Skip to main content
Log in

Radiation-induced cell death mechanisms

  • Review
  • Published:
Tumor Biology

Abstract

The main goal when treating malignancies with radiation therapy is to deprive tumor cells of their reproductive potential. One approach to achieve this is by inducing tumor cell apoptosis. Accumulating evidences suggest that induction of apoptosis alone is insufficient to account for the therapeutic effect of radiotherapy. It has become obvious in the last few years that inhibition of the proliferative capacity of malignant cells following irradiation, especially with solid tumors, can occur via alternative cell death modalities or permanent cell cycle arrests, i.e., senescence. In this review, apoptosis and mitotic catastrophe, the two major cell deaths induced by radiation, are described and dissected in terms of activating mechanisms. Furthermore, treatment-induced senescence and its relevance for the outcome of radiotherapy of cancer will be discussed. The importance of p53 for the induction and execution of these different types of cell deaths is highlighted. The efficiency of radiotherapy and radioimmunotherapy has much to gain by understanding the cell death mechanisms that are induced in tumor cells following irradiation. Strategies to use specific inhibitors that will manipulate key molecules in these pathways in combination with radiation might potentiate therapy and enhance tumor cell kill.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  1. Jonathan EC, Bernhard EJ, McKenna WG. How does radiation kill cells? Curr Opin Chem Biol. 1999;3:77–83.

    CAS  PubMed  Google Scholar 

  2. Hollstein M, Sidransky D, Vogelstein B, Harris CC. P53 mutations in human cancers. Science. 1991;253:49–53.

    CAS  PubMed  Google Scholar 

  3. Soussi T, Beroud C. Assessing tp53 status in human tumours to evaluate clinical outcome. Nat Rev Cancer. 2001;1:233–40.

    CAS  PubMed  Google Scholar 

  4. Soussi T, Lozano G. P53 mutation heterogeneity in cancer. Biochem Biophys Res Commun. 2005;331:834–42.

    CAS  PubMed  Google Scholar 

  5. Igney FH, Krammer PH. Death and anti-death: tumour resistance to apoptosis. Nat Rev Cancer. 2002;2:277–88.

    CAS  PubMed  Google Scholar 

  6. Eriksson D, Riklund Ahlstrom K, Johansson L, Stigbrand T. Radiation induced cell deaths. In: Stigbrand T, Carlsson J, Adams G, editors. Targeted radionuclide tumor therapy—biological aspects. New York: Springer; 2008.

    Google Scholar 

  7. Gewirtz DA, Holt SE, Elmore LW. Accelerated senescence: an emerging role in tumor cell response to chemotherapy and radiation. Biochem Pharmacol. 2008;76:947–57.

    CAS  PubMed  Google Scholar 

  8. Jones KR, Elmore LW, Jackson-Cook C, Demasters G, Povirk LF, Holt SE, et al. P53-dependent accelerated senescence induced by ionizing radiation in breast tumour cells. Int J Radiat Biol. 2005;81:445–58.

    CAS  PubMed  Google Scholar 

  9. Lehmann BD, McCubrey JA, Jefferson HS, Paine MS, Chappell WH, Terrian DM. A dominant role for p53-dependent cellular senescence in radiosensitization of human prostate cancer cells. Cell Cycle. 2007;6:595–605.

    CAS  PubMed  Google Scholar 

  10. Ianzini F, Bertoldo A, Kosmacek EA, Phillips SL, Mackey MA. Lack of p53 function promotes radiation-induced mitotic catastrophe in mouse embryonic fibroblast cells. Cancer Cell Int. 2006;6:11.

    PubMed  Google Scholar 

  11. Efeyan A, Serrano M. P53: Guardian of the genome and policeman of the oncogenes. Cell Cycle. 2007;6:1006–10.

    CAS  PubMed  Google Scholar 

  12. Gottlieb TM, Oren M. P53 in growth control and neoplasia. Biochim Biophys Acta. 1996;1287:77–102.

    PubMed  Google Scholar 

  13. Lane DP. Cancer. P53, guardian of the genome. Nature. 1992;358:15–6.

    CAS  PubMed  Google Scholar 

  14. Marx J. Oncology. Recruiting the cell's own guardian for cancer therapy. Science. 2007;315:1211–3.

    CAS  PubMed  Google Scholar 

  15. Rodier F, Campisi J, Bhaumik D. Two faces of p53: aging and tumor suppression. Nucleic Acids Res. 2007;35:7475–84.

    CAS  PubMed  Google Scholar 

  16. Teodoro JG, Evans SK, Green MR. Inhibition of tumor angiogenesis by p53: a new role for the guardian of the genome. J Mol Med. 2007;85:1175–86.

