2,277
Views
7
CrossRef citations to date
0
Altmetric
Reviews

Low dose ionizing radiation and the immune response: what is the role of non-targeted effects?

ORCID Icon, &
Pages 1368-1382 | Received 05 May 2021, Accepted 22 Jul 2021, Published online: 17 Aug 2021

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 (90)Y; consequences for radiation induced bystander effects. Dose Response. 11(4):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(5):539–545.
  • Andor N, Maley CC, Ji HP. 2017. Genomic instability in cancer: teetering on the limit of tolerance. Cancer Res. 77(9):2179–2185.
  • Ashrafizadeh M, Farhood B, Eleojo Musa A, Taeb S, Najafi M. 2020. The interactions and communications in tumor resistance to radiotherapy: Therapy perspectives. Int Immunopharmacol. 87:106807.
  • Attarwala H. 2010. TGN1412: from discovery to disaster. J Young Pharm. 2(3):332–336.
  • 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 U S A. 98(2):473–478.
  • Azzam EI, Jay-Gerin J-P, Pain D. 2012. Ionizing radiation-induced metabolic oxidative stress and prolonged cell injury. Cancer Lett. 327(1-2):48–60.
  • Azzam EI, Little JB. 2004. The radiation-induced bystander effect: evidence and significance. Hum Exp Toxicol. 23(2):61–65.
  • Barbero F, Thomas JA, Bonelli S, Balletto E, Schönrogge K. 2009. Queen ants make distinctive sounds that are mimicked by a butterfly social parasite. Science. 323(5915):782–785.
  • Baverstock K, Karotki AV. 2011. Towards a unifying theory of late stochastic effects of ionizing radiation. Mutat Res. 718(1-2):1–9.
  • Beck HL, Till JE, Grogan HA, Aanenson JW, Mohler HJ, Mohler SS, Voillequé PG. 2017. Red bone marrow and male breast doses for a cohort of atomic veterans. Rare. 187(2):221–228.
  • Bertucci A, Pocock RDJ, Randers-Pehrson G, Brenner DJ. 2009. Microbeam Irradiation of the C. elegans Nematode. J Radiat Res. 50 Suppl A(0 0):A49–A54.
  • Bogdándi EN, Balogh A, Felgyinszki N, Szatmári T, Persa E, Hildebrandt G, Sáfrány G, Lumniczky K. 2010. Effects of low-dose radiation on the immune system of mice after total-body irradiation. Radiat Res. 174(4):480–489.
  • Branch P, Bicknell DC, Rowan A, Bodmer WF, Karran P. 1995. Immune surveillance in colorectal carcinoma. Nat Genet. 9(3):231–232.
  • Calabrese E, Dhawan G, Kapoor R, Kozumbo W. 2019. Radiotherapy treatment of human inflammatory diseases and conditions: optimal dose. Hum Exp Toxicol. 38(8):888–898.
  • Calabrese EJ. 2005. Paradigm lost, paradigm found: the re-emergence of hormesis as a fundamental dose response model in the toxicological sciences. Environ Pollut. 138(3):379–411.
  • Calabrese EJ. 2017. Flaws in the LNT single-hit model for cancer risk: an historical assessment. Environ Res. 158:773–788.
  • Calabrese EJ. 2018. From Muller to mechanism: how LNT became the default model for cancer risk assessment. Environ Pollut. 241:289–302.
  • Calabrese EJ, Dhawan G. 2013. How radiotherapy was historically used to treat pneumonia: could it be useful today? Yale J Biol Med. 86(4):555–570.
  • Cheda A, Nowosielska EM, Wrembel-Wargocka J, Janiak MK. 2008. Production of cytokines by peritoneal macrophages and splenocytes after exposures of mice to low doses of X-rays. Radiat Environ Biophys. 47(2):275–283.
  • Choi B, Ghosh R, Gururani MA, Shanmugam G, Jeon J, Kim J, Park S-C, Jeong M-J, Han K-H, Bae D-W, et al. 2017. Positive regulatory role of sound vibration treatment in Arabidopsis thaliana against Botrytis cinerea infection. Sci Rep. 7(1):2527.
  • Chun SH, Park G-Y, Han YK, Kim SD, Kim JS, Lee CG, Yang K. 2012. Effect of low dose radiation on differentiation of bone marrow cells into dendritic cells. Dose Response. 11(3):374–384.
  • Clarke RH. 2000. Issues in the control of low-level radiation exposure . Med Confl Surviv. 16(4):411–422.
  • Clutton SM, Townsend KMS, Walker C, Ansell JD, Wright EG. 1996. Radiation-induced genomic instability and persisting oxidative stress in primary bone marrow cultures. Carcinogenesis. 17(8):1633–1639.
