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:

Living on a break: cellular senescence as a DNA-damage response

Key Points

  • Cells respond to the perception of DNA damage by arresting cell-cycle progression and attempting repair: collectively these actions are known as the DNA-damage response (DDR). In mammals, proliferation is not resumed until DNA damage is fixed.

  • Cellular senescence is a condition in which cells, despite being alive, are unable to proliferate further. This is a stress response, and therefore is different from quiescence or terminal differentiation.

  • Replicative senescence limits the proliferation of normal human cells. Proliferation that is associated with progressive telomere shortening leads to senescence establishment when critically short telomeres are recognized as DNA damage and trigger a DDR.

  • Mammalian ageing is associated with the progressive accumulation of senescent cells and DDR accumulation in stem or progenitor cells and more differentiated cells. In the skin of primates, DDR markers associate with the telomeres.

  • Oncogene activation also causes DDR activation and cellular senescence. Oncogene-induced DNA damage is caused by altered DNA replication, and oncogene-induced senescence is a barrier to cancer. Senescent cells can be observed in vivo in preneoplastic lesions.

  • Collectively, mounting evidence indicates that senescence, triggered by different stimuli, is the outcome of a protracted DDR.

Abstract

Cellular senescence is associated with ageing and cancer in vivo and has a proven tumour-suppressive function. Common to both ageing and cancer is the generation of DNA damage and the engagement of the DNA-damage response pathways. In this Review, the diverse mechanisms that lead to DNA-damage generation and the activation of DNA-damage-response signalling pathways are discussed, together with the evidence for their contribution to the establishment and maintenance of cellular senescence in the context of organismal ageing and cancer development.

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: The DNA-damage response.
Figure 2: DDR inhibition at telomeres.
Figure 3: Ras expression leads to a biphasic response.

Similar content being viewed by others

References

  1. Shiloh, Y. The ATM-mediated DNA-damage response: taking shape. Trends Biochem. Sci. 37, 402–410 (2006).

    Article  CAS  Google Scholar 

  2. Taylor, R. C., Cullen, S. P. & Martin, S. J. Apoptosis: controlled demolition at the cellular level. Nature Rev. Mol. Cell Biol. 9, 231–241 (2008).

    Article  CAS  Google Scholar 

  3. Zou, L. Single- and double-stranded DNA: building a trigger of ATR-mediated DNA damage response. Genes Dev. 21, 879–885 (2007).

    Article  CAS  PubMed  Google Scholar 

  4. Celeste, A. et al. Genomic instability in mice lacking histone H2AX. Science 296, 922–927 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Lou, Z., Minter-Dykhouse, K., Wu, X. & Chen, J. MDC1 is coupled to activated CHK2 in mammalian DNA damage response pathways. Nature 421, 957–961 (2003).

    Article  CAS  PubMed  Google Scholar 

  6. Goldberg, M. et al. MDC1 is required for the intra-S-phase DNA damage checkpoint. Nature 421, 952–956 (2003).

    Article  CAS  PubMed  Google Scholar 

  7. Stewart, G. S., Wang, B., Bignell, C. R., Taylor, A. M. R. & Elledge, S. J. MDC1 is a mediator of the mammalian DNA damage checkpoint. Nature 421, 961–966 (2003).

    Article  CAS  PubMed  Google Scholar 

  8. Abraham, R. T. Checkpoint signalling: focusing on 53BP1. Nature Cell Biol. 4, E277–E279 (2002).

    Article  CAS  PubMed  Google Scholar 

  9. Lukas, C. et al. Mdc1 couples DNA double-strand break recognition by Nbs1 with its H2AX-dependent chromatin retention. EMBO J. 23, 2674–2683 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Stucki, M. et al. MDC1 directly binds phosphorylated histone H2AX to regulate cellular responses to DNA double-strand breaks. Cell 123, 1213–1226 (2005).

    Article  CAS  PubMed  Google Scholar 

  11. Lou, Z. et al. MDC1 maintains genomic stability by participating in the amplification of ATM-dependent DNA damage signals. Mol. Cell 21, 187–200 (2006).

