed  PubMed Central  Google Scholar 

  67. Tomás-Loba, A. et al. Telomerase reverse transcriptase delays aging in cancer-resistant mice. Cell 135, 609–622 (2008).

    PubMed  Google Scholar 

  68. Bucciantini, M. et al. Inherent toxicity of aggregates implies a common mechanism for protein misfolding diseases. Nature 416, 507–511 (2002).

    CAS  PubMed  Google Scholar 

  69. Balch, W.E., Morimoto, R.I., Dillin, A. & Kelly, J.W. Adapting proteostasis for disease intervention. Science 319, 916–919 (2008).

    CAS  PubMed  Google Scholar 

  70. Powers, E.T., Morimoto, R.I., Dillin, A., Kelly, J.W. & Balch, W.E. Biological and chemical approaches to diseases of proteostasis deficiency. Annu. Rev. Biochem. 78, 959–991 (2009).

    CAS  PubMed  Google Scholar 

  71. Taylor, R.C. & Dillin, A. Aging as an event of proteostasis collapse. Cold Spring Harb. Perspect. Biol. 3, a004440 (2011).

    PubMed  PubMed Central  Google Scholar 

  72. Moreno-Gonzalez, I. & Soto, C. Misfolded protein aggregates: mechanisms, structures and potential for disease transmission. Semin. Cell Dev. Biol. 22, 482–487 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Morimoto, R.I. & Cuervo, A.M. Protein homeostasis and aging: taking care of proteins from the cradle to the grave. J. Gerontol. A Biol. Sci. Med. Sci. 64A, 167–170 (2009).

    CAS  PubMed Central  Google Scholar 

  74. Rubinsztein, D.C., Mariño, G. & Kroemer, G. Autophagy and aging. Cell 146, 682–695 (2011).

    CAS  PubMed  Google Scholar 

  75. Tomaru, U. et al. Decreased proteasomal activity causes age-related phenotypes and promotes the development of metabolic abnormalities. Am. J. Pathol. 180, 963–972 (2012).

    CAS  PubMed  Google Scholar 

  76. Warr, M.R. et al. FOXO3A directs a protective autophagy program in haematopoietic stem cells. Nature 494, 323–327 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Cook, C. et al. Aging is not associated with proteasome impairment in UPS reporter mice. PLoS ONE 4, e5888 (2009).

    PubMed  PubMed Central  Google Scholar 

  78. Mortensen, M. et al. The autophagy protein Atg7 is essential for hematopoietic stem cell maintenance. J. Exp. Med. 208, 455–467 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Yilmaz, Ö.H. et al. mTORC1 in the Paneth cell niche couples intestinal stem-cell function to calorie intake. Nature 486, 490–495 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Laplante, M. & Sabatini, D.M. mTOR signaling in growth control and disease. Cell 149, 274–293 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Vilchez, D. et al. Increased proteasome activity in human embryonic stem cells is regulated by PSMD11. Nature 489, 304–308 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Murphy, C.T. et al. Genes that act downstream of DAF-16 to influence the lifespan of Caenorhabditis elegans. Nature 424, 277–283 (2003).

    CAS  PubMed  Google Scholar 

  83. Oh, S.W. et al. Identification of direct DAF-16 targets controlling longevity, metabolism and diapause by chromatin immunoprecipitation. Nat. Genet. 38, 251–257 (2006).

    PubMed  Google Scholar 

  84. Demontis, F. & Perrimon, N. FOXO/4E-BP signaling in Drosophila muscles regulates organism-wide proteostasis during aging. Cell 143, 813–825 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Tatar, M., Khazaeli, A.A. & Curtsinger, J.W. Chaperoning extended life. Nature 390, 30 (1997).

    CAS  PubMed  Google Scholar 

  86. Walker, G.A. & Lithgow, G.J. Lifespan extension in C. elegans by a molecular chaperone dependent upon insulin-like signals. Aging Cell 2, 131–139 (2003).

    CAS  PubMed  Google Scholar 

  87. Morley, J.F. & Morimoto, R.I. Regulation of longevity in Caenorhabditis elegans by heat shock factor and molecular chaperones. Mol. Biol. Cell 15, 657–664 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Chen, C., Liu, Y., Liu, Y. & Zheng, P. mTOR regulation and therapeutic rejuvenation of aging hematopoietic stem cells. Sci. Signal. 2, ra75 (2009).