  • Coates PJ, Lorimore SA, Wright EG. 2004. Damaging and protective cell signalling in the untargeted effects of ionizing radiation. Mutat Res. 568(1):5–20.
  • Cohen J, Vo NTK, Chettle DR, McNeill FE, Seymour CB, Mothersill CE. 2020. Quantifying biophoton emissions from human cells directly exposed to low-dose gamma radiation. Dose Response. 18(2):1559325820926763.
  • Crossland PM. 1956. Therapy of tinea capitis—the value of X-Ray epilation. Calif Med. 84(5):351–353.
  • Csaba G. 2019. Hormesis and immunity: a review. Acta Microbiol Immunol Hung. 66(2):155–168.
  • Das IJ, Kalapurakal JA, Mittal BB. 2021. Caution warranted for low-dose radiation therapy for Covid-19. Br J Radiol. 94(1117):20200466.
  • Del Castillo R, Martinez D, Sarria GJ, Pinillos L, Garcia B, Castillo L, Carhuactocto A, Giordano FA, Sarria GR. 2020. Low-dose radiotherapy for COVID-19 pneumonia treatment: case report, procedure, and literature review. Strahlenther Onkol. 196(12):1086–1093.
  • Demaria S, Ng B, Devitt ML, Babb JS, Kawashima N, Liebes L, Formenti SC. 2004. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int J Radiat Oncol Biol Phys. 58(3):862–870.
  • El-Saghire H, Michaux A, Thierens H, Baatout S. 2013. Low doses of ionizing radiation induce immune-stimulatory responses in isolated human primary monocytes. Int J Mol Med. 32(6):1407–1414.
  • Ermakov AV, Kostyuk SV, Konkova MS, Egolina NA, Malinovskaya EM, Veiko NN. 2008. Extracellular DNA fragments: factors of stress signaling between X‐irradiated and nonirradiated human lymphocytes. Ann N Y Acad Sci. 1137(1):41–46.
  • Farhood B, Khodamoradi E, Hoseini-Ghahfarokhi M, Motevaseli E, Mirtavoos-Mahyari H, Eleojo Musa A, Najafi M. 2020. TGF-β in radiotherapy: Mechanisms of tumor resistance and normal tissues injury. Pharmacol Res. 155:104745.
  • Feinendegen LE. 2005. Evidence for beneficial low level radiation effects and radiation hormesis. Br J Radiol. 78(925):3–7.
  • 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(2):149–159.
  • Folley JH, Borges W, Yamawaki T. 1952. Incidence of leukemia in survivors of the atomic bomb in Hiroshima and Nagasaki, Japan. Am J Med. 13(3):311–321.
  • Franchi N, Ballarin L. 2017. Immunity in protochordates: the tunicate perspective. Front Immunol. 8:674.
  • Fujiwara S, Carter RL, Akiyama M, Akahoshi M, Kodama K, Shimaoka K, Yamakido M. 1994. Autoantibodies and Immunoglobulins among Atomic Bomb Survivors. J Radiat Res. 137(1):89–95.
  • Frongia F, Forti L, Arru L. 2020. Sound perception and its effects in plants and algae. Plant Signal Behav. 15(12):1828674.
  • Gardner MJ. 1991. Father's occupational exposure to radiation and the raised level of childhood leukemia near the Sellafield nuclear plant. Environ Health Perspect. 94:5–7.
  • Gluzman DF, Zavelevich MP, Philchenkov AA, Koval SV, Bezhenar TO. 2021. Immunodeficiency-associated lymphoproliferative disorders and lymphoid neoplasms in post-COVID-19 pandemic era. Exp Oncol. 43(1):87–91.
  • Guo X, Sun J, Bian P, Chen L, Zhan F, Wang J, Xu A, Wang Y, Hei TK, Wu L. 2013. Radiation-Induced Bystander Signaling from Somatic Cells to Germ Cells in Caenorhabditis elegans. Radiat Res. 180(3):268–275.
  • Hanekamp YN, Giordano J, Hanekamp JC, Khan MK, Limper M, Venema CS, Vergunst SD, Verhoeff JJC, Calabrese EJ. 2020. Immunomodulation through low-dose radiation for severe COVID-19: lessons from the past and new developments. Dose Response. 18(3):1559325820956800.
  • 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. J Pharm Pharmacol. 60(8):943–950.
  • Heineke H. 1905. Experimentelle untersuchungen über die einwirkung der röntgenstrahlen auf das knochenmark, nebst einigen bemerkungen über die röntgentherapie der leukämie und pseudoleukämie und des sarcoms. langenbeck’s. Deutsche Zeitschrift f Chirurgie. 78(1-3):196–230.