    Article  CAS  PubMed  Google Scholar 

  12. Bekker-Jensen, S., Lukas, C., Melander, F., Bartek, J. & Lukas, J. Dynamic assembly and sustained retention of 53BP1 at the sites of DNA damage are controlled by Mdc1/NFBD1. J. Cell Biol. 170, 201–211 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Cortez, D., Guntuku, S., Qin, J. & Elledge, S. J. ATR and ATRIP: partners in checkpoint signaling. Science 294, 1713–1716 (2001).

    Article  CAS  PubMed  Google Scholar 

  14. Weiss, R. S., Matsuoka, S., Elledge, S. J. & Leder, P. Hus1 acts upstream of Chk1 in a mammalian DNA damage response pathway. Curr. Biol. 12, 73–77 (2002).

    Article  CAS  PubMed  Google Scholar 

  15. Kumagai, A., Lee, J., Yoo, H. Y. & Dunphy, W. G. TopBP1 activates the ATR–ATRIP complex. Cell 124, 943–955 (2006).

    Article  CAS  PubMed  Google Scholar 

  16. Liu, S. et al. Claspin operates downstream of TopBP1 to direct ATR signaling towards Chk1 activation. Mol. Cell. Biol. 26, 6056–6064 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Jazayeri, A. et al. ATM- and cell cycle-dependent regulation of ATR in response to DNA double-strand breaks. Nature Cell Biol. 8, 37–45 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Buscemi, G. et al. Activation of ATM and Chk2 kinases in relation to the amount of DNA strand breaks. Oncogene 23, 7691–7700 (2004).

    Article  CAS  PubMed  Google Scholar 

  19. Lukas, C., Falck, J., Bartkova, J., Bartek, J. & Lukas, J. Distinct spatiotemporal dynamics of mammalian checkpoint regulators induced by DNA damage. Nature Cell Biol. 5, 255–260 (2003).

    Article  CAS  PubMed  Google Scholar 

  20. Bekker-Jensen, S. et al. Spatial organization of the mammalian genome surveillance machinery in response to DNA strand breaks. J. Cell Biol. 173, 195–206 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Mailand, N. et al. Rapid destruction of human Cdc25A in response to DNA damage. Science 288, 1425–1429 (2000).

    Article  CAS  PubMed  Google Scholar 

  22. Turenne, G. A., Paul, P., Laflair, L. & Price, B. D. Activation of p53 transcriptional activity requires ATM's kinase domain and multiple N-terminal serine residues of p53. Oncogene 20, 5100–5110 (2001).

    Article  CAS  PubMed  Google Scholar 

  23. Deng, C., Zhang, P., Harper, J. W., Elledge, S. J. & Leder, P. Mice lacking p21CIP1/WAF1 undergo normal development, but are defective in G1 checkpoint control. Cell 82, 675–684 (1995).

    Article  CAS  PubMed  Google Scholar 

  24. Downey, M. & Durocher, D. γH2AX as a checkpoint maintenance signal. Cell Cycle 5, 1376–1381 (2006).

    Article  CAS  PubMed  Google Scholar 

  25. Yamauchi, M. et al. Growth of persistent foci of DNA damage checkpoint factors is essential for amplification of G1 checkpoint signaling. DNA Repair 7, 405–417 (2008).

    Article  CAS  PubMed  Google Scholar 

  26. Hozak, P. & Cook, P. R. Replication factories. Trends Cell Biol. 4, 48–52 (1994).

    Article  CAS  PubMed  Google Scholar 

  27. Aten, J. A. et al. Dynamics of DNA double-strand breaks revealed by clustering of damaged chromosome domains. Science 303, 92–95 (2004).

    Article  CAS  PubMed  Google Scholar 

  28. Soutoglou, E. et al. Positional stability of single double-strand breaks in mammalian cells. Nature Cell Biol. 9, 675–682 (2007).

    Article  CAS  PubMed  Google Scholar 

  29. Kruhlak, M. J. et al. Changes in chromatin structure and mobility in living cells at sites of DNA double-strand breaks. J. Cell. Biochem. 172, 823–834 (2006).

