Skip to main content
Log in

Intercellular communications-redox interactions in radiation toxicity; potential targets for radiation mitigation

  • Review
  • Published:
Journal of Cell Communication and Signaling Aims and scope

Abstract

Nowadays, using ionizing radiation (IR) is necessary for clinical, agricultural, nuclear energy or industrial applications. Accidental exposure to IR after a radiation terror or disaster poses a threat to human. In contrast to the old dogma of radiation toxicity, several experiments during the last two recent decades have revealed that intercellular signaling and communications play a key role in this procedure. Elevated level of cytokines and other intercellular signals increase oxidative damage and inflammatory responses via reduction/oxidation interactions (redox system). Intercellular signals induce production of free radicals and inflammatory mediators by some intermediate enzymes such as cyclooxygenase-2 (COX-2), nitric oxide synthase (NOS), NADPH oxidase, and also via triggering mitochondrial ROS. Furthermore, these signals facilitate cell to cell contact and increasing cell toxicity via cohort effect. Nitric oxide is a free radical with ability to act as an intercellular signal that induce DNA damage and changes in some signaling pathways in irradiated as well as non-irradiated adjacent cells. Targeting of these mediators by some anti-inflammatory agents or via antioxidants such as mitochondrial ROS scavengers opens a window to mitigate radiation toxicity after an accidental exposure. Experiments which have been done so far suggests that some cytokines such as IL-1β, TNF-α, TGF-β, IL-4 and IL-13 are some interesting targets that depend on irradiated organs and may help mitigate radiation toxicity. Moreover, animal experiments in recent years indicated that targeting of toll like receptors (TLRs) may be more useful for radioprotection and mitigation. In this review, we aimed to describe the role of intercellular interactions in oxidative injury, inflammation, cell death and killing effects of IR. Moreover, we described evidence on potential mitigation of radiation injury via targeting of these mediators.

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.

Fig. 1

Similar content being viewed by others

References

  • Ameziane-El-Hassani R, Talbot M, de Souza Dos Santos MC, Al Ghuzlan A, Hartl D, Bidart J-M, De Deken X, Miot F, Diallo I, De Vathaire F, Schlumberger M, Dupuy C (2015) NADPH oxidase DUOX1 promotes long-term persistence of oxidative stress after an exposure to irradiation. Proc Natl Acad Sci U S A 112:5051–5056

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Anscher MS (2010) Targeting the TGF-β1 Pathway to Prevent Normal Tissue Injury After Cancer Therapy. The Oncologist 15:350–359

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Anscher MS, Thrasher B, Rabbani Z, Teicher B, Vujaskovic Z (2006) Antitransforming growth factor-beta antibody 1D11 ameliorates normal tissue damage caused by high-dose radiation. Int J Radiat Oncol Biol Phys 65:876–881

    Article  CAS  PubMed  Google Scholar 

  • Archambeau JO, Tovmasyan A, Pearlstein RD, Crapo JD, Batinic-Haberle I (2013) Superoxide dismutase mimic, MnTE-2-PyP(5+) ameliorates acute and chronic proctitis following focal proton irradiation of the rat rectum. Redox Biol 1:599–607

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Autsavapromporn N, de Toledo SM, Little JB, Jay-Gerin JP, Harris AL, Azzam EI (2011) The role of gap junction communication and oxidative stress in the propagation of toxic effects among high-dose alpha-particle-irradiated human cells. Radiat Res 175:347–357

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Autsavapromporn N, de Toledo SM, Jay-Gerin JP, Harris AL, Azzam EI (2013a) Human cell responses to ionizing radiation are differentially affected by the expressed connexins. J Radiat Res 54:251–259

    Article  CAS  PubMed  Google Scholar 

  • Autsavapromporn N, Suzuki M, Plante I, Liu C, Uchihori Y, Hei TK, Azzam EI, Murakami T (2013b) Participation of gap junction communication in potentially lethal damage repair and DNA damage in human fibroblasts exposed to low- or high-LET radiation. Mutat Res 756:78–85

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • 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

    Article  CAS  PubMed  Google Scholar 

  • 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 98:473–478

    CAS  PubMed  PubMed Central  Google Scholar 

  • Azzam EI, de Toledo SM, Spitz DR, Little JB (2002) Oxidative metabolism modulates signal transduction and micronucleus formation in bystander cells from α-particle-irradiated normal human fibroblast cultures. Cancer Res 62:5436–5442

    CAS  PubMed  Google Scholar 

  • Barnaby F (1995) The effects of the atomic bombings of Hiroshima and Nagasaki. Med War 11:1–9

    Article  CAS  PubMed  Google Scholar 

  • Battino M, Bullon P, Wilson M, Newman H (1999) Oxidative injury and inflammatory periodontal diseases: the challenge of anti-oxidants to free radicals and reactive oxygen species. Crit Rev Oral Biol Med 10:458–476

    Article  CAS  PubMed  Google Scholar 

  • Berhane H, Shinde A, Kalash R, Xu K, Epperly MW, Goff J, Franicola D, Zhang X, Dixon T, Shields D, Wang H, Wipf P, Li S, Gao X, Greenberger JS (2014) Amelioration of radiation-induced oral cavity mucositis and distant bone marrow suppression in fanconi anemia Fancd2-/- (FVB/N) mice by intraoral GS-nitroxide JP4-039. Radiat Res 182:35–49