  • Hektoen L. 1918. Further studies on the effects of the roentgen ray on antibody-production. J Infect Dis. 22(1):28–33.
  • Hellweg CE. 2015. The Nuclear Factor κB pathway: a link to the immune system in the radiation response. Cancer Lett. 368(2):275–289.
  • Heyd R, Tselis N, Ackermann H, Röddiger SJ, Zamboglou N. 2007. Radiation therapy for painful heel spurs: results of a prospective randomized study. Strahlenther Onkol. 183(1):3–9.
  • 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(1):240–244.
  • Hsu W-L, Preston DL, Soda M, Sugiyama H, Funamoto S, Kodama K, Kimura A, Kamada N, Dohy H, Tomonaga M, et al. 2013. The incidence of leukemia, lymphoma, and multiple myeloma among atomic bomb survivors. Radiat Res. 179(3):1950–2001.
  • Ilnytskyy Y, Kovalchuk O. 2011. Non-targeted radiation effects-an epigenetic connection. Mutat Res. 714(1-2):113–125.
  • Jacobson LO, Simmons EL, Bethard WF, Marks EK, Robson MJ. 1950. The influence of the spleen on hematopoietic recovery after irradiation injury. Proc Soc Exp Biol Med. 73(3):455–459.
  • Janiak MK, Wincenciak M, Cheda A, Nowosielska EM, Calabrese EJ. 2017. Cancer immunotherapy: how low-level ionizing radiation can play a key role. Cancer Immunol Immunother. 66(7):819–832.
  • Jansen JThM, Broerse JJ, Zoetelief J, Klein C, Seegenschmiedt HM. 2005. Estimation of the carcinogenic risk of radiotherapy of benign diseases from shoulder to heel. Radiother Oncol. 76(3):270–277.
  • Jargin S. 2012. Hormesis and radiation safety norms. Hum Exp Toxicol. 31(7):671–675.
  • Jella KK, 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(2):138–145.
  • Jerne NK. 1973. The immune system. Sci Am. 229(1):52–63.
  • Joiner MC, Marples B, Lambin P, Short SC, Turesson I. 2001. Low-dose hypersensitivity: current status and possible mechanisms. Inter J Rad Oncol Biol Phys. 49(2):379–389.
  • Kadhim MA, Lorimore SA, Townsend KMS, Goodhead DT, Buckle VJ, Wright EG. 1995. Radiation-induced genomic instability: delayed cytogenetic aberrations and apoptosis in primary human bone marrow cells. Int J Radiat Biol. 67(3):287–293.
  • Kadhim MA, Macdonald DA, Goodhead DT, Lorimore SA, Marsden SJ, Wright EG. 1992. Transmission of chromosomal instability after plutonium alpha-particle irradiation. Nature. 355(6362):738–740.
  • Kadhim MA, Mayah A, Brooks SA. 2020. Does direct and indirect exposure to ionising radiation influence the metastatic potential of breast cancer cells. Cancers. 12(1):236.
  • Kalpana KB, Devipriya N, Thayalan K, Menon VP. 2010. Protection against X-ray radiation-induced cellular damage of human peripheral blood lymphocytes by an aminothiazole derivative of dendrodoine. Chem Biol Interact. 186(3):267–274.
  • Karran P. 1996. Microsatellite instability and DNA mismatch repair in human cancer. Semin Cancer Biol. 7(1):15–24.
  • Karran P, Bignami M. 1994. DNA damage tolerance, mismatch repair and genome instability. Bioessays. 16(11):833–839.
  • Kefayat A, Ghahremani F. 2020. Low dose radiation therapy for COVID-19 pneumonia: a double-edged sword. Radiother Oncol. 147:224–225.
  • Kim GJ, Fiskum GM, Morgan WF. 2006. A role for mitochondrial dysfunction in perpetuating radiation-induced genomic instability. Cancer Res. 66(21):10377–10383.
  • Klug F, Prakash H, Huber PE, Seibel T, Bender N, Halama N, Pfirschke C, Voss RH, Timke C, Umansky L, et al. 2013. Low-dose irradiation programs macrophage differentiation to an iNOS+/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell 24(5):589–602.
  • Kumar A, Dey AD, Behl T, Chadha S, Aggarwal V. 2021. Exploring the multifocal therapeutic approaches in COVID-19: a ray of hope. Int Immunopharmacol. 90:107156.
  • Kurd N, Robey EA. 2016. T-cell selection in the thymus: a spatial and temporal perspective. Immunol Rev. 271(1):114–126.
  • Lara PC, Burgos J, Macias D. 2020. Low dose lung radiotherapy for COVID-19 pneumonia. The rationale for a cost-effective anti-inflammatory treatment. Clin Transl Radiat Oncol. 23:27–29.