    CAS  Google Scholar 

  30. Bonilla, C. Y., Melo, J. A. & Toczyski, D. P. Colocalization of sensors is sufficient to activate the DNA damage checkpoint in the absence of damage. Mol. Cell 30, 267–276 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Di Leonardo, A., Linke, S. P., Clarkin, K. & Wahl, G. M. DNA damage triggers a prolonged p53-dependent G1 arrest and long-term induction of Cip1 in normal human fibroblasts. Genes Dev. 8, 2540–2551 (1994).

    Article  CAS  PubMed  Google Scholar 

  32. Campisi, J. & d'Adda di Fagagna, F. Cellular senescence: when bad things happen to good cells. Nature Rev. Mol. Cell Biol. 8, 729–740 (2007).

    Article  CAS  Google Scholar 

  33. Hayflick, L. & Moorhead, P. S. The serial cultivation of human diploid cell strains. Exp. Cell Res. 25, 585–621 (1961). How it all started.

    Article  CAS  PubMed  Google Scholar 

  34. Hayflick, L. The limited in vitro proliferation of human diploid cell strains. Exp. Cell Res. 37, 614–636 (1965).

    Article  CAS  PubMed  Google Scholar 

  35. Longhese, M. P. DNA damage response at functional and dysfunctional telomeres. Genes Dev. 22, 125–140 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. d' Adda di Fagagna, F., Teo, S.-H. & Jackson, S. P. Functional links between telomeres and proteins of the DNA-damage response. Genes Dev. 18, 1781–1799 (2004).

    Article  PubMed  Google Scholar 

  37. Karlseder, J. et al. The telomeric protein TRF2 binds the ATM kinase and can inhibit the ATM-dependent DNA damage response. PLoS Biol. 2, e240 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Denchi, E. L. & de Lange, T. Protection of telomeres through independent control of ATM and ATR by TRF2 and POT1. Nature 448, 1068–1071 (2007).

    Article  CAS  PubMed  Google Scholar 

  39. Harley, C. B., Futcher, A. B. & Greider, C. W. Telomeres shorten during ageing of human fibroblasts. Nature 345, 458–460 (1990). First article to suggest that telomere shortening dictates replicative senescence.

    Article  CAS  PubMed  Google Scholar 

  40. Takai, H., Smogorzewska, A. & de Lange, T. DNA damage foci at dysfunctional telomeres. Curr. Biol. 13, 1549–1556 (2003).

    Article  CAS  PubMed  Google Scholar 

  41. d' Adda di Fagagna, F. et al. A DNA damage checkpoint response in telomere-initiated senescence. Nature 426, 194–198 (2003).

    Article  CAS  PubMed  Google Scholar 

  42. Herbig, U., Jobling, W. A., Chen, B. P., Chen, D. J. & Sedivy, J. M. Telomere shortening triggers senescence of human cells through a pathway involving ATM, p53, and p21(CIP1), but not p16(INK4a). Mol. Cell 14, 501–513 (2004). References 41 and 42 were the first to show DDR activation at critically short telomeres in senescent cells.

    Article  CAS  PubMed  Google Scholar 

  43. 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).

    Article  CAS  PubMed  Google Scholar 

  44. Brown, J. P., Wei, W. & Sedivy, J. M. Bypass of senescence after disruption of p21CIP1/WAF1 gene in normal diploid human fibroblasts. Science 277, 831–834 (1997).

    Article  CAS  PubMed  Google Scholar 

  45. Gire, V., Roux, P., Wynford-Thomas, D., Brondello, J. M. & Dulic, V. DNA damage checkpoint kinase Chk2 triggers replicative senescence. EMBO J. 23, 2554–2563 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Bond, J. A., Wyllie, F. S. & Wynford-Thomas, D. Escape from senescence in human diploid fibroblasts induced directly by mutant p53. Oncogene 9, 1885–1889 (1994).

    CAS  PubMed  Google Scholar 

  47. Zhang, H., Pan, K.-H. & Cohen, S. N. Senescence-specific gene expression fingerprints reveal cell-type-dependent physical clustering of up-regulated chromosomal loci. Proc. Natl Acad. Sci. USA 100, 3251–3256 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Makarov, V. L., Hirose, Y. & Langmore, J. P. Long G tails at both ends of human chromosomes suggest a C strand degradation mechanism for telomere shortening. Cell 88, 657–666 (1997).