    CAS  PubMed  Google Scholar 

  17. Helton ES, Chen X. P53 modulation of the DNA damage response. J Cell Biochem. 2007;100:883–96.

    CAS  PubMed  Google Scholar 

  18. Aylon Y, Oren M. Living with p53, dying of p53. Cell. 2007;130:597–600.

    CAS  PubMed  Google Scholar 

  19. Gudkov AV, Komarova EA. The role of p53 in determining sensitivity to radiotherapy. Nat Rev Cancer. 2003;3:117–29.

    CAS  PubMed  Google Scholar 

  20. Harris SL, Levine AJ. The p53 pathway: positive and negative feedback loops. Oncogene. 2005;24:2899–908.

    CAS  PubMed  Google Scholar 

  21. Laptenko O, Prives C. Transcriptional regulation by p53: one protein, many possibilities. Cell Death Differ. 2006;13:951–61.

    CAS  PubMed  Google Scholar 

  22. Liebermann DA, Hoffman B, Vesely D. P53 induced growth arrest versus apoptosis and its modulation by survival cytokines. Cell Cycle. 2007;6:166–70.

    CAS  PubMed  Google Scholar 

  23. Das S, Boswell SA, Aaronson SA, Lee SW. P53 promoter selection: choosing between life and death. Cell Cycle. 2008;7:154–7.

    CAS  PubMed  Google Scholar 

  24. Das S, Raj L, Zhao B, Kimura Y, Bernstein A, Aaronson SA, et al. Hzf determines cell survival upon genotoxic stress by modulating p53 transactivation. Cell. 2007;130:624–37.

    CAS  PubMed  Google Scholar 

  25. Tanaka T, Ohkubo S, Tatsuno I, Prives C. Hcas/cse1l associates with chromatin and regulates expression of select p53 target genes. Cell. 2007;130:638–50.

    CAS  PubMed  Google Scholar 

  26. Erenpreisa J, Cragg MS. Cancer: a matter of life cycle? Cell Biol Int. 2007;31:1507–10.

    CAS  PubMed  Google Scholar 

  27. Ganem NJ, Pellman D. Limiting the proliferation of polyploid cells. Cell. 2007;131:437–40.

    CAS  PubMed  Google Scholar 

  28. Ganem NJ, Storchova Z, Pellman D. Tetraploidy, aneuploidy and cancer. Curr Opin Genet Dev. 2007;17:157–62.

    CAS  PubMed  Google Scholar 

  29. Storchova Z, Pellman D. From polyploidy to aneuploidy, genome instability and cancer. Nat Rev Mol Cell Biol. 2004;5:45–54.

    CAS  PubMed  Google Scholar 

  30. Dewey WC, Ling CC, Meyn RE. Radiation-induced apoptosis: relevance to radiotherapy. Int J Radiat Oncol Biol Phys. 1995;33:781–96.

    CAS  PubMed  Google Scholar 

  31. Verheij M, Bartelink H. Radiation-induced apoptosis. Cell Tissue Res. 2000;301:133–42.

    CAS  PubMed  Google Scholar 

  32. Radford IR, Murphy TK, Radley JM, Ellis SL. Radiation response of mouse lymphoid and myeloid cell lines. Part ii. Apoptotic death is shown by all lines examined. Int J Radiat Biol. 1994;65:217–27.

    CAS  PubMed  Google Scholar 

  33. Shinomiya N. New concepts in radiation-induced apoptosis: 'premitotic apoptosis' and 'postmitotic apoptosis'. J Cell Mol Med. 2001;5:240–53.

    CAS  PubMed  Google Scholar 

  34. Clarke AR, Purdie CA, Harrison DJ, Morris RG, Bird CC, Hooper ML, et al. Thymocyte apoptosis induced by p53-dependent and independent pathways. Nature. 1993;362:849–52.

    CAS  PubMed  Google Scholar 

  35. Lowe SW, Schmitt EM, Smith SW, Osborne BA, Jacks T. P53 is required for radiation-induced apoptosis in mouse thymocytes. Nature. 1993;362:847–9.

