1,449
Views
55
CrossRef citations to date
0
Altmetric
Review Article

History of bystander effects research 1905-present; what is in a name?

, , ORCID Icon &
Pages 696-707 | Received 12 Jul 2017, Accepted 24 Oct 2017, Published online: 29 Nov 2017

References

  • Ahmad SB, McNeill FE, Byun SH, Prestwich WV, Mothersill C, Seymour C, Armstrong A, Fernandez C. 2013. Ultra-violet light emission from hpv-g cells irradiated with low let radiation from 90Y; consequences for radiation induced bystander effects. Dose Response. 11:498–516.
  • Al-Mayah AHJ, Irons SL, Pink RC, Carter DRF, Kadhim MA. 2012. Possible role of exosomes containing RNA in mediating nontargeted effect of ionizing radiation. Radiat Res. 177:539–545.
  • Alper T, Mothersill C, Seymour CB. 1988. Lethal mutations attributable to misrepair of Q-Lesions. Int J Radiat Biol. 54:525–530.
  • Arbetter KR, Prakash UBS, Tazelaar HD, Douglas WW. 1999. Radiation-induced pneumonitis in the “nonirradiated” lung. Mayo Clin Proc. 74:27–36.
  • Audette-Stuart M, Kim SB, McMullin D, Festarini A, Yankovich TL, Carr J, Mulpuru S. 2011. Adaptive response in frogs chronically exposed to low doses of ionizing radiation in the environment. J Environ Radioact. 102:566–573.
  • Autsavapromporn N, Suzuki M, Funayama T, Usami N, Plante I, Yokota Y, Mutou Y, Ikeda H, Kobayashi K, Kobayashi Y. 2013. Gap junction communication and the propagation of bystander effects induced by microbeam irradiation in human fibroblast cultures: the impact of radiation quality. Radiat Res. 180:367–375.
  • Azzam EI, de Toledo SM, Gooding T, Little JB. 1998. Intercellular communication is involved in the bystander regulation of gene expression in human cells exposed to very low fluences of alpha particles. Radiat Res. 150:497–504.
  • Azzam EI, de Toledo SM, Little JB. 2001. Direct evidence for the participation of gap junction-mediated intercellular communication in the transmission of damage signals from alpha-particle irradiated to nonirradiated cells. Proc Natl Acad Sci USA. 98:473–478.
  • Azzam EI, de Toledo SM, Little JB. 2003. Oxidative metabolism, gap junctions and the ionizing radiation-induced bystander effect. Oncogene. 22:7050–7057.
  • Azzam EI, De Toledo SM, Little JB. 2004. Stress signaling from irradiated to non-irradiated cells. Curr Cancer Drug Targets. 4:53–64.
  • Azzam EI, de Toledo SM, Spitz DR, Little JB. 2002. Oxidative metabolism modulates signal transduction and micronucleus formation in bystander cells from alpha-particle-irradiated normal human fibroblast cultures. Cancer Res. 62:5436–5442.
  • Baskar R, Balajee AS, Geard CR, Hande MP. 2008. Isoform-specific activation of protein kinase c in irradiated human fibroblasts and their bystander cells. Int J Biochem Cell Biol. 40:125–134.
  • Beam CA, Mortimer RK, Wolfe RG, Tobias CA. 1954. The relation of radioresistance to budding in saccharomyces cerevisiae. Arch Biochem Biophys. 49:110–122.
  • Bertucci A, Pocock RDJ, Randers-Pehrson G, Brenner DJ. 2009. Microbeam irradiation of the C. elegans nematode. J Radiat Res. 50:A49–A54.
  • Blyth BJ, Sykes PJ. 2011. Radiation-induced bystander effects: what are they, and how relevant are they to human radiation exposures? Radiat Res. 176:139–157.
  • Brooks AL, Retherford JC, McClellan RO. 1974. Effect of239 PuO2 particle number and size on the frequency and distribution of chromosome aberrations in the liver of the Chinese Hamster. Radiat Res. 59:693–709.
  • Born R, Trott KR. 1988. Clonogenicity of the progeny of surviving cells after irradiation. Int J Radiat Biol Relat Stud Phys Chem Med. 53:319–330.