    Article  CAS  PubMed  Google Scholar 

  49. Deckbar, D. et al. Chromosome breakage after G2 checkpoint release. J. Cell Biol. 176, 749–755 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Huang, L. C., Clarkin, K. C. & Wahl, G. M. Sensitivity and selectivity of the DNA damage sensor responsible for activating p53-dependent G1 arrest. Proc. Natl Acad. Sci. USA 93, 4827–4832 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Meier, A. et al. Spreading of mammalian DNA-damage response factors studied by ChIP-chip at damaged telomeres. EMBO J. 26, 2707–2718 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Baird, D. M., Rowson, J., Wynford-Thomas, D. & Kipling, D. Extensive allelic variation and ultrashort telomeres in senescent human cells. Nature Genet. 33, 203–207 (2003).

    Article  CAS  PubMed  Google Scholar 

  53. Martens, U. M. et al. Short telomeres on human chromosome 17p. Nature Genet. 18, 76–80 (1998).

    Article  CAS  PubMed  Google Scholar 

  54. Herbig, U., Ferreira, M., Condel, L., Carey, D. & Sedivy, J. M. Cellular senescence in aging primates. Science 311, 1257 (2006).

    Article  CAS  PubMed  Google Scholar 

  55. Jeyapalan, J. C., Ferreira, M., Sedivy, J. M. & Herbig, U. Accumulation of senescent cells in mitotic tissue of aging primates. Mech. Ageing Dev. 128, 36–44 (2006). References 54 and 55 provide the first evidence of telomere-associated DDR activation in ageing primates.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Varley, H., Pickett, H. A., Foxon, J. L., Reddel, R. R. & Royle, N. J. Molecular characterization of inter-telomere and intra-telomere mutations in human ALT cells. Nature Genet. 30, 301–305 (2002).

    Article  PubMed  Google Scholar 

  57. Celli, G. B., Denchi, E. L. & de Lange, T. Ku70 stimulates fusion of dysfunctional telomeres yet protects chromosome ends from homologous recombination. Nature Cell Biol. 8, 855–890 (2006).

    Article  CAS  Google Scholar 

  58. Celli, G. B. & de Lange, T. DNA processing is not required for ATM-mediated telomere damage response after TRF2 deletion. Nature Cell Biol. 7, 712–718 (2005).

    Article  CAS  PubMed  Google Scholar 

  59. Bae, N. S. & Baumann, P. A RAP1/TRF2 complex inhibits nonhomologous end-joining at human telomeric DNA ends. Mol. Cell 26, 323–334 (2007).

    Article  CAS  PubMed  Google Scholar 

  60. Zou, Y., Sfeir, A., Gryaznov, S. M., Shay, J. W. & Wright, W. E. Does a sentinel or a subset of short telomeres determine replicative senescence? Mol. Biol. Cell 15, 3709–3718 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Capper, R. et al. The nature of telomere fusion and a definition of the critical telomere length in human cells. Genes Dev. 21, 2495–2508 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Prieur, A. & Peeper, D. S. Cellular senescence in vivo: a barrier to tumorigenesis. Curr. Opin. Cell Biol. 20, 150–155 (2008).

    Article  CAS  PubMed  Google Scholar 

  63. Jones, C. J. et al. Evidence for a telomere-independent “clock” limiting RAS oncogene-driven proliferation of human thyroid epithelial cells. Mol. Cell. Biol. 20, 5690–5699 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Di Micco, R. et al. Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication. Nature 444, 638–642 (2006).

    Article  CAS  PubMed  Google Scholar 

  65. Bartkova, J. et al. Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature 444, 633–637 (2006). References 64 and 65 demonstrate that oncogene activation leads to DDR activation and consequent senescence establishment.

    Article  CAS  PubMed  Google Scholar 

  66. Debidda, M., Williams, D. A. & Zheng, Y. Rac1 GTPase regulates cell genomic stability and senescence. J. Biol. Chem. 281, 38519–38528 (2006).

