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.

How stem cells age and why this makes us grow old

Key Points

  • Cancer and ageing appear to represent the failure or success, respectively, of vital tumour-suppressor mechanisms that rely on the activities of telomere shortening and the activation of the cyclin-dependent kinase inhibitor p16INK4a and the tumour suppressor p53.

  • Evidence suggests that these tumour-suppressor molecules exert pro-ageing and anti-cancer functions in discrete compartments of self-renewing cells (tissue-specific stem cells) that are present in adult mammalian tissues.

  • Adult stem cells appear to accumulate DNA damage with ageing as a result of extrinsic exposures, telomere shortening and/or replicative stress associated with homeostatic proliferation.

  • Accumulated DNA damage in stem-cell compartments can be oncogenic, or can engage tumour-suppressor mechanisms such as cellular senescence and apoptosis.

  • Mice with dysfunctional telomeres or increased activity of p53 or p16INK4a demonstrate an accelerated appearance of ageing phenotypes, whereas loss of p16INK4a can attenuate the decline in stem-cell function that is associated with physiological ageing.

  • Loci near the INK4/ARF locus on human chromosome 9p21 are associated with an increased incidence of ageing-associated conditions in humans such as frailty, type 2 diabetes mellitus and coronary artery disease.

Abstract

Recent data suggest that we age, in part, because our self-renewing stem cells grow old as a result of heritable intrinsic events, such as DNA damage, as well as extrinsic forces, such as changes in their supporting niches. Mechanisms that suppress the development of cancer, such as senescence and apoptosis, which rely on telomere shortening and the activities of p53 and p16INK4a, may also induce an unwanted consequence: a decline in the replicative function of certain stem-cell types with advancing age. This decreased regenerative capacity appears to contribute to some aspects of mammalian ageing, with new findings pointing to a 'stem-cell hypothesis' for human age-associated conditions such as frailty, atherosclerosis and type 2 diabetes.

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: How stem cells age.
Figure 2: Proliferation of β-cells with age.
Figure 3: Fates of damaged stem cells.
Figure 4: SNPs and age-related phenotypes at the INK4a/ARF/INK4b locus on human chromosome 9p21.

Similar content being viewed by others

References

  1. Kudlow, B. A., Kennedy, B. K. & Monnat, R. J. Jr. Werner and Hutchinson–Gilford progeria syndromes: mechanistic basis of human progeroid diseases. Nature Rev. Mol. Cell Biol. 8, 394–404 (2007).

    CAS  Google Scholar 

  2. Bryder, D., Rossi, D. J. & Weissman, I. L. Hematopoietic stem cells: the paradigmatic tissue-specific stem cell. Am. J. Pathol. 169, 338–346 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Morrison, S. J. & Kimble, J. Asymmetric and symmetric stem-cell divisions in development and cancer. Nature 441, 1068–1074 (2006).

    CAS  PubMed  Google Scholar 

  4. Sharpless, N. E. & DePinho, R. A. Telomeres, stem cells, senescence, and cancer. J. Clin. Invest. 113, 160–168 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Hodgson, G. S. & Bradley, T. R. In vivo kinetic status of hematopoietic stem and progenitor cells as inferred from labeling with bromodeoxyuridine. Exp. Hematol. 12, 683–687 (1984).

    CAS  PubMed  Google Scholar 

  6. Passegue, E., Wagers, A. J., Giuriato, S., Anderson, W. C. & Weissman, I. L. Global analysis of proliferation and cell cycle gene expression in the regulation of hematopoietic stem and progenitor cell fates. J. Exp. Med. 202, 1599–1611 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Rossi, D. J. et al. Cell intrinsic alterations underlie hematopoietic stem cell aging. Proc. Natl Acad. Sci. USA 102, 9194–9199 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Weissman, I. L. Stem cells: units of development, units of regeneration, and units in evolution. Cell 100, 157–168 (2000).

    CAS  PubMed  Google Scholar 

  9. Reya, T., Morrison, S. J., Clarke, M. F. & Weissman, I. L. Stem cells, cancer, and cancer stem cells. Nature 414, 105–111 (2001).

    CAS  PubMed  Google Scholar 

  10. Campisi, J. Cancer and ageing: rival demons? Nature Rev. Cancer 3, 339–349 (2003).

    CAS  Google Scholar 

  11. Linton, P. J. & Dorshkind, K. Age-related changes in lymphocyte development and function. Nature Immunol. 5, 133–139 (2004).