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • 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

    Article  CAS  PubMed  Google Scholar 

  • Boerma M, Wang J, Sridharan V, Herbert JM, Hauer-Jensen M (2013) Pharmacological induction of transforming growth factor-beta1 in rat models enhances radiation injury in the intestine and the heart. PLoS One 8:e70479

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Boerma M, Sridharan V, Mao XW, Nelson GA, Cheema AK, Koturbash I, Singh SP, Tackett AJ, Hauer-Jensen M (2016) Effects of ionizing radiation on the heart. Mutat Res 770:319–327

    Article  CAS  PubMed Central  Google Scholar 

  • Brand MD, Affourtit C, Esteves TC, Green K, Lambert AJ, Miwa S, Pakay JL, Parker N (2004) Mitochondrial superoxide: production, biological effects, and activation of uncoupling proteins. Free Radic Biol Med 37:755–767

    Article  CAS  PubMed  Google Scholar 

  • Burdelya LG, Krivokrysenko VI, Tallant TC, Strom E, Gleiberman AS, Gupta D, Kurnasov OV, Fort FL, Osterman AL, Didonato JA, Feinstein E, Gudkov AV (2008) An agonist of toll-like receptor 5 has radioprotective activity in mouse and primate models. Science 320:226–230

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Calabrese V, Mancuso C, Calvani M, Rizzarelli E, Butterfield DA, Stella AM (2007) Nitric oxide in the central nervous system: neuroprotection versus neurotoxicity. Nat Rev Neurosci 8:766–775

    Article  CAS  PubMed  Google Scholar 

  • Cali B, Ceolin S, Ceriani F, Bortolozzi M, Agnellini AH, Zorzi V, Predonzani A, Bronte V, Molon B, Mammano F (2015) Critical role of gap junction communication, calcium and nitric oxide signaling in bystander responses to focal photodynamic injury. Oncotarget 6:10161–10174

    Article  PubMed  PubMed Central  Google Scholar 

  • Chai Y, Calaf G, Zhou H, Ghandhi S, Elliston C, Wen G, Nohmi T, Amundson S, Hei T (2012) Radiation induced COX-2 expression and mutagenesis at non-targeted lung tissues of gpt delta transgenic mice. Br J Cancer 108:91–98

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Chai Y, Lam R, Calaf G, Zhou H, Amundson S, Hei T (2013) Radiation-induced non-targeted response in vivo: role of the TGFβ-TGFBR1-COX-2 signalling pathway. Br J Cancer 108:1106–1112

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Chang J, Feng W, Wang Y, Luo Y, Allen AR, Koturbash I, Turner J, Stewart B, Raber J, Hauer-Jensen M, Zhou D, Shao L (2015) Whole-body proton irradiation causes long-term damage to hematopoietic stem cells in mice. Radiat Res 183:240–248

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Chaudhry MA, Omaruddin RA (2012) Differential regulation of microRNA expression in irradiated and bystander cells. Mol Biol (Mosk) 46:634–643

    Article  CAS  Google Scholar 

  • Cheki M, Yahyapour R, Farhood B, Rezaeyan A, Shabeeb D, Amini P, Rezapoor S, Najafi M (2018) COX-2 in Radiotherapy; a potential target for radioprotection and radiosensitization. Curr Mol Pharmacol. https://doi.org/10.2174/1874467211666180219102520

  • Cho HJ, Lee WH, Hwang OMH, Sonntag WE, Lee YW (2017) Role of NADPH oxidase in radiation-induced pro-oxidative and pro-inflammatory pathways in mouse brain. Int J Radiat Biol 93:1257–1266

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Chung SI, Horton JA, Ramalingam TR, White AO, Chung EJ, Hudak KE, Scroggins BT, Arron JR, Wynn TA, Citrin DE (2016) IL-13 is a therapeutic target in radiation lung injury. Sci Rep 6

  • Collins-Underwood JR, Zhao W, Kooshki M, Robbins M (2007) Modulation of NADPH oxidase by ionizing radiation and its role in radiation-induced oxidative stress and inflammation in brain endothelium. Cancer Res 67:1381–1381

    Google Scholar 

  • Dancea HC, Shareef MM, Ahmed MM (2009) Role of Radiation-induced TGF-beta Signaling in Cancer Therapy. Mol Cell Pharmacol 1

  • Dayal D, Martin SM, Owens KM, Aykin-Burns N, Zhu Y, Boominathan A, Pain D, Limoli CL, Goswami PC, Domann FE, Spitz DR (2009) Mitochondrial complex II dysfunction can contribute significantly to genomic instability after exposure to ionizing radiation. Radiat Res 172:737–745

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • de Toledo SM, Buonanno M, Harris AL, Azzam EI (2017) Genomic instability induced in distant progeny of bystander cells depends on the connexins expressed in the irradiated cells. Int J Radiat Biol 93:1182–1194

