784
Views
40
CrossRef citations to date
0
Altmetric
RADIATION HORMESIS: PUTATIVE MECHANISMS

Radiation hormesis: Autophagy and other cellular mechanisms

Pages 619-628 | Received 11 Mar 2012, Accepted 30 May 2012, Published online: 28 Jun 2012

References

  • Ahmed KM, Nantajit D, Fan M, Murley JS, Grdina DJ, Li JJ. 2009. Coactivation of ATM/ERK/NF-kappaB in the low-dose radiation-induced radioadaptive response in human skin keratinocytes. Free Radical Biology and Medicine 46:1543–1550.
  • Apostolova N, Blas-Garcia A, Esplugues JV. 2011. Mitochondria sentencing about cellular life and death: A matter of oxidative stress. Current Pharmaceutical Design 17:4047–4060.
  • Averbeck D. 2010. Non-targeted effects as a paradigm breaking evidence. Mutation Research 687:7–12.
  • Aypar U, Morgan WF, Baulch JE. 2011. Radiation-induced genomic instability: Are epigenetic mechanisms the missing link? International Journal of Radiation Biology 87:179–191.
  • Azad MB, Chen Y, Gibson SB. 2009. Regulation of autophagy by reactive oxygen species (ROS): Implications for cancer progression and treatment. Antioxidants & Redox Signaling 11:77–790.
  • Azzam EI. 2011. Exposure to low level environmental agents: The induction of hormesis. Mutation Research 726: 89–90.
  • Balaban RS, Nemoto S, Finkel T. 2005. Mitochondria, oxidants, and aging. Cell 120:483–495.
  • Barquinero JF, Barrios L, Caballin MR, Miro R, Ribas M, Subias A, Egozcue J. 1995. Occupational exposure to radiation induces an adaptive response in human lymphocytes. International Journal of Radiation Biology 67:187–191.
  • Benard G, Bellance N, Jose C, Melser S, Nouette-Gaulain K, Rossignol R. 2010. Multi-site control and regulation of mitochondrial energy production. Biochimica et Biophysica Acta 1797:698–709.
  • Bhattacharjee D. 1996. Role of radioadaptation on radiation-induced thymic lymphoma in mice. Mutation Research 358:231–235.
  • Boothman DA, Odegaard E, Yang CR, Hosley K, Mendonca MS. 1998. Molecular analyses of adaptive survival responses (ASRs): Role of ASRs in radiotherapy. Human and Experimental Toxicology 17:448–453.
  • Bjelakovic G, Nikolova D, Simonetti RG, Gluud C. 2008a. Antioxidant supplements for preventing gastrointestinal cancers. Cochrane Database of Systematic Reviews CD004183.
  • Bjelakovic G, Nikolova D, Gluud LL, Simonetti RG, Gluud C. 2008b. Antioxidant supplements for prevention of mortality in healthy participants and patients with various diseases. Cochrane Database of Systematic Reviews CD007176.
  • Broome EJ, Brown DL, Mitchel RE. 2002. Dose responses for adaption to low doses of (60)Co gamma rays and (3)H beta particles in normal human fibroblasts. Radiation Research 158:181–186.
  • Calabrese EJ, Iavicoli I, Calabrese V. 2012a. Hormesis: Why it is important to biogerontologists. Biogerontology 13:215–235.
  • Calabrese EJ, McCarthy ME, Kenyon E. 1987. The occurrence of chemically induced hormesis. Health Physics 52:531–541.
  • Calabrese EJ, Stanek EJ, III, Nascarella MA. 2011. Evidence for hormesis in mutagenicity dose-response relationships. Mutation Research 726: 91–97.
  • Calabrese V, Cornelius C, Dinkova-Kostova AT, Iavicoli I, Di PR, Koverech A, Cuzzocrea S, Rizzarelli E, Calabrese EJ. 2012b. Cellular stress responses, hormetic phytochemicals and vitagenes in aging and longevity. Biochimica et Biophysica Acta 1822:753–783.
  • Chakrabarti A, Chen AW, Varner JD. 2011. A review of the mammalian unfolded protein response. Biotechnology and Bioengineering 108:2777–2793.
  • Chen Y, Azad MB, Gibson SB. 2009. Superoxide is the major reactive oxygen species regulating autophagy. Cell Death & Differentiation 16:1040–1052.