  • Brown DC, Trott KR. 1994. Clonal heterogeneity in the progeny of HeLa cells which survive x-irradiation. Int J Radiat Biol. 66:151–155.
  • Camphausen K, Moses MA, Ménard C, Sproull M, Beecken WD, Folkman J, O’Reilly MS. 2003. Radiation abscopal antitumor effect is mediated through p53. Cancer Res. 63:1990–1993.
  • Chang WP, Little JB. 1991. Delayed reproductive death in x-irradiated Chinese Hamster ovary cells. Int J Radiat Biol. 60:483–496.
  • Chang WP, Little JB. 1992a. Evidence that DNA double-strand breaks initiate the phenotype of delayed reproductive death in Chinese Hamster ovary cells. Radiat Res. 131:53–59.
  • Chang WP, Little JB. 1992b. Persistently elevated frequency of spontaneous mutations in progeny of CHO clones surviving X-irradiation: association with delayed reproductive death phenotype. Mutat Res Mol Mech Mutagen. 270:191–199.
  • Coates PJ, Rundle JK, Lorimore SA, Wright EG. 2008. Indirect macrophage responses to ionizing radiation: implications for genotype-dependent bystander signaling. Cancer Res. 68:450–456.
  • Delbruck M. 1940. Radiation and the hereditary mechanism. Am Nat. 74:350–362.
  • Demoise CF, Conrad RA. 1972. Effects of age and radiation exposure on chromosomes in a Marshall Island population. J Gerontol. 27:197–201.
  • DeVeaux LC, Durtschi LS, Case JG, Wells DP. 2006. Bystander effects in unicellular organisms. Mutat Res. 597:78–86.
  • Emerit I. 1994. Reactive oxygen species, chromosome mutation, and cancer: possible role of clastogenic factors in carcinogenesis. Free Radic Biol Med. 16:99–109.
  • Faguet GB, Reichard SM, Welter DA. 1984. Radiation-induced clastogenic plasma factors. Cancer Genet Cytogenet. 12:73–83.
  • Fazzari J, Mersov A, Smith R, Seymour C, Mothersill C. 2012. Effect of 5-hydroxytryptamine (serotonin) receptor inhibitors on the radiation-induced bystander effect. Int J Radiat Biol. 88:786–790.
  • Fernandez-Palomo C, Schültke E, Bräuer-Krisch E, Laissue JA, Blattmann H, Seymour C, Mothersill C. 2016. Investigation of abscopal and bystander effects in immunocompromised mice after exposure to pencilbeam and microbeam synchrotron radiation. Health Phys. 111:149–159.
  • Fernandez-Palomo C, Schültke E, Smith R, Bräuer-Krisch E, Laissue J, Schroll C, Fazzari J, Seymour C, Mothersill C. 2013. Bystander effects in tumor-free and tumor-bearing rat brains following irradiation by synchrotron X-rays. Int J Radiat Biol. 89:445–453.
  • Goh K. 1975. Total-body irradiation and human chromosomes. IV. Cytogenetic follow-up studies 8 and 10 1/2 years after total-body irradiation. Radiat Res. 62:364–373.
  • Goh KO, Sumner H. 1968. Breaks in normal human chromosomes: are they induced by a transferable substance in the plasma of persons exposed to total-body irradiation? Radiat Res. 35:171–181.
  • Guan XJ, Bonney WJ, Ruch RJ. 1995. Changes in gap junction permeability, gap junction number, and Connexin43 expression in Lindane-treated rat liver epithelial cells. Toxicol Appl Pharmacol. 130:79–86.
  • Hamada N, Matsumoto H, Hara T, Kobayashi Y. 2007. Intercellular and intracellular signaling pathways mediating ionizing radiation-induced bystander effects. J Radiat Res. 48:87–95.
  • Hatzi VI, Laskaratou DA, Mavragani IV, Nikitaki Z, Mangelis A, Panayiotidis MI, Pantelias GE, Terzoudi GI, Georgakilas AG. 2015. Non-targeted radiation effects in vivo: a critical glance of the future in radiobiology. Cancer Lett. 356:34–42.
  • Hei TK, Zhou H, Ivanov VN. 2008. Mechanism of radiation-induced bystander effects: a unifying model. J Pharm Pharmacol. 60:943–950.