    Article  CAS  PubMed  Google Scholar 

  67. Xu, M. et al. β-catenin expression results in p53-independent DNA damage and oncogene-induced senescence in prelymphomagenic thymocytes in vivo. Mol. Cell. Biol. 28, 1713–1723 (2008).

    Article  CAS  PubMed  Google Scholar 

  68. Gorrini, C. et al. Tip60 is a haplo-insufficient tumour suppressor required for an oncogene-induced DNA damage response. Nature 448, 1063–1067 (2007).

    Article  CAS  PubMed  Google Scholar 

  69. Moiseeva, O., Mallette, F. A., Mukhopadhyay, U. K., Moores, A. & Ferbeyre, G. DNA damage signaling and p53-dependent senescence after prolonged β-interferon stimulation. Mol. Biol. Cell 17, 1583–1592 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Mallette, F. A., Gaumont-Leclerc, M.-F. & Ferbeyre, G. The DNA damage signaling pathway is a critical mediator of oncogene-induced senescence. Genes Dev. 21, 43–48 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Grandori, C. et al. Werner syndrome protein limits MYC-induced cellular senescence. Genes Dev. 17, 1569–1574 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Di Micco, R., Fumagalli, M. & d' Adda di Fagagna, F. Breaking news: high-speed race ends in arrest — how oncogenes induce senescence. Trends Cell Biol. 17, 529–536 (2007).

    Article  CAS  PubMed  Google Scholar 

  73. Michaloglou, C. et al. BRAFE600-associated senescence-like cell cycle arrest of human naevi. Nature 436, 720–724 (2005). Reports that skin naevi are senescent melanocytes.

    Article  CAS  PubMed  Google Scholar 

  74. Dankort, D. et al. A new mouse model to explore the initiation, progression, and therapy of BRAFV600E-induced lung tumors. Genes Dev. 21, 379–384 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Denchi, E. L., Attwooll, C., Pasini, D. & Helin, K. Deregulated E2F activity induces hyperplasia and senescence-like features in the mouse pituitary gland. Mol. Cell. Biol. 25, 2660–2672 (2005).

    Article  CAS  PubMed Central  Google Scholar 

  76. Dominguez-Sola, D. et al. Non-transcriptional control of DNA replication by c-Myc. Nature 448, 445–451 (2007). Reports that the MYC proto-oncogene is a direct regulator of DNA replication.

    Article  CAS  PubMed  Google Scholar 

  77. Kastan, M. B. & Bartek, J. Cell-cycle checkpoints and cancer. Nature 432, 316–323 (2004).

    Article  CAS  PubMed  Google Scholar 

  78. Ge, X. Q., Jackson, D. A. & Blow, J. J. Dormant origins licensed by excess Mcm2–7 are required for human cells to survive replicative stress. Genes Dev. 21, 3331–3341 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Vaziri, C. et al. A p53-dependent checkpoint pathway prevents rereplication. Mol. Cell 11, 997–1008 (2003).

    Article  CAS  PubMed  Google Scholar 

  80. Gorgoulis, V. G. et al. Activation of the DNA damage checkpoint and genomic instability in human precancerous lesions. Nature 434, 907–913 (2005).

    Article  CAS  PubMed  Google Scholar 

  81. Tsantoulis, P. K. et al. Oncogene-induced replication stress preferentially targets common fragile sites in preneoplastic lesions. A genome-wide study. Oncogene 27, 3256–3264 (2007).

    Article  CAS  PubMed  Google Scholar 

  82. Zhang, R. & Adams, P. D. Heterochromatin and its relationship to cell senescence and cancer therapy. Cell Cycle 6, 784–789 (2007).

    Article  CAS  PubMed  Google Scholar 

  83. Narita, M. et al. Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell 113, 703–716 (2003). The first report to describe global heterochromatinization of senescent nuclei.

    Article  CAS  PubMed  Google Scholar 

  84. Shimada, M. et al. Chk1 is a histone H3 threonine 11 kinase that regulates DNA damage-induced transcriptional repression. Cell 132, 221–232 (2008).

    Article  CAS  PubMed  Google Scholar 

  85. Downs, J. A., Nussenzweig, M. C. & Nussenzweig, A. Chromatin dynamics and the preservation of genetic information. Nature 447, 951–958 (2007).