    CAS  PubMed  Google Scholar 

  36. Riedl SJ, Shi Y. Molecular mechanisms of caspase regulation during apoptosis. Nat Rev Mol Cell Biol. 2004;5:897–907.

    CAS  PubMed  Google Scholar 

  37. Timmer JC, Salvesen GS. Caspase substrates. Cell Death Differ. 2007;14:66–72.

    CAS  PubMed  Google Scholar 

  38. Jin Z, El-Deiry WS. Overview of cell death signaling pathways. Cancer Biol Ther. 2005;4:139–63.

    Article  CAS  PubMed  Google Scholar 

  39. Alvarez S, Drane P, Meiller A, Bras M, Deguin-Chambon V, Bouvard V, et al. A comprehensive study of p53 transcriptional activity in thymus and spleen of gamma irradiated mouse: high sensitivity of genes involved in the two main apoptotic pathways. Int J Radiat Biol. 2006;82:761–70.

    CAS  PubMed  Google Scholar 

  40. Findley HW, Gu L, Yeager AM, Zhou M. Expression and regulation of bcl-2, bcl-xl, and bax correlate with p53 status and sensitivity to apoptosis in childhood acute lymphoblastic leukemia. Blood. 1997;89:2986–93.

    CAS  PubMed  Google Scholar 

  41. Kobayashi T, Ruan S, Jabbur JR, Consoli U, Clodi K, Shiku H, et al. Differential p53 phosphorylation and activation of apoptosis-promoting genes bax and fas/apo-1 by irradiation and ara-c treatment. Cell Death Differ. 1998;5:584–91.

    CAS  PubMed  Google Scholar 

  42. Zhan Q, Fan S, Bae I, Guillouf C, Liebermann DA, O'Connor PM, et al. Induction of bax by genotoxic stress in human cells correlates with normal p53 status and apoptosis. Oncogene. 1994;9:3743–51.

    CAS  PubMed  Google Scholar 

  43. Erlacher M, Michalak EM, Kelly PN, Labi V, Niederegger H, Coultas L, et al. Bh3-only proteins puma and bim are rate-limiting for gamma-radiation- and glucocorticoid-induced apoptosis of lymphoid cells in vivo. Blood. 2005;106:4131–8.

    CAS  PubMed  Google Scholar 

  44. Michalak EM, Villunger A, Adams JM, Strasser A. In several cell types tumour suppressor p53 induces apoptosis largely via puma but noxa can contribute. Cell Death Differ. 2008;15:1019–29.

    CAS  PubMed  Google Scholar 

  45. Jeffers JR, Parganas E, Lee Y, Yang C, Wang J, Brennan J, et al. Puma is an essential mediator of p53-dependent and -independent apoptotic pathways. Cancer Cell. 2003;4:321–8.

    CAS  PubMed  Google Scholar 

  46. Villunger A, Michalak EM, Coultas L, Mullauer F, Bock G, Ausserlechner MJ, et al. P53- and drug-induced apoptotic responses mediated by bh3-only proteins puma and noxa. Science. 2003;302:1036–8.

    CAS  PubMed  Google Scholar 

  47. Lin Y, Ma W, Benchimol S. Pidd, a new death-domain-containing protein, is induced by p53 and promotes apoptosis. Nat Genet. 2000;26:122–7.

    CAS  PubMed  Google Scholar 

  48. Fei P, El-Deiry WS. P53 and radiation responses. Oncogene. 2003;22:5774–83.

    CAS  PubMed  Google Scholar 

  49. Haldar S, Negrini M, Monne M, Sabbioni S, Croce CM. Down-regulation of bcl-2 by p53 in breast cancer cells. Cancer Res. 1994;54:2095–7.

    CAS  PubMed  Google Scholar 

  50. Miyashita T, Harigai M, Hanada M, Reed JC. Identification of a p53-dependent negative response element in the bcl-2 gene. Cancer Res. 1994;54:3131–5.

    CAS  PubMed  Google Scholar 

  51. Hoffman WH, Biade S, Zilfou JT, Chen J, Murphy M. Transcriptional repression of the anti-apoptotic survivin gene by wild type p53. J Biol Chem. 2002;277:3247–57.

    CAS  PubMed  Google Scholar 

  52. Zhou M, Gu L, Li F, Zhu Y, Woods WG, Findley HW. DNA damage induces a novel p53-survivin signaling pathway regulating cell cycle and apoptosis in acute lymphoblastic leukemia cells. J Pharmacol Exp Ther. 2002;303:124–31.