  • Laurier D, Richardson DB, Cardis E, Daniels RD, Gillies M, O'Hagan J, Hamra GB, Haylock R, Leuraud K, Moissonnier M, et al. 2017. The international nuclear workers study (Inworks): a collaborative epidemiological study to improve knowledge about health effects of protracted low-dose exposure. Radiat Prot Dosimetry. 173(1-3):21–25.
  • 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(3):e0173685.
  • Le M, McNeill FE, Seymour C, Rainbow AJ, Mothersill CE. 2015. An observed effect of ultraviolet radiation emitted from beta-irradiated HaCaT Cells upon non-beta-irradiated bystander cells. Radiation Res. 183(3):279–290.
  • Le M, McNeill FE, Seymour CB, Rusin A, Diamond K, Rainbow AJ, Murphy J, Mothersill CE. 2018. Modulation of oxidative phosphorylation (OXPHOS) by radiation- induced biophotons. Environ Res. 163:80–87.
  • Litman GW, Cannon JP, Dishaw LJ. 2005. Reconstructing immune phylogeny: new perspectives. Nat Rev Immunol. 5(11):866–879.
  • Litman GW, Rast JP, Shamblott MJ, Haire RN, Hulst M, Roess W, Litman RT, Hinds-Frey KR, Zilch A, Amemiya CT. 1993. Phylogenetic diversification of immunoglobulin genes and the antibody repertoire. Mol Biol Evol. 10(1):60–72.
  • Little MP. 2010. Do non-targeted effects increase or decrease low dose risk in relation to the linear-non-threshold (LNT) model? Mutat Res. 687(1-2):17–27.
  • Littlefield LG, Hollowell JG, Pool WH. 1969. Chromosomal aberrations induced by plasma from irradiated patients: an indirect effect of X radiation. Further observations and studies of a control population. Radiology. 93(4):879–886.
  • Liu S-Z. 2003. On Radiation Hormesis Expressed in the Immune System. Crit Rev Toxicol. 33(3-4):431–441.
  • Liu Y, Sawalha AH, Lu Q. 2021. COVID-19 and autoimmune diseases. Curr Opin Rheumatol. 33(2):155–162.
  • Lord BI. 1999. Transgenerational susceptibility to leukaemia induction resulting from preconception, paternal irradiation. Int J Radiat Biol. 75(7):801–810.
  • Lorimore SA, Chrystal JA, Robinson JI, Coates PJ, Wright EG. 2008. Chromosomal instability in unirradiated hemaopoietic cells induced by macrophages exposed in vivo to ionizing radiation. Cancer Res. 68(19):8122–8126.
  • Lyng FM, Howe OL, McClean B. 2011. Reactive oxygen species-induced release of signalling factors in irradiated cells triggers membrane signalling and calcium influx in bystander cells. Int J Radiat Biol. 87(7):683–695.
  • 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(4):400–409.
  • Lyng FM, Seymour CB, Mothersill C. 2000. Production of a signal by irradiated cells which leads to a response in unirradiated cells characteristic of initiation of apoptosis. Br J Cancer. 83(9):1223–1230.
  • Lyng FM, Seymour CB, Mothersill C. 2001. Oxidative stress in cells exposed to low levels of ionizing radiation. Biochem Soc Transac. 29(2):350–353.
  • Lyng FM, Seymour CB, Mothersill C. 2002. 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(4):365–370.
  • Maguire P, Mothersill C, McClean B, Seymour C, Lyng FM. 2007. Modulation of radiation responses by pre-exposure to irradiated cell conditioned medium. Radiat Res. 167(4):485–492.
  • Maiese A, Manetti AC, La Russa R, Di Paolo M, Turillazzi E, Frati P, Fineschi V. 2021. Autopsy findings in COVID-19-related deaths: a literature review. Forensic Sci Med Pathol. 17(2):279–296.
  • 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(4 Suppl 1):55–63.
  • Matarèse BFE, Lad J, Seymour C, Schofield PN, Mothersill C. 2020. Bio-acoustic signaling; exploring the potential of sound as a mediator of low-dose radiation and stress responses in the environment. Inter J Rad Biol. 0(0):1–15.
  • McBride WH, Chiang C-S, Olson JL, Wang C-C, Hong J-H, Pajonk F, Dougherty GJ, Iwamoto KS, Pervan M, Liao Y-P. 2004. A sense of danger from radiation. Radiat Res. 162(1):1–19.
  • McBride WH, Ganapathy E, Lee M-H, Nesseler JP, Nguyen C, Schaue D. 2017. A perspective on the impact of radiation therapy on the immune rheostat. Br J Radiol. 90(1078):20170272.