    PubMed  PubMed Central  Google Scholar 

  89. Zhang, C. & Cuervo, A.M. Restoration of chaperone-mediated autophagy in aging liver improves cellular maintenance and hepatic function. Nat. Med. 14, 959–965 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Cummings, C.J. et al. Over-expression of inducible HSP70 chaperone suppresses neuropathology and improves motor function in SCA1 mice. Hum. Mol. Genet. 10, 1511–1518 (2001).

    CAS  PubMed  Google Scholar 

  91. Feng, Y. et al. Heat shock improves Sca-1+ stem cell survival and directs ischemic cardiomyocytes toward a prosurvival phenotype via exosomal transfer: a critical role for HSF1/miR-34a/HSP70 pathway. Stem Cells 32, 462–472 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. McArdle, A., Dillmann, W.H., Mestril, R., Faulkner, J.A. & Jackson, M.J. Overexpression of HSP70 in mouse skeletal muscle protects against muscle damage and age-related muscle dysfunction. FASEB J. 18, 355–357 (2004).

    CAS  PubMed  Google Scholar 

  93. Bratic, A. & Larsson, N.-G.G. The role of mitochondria in aging. J. Clin. Invest. 123, 951–957 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Miquel, J., Economos, A.C., Fleming, J. & Johnson, J.E. Mitochondrial role in cell aging. Exp. Gerontol. 15, 575–591 (1980).

    CAS  PubMed  Google Scholar 

  95. Zheng, W., Khrapko, K., Coller, H.A., Thilly, W.G. & Copeland, W.C. Origins of human mitochondrial point mutations as DNA polymerase gamma-mediated errors. Mutat. Res. 599, 11–20 (2006).

    CAS  PubMed  Google Scholar 

  96. Trifunovic, A. et al. Premature ageing in mice expressing defective mitochondrial DNA polymerase. Nature 429, 417–423 (2004).

    CAS  PubMed  Google Scholar 

  97. Kujoth, G.C. et al. Mitochondrial DNA mutations, oxidative stress, and apoptosis in mammalian aging. Science 309, 481–484 (2005).

    CAS  PubMed  Google Scholar 

  98. Ahlqvist, K.J. et al. Somatic progenitor cell vulnerability to mitochondrial DNA mutagenesis underlies progeroid phenotypes in Polg mutator mice. Cell Metab. 15, 100–109 (2012).

    CAS  PubMed  Google Scholar 

  99. Fox, R.G., Magness, S., Kujoth, G.C., Prolla, T.A. & Maeda, N. Mitochondrial DNA polymerase editing mutation, PolgD257A, disturbs stem-progenitor cell cycling in the small intestine and restricts excess fat absorption. Am. J. Physiol. Gastrointest. Liver Physiol. 302, G914–G924 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Taylor, R.W. et al. Mitochondrial DNA mutations in human colonic crypt stem cells. J. Clin. Invest. 112, 1351–1360 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. McDonald, S.A. et al. Mechanisms of field cancerization in the human stomach: the expansion and spread of mutated gastric stem cells. Gastroenterology 134, 500–510 (2008).

    CAS  PubMed  Google Scholar 

  102. Fellous, T.G. et al. Locating the stem cell niche and tracing hepatocyte lineages in human liver. Hepatology 49, 1655–1663 (2009).

    CAS  PubMed  Google Scholar 

  103. Norddahl, G.L. et al. Accumulating mitochondrial DNA mutations drive premature hematopoietic aging phenotypes distinct from physiological stem cell aging. Cell Stem Cell 8, 499–510 (2011).

    CAS  PubMed  Google Scholar 

  104. Bonawitz, N.D., Chatenay-Lapointe, M., Pan, Y. & Shadel, G.S. Reduced TOR signaling extends chronological life span via increased respiration and upregulation of mitochondrial gene expression. Cell Metab. 5, 265–277 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Holzenberger, M. et al. IGF-1 receptor regulates lifespan and resistance to oxidative stress in mice. Nature 421, 182–187 (2003).