  • Chen Y, Gibson SB. 2008. Is mitochondrial generation of reactive oxygen species a trigger for autophagy? Autophagy 4:246–248.
  • Chen Y, Klionsky DJ. 2011. The regulation of autophagy – unanswered questions. Journal of Cell Science 124:161–170.
  • Daiber A. Redox signaling (cross-talk) from and to mitochondria involves mitochondrial pores. and reactive oxygen species . 2010.Biochimica et Biophysica Acta 1797:897–906.
  • Dalby KN, Tekedereli I, Lopez-Berestein G, Ozpolat B. 2010. Targeting the prodeath and prosurvival functions of autophagy as novel therapeutic strategies in cancer. Autophagy 6:322–329.
  • Dauer LT, Brooks AL, Hoel DG, Morgan WF, Stram D, Tran P. 2010. Review and evaluation of updated research on the health effects associated with low-dose ionising radiation. Radiation Protection & Dosimetry 140:103–136.
  • Dewaele M, Maes H, Agostinis P. 2010. ROS-mediated mechanisms of autophagy stimulation and their relevance in cancer therapy. Autophagy 26:838–854.
  • Druesne-Pecollo N, Latino-Martel P, Norat T, Barrandon E, Bertrais S, Galan P, Hercberg S. 2010. Beta-carotene supplementation and cancer risk: A systematic review and metaanalysis of randomized controlled trials. International Journal of Cancer 127:172–184.
  • Fan M, Ahmed KM, Coleman MC, Spitz DR, Li JJ. 2007. Nuclear factor-kappaB and manganese superoxide dismutase mediate adaptive in low-dose irradiated mouse skin epithelial cells. Cancer Research 67:3220–3228.
  • Faust F, Kassie F, Knasmuller S, Boedecker RH, Mann M, Mersch-Sundermann V. 2004. The use of the alkaline comet assay with lymphocytes in human biomonitoring studies. Mutation Research 566:209–229.
  • Feinendegen LE. 2002. Reactive oxygen species in cell responses to toxic agents. Human & Experimental Toxicology 21:85–90.
  • Feinendegen LE. 2005. Evidence for beneficial low level radiation effects radioresistance and radiation hormesis. British Journal of Radiology 78:3–7.
  • Ghiassi-Nejad M, Mortazavi SM, Cameron JR, Niroomand-rad A, Karam PA. 2002. Very high background radiation areas of Ramsar, Iran: Preliminary biological studies. Health Physics 82:87–93.
  • Goodhead DT. 2010. New radiobiological, radiation risk and radiation protection paradigms. Mutation Research 687:13–16.
  • Goodhead DT. 2011. Panel discussion: Do non-targeted effects impact the relation between microdosimetry and risk? Radiation Protection & Dosimetry 143:554–556.
  • Gottlieb RA, Carreira RS. 2010. Autophagy in health and disease. 5. Mitophagy as a way of life. American Journal of Physiology – Cell Physiology 299:C203–210.
  • Green DR, Galluzzi L, Kroemer G. 2011. Mitochondria and the autophagy-inflammation-cell death axis in organismal aging. Science 333:1109–1112.
  • Greer EL, Brunet A. 2005. FOXO transcription factors at the interface between longevity and tumor suppression. Oncogene 24:7410–7425.
  • Greer EL, Brunet A. 2008. FOXO transcription factors in ageing and cancer. Acta Physiologica 192:19–28.
  • Grivennikova VG, Vinogradov AD. 2006. Generation of superoxide by the mitochondrial Complex I. Biochimica et Biophysica Acta 1757:553–561.
  • Hamada N, Matsumoto H, Hara T, Kobayashi Y. 2007. Intercellular and intracellular signaling pathways mediating ionizing radiation-induced bystander effects. Journal of Radiation Research (Tokyo) 48:87–95.
  • Hamanaka RB, Chandel NS. 2010. Mitochondrial reactive oxygen species regulate cellular signaling and dictate biological outcomes. Trends in Biochemical Sciences 35:505–513.
  • Harman D. 2003. The free radical theory of aging. Antioxidants & Redox Signaling 5:557–561.