  • McKeown SR, Hatfield P, Prestwich RJ, Shaffer RE, Taylor RE. 2015. Radiotherapy for benign disease; assessing the risk of radiation-induced cancer following exposure to intermediate dose radiation. Br J Radiol. 88(1056):20150405.
  • McMahon SJ. 2018. The linear quadratic model: usage, interpretation and challenges. Phys Med Biol. 64(1):01TR01.
  • Melief CJM. 2003. Mini-review: Regulation of cytotoxic T lymphocyte responses by dendritic cells: peaceful coexistence of cross-priming and direct priming? Eur J Immunol. 33(10):2645–2654.
  • Mitchel REJ. 2006. Low Doses of Radiation Reduce Risk In Vivo. Dose Response. 5(1):1–10.
  • Moattari CR, Granstein RD. 2021. Neuropeptides and neurohormones in immune, inflammatory and cellular responses to ultraviolet radiation. Acta Physiol (Oxf). 232(1):e13644.
  • Mohammadi A, Blesso CN, Barreto GE, Banach M, Majeed M, Sahebkar A. 2019. Macrophage plasticity, polarization and function in response to curcumin, a diet-derived polyphenol, as an immunomodulatory agent. J Nutr Biochem. 66:1–16.
  • Mole RH. 1953. Whole Body irradiation; radiobiology or medicine? Br J Radiol. 26(305):234–241.
  • Montero A, Arenas M, Algara M. 2021. Low-dose radiation therapy: could it be a game-changer for COVID-19? Clin Transl Oncol. 23(1):1–4.
  • Morgan WF, Bair WJ. 2013. Issues in low dose radiation biology: the controversy continues. a perspective. Radiat Res. 179(5):501–510.
  • Morgan WF, Sowa MB. 2015. Non-targeted effects induced by ionizing radiation: mechanisms and potential impact on radiation induced health effects. Cancer Lett. 356(1):17–21.
  • Morrissy AS, Garzia L, Shih DJH, Zuyderduyn S, Huang X, Skowron P, Remke M, Cavalli FMG, Ramaswamy V, Lindsay PE, et al. 2016. Divergent clonal selection dominates medulloblastoma at recurrence. Nature. 529(7586):351–357.
  • Mosse IB. 2012. Genetic effects of ionizing radiation-some questions with no answers. J Environ Radioact. 112:70–75.
  • Mothersill C, Rusin A, Fernandez-Palomo C, Seymour C. 2018. History of bystander effects research 1905-present; what is in a name? Int J Radiat Biol. 94(8):696–707.
  • Mothersill C, Rusin A, Seymour C. 2017. Low doses and non-targeted effects in environmental radiation protection; where are we now and where should we go? Environ Res. 159:484–490.
  • Mothersill C, Seymour C. 1997. Lethal mutations and genomic instability. Int J Radiat Biol. 71(6):751–758.
  • Mothersill C, Seymour C. 2009. Communication of ionising radiation signals-a tale of two fish. Int J Radiat Biol. 85(11):909–919.
  • Mothersill C, Seymour C. 2013. Radiation-induced bystander effects and stress-induced mutagenesis. In: Mittelman D, editor. Stress-induced mutagenesis. New York (NY): Springer; p. 199–222.
  • 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(3):256–262.
  • Mothersill C, Seymour CB, Joiner MC. 2002. Relationship between radiation-induced low-dose hypersensitivity and the bystander effect. Rare. 157(5):526–532.
  • Mothersill C, Smith RW, Fazzari J, McNeill F, Prestwich W, Seymour CB. 2012. Evidence for a physical component to the radiation-induced bystander effect? Int J Radiat Biol. 88(8):583–591.
  • Mothersill C, Stamato TD, Perez ML, Cummins R, Mooney R, Seymour CB. 2000. Involvement of energy metabolism in the production of ‘bystander effects’ by radiation. Br J Cancer. 82(10):1740–1746.
  • Mottram ME, Hill HA. 1949. Radiation therapy of ringworm of the scalp. Calif Med. 70(3):189–193.
  • Muecke R, Micke O, Reichl B, Heyder R, Prott F-J, Seegenschmiedt MH, Glatzel M, Schneider O, Schäfer U, Kundt G. 2007. Demographic, clinical and treatment related predictors for event-free probability following low-dose radiotherapy for painful heel spurs – a retrospective multicenter study of 502 patients. Acta Oncologica. 46(2):239–246.
  • Müller WE, Blumbach B, Müller IM. 1999. Evolution of the innate and adaptive immune systems: relationships between potential immune molecules in the lowest metazoan phylum (Porifera) and those in vertebrates. Transplantation. 68(9):1215–1227.