    Article  CAS  PubMed  Google Scholar 

  • Dorr H, Meineke V (2011) Acute radiation syndrome caused by accidental radiation exposure - therapeutic principles. BMC Med 9:126

    Article  PubMed  PubMed Central  Google Scholar 

  • Douple EB, Mabuchi K, Cullings HM, Preston DL, Kodama K, Shimizu Y, Fujiwara S, Shore RE (2011) Long-term radiation-related health effects in a unique human population: lessons learned from the atomic bomb survivors of Hiroshima and Nagasaki. Disaster Med Public Health Prep 5(Suppl 1):S122–S133

    Article  PubMed  PubMed Central  Google Scholar 

  • Eldabaje R, Le DL, Huang W, Yang LX (2015) Radiation-associated Cardiac Injury. Anticancer Res 35:2487–2492

    PubMed  Google Scholar 

  • Fadok VA, Bratton DL, Konowal A, Freed PW, Westcott JY, Henson PM (1998) Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-beta, PGE2, and PAF. J Clin Invest 101:890–898

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Fang Z, Xu A, Wu L, Hei TK, Hong M (2016) The role of protein kinase C alpha translocation in radiation-induced bystander effect. Sci Rep 6:25817

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Fardid R, A S, Mosleh-Shirazi MA, Sharifzadeh S, Okhovat MA, Najafi M, Rezaeyan A, Abaszadeh A (2017) Melatonin ameliorates the production of COX-2, iNOS, and the formation of 8-OHdG in non-targeted lung tissue after pelvic irradiation. Cell J 19:324–331

    PubMed  PubMed Central  Google Scholar 

  • Flechsig P, Dadrich M, Bickelhaupt S, Jenne J, Hauser K, Timke C, Peschke P, Hahn EW, Grone HJ, Yingling J, Lahn M, Wirkner U, Huber PE (2012) LY2109761 attenuates radiation-induced pulmonary murine fibrosis via reversal of TGF-beta and BMP-associated proinflammatory and proangiogenic signals. Clin Cancer Res 18:3616–3627

    Article  CAS  PubMed  Google Scholar 

  • Gauter-Fleckenstein B, Fleckenstein K, Owzar K, Jiang C, Reboucas JS, Batinic-Haberle I, Vujaskovic Z (2010a) Early and late administration of MnTE-2-PyP5+ in mitigation and treatment of radiation-induced lung damage. Free Radic Biol Med 48:1034–1043

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Gauter-Fleckenstein B, Fleckenstein K, Owzar K, Jiang C, Rebouças JS, Batinic-Haberle I, Vujaskovic Z (2010b) Early and late administration of MnTE-2-PyP 5+ in mitigation and treatment of radiation-induced lung damage. Free Radic Biol Med 48:1034–1043

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Ghandhi SA, Yaghoubian B, Amundson SA (2008) Global gene expression analyses of bystander and alpha particle irradiated normal human lung fibroblasts: Synchronous and differential responses. BMC Med Genet 1:63–63

    Google Scholar 

  • Ghobadi A, Shirazi A, Najafi M, Kahkesh MH, Rezapoor S (2017) Melatonin ameliorates radiation-induced oxidative stress at targeted and nontargeted lung tissue. J Med Phys 42(4):241–244

  • Ghosh S, Maurya DK, Krishna M (2008) Role of iNOS in bystander signaling between macrophages and lymphoma cells. Int J Radiat Oncol Biol Phys 72:1567–1574

    Article  CAS  PubMed  Google Scholar 

  • Goff JP, Epperly MW, Dixon T, Wang H, Franicola D, Shields D, Wipf P, Li S, Gao X, Greenberger JS (2011) Radiobiologic effects of GS-nitroxide (JP4-039) on the hematopoietic syndrome. In Vivo 25:315–323

    PubMed  Google Scholar 

  • Golden E, Pellicciotta I, Demaria S, Barcellos-Hoff MH, Formenti S (2012) The convergence of radiation and immunogenic cell death signaling pathways. Front Oncol 2

  • Grammaticos P, Giannoula E, Fountos GP (2013) Acute radiation syndrome and chronic radiation syndrome. Hell J Nucl Med 16:56–59

    PubMed  Google Scholar 

  • Groves AM, Johnston CJ, Misra RS, Williams JP, Finkelstein JN (2016) Effects of IL-4 on pulmonary fibrosis and the accumulation and phenotype of macrophage subpopulations following thoracic irradiation. Int J Radiat Biol 92:754–765

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Guo J, Chen Y, Lei X, Xu Y, Liu Z, Cai J, Gao F, Yang Y (2017) Monophosphoryl lipid a attenuates radiation injury through TLR4 activation. Oncotarget 8:86031–86042

    PubMed  PubMed Central  Google Scholar 

  • Han W, Wu L, Chen S, Bao L, Zhang L, Jiang E, Zhao Y, Xu A, Hei TK, Yu Z (2007) Constitutive nitric oxide acting as a possible intercellular signaling molecule in the initiation of radiation-induced DNA double strand breaks in non-irradiated bystander cells. Oncogene 26:2330–2339