    CAS  PubMed  Google Scholar 

  106. Nakada, D., Saunders, T.L. & Morrison, S.J. Lkb1 regulates cell cycle and energy metabolism in haematopoietic stem cells. Nature 468, 653–658 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Gross, D.N., van den Heuvel, A.P. & Birnbaum, M.J. The role of FoxO in the regulation of metabolism. Oncogene 27, 2320–2336 (2008).

    CAS  PubMed  Google Scholar 

  108. Peserico, A. et al. A novel AMPK-dependent FoxO3A-SIRT3 intramitochondrial complex sensing glucose levels. Cell. Mol. Life Sci. 70, 2015–2029 (2013).

    CAS  PubMed  Google Scholar 

  109. Rera, M. et al. Modulation of longevity and tissue homeostasis by the Drosophila PGC-1 homolog. Cell Metab. 14, 623–634 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Cerletti, M., Jang, Y.C., Finley, L.W., Haigis, M.C. & Wagers, A.J. Short-term calorie restriction enhances skeletal muscle stem cell function. Cell Stem Cell 10, 515–519 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Piper, M.D. & Bartke, A. Diet and aging. Cell Metab. 8, 99–104 (2008).

    CAS  PubMed  Google Scholar 

  112. Lee, J., Duan, W. & Mattson, M.P. Evidence that brain-derived neurotrophic factor is required for basal neurogenesis and mediates, in part, the enhancement of neurogenesis by dietary restriction in the hippocampus of adult mice. J. Neurochem. 82, 1367–1375 (2002).

    CAS  PubMed  Google Scholar 

  113. Narala, S.R. et al. SIRT1 acts as a nutrient-sensitive growth suppressor and its loss is associated with increased AMPK and telomerase activity. Mol. Biol. Cell 19, 1210–1219 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Yang, S.-A.A. et al. FOXO/Fringe is necessary for maintenance of the germline stem cell niche in response to insulin insufficiency. Dev. Biol. 382, 124–135 (2013).

    CAS  PubMed  Google Scholar 

  115. Schulz, T.J. et al. Glucose restriction extends Caenorhabditis elegans life span by inducing mitochondrial respiration and increasing oxidative stress. Cell Metab. 6, 280–293 (2007).

    CAS  PubMed  Google Scholar 

  116. Gredilla, R., Sanz, A., Lopez-Torres, M. & Barja, G. Caloric restriction decreases mitochondrial free radical generation at complex I and lowers oxidative damage to mitochondrial DNA in the rat heart. FASEB J. 15, 1589–1591 (2001).

    CAS  PubMed  Google Scholar 

  117. Sohal, R.S., Agarwal, S., Candas, M., Forster, M.J. & Lal, H. Effect of age and caloric restriction on DNA oxidative damage in different tissues of C57BL/6 mice. Mech. Ageing Dev. 76, 215–224 (1994).

    CAS  PubMed  Google Scholar 

  118. Gomes, A.P. et al. Declining NAD+ induces a pseudohypoxic state disrupting nuclear-mitochondrial communication during aging. Cell 155, 1624–1638 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Hur, J.H. et al. Increased longevity mediated by yeast NDI1 expression in Drosophila intestinal stem and progenitor cells. Aging 5, 662–681 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Sousa-Victor, P. et al. Geriatric muscle stem cells switch reversible quiescence into senescence. Nature 506, 316–321 (2014).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Boyle, M., Wong, C., Rocha, M. & Jones, D.L. Decline in self-renewal factors contributes to aging of the stem cell niche in the Drosophila testis. Cell Stem Cell 1, 470–478 (2007).

    CAS  PubMed  Google Scholar 

  123. Florian, M.C. et al. Cdc42 activity regulates hematopoietic stem cell aging and rejuvenation. Cell Stem Cell 10, 520–530 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Geiger, H., de Haan, G. & Florian, M.C. The ageing haematopoietic stem cell compartment. Nat. Rev. Immunol. 13, 376–389 (2013).

    CAS  PubMed  Google Scholar 

  125. Campisi, J. Cancer, aging and cellular senescence. In Vivo 14, 183–188 (2000).

    CAS  PubMed  Google Scholar 

  126. Campisi, J. Senescent cells, tumor suppression, and organismal aging: good citizens, bad neighbors. Cell 120, 513–522 (2005).