  • Müller WEG. 2001. Review: How was metazoan threshold crossed? The hypothetical Urmetazoa. Compar Biochem Physiol Part A Mol Integr Physiol. 129(2-3):433–460.
  • Müller WEG, Wiens M, Gamulin AT, Schröder V, Müller Im HC. 2004. Bauplan of Urmetazoa: basis for genetic complexity of metazoa. Int Rev Cytol. 235:53–92.
  • Murphy JB, Liu JH, Sturm E. 1922. Studies on X-ray effects : ix. the action of serum from x-rayed animals on lymphoid cells in vitro. J Exp Med. 35(3):373–384.
  • Murphy JB, Morton JJ. 1915. The lymphocyte in natural and induced resistance to transplanted cancer : II. studies in lymphoid activity. J Exp Med. 22(2):204–211.
  • Murphy JB, Taylor HD. 1918. The lymphocyte in natural and induced resistance to transplanted cancer : III. the effect of X-rays on artificially induced immunity. J Exp Med. 28(1):1–9.
  • Nagasawa H, Little JB. 1992. Induction of sister chromatid exchanges by extremely low doses of α-particles. Cancer Res. 52(22):6394–6396.
  • Nakahara W, Murphy JB. 1920. Studies on X-ray effects. J Exp Med. 31(1):13–29.
  • Nakahira K, Haspel JA, Rathinam VA, Lee S-J, Dolinay T, Lam HC, Englert JA, Rabinovitch M, Cernadas M, Kim HP, et al. 2011. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome . Nat Immunol. 12(3):222–230.
  • Nakanishi M, Tanaka K, Shintani T, Takahashi T, Kamada N. 1999. Chromosomal instability in acute myelocytic leukemia and myelodysplastic syndrome patients among atomic bomb survivors. JRR. 40(2):159–167.
  • Nicholson LB. 2016. The immune system. Essays Biochem. 60(3):275–301.
  • Nicolò P, Matteo C, Daniela P, Aiti V. 2016. Chapter 13 - inflammatory response of the ascidian ciona intestinalis. In: Ballarin L, Cammarata M, editors. Lessons in immunity. London (UK): Academic Press; p. 177–192.
  • Niewald M, Seegenschmiedt MH, Micke O, Graeber S, Muecke R, Schaefer V, Scheid C, Fleckenstein J, Licht N, Ruebe C. 2012. Randomized, multicenter trial on the effect of radiation therapy on plantar fasciitis (painful heel spur) comparing a standard dose with a very low dose: mature results after 12 months’ follow-up. Inter J Rad Oncol Biol Phys. 84(4):e455–e462.
  • Nikitaki Z, Mavragani IV, Laskaratou DA, Gika V, Moskvin VP, Theofilatos K, Vougas K, Stewart RD, Georgakilas AG. 2016. Systemic mechanisms and effects of ionizing radiation: a new 'old' paradigm of how the bystanders and distant can become the players. Semin Cancer Biol. 37-38:77–95.
  • Nowosielska EM, Cheda A, Wrembel-Wargocka J, Janiak MK. 2012. Effect of low doses of low-let radiation on the innate anti-tumor reactions in radioresistant and radiosensitive mice. Dose Response. 10(4):500–515.
  • Nugent SME, Mothersill CE, Seymour C, McClean B, Lyng FM, Murphy JEJ. 2007. Increased mitochondrial mass in cells with functionally compromised mitochondria after exposure to both direct gamma radiation and bystander factors. Radiat Res. 168(1):134–142.
  • Olivieri G, Bodycote J, Wolff S. 1984. Adaptive response of human lymphocytes to low concentrations of radioactive thymidine. Science. 223(4636):594–597.
  • Padgham M. 2004. Reverberation and frequency attenuation in forests-implications for acoustic communication in animals. J Acoust Soc Am. 115(1):402–410.
  • 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(1):179–189.
  • Portess DI, Bauer G, Hill MA, O'Neill P. 2007. Low-dose irradiation of nontransformed cells stimulates the selective removal of precancerous cells via intercellular induction of apoptosis. Cancer Res. 67(3):1246–1253.
  • Prasanna A, Ahmed MM, Mohiuddin M, Coleman CN. 2014. Exploiting sensitization windows of opportunity in hyper and hypo-fractionated radiation therapy. J Thorac Dis. 6(4):287–302.
  • Prasanna PG, Woloschak GE, DiCarlo AL, Buchsbaum JC, Schaue D, Chakravarti A, Cucinotta FA, Formenti SC, Guha C, Hu DJ, et al. 2020. Low-Dose Radiation Therapy (LDRT) for COVID-19: benefits or risks? Radiat Res. 194(5):452–464.