    CAS  Google Scholar 

  12. Lichtman, M. A. & Rowe, J. M. The relationship of patient age to the pathobiology of the clonal myeloid diseases. Semin. Oncol. 31, 185–197 (2004).

    PubMed  Google Scholar 

  13. Beghe, C., Wilson, A. & Ershler, W. B. Prevalence and outcomes of anemia in geriatrics: a systematic review of the literature. Am. J. Med. 116 (Suppl 7A), 3S–10S (2004).

    PubMed  Google Scholar 

  14. Guralnik, J. M., Eisenstaedt, R. S., Ferrucci, L., Klein, H. G. & Woodman, R. C. Prevalence of anemia in persons 65 years and older in the United States:evidence for a high rate of unexplained anemia. Blood 104, 2263–2268 (2004).

    CAS  PubMed  Google Scholar 

  15. Appelbaum, F. R. et al. Age and acute myeloid leukemia. Blood 107, 3481–3485 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Brunello, A. et al. Adjuvant chemotherapy for elderly patients (> or = 70 years) with early high-risk breast cancer: a retrospective analysis of 260 patients. Ann. Oncol. 16, 1276–1282 (2005).

    CAS  PubMed  Google Scholar 

  17. Lenhoff, S. et al. Impact of age on survival after intensive therapy for multiple myeloma: a population-based study by the Nordic Myeloma Study Group. Br. J. Haematol. 133, 389–396 (2006).

    PubMed  Google Scholar 

  18. Kollman, C. et al. Donor characteristics as risk factors in recipients after transplantation of bone marrow from unrelated donors: the effect of donor age. Blood 98, 2043–2051 (2001).

    CAS  PubMed  Google Scholar 

  19. Ash, R. C. et al. Bone marrow transplantation from related donors other than HLA-identical siblings: effect of T cell depletion. Bone Marrow Transplant. 7, 443–452 (1991).

    CAS  PubMed  Google Scholar 

  20. Castro-Malaspina, H. et al. Unrelated donor marrow transplantation for myelodysplastic syndromes: outcome analysis in 510 transplants facilitated by the National Marrow Donor Program. Blood 99, 1943–1951 (2002).

    CAS  PubMed  Google Scholar 

  21. Buckner, C. D. et al. Marrow harvesting from normal donors. Blood 64, 630–634 (1984).

    CAS  PubMed  Google Scholar 

  22. Yakoub-Agha, I. et al. Allogeneic marrow stem-cell transplantation from human leukocyte antigen-identical siblings versus human leukocyte antigen-allelic-matched unrelated donors (10/10) in patients with standard-risk hematologic malignancy: a prospective study from the French Society of Bone Marrow Transplantation and Cell Therapy. J. Clin. Oncol. 24, 5695–5702 (2006).

    PubMed  Google Scholar 

  23. Morrison, S. J., Wandycz, A. M., Akashi, K., Globerson, A. & Weissman, I. L. The aging of hematopoietic stem cells. Nature Med. 2, 1011–1016 (1996).

    CAS  PubMed  Google Scholar 

  24. Sudo, K., Ema, H., Morita, Y. & Nakauchi, H. Age-associated characteristics of murine hematopoietic stem cells. J. Exp. Med. 192, 1273–1280 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Pearce, D. J., Anjos-Afonso, F., Ridler, C. M., Eddaoudi, A. & Bonnet, D. Age-dependent increase in side population distribution within hematopoiesis: implications for our understanding of the mechanism of aging. Stem Cells 25, 828–835 (2006).

    PubMed  Google Scholar 

  26. de Haan, G., Nijhof, W. & Van Zant, G. Mouse strain-dependent changes in frequency and proliferation of hematopoietic stem cells during aging: correlation between lifespan and cycling activity. Blood 89, 1543–1550 (1997).

    CAS  PubMed  Google Scholar 

  27. de Haan, G. & Van Zant, G. Dynamic changes in mouse hematopoietic stem cell numbers during aging. Blood 93, 3294–3301 (1999).

    CAS  PubMed  Google Scholar 

  28. Harrison, D. E. Mouse erythropoietic stem cell lines function normally 100 months: loss related to number of transplantations. Mech. Ageing Dev. 9, 427–433 (1979).

    CAS  PubMed  Google Scholar 

  29. Chen, J., Astle, C. M. & Harrison, D. E. Genetic regulation of primitive hematopoietic stem cell senescence. Exp. Hematol. 28, 442–450 (2000).