  • Hei TK, Zhou H, Ivanov VN, Hong M, Lieberman HB, Brenner DJ, Amundson SA, Geard CR. 2008. Mechanism of radiation-induced bystander effects: A unifying model. Journal of Pharmacy and Pharmacology 60:943–950.
  • Hendry JH, Simon SL, Wick A, Sohrabi M, Burkart W, Cardis E, Laurier D, Tirmarche M, Hayata I. 2009. Human exposure to high natural background radiation: What can it teach us about radiation risks? Journal of Radiological Protection 29:A29–42.
  • Horie K, Kubo K, Yonezawa M. 2002. p53 dependency of radio-adaptive responses in endogenous spleen colonies and peripheral blood-cell counts in C57BL mice. Journal of Radiation Research 43:353–360.
  • International Commission on Radiological Protection (ICRP) publication 103. 2007. The 2007 recommendations of the ICRP. Annex A. Biological and Epidemiological Information on Health Risks Attributable to Ionising Radiation. Annals of ICRP 37:137–332.
  • Ilnytskyy Y, Kovalchuk O. 2011. Non-targeted radiation effects – an epigenetic connection. Mutation Research 714:113–125.
  • Ina Y, Tanooka H, Yamada T, Sakai K. 2005. Suppression of thymic lymphoma induction by life-long low-dose-rate irradiation accompanied by immune activation in C57BL/6 mice. Radiation Research 163:153–158.
  • Ito M, Shibamoto Y, Ayakawa S, Tomita N, Sugie C, Ogino H. 2007. Low-dose whole-body irradiation induced radioadaptive response in C57BL/6 mice. Journal of Radiation Research (Tokyo) 48:455–460.
  • Jaworowski Z. 2010. Radiation hormesis – a remedy for fear. Human & Experimental Toxicology 29:263–270.
  • Jiang F, Zhang Y, Dusting GJ. 2011. NADPH oxidase-mediated redox signaling: Roles in cellular stress response, stress tolerance, and tissue repair. Pharmacological Reviews 63:218–242.
  • Jolly D, Meyer J. 2009. A brief review of radiation hormesis. Australasian Physical & Engineering Sciences in Medicine 32:180–187.
  • Juarez JC, Manuia M, Burnett ME, Betancourt O, Boivin B, Shaw DE, Tonks NK, Mazar AP, Donate F. 2008. Superoxide dismutase 1 (SOD1) is essential for H2O2-mediated oxidation and inactivation of phosphatases in growth factor signaling . Proceedings of the National Academy of Sciences of the USA 105:7147–7152.
  • Kim H, Bernard ME, Flickinger J, Epperly MW, Wang H, Dixon TM, Shields D, Houghton F, Zhang X, Greenberger JS. 2011. The autophagy-inducing drug carbamazepine is a radiation protector and mitigator. International Journal of Radiation Biology 87: 1052–1060.
  • Kimmelman AC. 2011. The dynamic nature of autophagy in cancer. Genes & Development 25:1999–2010.
  • Klaunig JE, Kamendulis LM, Hocevar BA. 2010. Oxidative stress and oxidative damage in carcinogenesis. Toxicologic Pathology 38:96–109.
  • Klaunig JE, Wang Z, Pu X, Zhou S. 2011. Oxidative stress and oxidative damage in chemical carcinogenesis. Toxicology and Applied Pharmacology 254:86–99.
  • Kloet DE, Burgering BM. 2011. The PKB /FOXO switch in aging and cancer. Biochimica et Biophysica Acta 1813:1926–1937.
  • Kourtis N, Tavernarakis N. 2011. Cellular stress response pathways and ageing: Intricate molecular relationships. EMBO Journal 30:2520–2531.
  • Kovalchuk O, Baulch JE. 2008. Epigenetic changes and nontargeted radiation effects – is there a link? Environmental and Molecular Mutagenesis 49:16–25.
  • Kroemer G, Marino G, Levine B. 2010. Autophagy and the integrated stress response. Molecular Cell 40:280–293.
  • Kumar PR, Cheriyan VD, Seshadri M. 2012. Evaluation of spontaneous DNA damage in lymphocytes of healthy adult individuals from high-level natural radiation areas of Kerala in India. Radiation Research 177:643–650.