    Article  CAS  PubMed  Google Scholar 

  86. Ziv, Y. et al. Chromatin relaxation in response to DNA double-strand breaks is modulated by a novel ATM- and KAP-1 dependent pathway. Nature Cell Biol. 8, 870–876 (2006).

    Article  CAS  PubMed  Google Scholar 

  87. Murr, R. et al. Histone acetylation by Trrap–Tip60 modulates loading of repair proteins and repair of DNA double-strand breaks. Nature Cell Biol. 8, 91–99 (2006).

    Article  CAS  PubMed  Google Scholar 

  88. Dimri, G. P. et al. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc. Natl Acad. Sci. USA 92, 9363–9367 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Lee, B. Y. et al. Senescence-associated beta-galactosidase is lysosomal β-galactosidase. Aging Cell 5, 187–195 (2006).

    Article  CAS  PubMed  Google Scholar 

  90. Campisi, J. From cells to organisms: can we learn about aging from cells in culture? Exp. Gerontol. 36, 607–618 (2001).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  92. Nijnik, A. et al. DNA repair is limiting for haematopoietic stem cells during ageing. Nature 447, 686–690 (2007). References 91 and 92 demonstrate that stem cells accumulate DDR markers during normal ageing.

    Article  CAS  PubMed  Google Scholar 

  93. Aladjem, M. I. Replication in context: dynamic regulation of DNA replication patterns in metazoans. Nature Rev. Genet. 8, 588–600 (2007).

    Article  CAS  PubMed  Google Scholar 

  94. Hong, Y. & Stambrook, P. J. Restoration of an absent G1 arrest and protection from apoptosis in embryonic stem cells after ionizing radiation. Proc. Natl Acad. Sci. USA 101, 14443–14448 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Beausejour, C. M. et al. Reversal of human cellular senescence: roles of the p53 and p16 pathways. EMBO J. 22, 4212–4222 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Bracken, A. P. et al. The Polycomb group proteins bind throughout the INK4A-ARF locus and are disassociated in senescent cells. Genes Dev. 21, 525–530 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Braig, M. et al. Oncogene-induced senescence as an initial barrier in lymphoma development. Nature 436, 660–665 (2005). Shows that heterochromatin formation is an in vivo barrier to cancer development.

    Article  CAS  PubMed  Google Scholar 

  101. Collado, M. et al. Tumour biology: senescence in premalignant tumours. Nature 436, 642 (2005).

    Article  CAS  PubMed  Google Scholar 

  102. Chen, Z. et al. Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature 436, 725–730 (2005). References 101 and 102 demonstrate that senescent cells accumulate in vivo in mouse models of oncogene activation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Nuciforo, P. G., Luise, C., Capra, M., Pelosi, G. & d'Adda di Fagagna, F. Complex engagement of DNA damage response pathways in human cancer and in lung tumor progression. Carcinogenesis 28, 2082–2088 (2007).

    Article  CAS  PubMed  Google Scholar 

  104. Bartkova, J. et al. DNA damage response as a candidate anti-cancer barrier in early human tumorigenesis. Nature 434, 864–870 (2005). References 80 and 104 are landmark papers that report that DDR signalling is activated in early preneoplastic lesions in humans.

    Article  CAS  PubMed  Google Scholar 

  105. Greenman, C. et al. Patterns of somatic mutation in human cancer genomes. Nature 446, 153–158 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Aguilera, A. & Gomez-Gonzalez, B. Genome instability: a mechanistic view of its causes and consequences. Nature Rev. Genet. 9, 204–217 (2008).

    Article  CAS  PubMed  Google Scholar 

  107. Tort, F. et al. Retinoblastoma pathway defects show differential ability to activate the constitutive DNA damage response in human tumorigenesis. Cancer Res. 66, 10258–10263 (2006).

    Article  CAS  PubMed  Google Scholar 

  108. Ventura, A. et al. Restoration of p53 function leads to tumour regression in vivo. Nature 445, 661–665 (2007).

    Article  CAS  PubMed  Google Scholar 

  109. Xue, W. et al. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature 445, 656–660 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Gasser, S., Orsulic, S., Brown, E. J. & Raulet, D. H. The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. 436, 1186–1190 (2005).