    CAS  PubMed  Google Scholar 

  30. Kamminga, L. M. et al. Impaired hematopoietic stem cell functioning after serial transplantation and during normal aging. Stem Cells 23, 82–92 (2005).

    CAS  PubMed  Google Scholar 

  31. Liang, Y., Van Zant, G. & Szilvassy, S. J. Effects of aging on the homing and engraftment of murine hematopoietic stem and progenitor cells. Blood 106, 1479–1487 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Wang, Y., Schulte, B. A., Larue, A. C., Ogawa, M. & Zhou, D. Total body irradiation selectively induces murine hematopoietic stem cell senescence. Blood 107, 358–366 (2006). An important study showing persistent proliferative defects and p16INK4a expression in HSCs after exposure to DNA-damaging agents.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Meng, A., Wang, Y., Van Zant, G. & Zhou, D. Ionizing radiation and busulfan induce premature senescence in murine bone marrow hematopoietic cells. Cancer Res. 63, 5414–5419 (2003).

    CAS  PubMed  Google Scholar 

  34. 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 

  35. Ito, K. et al. Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells. Nature Med. 12, 446–451 (2006). References 34 and 35 suggest roles for senescence-promoting molecules in HSC lifespan in the setting of impaired ATM function and increased ROS production.

    CAS  PubMed  Google Scholar 

  36. Boccadoro, M. et al. Oral melphalan at diagnosis hampers adequate collection of peripheral blood progenitor cells in multiple myeloma. Haematologica 87, 846–850 (2002).

    CAS  PubMed  Google Scholar 

  37. Knudsen, L. M., Rasmussen, T., Jensen, L. & Johnsen, H. E. Reduced bone marrow stem cell pool and progenitor mobilisation in multiple myeloma after melphalan treatment. Med. Oncol. 16, 245–254 (1999).

    CAS  PubMed  Google Scholar 

  38. Gardner, R. V., Astle, C. M. & Harrison, D. E. Hematopoietic precursor cell exhaustion is a cause of proliferative defect in primitive hematopoietic stem cells (PHSC) after chemotherapy. Exp. Hematol. 25, 495–501 (1997).

    CAS  PubMed  Google Scholar 

  39. Xing, Z. et al. Increased hematopoietic stem cell mobilization in aged mice. Blood 108, 2190–2197 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. 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 

  41. Wong, K. K. et al. Telomere dysfunction and ATM deficiency compromises organ homeostasis and accelerates ageing. Nature 421, 643–648 (2003). An important study showing that the effects of ATM loss seen in humans can be reproduced in mice if ATM deficiency is combined with telomere dysfunction, which implies that the ataxia telangiectasia syndrome partly results from telomere dysfunction in ATM-deficient cells.

    CAS  PubMed  Google Scholar 

  42. 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 

  43. Molofsky, A. V. et al. Increasing p16INK4a expression decreases forebrain progenitors and neurogenesis during ageing. Nature 443, 448–452 (2006). Shows that NSCs demonstrate a decrease in replicative function with ageing that is decreased in the setting of p16INK4a, implying that p16INK4a activation contributes to NSC ageing.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Enwere, E. et al. Aging results in reduced epidermal growth factor receptor signaling, diminished olfactory neurogenesis, and deficits in fine olfactory discrimination. J. Neurosci. 24, 8354–8365 (2004). A careful, functional description of NSC ageing.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Nishimura, E. K., Granter, S. R. & Fisher, D. E. Mechanisms of hair graying: incomplete melanocyte stem cell maintenance in the niche. Science 307, 720–724 (2005). An excellent study suggesting that greying, a paradigmatic ageing phenotype, appears to result from a defect in melanocyte stem-cell maintenance with ageing.

    CAS  PubMed  Google Scholar 

  46. Yoon, K. H. et al. Selective β-cell loss and α-cell expansion in patients with type 2 diabetes mellitus in Korea. J. Clin. Endocrinol. Metab. 88, 2300–2308 (2003).

    CAS  PubMed  Google Scholar 

  47. Butler, A. E. et al. β-cell deficit and increased β-cell apoptosis in humans with type 2 diabetes. Diabetes 52, 102–110 (2003).

    CAS  PubMed  Google Scholar 

  48. Meier, J. J. et al. Direct evidence of attempted β cell regeneration in an 89-year-old patient with recent-onset type 1 diabetes. Diabetologia 49, 1838–1844 (2006).