  • Leach JK, Van TG, Lin PS, Schmidt-Ullrich R, Mikkelsen RB. 2001. Ionizing radiation-induced, mitochondria-dependent generation of reactive oxygen/nitrogen. Cancer Research 61:3894–3901.
  • Lee J, Giordano S, Zhang J. 2012. Autophagy, mitochondria and oxidative stress: Cross-talk and redox signalling. Biochemical Journal 441:523–540.
  • Liang X, So YH, Cui J, Ma K, Xu X, Zhao Y, Cai L, Li W. 2011. The low-dose ionizing radiation stimulates cell proliferation via activation of the MAPK/ERK pathway in rat cultured mesenchymal stem cells. Journal of Radiation Research (Tokyo) 52:380–386.
  • Little JB, Lauriston S. 2006. Taylor lecture: Nontargeted effects of radiation: Implications for low-dose exposures. Health Physics 91:416–426.
  • Little MP. 2010. Do non-targeted effects increase or decrease low dose risk in relation to the linear-non-threshold (LNT) model? Mutation Research 687:17–27.
  • Little MP, Wakeford R, Tawn EJ, Bouffler SD, Berrington de GA. 2009. Risks associated with low doses and low dose rates of ionizing radiation: Why linearity may be (almost) the best we can do. Radiology 251:6–12.
  • Liu SZ. 2010. Biological effects of low level exposures to ionizing radiation: Theory and practice. Human & Experimental Toxicology 29:275–281.
  • Lundberg GD. 2008. Antioxidant supplements found not to improve human survival. Medscape Journal of Medicine 10:222.
  • Mai S, Muster B, Bereiter-Hahn J, Jendrach M. 2012. Autophagy proteins LC3B, ATG5 and ATG12 participate in quality control after mitochondrial damage and influence lifespan. Autophagy 8: c.
  • Maiuri MC, Zalckvar E, Kimchi A, Kroemer G. 2007. Self-eating and self-killing: Crosstalk between autophagy and apoptosis. Nature Reviews Molecular Cell Biology 8:741–752.
  • Makedonov GP, Tskhovrebova LV, Unzhakov SV, Semiachkina AN, Vasil'eva IM, Zasukhina GD. 1997. [Radioadaptive response in lymphocytes of children living in territories polluted by radionuclides as a result of the accident at the Chernobyl power plant]. Radiatsionnaia Biologiia, Radioecologiia (Moskva) 37:640–644.
  • Marchi S, Giorgi C, Suski JM, Agnoletto C, Bononi A, Bonora M, De ME, Missiroli S, Patergnani S, Poletti F, Rimessi A, Duszynski J, Wieckowski MR, Pinton P. 2012. Mitochondria-ros crosstalk in the control of cell death and aging. Journal of Signal Transduction 2012:329635.
  • Marino G, Morselli E, Bennetzen MV, Eisenberg T, Megalou E, Schroeder S, Cabrera S, Benit P, Rustin P, Criollo A, Kepp O, Galluzzi L, Shen S, Malik SA, Maiuri MC, Horio Y, Lopez-Otin C, Andersen JS, Tavernarakis N, Madeo F, Kroemer G. 2011. Longevity-relevant regulation of autophagy at the level of the acetylproteome. Autophagy 7:647–649.
  • Martins I, Galluzzi L, Kroemer G. 2011. Hormesis, cell death and aging. Aging (Albany. NY) 3:821–828.
  • Masoomi JR, Mohammadi S, Amini M, Ghiassi-Nejad M. 2006. High background radiation areas of Ramsar in Iran: Evaluation of DNA damage by alkaline single cell gel electrophoresis (SCGE). Journal of Environmental Radioactivity 86:176–186.
  • Matsumoto H, Takahashi A, Ohnishi T. 2007. Nitric oxide radicals choreograph a radioadaptive response. Cancer Research 67:8574–8579.
  • Matsumoto H, Tomita M, Otsuka K, Hatashita M, Hamada N. 2011. Nitric oxide is a key molecule serving as a bridge between radiation-induced bystander and adaptive responses. Current Molecular Pharmacology 4:126–134.
  • Michalek RD, Nelson KJ, Holbrook BC, Yi JS, Stridiron D, Daniel LW, Fetrow JS, King SB, Poole LB, Grayson JM. 2007. The requirement of reversible cysteine sulfenic acid formation for T cell activation and function. Journal of Immunology 179:6456–6467.