  111. Acosta, J. C. et al. Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell 133, 1006–1018 (2008).

    Article  CAS  PubMed  Google Scholar 

  112. Kuilman, T. et al. Oncogene-induced cellular senescence relayed by a cell-autonomous interleukin-dependent network. Cell 133, 1019–1031 (2008).

    Article  CAS  PubMed  Google Scholar 

  113. Bakkenist, C. J., Drissi, R., Wu, J., Kastan, M. B. & Dome, J. S. Disappearance of the telomere dysfunction-induced stress response in fully senescent cells. Cancer Res. 64, 3748–3752 (2004).

    Article  CAS  PubMed  Google Scholar 

  114. Riballo, E. et al. A pathway of double-strand break rejoining dependent upon ATM, Artemis, and proteins locating to γ-H2AX foci. Mol. Cell 16, 715–724 (2004).

    Article  CAS  PubMed  Google Scholar 

  115. Branzei, D. & Foiani, M. The Rad53 signal transduction pathway: replication fork stabilization, DNA repair, and adaptation. Exp. Cell Res. 312, 2654–2659 (2006).

    Article  CAS  PubMed  Google Scholar 

  116. Cosme-Blanco, W. et al. Telomere dysfunction suppresses spontaneous tumorigenesis in vivo by initiating p53-dependent cellular senescence. EMBO Rep. 8, 497–503 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Feldser, D. M. & Greider, C. W. Short telomeres limit tumor progression in vivo by inducing senescence. Cancer Cell 11, 461–469 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Matheu, A. et al. Delayed ageing through damage protection by the Arf/p53 pathway. Nature 448, 375–379 (2007).

    Article  CAS  PubMed  Google Scholar 

  119. Sherr, C. J. Divorcing ARF and p53: an unsettled case. Nature Rev. Cancer 6, 663–673 (2006).

    Article  CAS  Google Scholar 

  120. Ferbeyre, G. et al. Oncogenic ras and p53 cooperate to induce cellular senescence. Mol. Cell. Biol. 22, 3497–3508 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Vijg, J. A. N., Busuttil, R. A., Bahar, R. & Dolle, M. E. T. Aging and genome maintenance. Ann. NY Acad. Sci. 1055, 35–47 (2005).

    Article  CAS  PubMed  Google Scholar 

  122. Toledo, L. I., Murga, M., Gutierrez-Martinez, P., Soria, R. & Fernandez-Capetillo, O. ATR signaling can drive cells into senescence in the absence of DNA breaks. Genes Dev. 22, 297–302 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Celeste, A. et al. Histone H2AX phosphorylation is dispensable for the initial recognition of DNA breaks. Nature Cell Biol. 5, 675–679 (2003).

    Article  CAS  PubMed  Google Scholar 

  124. Mirzoeva, O. K. & Petrini, J. H. J. DNA damage-dependent nuclear dynamics of the Mre11 complex. Mol. Cell. Biol. 21, 281–288 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Turner, J. M. A. et al. BRCA1, histone H2AX phosphorylation, and male meiotic sex chromosome inactivation. Curr. Biol. 14, 2135–2142 (2004).

    Article  CAS  PubMed  Google Scholar 

  126. Bodnar, A. G. et al. Extension of life-span by introduction of telomerase into normal human cells. Science 279, 349–352 (1998). Conclusive evidence that telomere maintenance is the limiting factor that restrains cell proliferation of normal human fibroblasts.

    Article  CAS  PubMed  Google Scholar 

  127. Forsyth, N. R., Evans, A. P., Shay, J. W. & Wright, W. E. Developmental differences in the immortalization of lung fibroblasts by telomerase. Aging Cell 2, 235–243 (2003).

    Article  CAS  PubMed  Google Scholar 

  128. Ramirez, R. D. et al. Putative telomere-independent mechanisms of replicative aging reflect inadequate growth conditions. Genes Dev. 15, 398–403 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Parrinello, S. et al. Oxygen sensitivity severely limits the replicative lifespan of murine fibroblasts. Nature Cell Biol. 5, 741–747 (2003).