    Article  CAS  PubMed  Google Scholar 

  • Hardee ME, Marciscano AE, Medina-Ramirez CM, Zagzag D, Narayana A, Lonning SM, Barcellos-Hoff MH (2012) Resistance of glioblastoma-initiating cells to radiation mediated by the tumor microenvironment can be abolished by inhibiting transforming growth factor-beta. Cancer Res 72:4119–4129

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Havaki S, Kotsinas A, Chronopoulos E, Kletsas D, Georgakilas A, Gorgoulis VG (2015) The role of oxidative DNA damage in radiation induced bystander effect. Cancer Lett 356:43–51

    Article  CAS  PubMed  Google Scholar 

  • He J, Jiang B-H (2016) Interplay between Reactive oxygen Species and MicroRNAs in Cancer. Curr Pharmacol Rep 2:82–90

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Hildebrandt G, Loppnow G, Jahns J, Hindemith M, Anderegg U, Saalbach A, Kamprad F (2003) Inhibition of the iNOS pathway in inflammatory macrophages by low-dose X-irradiation in vitro. Is there a time dependence? Strahlenther Onkol 179:158–166

    Article  PubMed  Google Scholar 

  • Hong C-W, Kim Y-M, Pyo H, Lee J-H, Kim S, Lee S, Noh JM (2013) Involvement of inducible nitric oxide synthase in radiation-induced vascular endothelial damage. J Radiat Res 54:1036–1042

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Horton JA, Li F, Chung EJ, Hudak K, White A, Krausz K, Gonzalez F, Citrin D (2013) Quercetin inhibits radiation-induced skin fibrosis. Radiat Res 180:205–215

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Hosseinimehr SJ, Fathi M, Ghasemi A, Shiadeh SN, Pourfallah TA (2017) Celecoxib mitigates genotoxicity induced by ionizing radiation in human blood lymphocytes. Res Pharm Sci 12:82–87

    Article  PubMed  PubMed Central  Google Scholar 

  • Huaux F, Liu T, McGarry B, Ullenbruch M, Phan SH (2003) Dual roles of IL-4 in lung injury and fibrosis. J Immunol 170:2083–2092

    Article  CAS  PubMed  Google Scholar 

  • Jang CW, Chen CH, Chen CC, Chen JY, Su YH, Chen RH (2002) TGF-beta induces apoptosis through Smad-mediated expression of DAP-kinase. Nat Cell Biol 4:51–58

    Article  CAS  PubMed  Google Scholar 

  • Jiang J, Belikova NA, Hoye AT, Zhao Q, Epperly MW, Greenberger JS, Wipf P, Kagan VE (2008) A mitochondria-targeted nitroxide/hemigramicidin S conjugate protects mouse embryonic cells against gamma irradiation. Int J Radiat Oncol Biol Phys 70:816–825

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Jiang J, Stoyanovsky DA, Belikova NA, Tyurina YY, Zhao Q, Tungekar MA, Kapralova V, Huang Z, Mintz AH, Greenberger JS, Kagan VE (2009) A mitochondria-targeted triphenylphosphonium-conjugated nitroxide functions as a radioprotector/mitigator. Radiat Res 172:706–717

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Jiang Y, Chen X, Tian W, Yin X, Wang J, Yang H (2014) The role of TGF-β1–miR-21–ROS pathway in bystander responses induced by irradiated non-small-cell lung cancer cells. Br J Cancer 111:772–780

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Kamiya K, Ozasa K, Akiba S, Niwa O, Kodama K, Takamura N, Zaharieva EK, Kimura Y, Wakeford R (2015) Long-term effects of radiation exposure on health. Lancet 386:469–478

    Article  CAS  PubMed  Google Scholar 

  • Karki R, Igwe OJ (2013) Toll-like receptor 4-mediated nuclear factor kappa B activation is essential for sensing exogenous oxidants to propagate and maintain oxidative/nitrosative cellular stress. PloS one 8

  • Khayyal MT, El-Ghazaly MA, El-Hazek RM, Nada AS (2009) The effects of celecoxib, a COX-2 selective inhibitor, on acute inflammation induced in irradiated rats. Inflammopharmacology 17:255–266

    Article  CAS  PubMed  Google Scholar 

  • Klune JR, Dhupar R, Cardinal J, Billiar TR, Tsung A (2008) HMGB1: endogenous danger signaling. Mol Med 14:476–484

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Koh RY, Lim CL, Uhal BD, Abdullah M, Vidyadaran S, Ho CC, Seow HF (2015) Inhibition of transforming growth factor-beta via the activin receptor-like kinase-5 inhibitor attenuates pulmonary fibrosis. Mol Med Rep 11:3808–3813

    Article  CAS  PubMed  Google Scholar 

  • Kostyuk SV, Ermakov AV, Alekseeva AY, Smirnova TD, Glebova KV, Efremova LV, Baranova A, Veiko NN (2012) Role of extracellular DNA oxidative modification in radiation induced bystander effects in human endotheliocytes. Mutat Res 729:52–60