    CAS  PubMed  Google Scholar 

  49. Krishnamurthy, J. et al. p16INK4a induces an age-dependent decline in islet regenerative potential. Nature 443, 453–457 (2006). This study, similar to reference 43, shows that p16INK4a loss can ameliorate an ageing phenotype, in this case in the pancreatic islet.

    CAS  PubMed  Google Scholar 

  50. Conboy, I. M. et al. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature 433, 760–764 (2005). Using parabiosis, this study provides striking evidence that muscle satellite-cell ageing is cell non-autonomous.

    CAS  PubMed  Google Scholar 

  51. Ju, Z. et al. Telomere dysfunction induces environmental alterations limiting hematopoietic stem cell function and engraftment. Nature Med. 13, 742–747 (2007). This paper shows that telomere dysfunction limits HSC niche function, suggesting a cause of extrinsic ageing in HSCs.

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  53. Navarro, S. et al. Hematopoietic dysfunction in a mouse model for Fanconi anemia group D1. Mol. Ther. 14, 525–535 (2006).

    CAS  PubMed  Google Scholar 

  54. Reese, J. S., Liu, L. & Gerson, S. L. Repopulating defect of mismatch repair-deficient hematopoietic stem cells. Blood 102, 1626–1633 (2003).

    CAS  PubMed  Google Scholar 

  55. Prasher, J. M. et al. Reduced hematopoietic reserves in DNA interstrand crosslink repair-deficient Ercc1−/− mice. EMBO J. 24, 861–871 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  57. Morales, M. et al. The Rad50S allele promotes ATM-dependent DNA damage responses and suppresses ATM deficiency: implications for the Mre11 complex as a DNA damage sensor. Genes Dev. 19, 3043–3054 (2005). A striking finding showing that an engineered Rad50S allele, which is associated with bone marrow hypoplasia, can be rescued by ATM deficiency. This result indicates that the Rad50S allele is hypermorphic, and suggests that activation of the DNA-damage response per se can lead to stem-cell dysfunction.

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Rossi, D. J. et al. Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age. Nature 447, 725–729 (2007). An interesting study showing the accumulation of H2AX foci in HSCs with murine ageing, and an intrinsic role for several DNA-repair mechanisms in HSCs to prevent stem-cell ageing of this compartment.

    CAS  PubMed  Google Scholar 

  59. Rudolph, K. L. et al. Longevity, stress response, and cancer in aging telomerase-deficient mice. Cell 96, 701–712 (1999). Classical study that established the importance of telomere maintenance in age-related processes and lifespan in mammals.

    CAS  PubMed  Google Scholar 

  60. Ruzankina, Y. et al. Deletion of the developmentally essential gene Atr in adult mice leads to age-related phenotypes and stem cell loss. Cell Stem Cell 1, 113–126 (2007). A recent demonstration that homeostatic proliferation, in the absence of external DNA-damaging agents, can promote stem-cell dysfunction and ageing.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Brown, E. J. & Baltimore, D. ATR disruption leads to chromosomal fragmentation and early embryonic lethality. Genes Dev. 14, 397–402 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Blasco, M. A. et al. Telomere shortening and tumor formation by mouse cells lacking telomerase RNA. Cell 91, 25–34 (1997).

    CAS  PubMed  Google Scholar 

  63. Lee, H. W. et al. Essential role of mouse telomerase in highly proliferative organs. Nature 392, 569–574 (1998).

    CAS  PubMed  Google Scholar 

  64. Herrera, E. et al. Disease states associated with telomerase deficiency appear earlier in mice with short telomeres. EMBO J. 18, 2950–2960 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Allsopp, R. C., Morin, G. B., DePinho, R., Harley, C. B. & Weissman, I. L. Telomerase is required to slow telomere shortening and extend replicative lifespan of HSCs during serial transplantation. Blood 102, 517–520 (2003). This paper shows that telomere dysfunction can limit HSC lifespan in serial transplantation.

    CAS  PubMed  Google Scholar 

  66. Choudhury, A. R. et al. Cdkn1a deletion improves stem cell function and lifespan of mice with dysfunctional telomeres without accelerating cancer formation. Nature Genet. 39, 99–105 (2007). This paper demonstrates that p21CIP is not required for the anti-cancer effects of p53 mediated in response to telomere dysfunction, but is important for the pro-ageing effects of p53 in HSCs and other stem cells in response to telomere dysfunction.

    CAS  PubMed  Google Scholar 

  67. Chin, L. et al. p53 deficiency rescues the adverse effects of telomere loss and cooperates with telomere dysfunction to accelerate carcinogenesis. Cell 97, 527–538 (1999).