  • Miura Y. 2004. Oxidative stress, radiation-adaptive responses and aging. Journal of Radiation Research 45:357–372.
  • Miura Y, Abe K, Urano S, Furuse T, Noda Y, Tatsumi K, Suzuki S. 2002. Adaptive response and the influence of ageing: Effects of low-dose irradiation on cell growth of cultured glial cells. International Journal of Radiation Biology 78:913–921.
  • Miura Y, Kano M, Yamada M, Nishine T, Urano S, Suzuki S, Endo T, Toda T. 2007. Proteomic study on X-irradiation-responsive proteins and ageing: Search for responsible proteins for radiation adaptive response. Journal of Biochemistry 142:145–155.
  • Mohammadi S, Taghavi-Dehaghani M, Gharaati MR, Masoomi R, Ghiassi-Nejad M. 2006. Adaptive response of blood lymphocytes of inhabitants residing in high background radiation areas of ramsar-micronuclei, apoptosis and comet assays. Journal of Radiation Research (Tokyo) 47:279–285.
  • Monfared AS, Hajian K, Hosseini R, Nasir A. 2010. Association between local external gamma rays and frequency of cancer in Babol, Iran. Dose Response 8:368–377.
  • Morgan WF. 2003. Is there a common mechanism underlying genomic instability, bystander effects and other nontargeted effects of exposure to ionizing radiation? Oncogene 22:7094–7099.
  • Morgan WF, Sowa MB. 2009. Non-targeted effects of ionizing radiation: Implications for risk assessment and the radiation dose response profile. Health Physics 97:426–432.
  • Morselli E, Galluzzi L, Kepp O, Criollo A, Maiuri MC, Tavernarakis N, Madeo F, Kroemer G. 2009a. Autophagy mediates pharmacological lifespan extension by spermidine and resveratrol. Aging (Albany.NY) 1:961–970.
  • Morselli E, Galluzzi L, Kepp O, Vicencio JM, Criollo A, Maiuri MC, Kroemer G. 2009b. Anti- and pro-tumor functions of autophagy. Biochimica et Biophysica Acta 1793:1524–1532.
  • Morselli E, Maiuri MC, Markaki M, Megalou E, Pasparaki A, Palikaras K, Criollo A, Galluzzi L, Malik SA, Vitale I, Michaud M, Madeo F, Tavernarakis N, Kroemer G. 2010. The life span-prolonging effect of sirtuin-1 is mediated by autophagy. Autophagy 6:186–188.
  • Morselli E, Galluzzi L, Kepp O, Marino G, Michaud M, Vitale I, Maiuri MC, Kroemer G. 2011a. Oncosuppressive functions of autophagy. Antioxidants & Redox Signaling 14:2251–2269.
  • Morselli E, Marino G, Bennetzen MV, Eisenberg T, Megalou E, Schroeder S, Cabrera S, Benit P, Rustin P, Criollo A, Kepp O, Galluzzi L, Shen S, Malik SA, Maiuri MC, Horio Y, Lopez-Otin C, Andersen JS, Tavernarakis N, Madeo F, Kroemer G. 2011b. Spermidine and resveratrol induce autophagy by distinct pathways converging on the acetylproteome. Journal of Cell Biology 192:615–629.
  • Mothersill C, Seymour C. 2004. Radiation-induced bystander effects and adaptive responses – the Yin and Yang of low dose radiobiology? Mutation Research 568:121–128.
  • Mothersill C, Seymour C. 2012. Changing paradigms in radiobiology. Mutation Research 750:85–95.
  • Muller FL, Liu Y, Van RH. 2004. Complex III releases superoxide to both sides of the inner mitochondrial membrane. Journal of Biological Chemistry 279:49064–49073.
  • Murley JS, Baker KL, Miller RC, Darga TE, Weichselbaum RR, Grdina DJ. 2011a. SOD2-mediated adaptive responses induced by low-dose ionizing radiation via TNF signaling and amifostine. Free Radical Biology and Medicine 51:1918–1925.
  • Murley JS, Kataoka Y, Miller RC, Li JJ, Woloschak G, Grdina DJ. 2011b. SOD2-mediated effects induced by WR1065 and low-dose ionizing radiation on micronucleus formation in RKO human colon carcinoma cells. Radiation Research 175:57–65.