    Article  CAS  PubMed  Google Scholar 

  • Krivokrysenko VI, Toshkov IA, Gleiberman AS, Krasnov P, Shyshynova I, Bespalov I, Maitra RK, Narizhneva NV, Singh VK, Whitnall MH, Purmal AA, Shakhov AN, Gudkov AV, Feinstein E (2015) The Toll-Like Receptor 5 Agonist Entolimod Mitigates Lethal Acute Radiation Syndrome in Non-Human Primates. PLoS One 10:e0135388

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P, Vandenabeele P (2012) Immunogenic cell death and DAMPs in cancer therapy. Nat Rev Cancer 12:860–875

    Article  CAS  PubMed  Google Scholar 

  • Kurkjian CJ, Guo H, Montgomery ND, Cheng N, Yuan H, Merrill JR, Sempowski GD, Brickey WJ, Ting JP (2017) The Toll-Like Receptor 2/6 Agonist, FSL-1 Lipopeptide, Therapeutically Mitigates Acute Radiation Syndrome. Sci Rep 7:17355

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Leach JK, Van Tuyle G, Lin P-S, Schmidt-Ullrich R, Mikkelsen RB (2001) Ionizing Radiation-induced, Mitochondria-dependent Generation of Reactive Oxygen/Nitrogen. Cancer Res 61:3894–3901

    CAS  PubMed  Google Scholar 

  • Lee JC, Krochak R, Blouin A, Kanterakis S, Chatterjee S, Arguiri E, Vachani A, Solomides CC, Cengel KA, Christofidou-Solomidou M (2009) Dietary flaxseed prevents radiation-induced oxidative lung damage, inflammation and fibrosis in a mouse model of thoracic radiation injury. Cancer Biol Ther 8:47–53

    Article  CAS  PubMed  Google Scholar 

  • Li G, Tang D, Lotze MT (2013) Menage a Trois in stress: DAMPs, redox and autophagy. Semin Cancer Biol 23:380–390

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • 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 20:7085–7095

    Article  CAS  PubMed  Google Scholar 

  • Mahmood J, Jelveh S, Zaidi A, Doctrow SR, Hill RP (2013) Mitigation of radiation-induced lung injury with EUK-207 and genistein: effects in adolescent rats. Radiat Res 179:125–134

    Article  CAS  PubMed  Google Scholar 

  • Maier P, Hartmann L, Wenz F, Herskind C (2016) Cellular Pathways in Response to Ionizing Radiation and Their Targetability for Tumor Radiosensitization. Int J Mol Sci 17:102

    Article  CAS  PubMed Central  Google Scholar 

  • Martin M, Lefaix J-L, Delanian S (2000) TGF-β1 and radiation fibrosis: a master switch and a specific therapeutic target? Int J Radiat Oncol Biol Phys 47:277–290

    Article  CAS  PubMed  Google Scholar 

  • Medhora M, Gao F, Jacobs ER, Moulder JE (2012) Radiation damage to the lung: mitigation by angiotensin-converting enzyme (ACE) inhibitors. Respirology 17:66–71

    Article  PubMed  PubMed Central  Google Scholar 

  • Melisi D, Ishiyama S, Sclabas GM, Fleming JB, Xia Q, Tortora G, Abbruzzese JL, Chiao PJ (2008) LY2109761, a novel transforming growth factor beta receptor type I and type II dual inhibitor, as a therapeutic approach to suppressing pancreatic cancer metastasis. Mol Cancer Ther 7:829–840

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Mi S, Li Z, Liu H, Hu ZW, Hua F (2012) Blocking IL-17A protects against lung injury-induced pulmonary fibrosis through promoting the activation of p50NF-kappaB. Yao Xue Xue Bao 47:739–744

    CAS  PubMed  Google Scholar 

  • Mittra I, Khare NK, Raghuram GV, Chaubal R, Khambatti F, Gupta D, Gaikwad A, Prasannan P, Singh A, Iyer A, Singh A, Upadhyay P, Nair NK, Mishra PK, Dutt A (2015) Circulating nucleic acids damage DNA of healthy cells by integrating into their genomes. J Biosci 40:91–111

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Mittra I, Samant U, Sharma S, Raghuram GV, Saha T, Tidke P, Pancholi N, Gupta D, Prasannan P, Gaikwad A, Gardi N, Chaubal R, Upadhyay P, Pal K, Rane B, Shaikh A, Salunkhe S, Dutt S, Mishra PK, Khare NK, Nair NK, Dutt A (2017) Cell-free chromatin from dying cancer cells integrate into genomes of bystander healthy cells to induce DNA damage and inflammation. Cell Death Discov 3:17015

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • 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

    Article  CAS  PubMed  Google Scholar 

  • Mothersill C, Seymour C (2012) Are Epigenetic Mechanisms Involved in Radiation-Induced Bystander Effects? Front Genet 3:74

    Article  PubMed  PubMed Central  Google Scholar 

  • 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 82:1740–1746

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Moulder JE (2003) Pharmacological intervention to prevent or ameliorate chronic radiation injuries. Semin Radiat Oncol 13:73–84

    Article  PubMed  Google Scholar 

  • Moulder JE (2004) Post-irradiation approaches to treatment of radiation injuries in the context of radiological terrorism and radiation accidents: a review. Int J Radiat Biol 80:3–10