  • Hei TK, Zhou H, Chai Y, Ponnaiya B, Ivanov VN. 2011. Radiation induced non-targeted response: mechanism and potential clinical implications. Curr Mol Pharmacol. 4:96–105.
  • Heineke H. 1905. Experimental investigations on the effects of X-rays on the bone marrow, with some remarks on the radiotherapy of leukemia and pseudoleukemia and sarcoms. Deutsche Zeitschrift Für Chirurgie. 78:196–230.
  • Hevesy G. 1945. On the effect of roentgen rays on cellular division. Rev Mod Phys. 17:102–111.
  • Hollowell JG, Littlefield LG. 1968. Chromosome damage induced by plasma of X-rayed patients: an indirect effect of X-ray. Proc Soc Exp Biol Med. 129:240–244.
  • Hollowell JGJ, Littlefield LG. 1967. Chromosome aberrations induced by plasma from irradiated patients: a brief report. J S C Med Assoc. 63:442.
  • Hu B, Shen B, Su Y, Geard CR, Balajee AS. 2009. Protein kinase C epsilon is involved in ionizing radiation induced bystander response in human cells. Int J Biochem Cell Biol. 41:2413–2421.
  • Iyer R, Lehnert BE. 2000. Effects of ionizing radiation in targeted and nontargeted cells. Arch Biochem Biophys. 376:14–25.
  • Jolles B. 1941. X-ray skin reactions and the protective role of normal tissues. Br J Radiol. 14:110–112.
  • Jolles B. 1949. A diffusible substance in irradiated tissues? Nature. 164:63–64.
  • Kadhim MA, Macdonald DA, Goodhead DT, Lorimore SA, Marsden SJ, Wright EG. 1992. Transmission of chromosomal instability after plutonium alpha-particle irradiation. Nature. 355:738–740.
  • Kashino G, Prise KM, Schettino G, Folkard M, Vojnovic B, Michael BD, Suzuki K, Kodama S, Watanabe M. 2004. Evidence for induction of DNA double strand breaks in the bystander response to targeted soft X-rays in CHO cells. Mutat Res Mol Mech Mutagen. 556:209–215.
  • Khan MA, Hill RP, Van Dyk J. 1998. Partial volume rat lung irradiation: an evaluation of early DNA damage. Int J Radiat Oncol Biol Phys. 40:467–476.
  • Khan MA, Van Dyk J, Yeung IWT, Hill RP. 2003. Partial volume rat lung irradiation; assessment of early DNA damage in different lung regions and effect of radical scavengers. Radiother Oncol. 66:95–102.
  • Koturbash I, Zemp FJ, Kutanzi K, Luzhna L, Loree J, Kolb B, Kovalchuk O. 2008. Sex-specific microRNAome deregulation in the shielded bystander spleen of cranially exposed mice. Cell Cycle. 7:1658–1667.
  • Kumar Jella K, Rani S, O’driscoll L, McClean B, Byrne HJ, Lyng FM. 2014. Exosomes are involved in mediating radiation induced bystander signaling in human keratinocyte cells. Radiat Res. 181:138–145.
  • Lacassagne A. 1928. The direct and indirect action of radiation on cancer tissues. Radiology. 11:393–402.
  • Le M, Fernandez-Palomo C, McNeill FE, Seymour CB, Rainbow AJ, Mothersill CE. 2017. Exosomes are released by bystander cells exposed to radiation-induced biophoton signals: reconciling the mechanisms mediating the bystander effect. PLoS One. 12:e0173685.
  • Le M, Mothersill CE, Seymour CB, Ahmad SB, Armstrong A, Rainbow AJ, McNeill FE. 2015a. Factors affecting ultraviolet-a photon emission from beta-irradiated human keratinocyte cells. Phys Med Biol. 60:6371–6389.
  • Le M, McNeill FE, Seymour C, Rainbow AJ, Mothersill CE. 2015b. An observed effect of ultraviolet radiation emitted from beta-irradiated HaCaT cells upon non-beta-irradiated bystander cells. Radiat Res. 183:279–290.
  • Lea DE. 1947. Actions of radiations on living cells. 1st ed. Cambridge (UK): Cambridge University Press.