  • Murphy MP. 2009. How mitochondria produce reactive oxygen species. Biochemical Journal 417:1–13.
  • Myatt SS, Brosens JJ, Lam EW. 2011. Sense and sensitivity: FOXO and ROS in cancer development and treatment. Antioxidants & Redox Signaling 14:675–687.
  • Némethová G, Kalina I, Raceková N. 1995. The adaptive response of peripheral blood lymphocytes to low doses of mutagenic agents in patients with ataxia telangiectasia. Mutation Research 348: 101–104.
  • Odegaard E, Yang CR, Boothman DA. 1998. DNA-dependent protein kinase does not play a role in adaptive survival responses to ionizing radiation. Environmental Health Perspectives 106(Suppl. 1): 301–305.
  • O'Donnell MA, Ting AT. 2010. Chronicles of a death foretold: Dual sequential cell death checkpoints in TNF signaling. Cell Cycle 9:1065–1071.
  • Okamoto K, Kondo-Okamoto N. 2012. Mitochondria and autophagy: Critical interplay between the two homeostats. Biochimica et Biophysica Acta 2011 , 1820:595–600.
  • Paglin S, Hollister T, Delohery T, Hackett N, McMahill M, Sphicas E, Domingo D, Yahalom J. 2001. A novel response of cancer cells to radiation involves autophagy and formation of acidic vesicles. Cancer Research 61:439–444.
  • Paglin S, Lee NY, Nakar C, Fitzgerald M, Plotkin J, Deuel B, Hackett N, McMahill M, Sphicas E, Lampen N, Yahalom J. 2005. Rapamycin-sensitive pathway regulates mitochondrial membrane potential, autophagy, and survival in irradiated MCF-7 cells. Cancer Research 65:11061–11070.
  • Paglin S, Yahalom J. 2006. Pathways that regulate autophagy and their role in mediating tumor response to treatment. Autophagy 2: 291–293.
  • Parsons PA. 2009. Prospectus. Survival across the fitness-stress continuum under the ecological stress theory of aging: Caloric restriction and ionizing radiation. Dose Response 8:4–9.
  • Patlak M. 2002. Targeting leukemia: From bench to bedside. FASEB Journal 16:273E–273e.
  • Paulsen CE, Carroll KS. 2010. Orchestrating redox signaling networks through regulatory cysteine switches. ACS Chemical Biology 5:47–62.
  • Paulsen CE, Truong TH, Garcia FJ, Homann A, Gupta V, Leonard SE, Carroll KS. 2011. Peroxide-dependent sulfenylation of the EGFR catalytic site enhances kinase activity. Nature Chemical Biology 8:57–64.
  • Pazhanisamy SK, Li H, Wang Y, Batinic-Haberle I, Zhou D. 2011. NADPH oxidase inhibition attenuates total body irradiation-induced haematopoietic genomic instability. Mutagenesis 26:431–435.
  • Pelevina II, Nikolaev VA, Gotlib VI, Afanas'ev GG, Kozlova LE, Serebrianyi AM, Tereshchenko DG, Tronov VA, Khramtsova EA. 1994 [The adaptive reaction of the blood lymphocytes in persons subjected to chronic radiation exposure at low doses]. Radiatsionnaia Biologiia, Radioecologiia (Moskva) 34:805–817.
  • Pelevina II, Gotlib VI, Kudriashova OV, Serebrianyi AM, Afanas'ev GG. 1996. [Genomic instability after exposure to radiation at low doses (in the 10-kilometer zone of the accident at the Chernobyl Atomic Electric Power Station and under laboratory conditions]. Radiatsionnaia Biologiia, Radioecologiia (Moskva) 36:546–560.
  • Piantadosi CA, Suliman HB. 2012. Transcriptional control of mitochondrial biogenesis and its interface with inflammatory processes. Biochimica et Biophysica Acta 2012 , Epub ahead of print.
  • Riabchenko NI, Antoshchina MM, Nasonova VA, Fesenko EV. 1995. [The cytogenetic analysis of the adaptive response in the lymphocytes of donors living in areas with different levels of radioactive contamination]. Radiatsionnaia Biologiia, Radioecologiia (Moskva) 35:670–675.