    Article  CAS  PubMed  Google Scholar 

  • Multhoff G, Radons J (2012) Radiation, inflammation, and immune responses in cancer. Front Oncol 2

  • Najafi M, Fardid R, Hadadi G, Fardid M (2014) The mechanisms of radiation-induced bystander effect. J Biomed Phys Eng 4:163–172

    CAS  PubMed  PubMed Central  Google Scholar 

  • Najafi M, Fardid R, Takhshid MA (2016) Radiation-Induced Oxidative Stress at Out-of-Field. Cell J 18:340–345

    PubMed  PubMed Central  Google Scholar 

  • Najafi M, Salajegheh A, Rezaeyan A (2017a) Bystander Effect and Second Primary Cancers following Radiotherapy: What are its Significances? J Med Phys 42:55–56

    Article  PubMed  PubMed Central  Google Scholar 

  • Najafi M, Shirazi A, Motevaseli E, Rezaeyan AH, Salajegheh A, Rezapoor S (2017b) Melatonin as an anti-inflammatory agent in radiotherapy. Inflammopharmacology 25:403–413

    Article  CAS  PubMed  Google Scholar 

  • Najafi M, Motevaseli E, Shirazi A, Geraily G, Rezaeyan A, Norouzi F, Rezapoor S, Abdollahi H (2018a) Mechanisms of inflammatory responses to radiation and normal tissues toxicity: clinical implications. Int J Radiat Biol 94:335–356

  • Najafi M, Cheki M, Rezapoor S, Geraily G, Motevaseli E, Carnovale C, Clementi E, Shirazi A (2018b) Metformin: prevention of genomic instability and cancer: a review. Mutat Res Genet Toxicol Environ Mutagen 827:1–8. https://doi.org/10.1016/j.mrgentox.2018.01.007

  • Nishioka A, Ogawa Y, Mima T, Jin YJ, Sonobe H, Kariya S, Kubota K, Yoshida S, Ueno H (2004) Histopathologic amelioration of fibroproliferative change in rat irradiated lung using soluble transforming growth factor-beta (TGF-beta) receptor mediated by adenoviral vector. Int J Radiat Oncol Biol Phys 58:1235–1241

    Article  CAS  PubMed  Google Scholar 

  • Nishioka A, Ogawa Y, Kariya S, Hamada N, Nogami M, Inomata T, Ueno H (2015) Reduction of fibroproliferative changes in irradiated rat lung with soluble transforming growth factor-beta receptor. Mol Med Rep 11:2659–2663

    Article  CAS  PubMed  Google Scholar 

  • Oikonomou N, Harokopos V, Zalevsky J, Valavanis C, Kotanidou A, Szymkowski DE (2006) Soluble TNF mediates the transition from pulmonary inflammation to fibrosis. PLoS One 1

  • Park JS, Gamboni-Robertson F, He Q, Svetkauskaite D, Kim JY, Strassheim D, Sohn JW, Yamada S, Maruyama I, Banerjee A, Ishizaka A, Abraham E (2006) High mobility group box 1 protein interacts with multiple Toll-like receptors. Am J Physiol Cell Physiol 290:C917–C924

    Article  CAS  PubMed  Google Scholar 

  • 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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Peter RU, Braun-Falco O, Birioukov A, Hacker N, Kerscher M, Peterseim U, Ruzicka T, Konz B, Plewig G (1994) Chronic cutaneous damage after accidental exposure to ionizing radiation: the Chernobyl experience. J Am Acad Dermatol 30:719–723

    Article  CAS  PubMed  Google Scholar 

  • Piccinini A, Midwood K (2010) DAMPening inflammation by modulating TLR signalling. Mediat Inflamm 2010

  • Pietrofesa R, Turowski J, Tyagi S, Dukes F, Arguiri E, Busch TM, Gallagher-Colombo SM, Solomides CC, Cengel KA, Christofidou-Solomidou M (2013) Radiation mitigating properties of the lignan component in flaxseed. BMC Cancer 13:179

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Piwkowska A, Rogacka D, Jankowski M, Angielski S (2013) Metformin reduces NAD(P)H oxidase activity in mouse cultured podocytes through purinergic dependent mechanism by increasing extracellular ATP concentration. Acta Biochim Pol 60:607–612

    PubMed  Google Scholar 

  • Prise KM, O'Sullivan JM (2009) Radiation-induced bystander signalling in cancer therapy. Nat Rev Cancer 9:351–360

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Prise KM, Schettino G, Folkard M, Held KD (2005) New insights on cell death from radiation exposure. Lancet Oncol 6:520–528

    Article  CAS  PubMed  Google Scholar 

  • Rabender C, Mezzaroma E, Mauro AG, Mullangi R, Abbate A, Anscher M, Hart B, Mikkelsen R (2016) IPW-5371 Proves Effective as a Radiation Countermeasure by Mitigating Radiation-Induced Late Effects. Radiat Res 186:478–488