  • Liu ZF, Lyun SH, McNeill FE, Mothersill CE, Seymour CB, Prestwich WV. 2007. Fluence and dose measurements for an accelerator neutron beam. Nucl Instrum Meth B. 263:326–328.
  • Liu ZF, Mothersill CE, McNeill FE, Lyng FM, Byun SH, Seymour CB, Prestwich WV. 2006. A dose threshold for a medium transfer bystander effect for a human skin cell line. Radiat Res. 166:19–23.
  • Lorimore SA, Coates PJ, Scobie GE, Milne G, Wright EG. 2001. Inflammatory-type responses after exposure to ionizing radiation in vivo: a mechanism for radiation-induced bystander effects? Oncogene. 20:7085.
  • Lyng FM, Desplanques M, Jella KK, Garcia A, McClean B. 2012. The importance of serum serotonin levels in the measurement of radiation-induced bystander cell death in HaCaT cells. Int J Radiat Biol. 88:770–772.
  • Lyng FM, Maguire P, McClean B, Seymour C, Mothersill C. 2006. The involvement of calcium and MAP kinase signaling pathways in the production of radiation-induced bystander effects. Radiat Res. 165:400–409.
  • Lyng FM, OReilly S, Cottell DC, Seymour CB, Mothersill C. 1996. Persistent expression of morphological abnormalities in the distant progeny of irradiated cells. Radiat Environ Biophys. 35:273–283.
  • Lyng FM, Seymour CB, Mothersill C. 2001. Oxidative stress in cells exposed to low levels of ionizing radiation. Biochem Soc Trans. 29:350–353.
  • Lyng FM, Seymour CB, Mothersill C. 2002a. Early events in the apoptotic cascade initiated in cells treated with medium from the progeny of irradiated cells. Radiat Prot Dosimetry. 99:169–172.
  • Lyng FM, Seymour CB, Mothersill C. 2002b. Initiation of apoptosis in cells exposed to medium from the progeny of irradiated cells: a possible mechanism for bystander-induced genomic instability? Radiat Res. 157:365–370.
  • Mancuso M, Pasquali E, Leonardi S, Tanori M, Rebessi S, Di Majo V, Pazzaglia S, Toni MP, Pimpinella M, Covelli V, et al. 2008. Oncogenic bystander radiation effects in patched heterozygous mouse cerebellum. Proc Natl Acad Sci USA. 105:12445–12450.
  • Marozik P, Mothersill C, Seymour CB, Mosse I, Melnov S. 2007. Bystander effects induced by serum from survivors of the chernobyl accident. Exp Hematol. 35:55–63.
  • Martín C, Romero S, Sánchez-Payá J, Massutí B, Arriero JM, Hernández L. 1999. Bilateral lymphocytic alveolitis: a common reaction after unilateral thoracic irradiation. Eur Respir J. 13:727–732.
  • Matsumoto H, Hayashi S, Hatashita M, Ohnishi K, Shioura H, Ohtsubo T, Kitai R, Ohnishi T, Kano E. 2001. Induction of radioresistance by a nitric oxide-mediated bystander effect. Radiat Res. 155:387–396.
  • McNaughton SJ. 1930. Radiation and genetics. Am Nat. 64:220–251.
  • McMahon SJ, McGarry CK, Butterworth KT, Jain S, O’Sullivan JM, Hounsell AR, Prise KM. 2015. Cellular signalling effects in high precision radiotherapy. Phys Med Biol. 60:4551.
  • Mole RH. 1953. Whole body irradiation; radiobiology or medicine? Br J Radiol. 26:234–241.
  • Morgan GW, Pharm B, Breit SN. 1995. Radiation and the lung: a reevaluation of the mechanisms mediating pulmonary injury. Int J Radiat Oncol Biol Phys. 31:361–369.
  • Morgan WF. 2003a. Non-targeted and delayed effects of exposure to ionizing radiation: i. radiation-induced genomic instability and bystander effects in vitro. Radiat Res. 159:567–580.
  • Morgan WF. 2003b. Non-targeted and delayed effects of exposure to ionizing radiation: ii. radiation-induced genomic instability and bystander effects in vivo, clastogenic factors and transgenerational effects. Radiat Res. 159:581–596.