    CAS  PubMed  Google Scholar 

  127. Coppé, J.-P.P., Desprez, P.-Y.Y., Krtolica, A. & Campisi, J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu. Rev. Pathol. 5, 99–118 (2010).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  129. Attema, J.L., Pronk, C.J., Norddahl, G.L., Nygren, J.M. & Bryder, D. Hematopoietic stem cell ageing is uncoupled from p16 INK4A-mediated senescence. Oncogene 28, 2238–2243 (2009).

    CAS  PubMed  Google Scholar 

  130. Cheng, T. et al. Hematopoietic stem cell quiescence maintained by p21cip1/waf1. Science 287, 1804–1808 (2000).

    CAS  PubMed  Google Scholar 

  131. Kippin, T.E., Martens, D.J. & van der Kooy, D. p21 loss compromises the relative quiescence of forebrain stem cell proliferation leading to exhaustion of their proliferation capacity. Genes Dev. 19, 756–767 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. 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 

  133. Hamanaka, R.B. & Chandel, N.S. Mitochondrial reactive oxygen species regulate cellular signaling and dictate biological outcomes. Trends Biochem. Sci. 35, 505–513 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. 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 

  135. Copelan, E.A. Hematopoietic stem-cell transplantation. N. Engl. J. Med. 354, 1813–1826 (2006).

    CAS  PubMed  Google Scholar 

  136. Salani, S. et al. Generation of skeletal muscle cells from embryonic and induced pluripotent stem cells as an in vitro model and for therapy of muscular dystrophies. J. Cell. Mol. Med. 16, 1353–1364 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Chhabra, P. & Brayman, K.L. Stem cell therapy to cure type 1 diabetes: from hype to hope. Stem Cells Transl. Med. 2, 328–336 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Kim, S.U. & de Vellis, J. Stem cell-based cell therapy in neurological diseases: a review. J. Neurosci. Res. 87, 2183–2200 (2009).

    CAS  PubMed  Google Scholar 

  139. 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 

  140. Cosgrove, B.D. et al. Rejuvenation of the muscle stem cell population restores strength to injured aged muscles. Nat. Med. 20, 255–264 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Tabebordbar, M., Wang, E. & Wagers, A.J. Skeletal muscle degenerative diseases and strategies for therapeutic muscle repair. Annu. Rev. Pathol. 8, 441–475 (2013).

    CAS  PubMed  Google Scholar 

  142. Bernet, J.D. et al. p38 MAPK signaling underlies a cell-autonomous loss of stem cell self-renewal in skeletal muscle of aged mice. Nat. Med. 20, 265–271 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Ding, Q. et al. Enhanced efficiency of human pluripotent stem cell genome editing through replacing TALENs with CRISPRs. Cell Stem Cell 12, 393–394 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Ding, Q. et al. A TALEN genome-editing system for generating human stem cell-based disease models. Cell Stem Cell 12, 238–251 (2013).

    CAS  PubMed  Google Scholar 

  145. Xu, Y. et al. Integrating haplotypes and single genetic variability effects of the Pax7 gene on growth traits in two cattle breeds. Genome 56, 9–15 (2013).

    CAS  PubMed  Google Scholar 

  146. Borchin, B., Chen, J. & Barberi, T. Derivation and FACS-mediated purification of PAX3+/PAX7+ skeletal muscle precursors from human pluripotent stem cells. Stem Cell Reports 1, 620–631 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Darabi, R. et al. Human ES- and iPS-derived myogenic progenitors restore DYSTROPHIN and improve contractility upon transplantation in dystrophic mice. Cell Stem Cell 10, 610–619 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Kobayashi, Y. et al. Pre-evaluated safe human iPSC-derived neural stem cells promote functional recovery after spinal cord injury in common marmoset without tumorigenicity. PLoS ONE 7, e52787 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Nakamura, M. & Okano, H. Cell transplantation therapies for spinal cord injury focusing on induced pluripotent stem cells. Cell Res. 23, 70–80 (2013).