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Rajagopalan MS, Gupta K, Epperly MW, Franicola D, Zhang X, Wang H, Zhao H, Tyurin VA, Pierce JG, Kagan VE, Wipf P, Kanai AJ, Greenberger JS (2009) The mitochondria-targeted nitroxide JP4-039 augments potentially lethal irradiation damage repair. In: In Vivo, vol 23, pp 717–726

    Google Scholar 

  • Reeves A, Zagurovskaya M, Gupta S, Shareef MM, Mohiuddin M, Ahmed MM (2007) Inhibition of transforming growth factor-beta signaling in normal lung epithelial cells confers resistance to ionizing radiation. Int J Radiat Oncol Biol Phys 68:187–195

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Robbins ME, Zhao W (2004) Chronic oxidative stress and radiation-induced late normal tissue injury: a review. Int J Radiat Biol 80:251–259

    Article  CAS  PubMed  Google Scholar 

  • Rube CE, Uthe D, Schmid KW, Richter KD, Wessel J, Schuck A, Willich N, Rube C (2000) Dose-dependent induction of transforming growth factor beta (TGF-beta) in the lung tissue of fibrosis-prone mice after thoracic irradiation. Int J Radiat Oncol Biol Phys 47:1033–1042

    Article  CAS  PubMed  Google Scholar 

  • Rwigema JC, Beck B, Wang W, Doemling A, Epperly MW, Shields D, Goff JP, Franicola D, Dixon T, Frantz MC, Wipf P, Tyurina Y, Kagan VE, Wang H, Greenberger JS (2011) Two strategies for the development of mitochondrion-targeted small molecule radiation damage mitigators. Int J Radiat Oncol Biol Phys 80:860–868

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Sakai Y, Yamamori T, Yoshikawa Y, Bo T, Suzuki M, Yamamoto K, Ago T, Inanami O (2018) NADPH oxidase 4 mediates ROS production in radiation-induced senescent cells and promotes migration of inflammatory cells. Free Radic Res 52:92–102

    Article  CAS  PubMed  Google Scholar 

  • Schuster N, Krieglstein K (2002) Mechanisms of TGF-beta-mediated apoptosis. Cell Tissue Res 307:1–14

    Article  CAS  PubMed  Google Scholar 

  • Shakhov AN, Singh VK, Bone F, Cheney A, Kononov Y, Krasnov P, Bratanova-Toshkova TK, Shakhova VV, Young J, Weil MM, Panoskaltsis-Mortari A, Orschell CM, Baker PS, Gudkov A, Feinstein E (2012) Prevention and mitigation of acute radiation syndrome in mice by synthetic lipopeptide agonists of Toll-like receptor 2 (TLR2). PLoS One 7:e33044

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Shinde A, Berhane H, Rhieu BH, Kalash R, Xu K, Goff J, Epperly MW, Franicola D, Zhang X, Dixon T, Shields D, Wang H, Wipf P, Parmar K, Guinan E, Kagan V, Tyurin V, Ferris RL, Zhang X, Li S, Greenberger JS (2016) Intraoral Mitochondrial-Targeted GS-Nitroxide, JP4-039, Radioprotects Normal Tissue in Tumor-Bearing Radiosensitive Fancd2(-/-) (C57BL/6) Mice. Radiat Res 185:134–150

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Szondy Z, Sarang Z, Kiss B, Garabuczi E, Koroskenyi K (2017) Anti-inflammatory Mechanisms Triggered by Apoptotic Cells during Their Clearance. Front Immunol 8:909

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Tang D, Shi Y, Kang R, Li T, Xiao W, Wang H, Xiao X (2007) Hydrogen peroxide stimulates macrophages and monocytes to actively release HMGB1. J Leukoc Biol 81:741–747

    Article  CAS  PubMed  Google Scholar 

  • 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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Tian W, Yin X, Wang L, Wang J, Zhu W, Cao J, Yang H (2015) The key role of miR-21-regulated SOD2 in the medium-mediated bystander responses in human fibroblasts induced by alpha-irradiated keratinocytes. Mutat Res 780:77–85

    Article  CAS  PubMed  Google Scholar 

  • Tomita M, Matsumoto H, Funayama T, Yokota Y, Otsuka K, Maeda M, Kobayashi Y (2015) Nitric oxide-mediated bystander signal transduction induced by heavy-ion microbeam irradiation. Life Sci Space Res (Amst) 6:36–43

    Article  Google Scholar 

  • Toshkov IA, Gleiberman AS, Mett VL, Hutson AD, Singh AK, Gudkov AV, Burdelya LG (2017) Mitigation of Radiation-Induced Epithelial Damage by the TLR5 Agonist Entolimod in a Mouse Model of Fractionated Head and Neck Irradiation. Radiat Res 187:570–580

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Valentin J (2005) Protecting people against radiation exposure in the event of a radiological attack. A report of The International Commission on Radiological Protection. Ann ICRP 35(1-110):iii–iiv

    Article  Google Scholar 

  • Wang B-Z, Wang L-P, Han H, Cao F-L, Li G-Y, Xu J-L, Wang X-W, Wang L-X (2014a) Interleukin-17A antagonist attenuates radiation-induced lung injuries in mice. Exp Lung Res 40:77–85

    Article  CAS  PubMed  Google Scholar 

  • Wang, L. P., Wang, Y. W., Wang, B. Z., Sun, G. M., Wang, X. Y. & Xu, J. L. 2014b. Expression of interleukin-17A in lung tissues of irradiated mice and the influence of dexamethasone. ScientificWorldJournal, 2014, 251067.