  • Prise K, Folkard M, D. Michael (INVITED) B. 2003. A review of the bystander effect and its implications for low-dose exposure. Radiat Prot Dosimetry. 104(4):347–355.
  • Prise KV, Belyakov OC, Newman H, Patel S, Schettino G, Folkard MD, Michael (INVITED) B. 2002. Non-targeted Effects of radiation: bystander responses in cell and tissue models. Radiat Prot Dosimetry. 99(1-4):223–226.
  • Pusey WA, Caldwell E. 1903. The practical application of the Röntgen rays in therapeutics and diagnosis. Philadelphia: Saunders.
  • Rinkevich B, Weissman IL. 1992. Allogeneic resorption in colonial protochordates: consequences of nonself recognition. Dev Comp Immunol. 16(4):275–286.
  • Rödel F, Keilholz L, Herrmann M, Sauer R, Hildebrandt G. 2007. Radiobiological mechanisms in inflammatory diseases of low-dose radiation therapy. Int J Radiat Biol. 83(6):357–366.
  • Rusin A, Seymour C, Mothersill C. 2018. Chronic fatigue and immune deficiency syndrome (CFIDS), cellular metabolism, and ionizing radiation: a review of contemporary scientific literature and suggested directions for future research. Int J Radiat Biol. 94(3):212–228.
  • Russ S, Chambers H, Scott GM, Mottram JC. 1919. Experimental studies with small doses of x rays. The Lancet. 193(4991):692–695.
  • Ryan LA, Seymour CB, O'Neill-Mehlenbacher A, Mothersill CE. 2008. Radiation-induced adaptive response in fish cell lines. J Environ Radioact. 99(4):739–747.
  • Ryan MT, Poston JW. 2006. A half century of health physics: 50th anniversary of the health physics society. Baltimore (MD): Lippincott Williams & Wilkins.
  • Salomaa S, Bouffler SD, Atkinson MJ, Cardis E, Hamada N. 2020. Is there any supportive evidence for low dose radiotherapy for COVID-19 pneumonia? Int J Radiat Biol. 96(10):1228–1235.
  • Schaue D. 2017. A century of radiation therapy and adaptive immunity. Front Immunol. 8:431.
  • Schaue D, Kachikwu EL, McBride WH. 2012. Cytokines in radiobiological responses: a review. Radiat Res. 178(6):505–523.
  • Schaue D, Marples B, Trott KR. 2002. The effects of low-dose X-irradiation on the oxidative burst in stimulated macrophages. Null. 78(7):567–576.
  • Schirrmacher V. 2020. Mitochondria at work: new insights into regulation and dysregulation of cellular energy supply and metabolism. Biomedicines. 8(11):526.
  • Schneider O, Stückle CA, Bosch E, Gott C, Adamietz IA. 2004. Effectiveness and prognostic factors of radiotherapy for painful plantar heel spurs. Strahlenther Onkol. 180(8):502–509.
  • Scott BR. 2008. It's time for a new low-dose-radiation risk assessment paradigm-one that acknowledges hormesis. Dose Response. 6(4):333–351.
  • Scott BR. 2017. Small radiation doses enhance natural barriers to cancer. J Am Physic Surg. 22(4):105.
  • Scott WR. 1939. X-ray therapy in the treatment of acute pneumonia. Radiology. 33(3):331–349.
  • Seymour CB, Mothersill C, Alper T. 1986. High yields of lethal mutations in somatic mammalian cells that survive ionizing radiation. Int J Radiat Biol Relat Stud Phys Chem Med. 50(1):167–179.
  • Shamblott MJ, Litman GW. 1989. Complete nucleotide sequence of primitive vertebrate immunoglobulin light chain genes. Proc Natl Acad Sci USA. 86(12):4684–4688.
  • 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(3):479–487.
  • Shimura T, Kunugita N. 2016. Mitochondrial reactive oxygen species-mediated genomic instability in low-dose irradiated human cells through nuclear retention of cyclin D1. Cell Cycle. 15(11):1410–1414.
  • Sisakht M, Darabian M, Mahmoodzadeh A, Bazi A, Shafiee SM, Mokarram P, Khoshdel Z. 2020. The role of radiation induced oxidative stress as a regulator of radio-adaptive responses. Int J Radiat Biol. 96(5):561–576.
  • Smith LC, Davidson EH. 1994. The echinoderm immune system. characters shared with vertebrate immune systems and characters arising later in deuterostome phylogeny. Ann N Y Acad Sci. 712(1):213–226.
  • Souto J. 1962. Tumour development in the rat induced by blood of irradiated animals. Nature. 195(4848):1317–1318.