  • Richardson RB. 2009. Ionizing radiation and aging: Rejuvenating an old idea. Aging (Albany, NY) 1:887–902.
  • Ristow M, Schmeisser S. 2011. Extending life span by increasing oxidative stress. Free Radical Biology and Medicine 51:327–336.
  • Rubinsztein DC, Marino G, Kroemer G. 2011. Autophagy and aging. Cell 146:682–695.
  • Ryan LR, Seymour CB, Joiner MC, Mothersill CE. 2009. Radiation-induced adaptive response is not seen in cell lines showing a bystander effect but is seen in lines showing HRS/IRR response. International Journal of Radiation Biology 85:87–95.
  • Salih DA, Brunet A. 2008. FoxO transcription factors in the maintenance of cellular homeostasis during aging. Current Opinion in Cell Biology 20:126–136.
  • Salminen A, Kaarniranta K. 2010. ER stress and hormetic regulation of the aging process. Ageing Research Reviews 9:211–217.
  • Sasaki MS, Ejima Y, Tachibana A, Yamada T, Ishizaki K, Shimizu T, Nomura T. 2002. DNA damage response pathway in radioadaptive response. Mutation Research 504:101–118.
  • Scherz-Shouval R, Elazar Z. 2011. Regulation of autophagy by ROS: Physiology and pathology. Trends in Biochemical Sciences 36: 30–38.
  • Shadley JD, Wiencke JK. 1989. Induction of the adaptive response by X-rays is dependent on radiation intensity. International Journal of Radiation Biology 56:107–118.
  • Shen S, Kepp O, Michaud M, Martins I, Minoux H, Metivier D, Maiuri MC, Kroemer RT, Kroemer G. 2011. Association and dissociation of autophagy, apoptosis and necrosis by systematic chemical study. Oncogene 30:4544–4556.
  • Shen S, Kepp O, Kroemer G. 2012. The end of autophagic cell death? Autophagy 2012, 8:1–3.
  • Spitz DR, Azzam EI, Li JJ, Gius D. 2004. Metabolic oxidation/ reduction reactions and cellular responses to ionizing radiation: A unifying concept in stress response biology. Cancer Metastasis Reviews 23:311–322.
  • Streffer C. 2004. Bystander effects, adaptive response and genomic instability induced by prenatal irradiation. Mutation Research 568:79–87.
  • Szumiel I. 2005. Adaptive response: stimulated DNA repair or decreased damage fixation? International Journal of Radiation Biology 81: 233–241.
  • Szumiel I. 2011. Autophagy, reactive oxygen species and the fate of mammalian cells. Free Radical Research 45:253–265.
  • Tapio S, Jacob V. 2007. Radioadaptive response revisited. Radiation and Environmental Biophysics 46:1–12.
  • Takahashi A, Matsumoto H, Ohnishi T. 2008. Hdm2 and nitric oxide radicals contribute to the p53-dependent radioadaptive response. International Journal of Radiation Oncology, Biology, Physics 71:550–558.
  • Thayer KA, Melnick R, Burns K, Davis D, Huff J. 2005. Fundamental flaws of hormesis for public health decisions. Environmental Health Perspectives 113:1271–1276.
  • Thierens H, Vral A, Barbe M, Meijlaers M, Baeyens A, Ridder LD. 2002. Chromosomal radiosensitivity study of temporary nuclear workers and the support of the adaptive response induced by occupational exposure. International Journal of Radiation Biology 78:1117–1126.
  • Tubiana M, Feinendegen LE, Yang C, Kaminski JM. 2009. The linear no-threshold relationship is inconsistent with radiation biologic and experimental data. Radiology 251:13–22.
  • Turrens JF. 2003. Mitochondrial formation of reactive oxygen species. Journal of Physiology 552:335–344.
  • Unizhakov SV, Vasil'eva IM, Meliksetova IA, Semiachkina AN, Baleva LS, Kazantseva LZ, Zasukhina GD. 1994. [The formation of an adaptive response in children exposed to the effect of low doses of radiation as a result of the accident at the Chernobyl Atomic Electric Power Station]. Radiatsionnaia Biologiia, Radioecologiia (Moskva) 34:827–831.