  • Mosse I, Marozik P, Seymour C, Mothersill C. 2006. The effect of melanin on the bystander effect in human keratinocytes. Mutat Res. 597:133–137.
  • Mothersill C, Seymour C. 1997. Medium from irradiated human epithelial cells but not human fibroblasts reduces the clonogenic survival of unirradiated cells. Int J Radiat Biol. 71:421–427.
  • Mothersill C, Seymour C. 2001. Radiation-induced bystander effects: past history and future directions. Radiat Res. 155:759–767.
  • Mothersill C, Seymour C. 2004. Radiation-induced bystander effects and adaptive responses-the Yin and Yang of low dose radiobiology? Mutat Res. 568:121–128.
  • Mothersill C, Seymour CB. 1998. Cell-cell contact during gamma irradiation is not required to induce a bystander effect in normal human keratinocytes: evidence for release during irradiation of a signal controlling survival into the medium. Radiat Res. 149:256–262.
  • Mothersill C, Seymour CB. 2002. Bystander and delayed effects after fractionated radiation exposure. Radiat Res. 158:626–633.
  • Mothersill C, Bucking C, Smith RW, Agnihotri N, O’Neill A, Kilemade M, Seymour CB. 2006. Communication of radiation-induced stress or bystander signals between fish in vivo. Environ Sci Technol. 40:6859–6864.
  • Mothersill C, Lyng F, Seymour C, Maguire P, Lorimore S, Wright E. 2005. Genetic factors influencing bystander signaling in murine bladder epithelium after low-dose irradiation in vivo. Radiat Res. 163:391–399.
  • Mothersill C, Moran G, McNeill F, Gow MD, Denbeigh J, Prestwich W, Seymour CB. 2007a. A role for bioelectric effects in the induction of bystander signals by ionizing radiation? Dose Response. 5:214–229.
  • Mothersill C, Seymour RJ, Seymour CB. 2006. Increased radiosensitivity in cells of two human cell lines treated with bystander medium from irradiated repair-deficient cells. Radiat Res. 165:26–34.
  • Mothersill C, Smith RW, Agnihotri N, Seymour CB. 2007b. Characterization of a radiation-induced stress response communicated in vivo between zebrafish. Environ Sci Technol. 41:3382–3387.
  • Mothersill C, Saroya R, Smith RW, Singh H, Seymour CB. 2010a. Serum serotonin levels determine the magnitude and type of bystander effects in medium transfer experiments. Radiat Res. 174:119–123.
  • Mothersill C, Seymour C. 2012. Changing paradigms in radiobiology. Mutat Res. 750:85–95.
  • Mothersill C, Smith RW, Saroya R, Denbeigh J, Rowe B, et al. 2010b. Irradiation of rainbow trout at early life stages results in legacy effects in adults. Int J Radiat Biol. 86:817–828.
  • Muller HJ. 1928. The production of mutations by X-rays. Proc Natl Acad Sci USA. 14:714–726.
  • Murphy JB, Morton JJ. 1915. The effect of roentgen rays on the rate of growth of spontaneous tumors in mice. J Exp Med. 22:800–803.
  • Murphy JEJ, Nugent S, Seymour C, Mothersill C. 2005. Mitochondrial DNA point mutations and a novel deletion induced by direct low-LET radiation and by medium from irradiated cells. Mutat Res Toxicol Environ Mutagen. 585:127–136.
  • Nagasawa H, Little JB. 1992. Induction of sister chromatid exchanges by extremely low doses of α-particles. Cancer Res. 52:6394–6396.
  • Narayanan PK, Goodwin EH, Lehnert BE. 1997. Alpha particles initiate biological production of superoxide anions and hydrogen peroxide in human cells. Cancer Res. 57:3963–3971.
  • Narayanan PK, LaRue KEA, Goodwin EH, Lehnert BE. 1999. Alpha particles induce the production of interleukin-8 by human cells. Radiat Res. 152:57–63.
  • Nickson JJ. 1950. The basic aspects of radiation effects on living systems. 1st ed. In: Morrison P, Burton M, Barron EG, Failla G, Patt HM, editors. New York (NY): Wiley.
  • O’Dowd C, Mothersill CE, Cairns MT, Austin B, McClean B, Lyng FM, Murphy JEJ. 2006. The release of bystander factor(s) from tissue explant cultures of rainbow trout (Onchorhynchus mykiss) after exposure to gamma radiation. Radiat Res. 166:611–617.