    CAS  PubMed  Google Scholar 

  53. Wang X. The expanding role of mitochondria in apoptosis. Genes Dev. 2001;15:2922–33.

    CAS  PubMed  Google Scholar 

  54. Jiang X, Wang X. Cytochrome c promotes caspase-9 activation by inducing nucleotide binding to apaf-1. J Biol Chem. 2000;275:31199–203.

    CAS  PubMed  Google Scholar 

  55. Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES, et al. Cytochrome c and datp-dependent formation of apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell. 1997;91:479–89.

    CAS  PubMed  Google Scholar 

  56. Rodriguez J, Lazebnik Y. Caspase-9 and apaf-1 form an active holoenzyme. Genes Dev. 1999;13:3179–84.

    CAS  PubMed  Google Scholar 

  57. Embree-Ku M, Venturini D, Boekelheide K. Fas is involved in the p53-dependent apoptotic response to ionizing radiation in mouse testis. Biol Reprod. 2002;66:1456–61.

    CAS  PubMed  Google Scholar 

  58. Sheard MA, Uldrijan S, Vojtesek B. Role of p53 in regulating constitutive and x-radiation-inducible cd95 expression and function in carcinoma cells. Cancer Res. 2003;63:7176–84.

    CAS  PubMed  Google Scholar 

  59. Sheard MA, Vojtesek B, Janakova L, Kovarik J, Zaloudik J. Up-regulation of fas (cd95) in human p53wild-type cancer cells treated with ionizing radiation. Int J Cancer. 1997;73:757–62.

    CAS  PubMed  Google Scholar 

  60. Burns TF, Bernhard EJ, El-Deiry WS. Tissue specific expression of p53 target genes suggests a key role for killer/dr5 in p53-dependent apoptosis in vivo. Oncogene. 2001;20:4601–12.

    CAS  PubMed  Google Scholar 

  61. Sheikh MS, Burns TF, Huang Y, Wu GS, Amundson S, Brooks KS, et al. P53-dependent and -independent regulation of the death receptor killer/dr5 gene expression in response to genotoxic stress and tumor necrosis factor alpha. Cancer Res. 1998;58:1593–8.

    CAS  PubMed  Google Scholar 

  62. Wu GS, Burns TF, McDonald 3rd ER, Jiang W, Meng R, Krantz ID, et al. Killer/dr5 is a DNA damage-inducible p53-regulated death receptor gene. Nat Genet. 1997;17:141–3.

    CAS  PubMed  Google Scholar 

  63. Kastan M. On the trail from p53 to apoptosis? Nat Genet. 1997;17:130–1.

    CAS  PubMed  Google Scholar 

  64. Scaffidi C, Fulda S, Srinivasan A, Friesen C, Li F, Tomaselli KJ, et al. Two cd95 (apo-1/fas) signaling pathways. EMBO J. 1998;17:1675–87.

    CAS  PubMed  Google Scholar 

  65. Taylor RC, Cullen SP, Martin SJ. Apoptosis: controlled demolition at the cellular level. Nat Rev Mol Cell Biol. 2007;9:231–41.

    Google Scholar 

  66. Luo X, Budihardjo I, Zou H, Slaughter C, Wang X. Bid, a bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors. Cell. 1998;94:481–90.

    CAS  PubMed  Google Scholar 

  67. Galluzzi L, Maiuri MC, Vitale I, Zischka H, Castedo M, Zitvogel L, et al. Cell death modalities: Classification and pathophysiological implications. Cell Death Differ. 2007;14:1237–43.

    CAS  PubMed  Google Scholar 

  68. Roninson IB, Broude EV, Chang BD. If not apoptosis, then what? Treatment-induced senescence and mitotic catastrophe in tumor cells. Drug Resist Updat. 2001;4:303–13.

    CAS  PubMed  Google Scholar 

  69. Bhattathiri NV, Bharathykkutty C, Prathapan R, Chirayathmanjiyil DA, Nair KM. Prediction of radiosensitivity of oral cancers by serial cytological assay of nuclear changes. Radiother Oncol. 1998;49:61–5.

    CAS  PubMed  Google Scholar 

  70. Bhattathiri NV, Bindu L, Remani P, Chandralekha B, Nair KM. Radiation-induced acute immediate nuclear abnormalities in oral cancer cells: serial cytologic evaluation. Acta Cytol. 1998;42:1084–90.