    CAS  PubMed  Google Scholar 

  150. Kuhn, H.G., Dickinson-Anson, H. & Gage, F.H. Neurogenesis in the dentate gyrus of the adult rat: age-related decrease of neuronal progenitor proliferation. J. Neurosci. 16, 2027–2033 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Maslov, A.Y., Barone, T.A., Plunkett, R.J. & Pruitt, S.C. Neural stem cell detection, characterization, and age-related changes in the subventricular zone of mice. J. Neurosci. 24, 1726–1733 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Morrison, S.J. & Spradling, A.C. Stem cells and niches: mechanisms that promote stem cell maintenance throughout life. Cell 132, 598–611 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Pan, L. et al. Stem cell aging is controlled both intrinsically and extrinsically in the Drosophila ovary. Cell Stem Cell 1, 458–469 (2007).

    CAS  PubMed  Google Scholar 

  154. Ryu, B.-Y.Y., Orwig, K.E., Oatley, J.M., Avarbock, M.R. & Brinster, R.L. Effects of aging and niche microenvironment on spermatogonial stem cell self-renewal. Stem Cells 24, 1505–1511 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Sato, T. et al. In vitro production of fertile sperm from murine spermatogonial stem cell lines. Nat. Commun. 2, 472 (2011).

    PubMed  Google Scholar 

  156. Sato, T. et al. Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature 469, 415–418 (2011).

    CAS  PubMed  Google Scholar 

  157. Schnoor, M. et al. Production of type VI collagen by human macrophages: a new dimension in macrophage functional heterogeneity. J. Immunol. 180, 5707–5719 (2008).

    CAS  PubMed  Google Scholar 

  158. Murphy, M.M., Lawson, J.A., Mathew, S.J., Hutcheson, D.A. & Kardon, G. Satellite cells, connective tissue fibroblasts and their interactions are crucial for muscle regeneration. Development 138, 3625–3637 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Joe, A.W. et al. Muscle injury activates resident fibro/adipogenic progenitors that facilitate myogenesis. Nat. Cell Biol. 12, 153–163 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. 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 

  161. Carlson, M.E. et al. Relative roles of TGF-b1 and Wnt in the systemic regulation and aging of satellite cell responses. Aging Cell 8, 676–689 (2009).

    CAS  PubMed  Google Scholar 

  162. Franceschi, C. et al. Inflamm-aging: an evolutionary perspective on immunosenescence. Ann. NY Acad. Sci. 908, 244–254 (2000).

    CAS  PubMed  Google Scholar 

  163. de Magalhães, J.P., Curado, J. & Church, G.M. Meta-analysis of age-related gene expression profiles identifies common signatures of aging. Bioinformatics 25, 875–881 (2009).

    PubMed  PubMed Central  Google Scholar 

  164. 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 

  165. Villeda, S.A. et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature 477, 90–94 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Katsimpardi, L. et al. Vascular and neurogenic rejuvenation of the aging mouse brain by young systemic factors. Science 344, 630–634 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  167. Ruckh, J.M. et al. Rejuvenation of regeneration in the aging central nervous system. Cell Stem Cell 10, 96–103 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Loffredo, F.S. et al. Growth differentiation factor 11 is a circulating factor that reverses age-related cardiac hypertrophy. Cell 153, 828–839 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Elabd, C. et al. Oxytocin is an age-specific circulating hormone that is necessary for muscle maintenance and regeneration. Nat. Commun. 5, 4082 (2014).

    CAS  PubMed  Google Scholar 

  170. Baker, D.J. et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 479, 232–236 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  171. Fraga, M.F. & Esteller, M. Epigenetics and aging: the targets and the marks. Trends Genet. 23, 413–418 (2007).

    CAS  PubMed  Google Scholar 

  172. Greer, E.L. et al. Members of the H3K4 trimethylation complex regulate lifespan in a germline-dependent manner in C. elegans. Nature 466, 383–387 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Han, S. & Brunet, A. Histone methylation makes its mark on longevity. Trends Cell Biol. 22, 42–49 (2012).

    PubMed  Google Scholar 

  174. Greer, E.L. et al. Transgenerational epigenetic inheritance of longevity in Caenorhabditis elegans. Nature 479, 365–371 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  175. Peleg, S. et al. Altered histone acetylation is associated with age-dependent memory impairment in mice. Science 328, 753–756 (2010).