    CAS  PubMed  Google Scholar 

  68. Artandi, S. E. et al. Telomere dysfunction promotes non-reciprocal translocations and epithelial cancers in mice. Nature 406, 641–645 (2000). References 67 and 68 provide insights into the mechanisms that underlie the intimate link between cancer and ageing, particularly why aged individuals develop epithelial cancers and why such cancers emerge with radically altered cytogenetic profiles.

    CAS  PubMed  Google Scholar 

  69. Kamijo, T. et al. Tumor suppression at the mouse INK4a locus mediated by the alternative reading frame product p19ARF. Cell 91, 649–659 (1997).

    CAS  PubMed  Google Scholar 

  70. Stein, G. H., Drullinger, L. F., Soulard, A. & Dulic, V. Differential roles for cyclin-dependent kinase inhibitors p21 and p16 in the mechanisms of senescence and differentiation in human fibroblasts. Mol. Cell. Biol. 19, 2109–2117 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Alcorta, D. A. et al. Involvement of the cyclin-dependent kinase inhibitor p16 (INK4a) in replicative senescence of normal human fibroblasts. Proc. Natl Acad. Sci. USA 93, 13742–13747 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Campisi, J. Suppressing cancer: the importance of being senescent. Science 309, 886–887 (2005).

    CAS  PubMed  Google Scholar 

  73. Kim, W. Y. & Sharpless, N. E. The regulation of INK4/ARF in cancer and aging. Cell 127, 265–275 (2006).

    CAS  PubMed  Google Scholar 

  74. Sone, H. & Kagawa, Y. Pancreatic β cell senescence contributes to the pathogenesis of type 2 diabetes in high-fat diet-induced diabetic mice. Diabetologia 48, 58–67 (2005).

    CAS  PubMed  Google Scholar 

  75. Krishnamurthy, J. et al. Ink4a/Arf expression is a biomarker of aging. J. Clin. Invest. 114, 1299–1307 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Edwards, M. G. et al. Gene expression profiling of aging reveals activation of a p53-mediated transcriptional program. BMC Genomics 8, e80 (2007).

    Google Scholar 

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

    CAS  PubMed  Google Scholar 

  78. Chen, J., Astle, C. M. & Harrison, D. E. Hematopoietic senescence is postponed and hematopoietic stem cell function is enhanced by dietary restriction. Exp. Hematol. 31, 1097–1103 (2003). A provocative description of the effects of caloric restriction on HSC function with ageing.

    CAS  PubMed  Google Scholar 

  79. Chen, Q. & Ames, B. N. Senescence-like growth arrest induced by hydrogen peroxide in human diploid fibroblast F65 cells. Proc. Natl Acad. Sci. USA 91, 4130–4134 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Chen, J. H. et al. Loss of proliferative capacity and induction of senescence in oxidatively stressed human fibroblasts. J. Biol. Chem. 279, 49439–49446 (2004).

    CAS  PubMed  Google Scholar 

  81. Takahashi, A. et al. Mitogenic signalling and the p16(INK4a)-Rb pathway cooperate to enforce irreversible cellular senescence. Nature Cell Biol. 8, 1291–1297 (2006).

    CAS  PubMed  Google Scholar 

  82. Tothova, Z. et al. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell 128, 325–339 (2007). A detailed genetic study that establishes the importance of FOXO-dependent regulation of intracellular ROS in the maintenance of HSCs.

    CAS  PubMed  Google Scholar 

  83. 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). Together with references 43, 49 and 77, this study suggests that p16INK4a loss can ameliorate a murine ageing phenotype, in this case in the HSCs.

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Tyner, S. D. et al. p53 mutant mice that display early ageing-associated phenotypes. Nature 415, 45–53 (2002).

    CAS  PubMed  Google Scholar 

  85. Maier, B. et al. Modulation of mammalian life span by the short isoform of p53. Genes Dev. 18, 306–319 (2004). References 84 and 85 demonstrate that excess p53 activation can markedly accelerate the development of ageing-associated phenotypes in mice.

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Orsted, D. D., Bojesen, S. E., Tybjaerg-Hansen, A. & Nordestgaard, B. G. Tumor suppressor p53 Arg72Pro polymorphism and longevity, cancer survival, and risk of cancer in the general population. J. Exp. Med. 204, 1295–1301 (2007).