    CAS  PubMed  Google Scholar 

  71. Eriksson D, Joniani HM, Sheikholvaezin A, Lofroth PO, Johansson L. Combined low dose radio- and radioimmunotherapy of experimental hela hep 2 tumours. Eur J Nucl Med Mol Imaging. 2003;30:895–906.

    CAS  PubMed  Google Scholar 

  72. Eriksson D, Lofroth PO, Johansson L, Riklund KA, Stigbrand T. Cell cycle disturbances and mitotic catastrophes in hela hep2 cells following 2.5 to 10 gy of ionizing radiation. Clin Cancer Res. 2007;13:5501s–8.

    CAS  PubMed  Google Scholar 

  73. Somosy Z. Radiation response of cell organelles. Micron. 2000;31:165–81.

    CAS  PubMed  Google Scholar 

  74. Castedo M, Kroemer G. [mitotic catastrophe: a special case of apoptosis]. J Soc Biol. 2004;198:97–103.

    CAS  PubMed  Google Scholar 

  75. Erenpreisa J, Kalejs M, Ianzini F, Kosmacek EA, Mackey MA, Emzinsh D, et al. Segregation of genomes in polyploid tumour cells following mitotic catastrophe. Cell Biol Int. 2005;29:1005–11.

    CAS  PubMed  Google Scholar 

  76. Bourke E, Dodson H, Merdes A, Cuffe L, Zachos G, Walker M, et al. DNA damage induces chk1-dependent centrosome amplification. EMBO Rep. 2007;8:603–9.

    CAS  PubMed  Google Scholar 

  77. Dodson H, Wheatley SP, Morrison CG. Involvement of centrosome amplification in radiation-induced mitotic catastrophe. Cell Cycle. 2007;6:364–70.

    CAS  PubMed  Google Scholar 

  78. Kawamura K, Fujikawa-Yamamoto K, Ozaki M, Iwabuchi K, Nakashima H, Domiki C, et al. Centrosome hyperamplification and chromosomal damage after exposure to radiation. Oncology. 2004;67:460–70.

    CAS  PubMed  Google Scholar 

  79. Kawamura K, Morita N, Domiki C, Fujikawa-Yamamoto K, Hashimoto M, Iwabuchi K, et al. Induction of centrosome amplification in p53 sirna-treated human fibroblast cells by radiation exposure. Cancer Sci. 2006;97:252–8.

    CAS  PubMed  Google Scholar 

  80. Eriksson D, Lofroth PO, Johansson L, Riklund K, Stigbrand T. Apoptotic signalling in hela hep2 cells following 5 gy of cobalt-60 gamma radiation. Anticancer Res. 2009;29:4361–6.

    CAS  PubMed  Google Scholar 

  81. Loffler H, Lukas J, Bartek J, Kramer A. Structure meets function–centrosomes, genome maintenance and the DNA damage response. Exp Cell Res. 2006;312:2633–40.

    PubMed  Google Scholar 

  82. Eriksson D, Blomberg J, Lindgren T, Lofroth PO, Johansson L, Riklund K, et al. Iodine-131 induces mitotic catastrophes and activates apoptotic pathways in hela hep2 cells. Cancer Biother Radiopharm. 2008;23:541–9.

    CAS  PubMed  Google Scholar 

  83. Wang Y, Ji P, Liu J, Broaddus RR, Xue F, Zhang W. Centrosome-associated regulators of the g(2)/m checkpoint as targets for cancer therapy. Mol Cancer. 2009;8:8.

    PubMed  Google Scholar 

  84. Castedo M, Perfettini JL, Roumier T, Andreau K, Medema R, Kroemer G. Cell death by mitotic catastrophe: a molecular definition. Oncogene. 2004;23:2825–37.

    CAS  PubMed  Google Scholar 

  85. Hanashiro K, Kanai M, Geng Y, Sicinski P, Fukasawa K. Roles of cyclins a and e in induction of centrosome amplification in p53-compromised cells. Oncogene. 2008;27:5288–302.

    CAS  PubMed  Google Scholar 

  86. Fukasawa K. Oncogenes and tumour suppressors take on centrosomes. Nat Rev Cancer. 2007;7:911–24.

    CAS  PubMed  Google Scholar 

  87. Fukasawa K. P53, cyclin-dependent kinase and abnormal amplification of centrosomes. Biochim Biophys Acta. 2008;1786:15–23.

    CAS  PubMed  Google Scholar 

  88. Castedo M, Perfettini JL, Roumier T, Valent A, Raslova H, Yakushijin K, et al. Mitotic catastrophe constitutes a special case of apoptosis whose suppression entails aneuploidy. Oncogene. 2004;23:4362–70.