  • Spix C, Schmiedel S, Kaatsch P, Schulze-Rath R, Blettner M. 2008. Case-control study on childhood cancer in the vicinity of nuclear power plants in Germany 1980-2003. Eur J Cancer. 44(2):275–284.
  • Surinov BP, Isaeva VG, Dukhova NN. 2004. Postirradiation volatile secretions of mice: syngeneic and allogeneic immune and behavioral effects. Bull Exp Biol Med. 138(4):384–386.
  • Sutou S. 2018. Low-dose radiation from A-bombs elongated lifespan and reduced cancer mortality relative to un-irradiated individuals. Genes Environ. 40:26.
  • Szatmári T, Persa E, Kis E, Benedek A, Hargitai R, Sáfrány G, Lumniczky K. 2019. Extracellular vesicles mediate low dose ionizing radiation-induced immune and inflammatory responses in the blood. Int J Radiat Biol. 95(1):12–22.
  • Szumiel I. 2015. Ionizing radiation-induced oxidative stress, epigenetic changes and genomic instability: The pivotal role of mitochondria. Int J Radiat Biol. 91(1):1–12.
  • Taher M, Tik N, Susanti D. 2021. Drugs intervention study in COVID-19 management. Drug Metabol Personal Ther. 36(2):87–98.
  • Tang FR, Loke WK. 2015. Molecular mechanisms of low dose ionizing radiation-induced hormesis, adaptive responses, radioresistance, bystander effects, and genomic instability. Int J Radiat Biol. 91(1):13–27.
  • Taylor HD, Witherbee WD, Murphy JB. 1919. Studies on X-ray effects. J Exp Med. 29(1):53–73.
  • Till JE, Beck HL, Aanenson JW, Grogan HA, Mohler HJ, Mohler SS, Voillequé PG. 2018. Dosimetry associated with veterans who participated in nuclear weapons testing. Int J Radiat Biol . 0(0):1–9.
  • Tobaiqy M, Qashqary M, Al-Dahery S, Mujallad A, Hershan AA, Kamal MA, Helmi N. 2020. Therapeutic management of patients with COVID-19: a systematic review. Infect Prevent Prac. 2(3):100061.
  • Tuncay Cagatay S, Mayah A, Mancuso M, Giardullo P, Pazzaglia S, Saran A, Daniel A, Traynor D, Meade AD, Lyng F. 2020. Phenotypic and functional characteristics of exosomes derived from irradiated mouse organs and their role in the mechanisms driving non-targeted effects. Int J Mol Sci. 21(21):8389.
  • Ullrich SE, Byrne SN. 2012. The immunologic revolution: photoimmunology. J Invest Dermatol. 132(3 Pt 2):896–905.
  • United Nations Scientific Committee on the Effects of Atomic Radiation 2017. Sources, Effects and Risks of Ionizing Radiation, UNSCEAR 2012 Report: Report to the General Assembly, with Scientific Annexes A and B. Geneva: UN; [accessed 2021 Mar 19].
  • Valerie K, Yacoub A, Hagan MP, Curiel DT, Fisher PB, Grant S, Dent P. 2007. Radiation-induced cell signaling: inside-out and outside-in. Mol Cancer Ther. 6(3):789–801.
  • Wambersie A, Jr Caswell PMD, Menzel Hg RS. 2000. The International Commission on Radiation Units and Measurements (ICRU): Activities and Future Plans. ICRP Publication. 60:20–22.
  • Warthin AS. 1907. The changes produced in the kidneys by röntgen irradiation. Am J Med Sci (1827–1924). 133(5):736.
  • Watson GE, Lorimore SA, Macdonald DA, Wright EG. 2000. Chromosomal instability in unirradiated cells induced in vivo by a bystander effect of ionizing radiation. Cancer Res. 60(20):5608–5611.
  • Yahyapour R, Amini P, Rezapour S, Cheki M, Rezaeyan A, Farhood B, Shabeeb D, Musa AE, Fallah H, Najafi M. 2018. Radiation-induced inflammation and autoimmune diseases. Mil Med Res. 5(1):9.
  • York JM, Blevins NA, Meling DD, Peterlin MB, Gridley DS, Cengel KA, Freund GG. 2012. The biobehavioral and neuroimmune impact of low-dose ionizing radiation. Brain Behav Immun. 26(2):218–227.
  • Yoshida GJ, Saya H. 2021. Molecular pathology underlying the robustness of cancer stem cells. Regen Ther. 17:38–50.
  • Zahnreich S, Rösler H-P, Schwanbeck C, Karle H, Schmidberger H. 2020. Radiation-induced DNA double-strand breaks in peripheral leukocytes and therapeutic response of heel spur patients treated by orthovoltage X-rays or a linear accelerator. Strahlenther Onkol. 196(12):1116–1127.

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.