    Article  CAS  PubMed  Google Scholar 

  130. D'Amours, D. & Jackson, S. P. The MRE11 complex: at the crossroads of DNA repair and checkpoint signalling. Nature Rev. Mol. Cell Biol. 3, 317–327 (2002).

    Article  CAS  Google Scholar 

  131. Falck, J., Coates, J. & Jackson, S. P. Conserved modes of recruitment of ATM, ATR and DNA-PKcs to sites of DNA damage. Nature 434, 605–611 (2005).

    Article  CAS  PubMed  Google Scholar 

  132. Huyen, Y. et al. Methylated lysine 79 of histone H3 targets 53BP1 to DNA double-strand breaks. Nature 432, 406–411 (2004).

    Article  CAS  PubMed  Google Scholar 

  133. Smits, V. A., Reaper, P. M. & Jackson, S. P. Rapid PIKK-dependent release of Chk1 from chromatin promotes the DNA-damage checkpoint response. Curr. Biol. 16, 150–159 (2006).

    Article  CAS  PubMed  Google Scholar 

  134. Wang, F. et al. The POT1–TPP1 telomere complex is a telomerase processivity factor. Nature 445, 506–510 (2007).

    Article  CAS  PubMed  Google Scholar 

  135. Wu, L. et al. Pot1 deficiency initiates DNA damage checkpoint activation and aberrant homologous recombination at telomeres. Cell 126, 49–62 (2006).

    Article  CAS  PubMed  Google Scholar 

  136. Denchi, E. L. & de Lange, T. Protection of telomeres through independent control of ATM and ATR by TRF2 and POT1. Nature 448, 1068–1071 (2007).

    Article  CAS  PubMed  Google Scholar 

  137. Courtois-Cox, S. et al. A negative feedback signaling network underlies oncogene-induced senescence. Cancer Cell 10, 459–472 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Serrano, M., Lin, A. W., McCurrach, M. E., Beach, D. & Lowe, S. W. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88, 593–602 (1997). First report to describe oncogene-induced cellular senescence.

    Article  CAS  PubMed  Google Scholar 

  139. Zhu, J., Woods, D., McMahon, M. & Bishop, J. M. Senescence of human fibroblasts induced by oncogenic Raf. Genes Dev. 12, 2997–3007 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Lin, A. W. et al. Premature senescence involving p53 and p16 is activated in response to constitutive MEK/MAPK mitogenic signaling. Genes Dev. 12, 3008–3019 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Katakura, Y., Nakata, E., Miura, T. & Shirahata, S. Transforming growth factor β triggers two independent-senescence programs in cancer cells. Biochem. Biophys Res. Commun. 255, 110–115 (1999).

    Article  CAS  PubMed  Google Scholar 

  142. Zhang, H. & Cohen, S. N. Smurf2 up-regulation activates telomere-dependent senescence. Genes Dev. 18, 3028–3040 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Pearson, M. et al. PML regulates p53 acetylation and premature senescence induced by oncogenic Ras. Nature 406, 207–210 (2000).

    Article  CAS  PubMed  Google Scholar 

  144. Liontos, M. et al. Deregulated overexpression of hCdt1 and hCdc6 promotes malignant behavior. Cancer Res. 67, 10899–10909 (2007).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

F.d'A.d.F. is supported by the Associazione Italiana per la Ricerca sul Cancro, the Association for International Cancer Research, the Human Frontier Science Program, the FP6 and FP7 program of the European Union and the EMBO Young Investigator Program.

Author information

Authors and Affiliations

Authors

Related links

Related links

DATABASES

National Cancer Institute

Bladder cancer

lung squamous-cell carcinoma

non-small cell lung cancer

sarcoma

FURTHER INFORMATION

Fabrizio d'Adda di Fagagna's homepage

Rights and permissions

Reprints and permissions

About this article

Cite this article

d'Adda di Fagagna, F. Living on a break: cellular senescence as a DNA-damage response. Nat Rev Cancer 8, 512–522 (2008). https://doi.org/10.1038/nrc2440

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc2440

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