    CAS  PubMed  Google Scholar 

  89. Abend M. Reasons to reconsider the significance of apoptosis for cancer therapy. Int J Radiat Biol. 2003;79:927–41.

    CAS  PubMed  Google Scholar 

  90. Ruth AC, Roninson IB. Effects of the multidrug transporter p-glycoprotein on cellular responses to ionizing radiation. Cancer Res. 2000;60:2576–8.

    CAS  PubMed  Google Scholar 

  91. Jallepalli PV, Lengauer C. Chromosome segregation and cancer: cutting through the mystery. Nat Rev Cancer. 2001;1:109–17.

    CAS  PubMed  Google Scholar 

  92. Norbury CJ, Zhivotovsky B. DNA damage-induced apoptosis. Oncogene. 2004;23:2797–808.

    CAS  PubMed  Google Scholar 

  93. Zhivotovsky B, Orrenius S. Caspase-2 function in response to DNA damage. Biochem Biophys Res Commun. 2005;331:859–67.

    CAS  PubMed  Google Scholar 

  94. Weaver BA, Cleveland DW. Decoding the links between mitosis, cancer, and chemotherapy: the mitotic checkpoint, adaptation, and cell death. Cancer Cell. 2005;8:7–12.

    CAS  PubMed  Google Scholar 

  95. Yamada HY, Gorbsky GJ. Spindle checkpoint function and cellular sensitivity to antimitotic drugs. Mol Cancer Ther. 2006;5:2963–9.

    CAS  PubMed  Google Scholar 

  96. Rieder CL, Maiato H. Stuck in division or passing through: What happens when cells cannot satisfy the spindle assembly checkpoint. Dev Cell. 2004;7:637–51.

    CAS  PubMed  Google Scholar 

  97. Castedo M, Coquelle A, Vivet S, Vitale I, Kauffmann A, Dessen P, et al. Apoptosis regulation in tetraploid cancer cells. EMBO J. 2006;25:2584–95.

    CAS  PubMed  Google Scholar 

  98. Uetake Y, Sluder G. Cell cycle progression after cleavage failure: mammalian somatic cells do not possess a "Tetraploidy checkpoint". J Cell Biol. 2004;165:609–15.

    CAS  PubMed  Google Scholar 

  99. Blagosklonny MV, Demidenko ZN, Giovino M, Szynal C, Donskoy E, Herrmann RA, et al. Cytostatic activity of paclitaxel in coronary artery smooth muscle cells is mediated through transient mitotic arrest followed by permanent post-mitotic arrest: comparison with cancer cells. Cell Cycle. 2006;5:1574–9.

    CAS  PubMed  Google Scholar 

  100. Klein LE, Freeze BS, Smith 3rd AB, Horwitz SB. The microtubule stabilizing agent discodermolide is a potent inducer of accelerated cell senescence. Cell Cycle. 2005;4:501–7.

    CAS  PubMed  Google Scholar 

  101. Casenghi M, Mangiacasale R, Tuynder M, Caillet-Fauquet P, Elhajouji A, Lavia P, et al. P53-independent apoptosis and p53-dependent block of DNA rereplication following mitotic spindle inhibition in human cells. Exp Cell Res. 1999;250:339–50.

    CAS  PubMed  Google Scholar 

  102. Chang BD, Broude EV, Dokmanovic M, Zhu H, Ruth A, Xuan Y, et al. A senescence-like phenotype distinguishes tumor cells that undergo terminal proliferation arrest after exposure to anticancer agents. Cancer Res. 1999;59:3761–7.

    CAS  PubMed  Google Scholar 

  103. Chang BD, Swift ME, Shen M, Fang J, Broude EV, Roninson IB. Molecular determinants of terminal growth arrest induced in tumor cells by a chemotherapeutic agent. Proc Natl Acad Sci U S A. 2002;99:389–94.

    CAS  PubMed  Google Scholar 

  104. Elmore LW, Rehder CW, Di X, McChesney PA, Jackson-Cook CK, Gewirtz DA, et al. Adriamycin-induced senescence in breast tumor cells involves functional p53 and telomere dysfunction. J Biol Chem. 2002;277:35509–15.