    CAS  PubMed  Google Scholar 

  176. Krishnan, V. et al. Histone H4 lysine 16 hypoacetylation is associated with defective DNA repair and premature senescence in Zmpste24-deficient mice. Proc. Natl. Acad. Sci. USA 108, 12325–12330 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  177. Beerman, I. et al. Proliferation-dependent alterations of the DNA methylation landscape underlie hematopoietic stem cell aging. Cell Stem Cell 12, 413–425 (2013).

    CAS  PubMed  Google Scholar 

  178. Sun, D. et al. Epigenomic profiling of young and aged HSCs reveals concerted changes during aging that reinforce self-renewal. Cell Stem Cell 14, 673–688 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Gurdon, J.B. The developmental capacity of nuclei taken from intestinal epithelium cells of feeding tadpoles. J. Embryol. Exp. Morphol. 10, 622–640 (1962).

    CAS  PubMed  Google Scholar 

  180. Lanza, R.P. et al. Extension of cell life-span and telomere length in animals cloned from senescent somatic cells. Science 288, 665–669 (2000).

    CAS  PubMed  Google Scholar 

  181. Abramovich, A., Muradian, K.K. & Fraifeld, V.E. Have we reached the point for in vivo rejuvenation? Rejuvenation Res. 11, 489–492 (2008).

    PubMed  Google Scholar 

  182. Takahashi, K. & Yamanaka, S. Induced pluripotent stem cells in medicine and biology. Development 140, 2457–2461 (2013).

    CAS  PubMed  Google Scholar 

  183. Wahlestedt, M. et al. An epigenetic component of hematopoietic stem cell aging amenable to reprogramming into a young state. Blood 121, 4257–4264 (2013).

    CAS  PubMed  Google Scholar 

  184. Price, N.L. et al. SIRT1 is required for AMPK activation and the beneficial effects of resveratrol on mitochondrial function. Cell Metab. 15, 675–690 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Miller, R.A. Rebuttal to Hasty and Vijg: 'Accelerating aging by mouse reverse genetics: a rational approach to understanding longevity'. Aging Cell 3, 53–54 (2004).

    CAS  PubMed  Google Scholar 

  186. Burtner, C.R. & Kennedy, B.K. Progeria syndromes and ageing: what is the connection? Nat. Rev. Mol. Cell Biol. 11, 567–578 (2010).

    CAS  PubMed  Google Scholar 

  187. Hua, G. et al. Crypt base columnar stem cells in small intestines of mice are radioresistant. Gastroenterology 143, 1266–1276 (2012).

    CAS  PubMed  Google Scholar 

  188. Sotiropoulou, P.A. et al. Bcl-2 and accelerated DNA repair mediates resistance of hair follicle bulge stem cells to DNA-damage-induced cell death. Nat. Cell Biol. 12, 572–582 (2010).

    CAS  PubMed  Google Scholar 

  189. Cheung, H.-H.H. et al. Telomerase protects Werner syndrome lineage-specific stem cells from premature aging. Stem Cell Reports 2, 534–546 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Zhang, J. et al. A human iPSC model of Hutchinson Gilford progeria reveals vascular smooth muscle and mesenchymal stem cell defects. Cell Stem Cell 8, 31–45 (2011).

    CAS  PubMed  Google Scholar 

  191. Mitchell, J.R., Wood, E. & Collins, K. A telomerase component is defective in the human disease dyskeratosis congenita. Nature 402, 551–555 (1999).

    CAS  PubMed  Google Scholar 

  192. Jones, M. et al. Hematopoietic stem cells are acutely sensitive to Acd shelterin gene inactivation. J. Clin. Invest. 124, 353–366 (2014).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank all members of the Wagers laboratory for advice and comments during the preparation of this article. This work was funded in part by US National Cancer Institute grant T32CA-0216 from the Massachusetts General Hospital Department of Pathology (Y.D.L.), by US National Institutes of Health (NIH) grant T32DK007260 (J.O.), and by NIH grants 1R01 AG033053 and 5U01 HL100402 and the Paul F. Glenn Laboratories for the Biological Mechanisms of Aging (A.J.W.). A.J.W. is an Early Career Scientist of the Howard Hughes Medical Institute. Content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH or other funding agencies.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Amy J Wagers.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Oh, J., Lee, Y. & Wagers, A. Stem cell aging: mechanisms, regulators and therapeutic opportunities. Nat Med 20, 870–880 (2014). https://doi.org/10.1038/nm.3651

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3651

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