  • Unizhakov SV, L'vova GN, Chekova VV, Semiachkina AN, Baleva LS, Zasukhina GD, Kazantseva LZ. 1995. [DNA repair activity in children exposed to small doses of ionizing radiation as a result of the accident at the Chernobyl nuclear power station]. Genetika 31:1433–1437.
  • United Nations Scientific Committee on the Effects of Atomic Radiation (UNSCEAR). 1994. Report to the General Assembly with Scientific Annexes. Annex B: Adaptive responses to radiation in cells and organisms. pp. 185–272.
  • United Nations Scientific Committee on the Effects of Atomic Radiation (UNSCEAR). 2006. Report to the General Assembly with Scientific Annexes. >Effects of ionizing radiation. Vol. II, Annex C, Non-targeted and delayed effects of ionizing radiation. pp 3–79.
  • Vaiserman AM. 2010. Radiation hormesis: Historical perspective and implications for low-dose cancer risk assessment. Dose Response 8:172–191.
  • van Montfort RL, Congreve M, Tisi D, Carr R, Jhoti H. 2003. Oxidation state of the active-site cysteine in protein tyrosine phosphatase 1B. Nature 423:773–777.
  • Vendelbo MH, Nair KS. 2011. Mitochondrial longevity pathways. Biochimica et Biophysica Acta 1813:634–644.
  • Vurusaner B, Poli G, Basaga H. 2012. Tumor suppressor genes and ROS: Complex networks of interactions. Free Radical Biology and Medicine 52:7–18.
  • Wang K, Klionsky DJ. 2011. Mitochondria removal by autophagy. Autophagy 7:297–300.
  • Weber TA, Reichert AS. 2010. Impaired quality control of mitochondria: Aging from a new perspective. Experimental Gerontology 45:503–511.
  • Wirawan E, Vande WL, Kersse K, Cornelis S, Claerhout S, Vanoverberghe I, Roelandt R, De RR, Verspurten J, Declercq W, Agostinis P, Vanden BT, Lippens S, Vandenabeele P. 2010. Caspase-mediated cleavage of Beclin-1 inactivates Beclin-1-induced autophagy and enhances apoptosis by promoting the release of proapoptotic factors from mitochondria. Cell Death & Disease 1:e18.
  • Wirawan E, Vanden BT, Lippens S, Agostinis P, Vandenabeele P. 2012. Autophagy: For better or for worse. Cell Research 22:43–61.
  • Wojewodzka M, Kruszewski M, Szumiel I. 1997. Effect of signal transduction inhibition in adapted lymphocytes: Micronuclei frequency and DNA repair. International Journal of Radiation Biology 71:245–252.
  • Wu WK, Coffelt SB, Cho CH, Wang XJ, Lee CW, Chan FK, Yu J, Sung JJ. 2012. The autophagic paradox in cancer therapy. Oncogene 31: 939–953.
  • Wyllie AH. 1997. Apoptosis and carcinogenesis. European Journal of Cell Biology 73:189–197.
  • Yang Z, Klionsky DJ. 2010a. Mammalian autophagy: Core molecular machinery and signaling regulation. Current Opinion in Cell Biology 22:124–131.
  • Yang Z, Klionsky DJ. 2010b. Eaten alive: A history of macroautophagy. Nature Cell Biology 12:814–822.
  • Yen WL, Klionsky DJ. 2008. How to live long and prosper: Autophagy, mitochondria, and aging. Physiology (Bethesda) 23:248–262.
  • Zalckvar E, Yosef N, Reef S, Ber Y, Rubinstein AD, Mor I, Sharan R, Ruppin E, Kimchi A. 2010. A systems level strategy for analyzing the cell death network: Implication in exploring the apoptosis/autophagy connection. Cell Death & Differentiation 17:1244–1253.
  • Zhang Q, Pi J, Woods CG, Andersen ME. 2010. A systems biology perspective on Nrf2-mediated antioxidant response. Toxicology & Applied Pharmacology 244:84–97.
  • Zhou F, Yang Y, Xing D. 2011. Bcl-2 and Bcl-xL play important roles in the crosstalk between autophagy and apoptosis. FEBS Journal 278:403–413.
  • Zois CE, Koukourakis MI. 2009. Radiation-induced autophagy in normal and cancer cells: Towards novel cytoprotection and radio-sensitization policies? Autophagy 5:442–450.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.