    CAS  PubMed  Google Scholar 

  105. Han Z, Wei W, Dunaway S, Darnowski JW, Calabresi P, Sedivy J, et al. Role of p21 in apoptosis and senescence of human colon cancer cells treated with camptothecin. J Biol Chem. 2002;277:17154–60.

    CAS  PubMed  Google Scholar 

  106. Haq R, Brenton JD, Takahashi M, Finan D, Finkielsztein A, Damaraju S, et al. Constitutive p38hog mitogen-activated protein kinase activation induces permanent cell cycle arrest and senescence. Cancer Res. 2002;62:5076–82.

    CAS  PubMed  Google Scholar 

  107. Mirzayans R, Scott A, Cameron M, Murray D. Induction of accelerated senescence by gamma radiation in human solid tumor-derived cell lines expressing wild-type tp53. Radiat Res. 2005;163:53–62.

    CAS  PubMed  Google Scholar 

  108. Quick QA, Gewirtz DA. An accelerated senescence response to radiation in wild-type p53 glioblastoma multiforme cells. J Neurosurg. 2006;105:111–8.

    CAS  PubMed  Google Scholar 

  109. Roberson RS, Kussick SJ, Vallieres E, Chen SY, Wu DY. Escape from therapy-induced accelerated cellular senescence in p53-null lung cancer cells and in human lung cancers. Cancer Res. 2005;65:2795–803.

    CAS  PubMed  Google Scholar 

  110. Roninson IB. Tumor cell senescence in cancer treatment. Cancer Res. 2003;63:2705–15.

    CAS  PubMed  Google Scholar 

  111. Schmitt CA. Cellular senescence and cancer treatment. Biochim Biophys Acta. 2007;1775:5–20.

    CAS  PubMed  Google Scholar 

  112. Schmitt CA, Fridman JS, Yang M, Lee S, Baranov E, Hoffman RM, et al. A senescence program controlled by p53 and p16ink4a contributes to the outcome of cancer therapy. Cell. 2002;109:335–46.

    CAS  PubMed  Google Scholar 

  113. Schwarze SR, Fu VX, Desotelle JA, Kenowski ML, Jarrard DF. The identification of senescence-specific genes during the induction of senescence in prostate cancer cells. Neoplasia. 2005;7:816–23.

    CAS  PubMed  Google Scholar 

  114. te Poele RH, Okorokov AL, Jardine L, Cummings J, Joel SP. DNA damage is able to induce senescence in tumor cells in vitro and in vivo. Cancer Res. 2002;62:1876–83.

    Google Scholar 

  115. Bataini JP, Belloir C, Mazabraud A, Pilleron JP, Cartigny A, Jaulerry C, et al. Desmoid tumors in adults: The role of radiotherapy in their management. Am J Surg. 1988;155:754–60.

    CAS  PubMed  Google Scholar 

  116. Cox JD, Kline RW. Do prostatic biopsies 12 months or more after external irradiation for adenocarcinoma, stage iii, predict long-term survival? Int J Radiat Oncol Biol Phys. 1983;9:299–303.

    CAS  PubMed  Google Scholar 

  117. Shay JW, Roninson IB. Hallmarks of senescence in carcinogenesis and cancer therapy. Oncogene. 2004;23:2919–33.

    CAS  PubMed  Google Scholar 

  118. Deng Y, Chan SS, Chang S. Telomere dysfunction and tumour suppression: The senescence connection. Nat Rev Cancer. 2008;8:450–8.

    CAS  PubMed  Google Scholar 

  119. Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res. 1961;25:585–621.

    Google Scholar 

  120. Stein GH, Dulic V. Origins of g1 arrest in senescent human fibroblasts. Bioessays. 1995;17:537–43.

    CAS  PubMed  Google Scholar 

  121. Collado M, Serrano M. Senescence in tumours: evidence from mice and humans. Nat Rev Cancer. 2010;10:51–7.

    CAS  PubMed  Google Scholar 

  122. Rodier F, Coppe JP, Patil CK, Hoeijmakers WA, Munoz DP, Raza SR, et al. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat Cell Biol. 2009;11:973–9.

    CAS  PubMed  Google Scholar 

  123. Suzuki K, Mori I, Nakayama Y, Miyakoda M, Kodama S, Watanabe M. Radiation-induced senescence-like growth arrest requires tp53 function but not telomere shortening. Radiat Res. 2001;155:248–53.