    PubMed  PubMed Central  Google Scholar 

  87. Matheu, A. et al. Delayed ageing through damage protection by the Arf/p53 pathway. Nature 448, 375–379 (2007). This paper challenges the view that increased p53 activity is necessarily pro-ageing.

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  89. Melzer, D. et al. A common variant of the p16(INK4a) genetic region is associated with physical function in older people. Mech. Ageing Dev. 128, 370–377 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Scott, L. J. et al. A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. Science 316, 1341–1345 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Saxena, R. et al. Genome-wide association analysis identifies loci for type 2 diabetes and triglyceride levels. Science 316, 1331–1336 (2007).

    CAS  PubMed  Google Scholar 

  92. Zeggini, E. et al. Replication of genome-wide association signals in UK samples reveals risk loci for type 2 diabetes. Science 316, 1336–1341 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Helgadottir, A. et al. A common variant on chromosome 9p21 affects the risk of myocardial infarction. Science 316, 1491–1493 (2007).

    CAS  PubMed  Google Scholar 

  94. McPherson, R. et al. A common allele on chromosome 9 associated with coronary heart disease. Science 316, 1488–1491 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. The Wellcome Trust Case Control Consortium. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447, 661–678 (2007). References 89–95 describe the identification of 9p21 SNPs near the INK4/ARF locus as being associated with type 2 diabetes mellitus, atherosclerotic heart disease and frailty.

  96. Matthews, C. et al. Vascular smooth muscle cells undergo telomere-based senescence in human atherosclerosis: effects of telomerase and oxidative stress. Circ. Res. 99, 156–164 (2006).

    CAS  PubMed  Google Scholar 

  97. Chimenti, C. et al. Senescence and death of primitive cells and myocytes lead to premature cardiac aging and heart failure. Circ. Res. 93, 604–613 (2003).

    CAS  PubMed  Google Scholar 

  98. Urbanek, K. et al. Myocardial regeneration by activation of multipotent cardiac stem cells in ischemic heart failure. Proc. Natl Acad. Sci. USA 102, 8692–8697 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Kleinjan, D. A. & van Heyningen, V. Long-range control of gene expression: emerging mechanisms and disruption in disease. Am. J. Hum. Genet. 76, 8–32 (2005).

    CAS  PubMed  Google Scholar 

  100. Pasmant, E. et al. Characterization of a germ-line deletion, including the entire INK4/ARF locus, in a melanoma-neural system tumor family: identification of ANRIL, an antisense noncoding RNA whose expression coclusters with ARF. Cancer Res. 67, 3963–3969 (2007).

    CAS  PubMed  Google Scholar 

  101. Vulliamy, T. et al. The RNA component of telomerase is mutated in autosomal dominant dyskeratosis congenita. Nature 413, 432–435 (2001).

    CAS  PubMed  Google Scholar 

  102. Yamaguchi, H. et al. Mutations in TERT, the gene for telomerase reverse transcriptase, in aplastic anemia. N. Engl. J. Med. 352, 1413–1424 (2005).

    CAS  PubMed  Google Scholar 

  103. 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 

  104. Tsakiri, K. D. et al. Adult-onset pulmonary fibrosis caused by mutations in telomerase. Proc. Natl Acad. Sci. USA 104, 7552–7557 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Armanios, M. Y. et al. Telomerase mutations in families with idiopathic pulmonary fibrosis. N. Engl. J. Med. 356, 1317–1326 (2007). References 101–105 are important human studies describing the role of telomere dysfunction in human syndromes such as aplastic anaemia and idiopathic pulmonary fibrosis.

    CAS  PubMed  Google Scholar 

  106. Kitada, T., Seki, S., Kawakita, N., Kuroki, T. & Monna, T. Telomere shortening in chronic liver diseases. Biochem. Biophys. Res. Commun. 211, 33–39 (1995).

    CAS  PubMed  Google Scholar 

  107. Miura, N. et al. Progressive telomere shortening and telomerase reactivation during hepatocellular carcinogenesis. Cancer Genet. Cytogenet. 93, 56–62 (1997).

    CAS  PubMed  Google Scholar 

  108. Urabe, Y. et al. Telomere length in human liver diseases. Liver 16, 293–297 (1996).

    CAS  PubMed  Google Scholar 

  109. Wiemann, S. U. et al. Hepatocyte telomere shortening and senescence are general markers of human liver cirrhosis. FASEB J. 16, 935–942 (2002).

    CAS  PubMed  Google Scholar 

  110. Rudolph, K. L., Chang, S., Millard, M., Schreiber-Agus, N. & DePinho, R. A. Inhibition of experimental liver cirrhosis in mice by telomerase gene delivery. Science 287, 1253–1258 (2000).