  • O’Leary VB, Ovsepian SV, Carrascosa LG, Buske FA, Radulovic V, Niyazi M, Moertl S, Trau M, Atkinson MJ, Anastasov N. 2015. PARTICLE, a triplex-forming long ncRNA, regulates locus-specific methylation in response to low-dose irradiation. Cell Rep. 11:474–485.
  • Packard C. 1927. Biological effects of irradiation. Science. 69:x–xii.
  • Pampfer S, Streffer C. 1989. Increased chromosome aberration levels in cells from mouse fetuses after zygote x-irradiation. Int J Radiat Biol. 55:85–92.
  • Pant GS, Kamada N. 1977. Chromosome aberrations in normal leukocytes induced by the plasma of exposed individuals. Hiroshima J Med Sci. 26:149–154.
  • Parsons WB, Watkins CH, Pease GL, Childs DS. 1954. Changes in sternal marrow following roentgen-ray therapy to the spleen in chronic granulocytic leukemia. Cancer. 7:179–189.
  • Poncy JL, Walter C, Fritsch P, Masse R, Lafuma J. 1979. Delayed SCE frequency of rat bone marrow cells after radon inhalation. 19. Annual ranford life sciences symposium on pulmonary toxicology of respirable particle; October 22–24; Richland, USA.
  • Poon RC, Agnihotri N, Seymour C, Mothersill C. 2007. Bystander effects of ionizing radiation can be modulated by signaling amines. Environ Res. 105:200–211.
  • Prise KM, Belyakov OV, Newman HC, Patel S, Schettino G, Folkard M, Michael BD. 2002. Non-targeted effects of radiation: bystander responses in cell and tissue models. Radiat Prot Dosimetry. 99:223–226.
  • Prise KM, O’sullivan JM. 2009. Radiation-induced bystander signalling in cancer therapy. Nat Rev Cancer. 9:351–360.
  • Prise KM, Schettino G, Folkard M, Held KD. 2005. New insights on cell death from radiation exposure. Lancet Oncol. 6:520–528.
  • Robertson JA. 1978. The CANDU reactor system: an appropriate technology. Science. 199:657–664.
  • Saroya R, Smith R, Seymour C, Mothersill C. 2010. Injection of reserpine into zebrafish prevents fish to fish communication of radiation induced bystander signals: confirmation in vivo of a role for serotonin in the mechanism. Dose Response. 8:317–330.
  • Schettino G, Folkard M, Michael BD, Prise KM. 2005. Low-dose binary behavior of bystander cell killing after microbeam irradiation of a single cell with focused c(k) x rays. Radiat Res. 163:332–336.
  • Schettino G, Folkard M, Prise KM, Vojnovic B, Held KD, Michael BD. 2003. Low-dose studies of bystander cell killing with targeted soft X rays. Radiat Res. 160:505–511.
  • Seymour CB, Mothersill C. 2000. Relative contribution of bystander and targeted cell killing to the low-dose region of the radiation dose-response curve. Radiat Res. 153:508–511.
  • Seymour CB, Mothersill CE, Alper T. 1986. High yields of lethal mutations in somatic mammalian-cells that survive ionizing-radiation. Int J Radiat Biol. 50:167–179.
  • Shao C, Furusawa Y, Aoki M, Matsumoto H, Ando K. 2002. Nitric oxide-mediated bystander effect induced by heavy-ions in human salivary gland tumour cells. Int J Radiat Biol. 78:837–844.
  • Shao C, Stewart V, Folkard M, Michael BD, Prise KM. 2003. Nitric oxide-mediated signaling in the bystander response of individually targeted glioma cells. Cancer Res. 63:8437–8442.
  • Shao C, Lyng FM, Folkard M, Prise KM. 2006. Calcium fluxes modulate the radiation-induced bystander responses in targeted glioma and fibroblast cells. Radiat Res. 166:479–487.
  • Sharetskiĭ AN, Kharlamov VA, Surinov BP. 2011. Effect of radiation-induced bystander chemosignals of mice on the humoral immune response in spleen and lymph nodes of intact recipients. Radiats Biol Radioecol. 52:229–233.