    CAS  PubMed  Google Scholar 

  124. Dai CY, Enders GH. P16 ink4a can initiate an autonomous senescence program. Oncogene. 2000;19:1613–22.

    CAS  PubMed  Google Scholar 

  125. Steiner MS, Wang Y, Zhang Y, Zhang X, Lu Y. P16/mts1/ink4a suppresses prostate cancer by both prb dependent and independent pathways. Oncogene. 2000;19:1297–306.

    CAS  PubMed  Google Scholar 

  126. Sugrue MM, Shin DY, Lee SW, Aaronson SA. Wild-type p53 triggers a rapid senescence program in human tumor cells lacking functional p53. Proc Natl Acad Sci U S A. 1997;94:9648–53.

    CAS  PubMed  Google Scholar 

  127. Zhang H. Molecular signaling and genetic pathways of senescence: its role in tumorigenesis and aging. J Cell Physiol. 2007;210:567–74.

    CAS  PubMed  Google Scholar 

  128. Coppe JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol. 2010;5:99–118.

    CAS  PubMed  Google Scholar 

  129. Novakova Z, Hubackova S, Kosar M, Janderova-Rossmeislova L, Dobrovolna J, Vasicova P, et al. Cytokine expression and signaling in drug-induced cellular senescence. Oncogene. 2010;29:273–84.

    CAS  PubMed  Google Scholar 

  130. Xue W, Zender L, Miething C, Dickins RA, Hernando E, Krizhanovsky V, et al. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature. 2007;445:656–60.

    CAS  PubMed  Google Scholar 

  131. Bassett EA, Wang W, Rastinejad F, El-Deiry WS. Structural and functional basis for therapeutic modulation of p53 signaling. Clin Cancer Res. 2008;14:6376–86.

    CAS  PubMed  Google Scholar 

  132. Brown CJ, Lain S, Verma CS, Fersht AR, Lane DP. Awakening guardian angels: drugging the p53 pathway. Nat Rev Cancer. 2009;9:862–73.

    CAS  PubMed  Google Scholar 

  133. Wang W, El-Deiry WS. Restoration of p53 to limit tumor growth. Curr Opin Oncol. 2008;20:90–6.

    PubMed  Google Scholar 

  134. Wiman KG. Restoration of wild-type p53 function in human tumors: Strategies for efficient cancer therapy. Adv Cancer Res. 2007;97:321–38.

    CAS  PubMed  Google Scholar 

  135. Kawabe T. G2 checkpoint abrogators as anticancer drugs. Mol Cancer Ther. 2004;3:513–9.

    CAS  PubMed  Google Scholar 

  136. Lapenna S, Giordano A. Cell cycle kinases as therapeutic targets for cancer. Nat Rev Drug Discov. 2009;8:547–66.

    CAS  PubMed  Google Scholar 

  137. Gautschi O, Heighway J, Mack PC, Purnell PR, Lara Jr PN, Gandara DR. Aurora kinases as anticancer drug targets. Clin Cancer Res. 2008;14:1639–48.

    CAS  PubMed  Google Scholar 

  138. Kitzen JJ, de Jonge MJ, Verweij J. Aurora kinase inhibitors. Crit Rev Oncol Hematol. 2010;73:99–110.

    CAS  PubMed  Google Scholar 

  139. Mita AC, Mita MM, Nawrocki ST, Giles FJ. Survivin: Key regulator of mitosis and apoptosis and novel target for cancer therapeutics. Clin Cancer Res. 2008;14:5000–5.

    CAS  PubMed  Google Scholar 

  140. Kwon M, Godinho SA, Chandhok NS, Ganem NJ, Azioune A, Thery M, et al. Mechanisms to suppress multipolar divisions in cancer cells with extra centrosomes. Genes Dev. 2008;22:2189–203.

    CAS  PubMed  Google Scholar 

  141. Rello-Varona S, Vitale I, Kepp O, Senovilla L, Jemaa M, Metivier D, et al. Preferential killing of tetraploid tumor cells by targeting the mitotic kinesin eg5. Cell Cycle. 2009;8:1030–5.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Financial support from the Swedish Cancer Society, the County of Västerbotten, and the Medical Faculty at Umeå University for research related to the content of this chapter is acknowledged.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to David Eriksson.

Additional information

Part of this content was presented as the Abbott Award lecture at ISOBM in Amsterdam 2009.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Eriksson, D., Stigbrand, T. Radiation-induced cell death mechanisms. Tumor Biol. 31, 363–372 (2010). https://doi.org/10.1007/s13277-010-0042-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13277-010-0042-8

Keywords

Navigation