    CAS  PubMed  Google Scholar 

  111. Samani, N. J., Boultby, R., Butler, R., Thompson, J. R. & Goodall, A. H. Telomere shortening in atherosclerosis. Lancet 358, 472–473 (2001).

    CAS  PubMed  Google Scholar 

  112. Obana, N. et al. Telomere shortening of peripheral blood mononuclear cells in coronary disease patients with metabolic disorders. Intern. Med. 42, 150–153 (2003).

    CAS  PubMed  Google Scholar 

  113. Tabori, U., Nanda, S., Druker, H., Lees, J. & Malkin, D. Younger age of cancer initiation is associated with shorter telomere length in Li–Fraumeni syndrome. Cancer Res. 67, 1415–1418 (2007). In accordance with results in mice (see reference 67), this provocative human study suggests that telomere dysfunction accelerates tumour formation in the setting of p53 insufficiency.

    CAS  PubMed  Google Scholar 

  114. Wu, X. et al. Telomere dysfunction: a potential cancer predisposition factor. J. Natl Cancer Inst. 95, 1211–1218 (2003).

    CAS  PubMed  Google Scholar 

  115. Cawthon, R. M., Smith, K. R., O'Brien, E., Sivatchenko, A. & Kerber, R. A. Association between telomere length in blood and mortality in people aged 60 years or older. Lancet 361, 393–395 (2003).

    CAS  PubMed  Google Scholar 

  116. Frenck, R. W. Jr., Blackburn, E. H. & Shannon, K. M. The rate of telomere sequence loss in human leukocytes varies with age. Proc. Natl Acad. Sci. USA 95, 5607–5610 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Hemann, M. T., Strong, M. A., Hao, L. Y. & Greider, C. W. The shortest telomere, not average telomere length, is critical for cell viability and chromosome stability. Cell 107, 67–77 (2001).

    CAS  PubMed  Google Scholar 

  118. Rane, S. G. et al. Loss of Cdk4 expression causes insulin-deficient diabetes and Cdk4 activation results in b-islet cell hyperplasia. Nature Genet. 22, 44–52 (1999).

    CAS  PubMed  Google Scholar 

  119. Tsutsui, T. et al. Targeted disruption of CDK4 delays cell cycle entry with enhanced p27(Kip1) activity. Mol. Cell. Biol. 19, 7011–7019 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Nielsen, G. P. et al. Immunohistochemical survey of p16INK4A expression in normal human adult and infant tissues. Lab. Invest. 79, 1137–1143 (1999).

    CAS  PubMed  Google Scholar 

  121. Molofsky, A. V. et al. Bmi-1 dependence distinguishes neural stem cell self-renewal from progenitor proliferation. Nature 425, 962–967 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Bruggeman, S. W. et al. Ink4a and Arf differentially affect cell proliferation and neural stem cell self-renewal in Bmi1-deficient mice. Genes Dev. 19, 1438–1443 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Molofsky, A. V., He, S., Bydon, M., Morrison, S. J. & Pardal, R. Bmi-1 promotes neural stem cell self-renewal and neural development but not mouse growth and survival by repressing the p16Ink4a and p19Arf senescence pathways. Genes Dev. 19, 1432–1437 (2005). References 121–123 are careful characterizations of the effects of BMI1 loss on NSCs, and the role of p16INK4a and ARF in NSCs in the setting of BMI1 deficiency.

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Jacobs, J. J., Kieboom, K., Marino, S., DePinho, R. A. & van Lohuizen, M. The oncogene and Polycomb-group gene bmi-1 regulates cell proliferation and senescence through the ink4a locus. Nature 397, 164–168 (1999).

    CAS  PubMed  Google Scholar 

  125. Park, I. K. et al. Bmi-1 is required for maintenance of adult self-renewing haematopoietic stem cells. Nature 423, 302–305 (2003).

    CAS  PubMed  Google Scholar 

  126. Oguro, H. et al. Differential impact of INK4a and ARF on hematopoietic stem cells and their bone marrow microenvironment in BMI1-deficient mice. J. Exp. Med. 203, 2247–2253 (2006). References 125 and 126 are careful characterizations of the effects of BMI1 loss on HSCs, and the role of p16INK4a and ARF in the setting of BMI1 deficiency.