  • Shields L, Vega-Carrascal I, Singleton S, Lyng FM, McClean B. 2014. Cell survival and DNA damage in normal prostate cells irradiated out-of-field. Radiat Res. 182:499–506.
  • Smith RW, Wang J, Bucking CP, Mothersill CE, Seymour CB. 2007. Evidence for a protective response by the gill proteome of rainbow trout exposed to X-ray induced bystander signals. Proteomics. 7:4171–4180.
  • Smith RW, Seymour CB, Moccia RD, Hinton TG, Mothersill CE. 2013. The induction of a radiation-induced bystander effect in fish transcends taxonomic group and trophic level. Int J Radiat Biol. 89:225–233.
  • Smith RW, Seymour CB, Moccia RD, Mothersill CE. 2016. Irradiation of rainbow trout at early life stages results in trans-generational effects including the induction of a bystander effect in non-irradiated fish. Environ Res. 145:26–38.
  • Souto J. 1962. Tumour development in the rat induced by blood of irradiated animals. Nature. 195:1317–1318.
  • Strangeway TSP, Fell HB. 1927. A study of the direct and indirect action of X-rays upon the tissues of the embryonic fowl. Proc R Soc London Ser B Contain Pap a Biol Character. 102:9–29.
  • Surinov BP, Isaeva VG, Dukhova NN. 2004a. Communicative multiplication of secondary disorders in blood formula and immunity in groups of intact mice caused by volatile compounds excreted by irradiated animals. Radiats Biol Radioecol. 44:387–391.
  • Surinov BP, Isaeva VG, Dukhova NN. 2004b. Postirradiation volatile secretions of mice: syngeneic and allogeneic immune and behavioral effects. Bull Exp Biol Med. 138:384–386.
  • Surinov BP, Isaeva VG, Tokarev OI. 2001. Allelopathic activity of volatile secretions in irradiated animals. Radiats Biol Radioecol. 41:645–649.
  • Surinov BP, Karpova NA, Isaeva VG, Kulish I. 1997. Communicative behavioral effects and disorders of immunity. Zh Vyss Nerv Deiat Im I P Pavlova. 48:1073–1079.
  • Surinov BP, Karpova NA, Isaeva VG, Kulish IS. 2000. Post-stress state and communicative alterations of immunity and blood. Patol Fiziol Eksp Ter. 4:9–11.
  • Surinov VP, Karpova NA, Isaeva VG, Kulish I. 1998. Natural excretions of mice in the postradiation period and contact induction of immunodeficiencies. Radiats Biol Radioecol. 38:9.
  • Tartier L, Gilchrist S, Burdak-Rothkamm S, Folkard M, Prise KM. 2007. Cytoplasmic irradiation induces mitochondrial-dependent 53BP1 protein relocalization in irradiated and bystander cells. Cancer Res. 67:5872–5879.
  • Timofeeff-Ressovsky NW, Delbruck M. 1936. Radiation researches on visible mutations and the mutability of single genes in Drosophila. Z indukt Abstamm-u VererbLehre. 71:322.
  • Timoféeff-Ressovsky NW, Zimmer KG. 1939. Radiation genetics. Strahlentherapie. 66:684.
  • Trott KR. 2001. Non-targeted radiation effects in radiotherapy & roles of radiation-induced genomic instability and of the bystander effect in cancer cure by radiotherapy. Acta Oncol. 40:976–980.
  • Xu S, Wang J, Ding N, Hu W, Zhang X, Wang B, Hua J, Wei W, Zhu Q. 2015. Exosome-mediated microRNA transfer plays a role in radiation-induced bystander effect. RNA Biol. 12:1355–1363.
  • Zhou H, Ivanov VN, Gillespie J, Geard CR, Amundson SA, Brenner DJ, Yu Z, Lieberman HB, Hei TK. 2005. Mechanism of radiation-induced bystander effect: role of the cyclooxygenase-2 signaling pathway. Proc Natl Acad Sci USA. 102:14641–14646.
  • Zhou H, Ivanov VN, Lien Y-C, Davidson M, Hei TK. 2008. Mitochondrial function and nuclear factor-kappaB-mediated signaling in radiation-induced bystander effects. Cancer Res. 68:2233–2240.

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.