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank E. Sahin, J.-H. Paik, T. Letai and K. Mohlke for critical reading and advice on the manuscript. R.A.D. is a director, co-founder and scientific advisor of AVEO Pharmaceuticals, Inc. in Cambridge, Massachusetts, USA, and is an American Cancer Society Research Professor and an Ellison Medical Foundation Senior Scholar. This work was supported by grants from the Sidney Kimmel Foundation for Cancer Research (N.E.S.), the Ellison Medical Foundation (N.E.S. and R.A.D.), the American Federation of Aging Research (N.E.S.), the Burroughs Wellcome Fund (N.E.S.) and the US National Institutes of Health (R.A.D.). R.A.D. is supported by the LeBow Fund to Cure Myeloma and the Robert A. and Renee E. Belfer Foundation Institute for Innovative Cancer Science.

Author information

Authors and Affiliations

Authors

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

DATABASES

OMIM

ataxia telangiectasia

Hutchinson–Gilford syndrome

type 2 diabetes mellitus

Werner syndrome

FURTHER INFORMATION

Ronald A. DePinho's homepage

Norman E. Sharpless's homepage

Glossary

Forkhead transcription factor class O

(FOXO). One of a family of evolutionarily conserved transcription factors that are linked to lifespan regulation in lower systems and to stem-cell maintenance in mice. The FOXO proteins are thought to exert these effects by regulating the expression of genes involved in apoptosis, proliferation, glucose metabolism, DNA repair, regulation of reactive oxygen species and other diverse cellular processes.

Self-renewal

The capacity of replicating stem cells to generate daughter cells with the same biological and molecular profile that endows continued renewal potential. This can occur either asymmetrically when a stem cell produces another stem cell and a more differentiated daughter cell, or symmetrically when stem-cell division gives rise to two identical stem cells. Importantly, in mature organ systems, most cell-division activity that is responsible for tissue maintenance and expansion is not self-renewing.

Progeny

Along with progenitor cells, these are relatively undifferentiated cell types that are derived from asymmetric stem-cell division and lack the capacity to self-renew.

Senescence

A specialized form of growth arrest induced by various stressful stimuli including loss of telomere function, reactive oxygen species, some forms of DNA damage and activation of certain oncogenes or reactivation of tumour-suppressor genes. Senescence is characterized by several markers such as senescence-associated-β-galactosidase, alterations in chromatin structure (senescence-associated heterochromatic foci) and a marked increase in the secretion of several cytokines and other bioactive molecules (senescence-associated secretory phenotype).

Tissue-specific stem cell

A specialized cell found in many tissues of adults. These cells can replace themselves through self-renewal and are generally multipotent, in that they can give rise to progeny that can differentiate into multiple different cell types of the associated organ.

Multipotency

The ability to give rise to differentiated progeny of different specialized subtypes. However, some self-renewing cells (for example, pancreatic β-cells) have a narrow potential for differentiation, generating progeny similar to the parental cell. This type of self-renewing cell is termed a 'unipotent progenitor', which can be viewed as a special stem-cell subtype, at least in terms of long-term proliferative capacity. For convenience, in this Review the term 'stem cell' is applied to both types of adult self-renewing cells.

Telomere

A nucleoprotein complex at the end of chromosomes that maintains chromosomal integrity. It consists of many double-stranded 5′-TTAGGG-3′ repeats, a 3′-single-stranded overhang and associated telomere-binding proteins, which together generate a capped structure that is impervious to the actions of complexes that repair DNA damage.

Telomerase

A ribonucleoprotein complex that extends the ends of telomeres after replication by using telomerase reverse transcriptase (TERT) and an RNA template (TERC) that is part of the enzyme complex.

Single nucleotide polymorphism

A common, single-base difference in a gene among individuals within a species.

Frailty

A clinically validated, functional measure used in clinical geriatrics. It is scored as a continuous variable using a series of routine, easily measured tests such as gait speed. Frail individuals are less able to live independently, are more likely to harbour co-morbid illnesses, and exhibit increased mortality.

Linkage disequilibrium

(LD). A measure of genetic associations between alleles at different loci, which indicates whether allelic or marker associations on the same chromosome are more common than expected. Loci are generally considered to be in strong LD if their correlation is higher than a pre-defined cut-off (for example, 0.8).

Rights and permissions

Reprints and permissions

About this article

Cite this article

Sharpless, N., DePinho, R. How stem cells age and why this makes us grow old. Nat Rev Mol Cell Biol 8, 703–713 (2007). https://doi.org/10.1038/nrm2241

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrm2241

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