  • Wang L, He L, Bao G, He X, Fan S, Wang H (2016) Ionizing Radiation Induces HMGB1 Cytoplasmic Translocation and Extracellular Release. Guo Ji Fang She Yi Xue He Yi Xue Za Zhi 40:91–99

    PubMed  PubMed Central  Google Scholar 

  • Weyemi U, Redon CE, Aziz T, Choudhuri R, Maeda D, Parekh PR, Bonner MY, Arbiser JL, Bonner WM (2015) Inactivation of NADPH oxidases NOX4 and NOX5 protects human primary fibroblasts from ionizing radiation-induced DNA damage. Radiat Res 183:262–270

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Willis, J., Epperly, M. W., Fisher, R., Zhang, X., Shields, D., Hou, W., Wang, H., Li, S., Wipf, P., Parmar, K., Guinan, E., Steinman, J. & Greenberger, J. S. 2018. Amelioration of Head and Neck Radiation-Induced Mucositis and Distant Marrow Suppression in Fanca(-/-) and Fancg(-/-) Mice by Intraoral Administration of GS-Nitroxide (JP4-039). Radiat Res.

  • Wu Y, Doroshow JH (2014a) Abstract 5358: IL-4/IL-13 induce Duox2/DuoxA2 expression and reactive oxygen production in human pancreatic and colon cancer cells. Cancer Res 74:5358

    Google Scholar 

  • Wu, Y. & Doroshow, J. H. 2014b. IL-4/IL-13 induce Duox2/DuoxA2 expression and reactive oxygen production in human pancreatic and colon cancer cells. AACR.

  • Wu Q, Allouch A, Martins I, Modjtahedi N, Deutsch E, Perfettini J-L (2017) Macrophage biology plays a central role during ionizing radiation-elicited tumor response. Biomed J 40:200–211

    Article  PubMed  PubMed Central  Google Scholar 

  • Xavier S, Piek E, Fujii M, Javelaud D, Mauviel A, Flanders KC, Samuni AM, Felici A, Reiss M, Yarkoni S, Sowers A, Mitchell JB, Roberts AB, Russo A (2004) Amelioration of radiation-induced fibrosis: inhibition of transforming growth factor-beta signaling by halofuginone. J Biol Chem 279:15167–15176

    Article  CAS  PubMed  Google Scholar 

  • Xu S, Ding N, Pei H, Hu W, Wei W, Zhang X, Zhou G, Wang J (2014) MiR-21 is involved in radiation-induced bystander effects. RNA Biol 11:1161–1170

    Article  PubMed  PubMed Central  Google Scholar 

  • Xu W, Wang T, Xu S, Xu S, Wu L, Wu Y, Bian P (2015) Radiation-induced epigenetic bystander effects demonstrated in Arabidopsis thaliana. Radiat Res 183:511–524

    Article  CAS  PubMed  Google Scholar 

  • Yahyapour R, Amini P, Rezapoor S, Rezaeyan A, Farhood B, Cheki M, Fallah H, Najafi M (2017a) Targeting of inflammation for radiation protection and mitigation. Curr Mol Pharmacol. https://doi.org/10.2174/1874467210666171108165641

  • Yahyapour R, Motevaseli E, Rezaeyan A, Abdollahi H, Farhood B, Cheki M, Najafi M, Villa V (2017b) Mechanisms of radiation bystander and non-targeted effects: implications to radiation carcinogenesis and radiotherapy. Curr Radiopharm 11(1):34–45

  • Yahyapour R, Amini P, Rezapour S, Cheki M, Rezaeyan A, Farhood B, Shabeeb D, Musa AE, Fallah H, Najafi M (2018a) Radiation-induced inflammation and autoimmune diseases. Mil Med Res 5:9

    Article  PubMed  PubMed Central  Google Scholar 

  • Yahyapour R, Motevaseli E, Rezaeyan A, Abdollahi H, Farhood B, Cheki M, Rezapoor S, Shabeeb D, Musa AE, Najafi M, Villa V (2018b) Reduction-oxidation (redox) system in radiation-induced normal tissue injury: molecular mechanisms and implications in radiation therapeutics. Clin Transl Oncol. https://doi.org/10.1007/s12094-017-1828-6

  • Yahyapour R, Salajegheh A, Safari A, Abbasi S, Amini P, Rezaeyan A, Amraee A, Najafi M (2018c) Radiation-induced non-targeted effect and carcinogenesis; Implications in Clinical Radiotherapy. J Biomed Phys Eng. https://doi.org/10.22086/jbpe.v0i0.713

  • Yakovlev VA, Rabender CS, Sankala H, Gauter-Fleckenstein B, Fleckenstein K, Batinic-Haberle I, Jackson I, Vujaskovic Z, Anscher MS, Mikkelsen RB, Graves PR (2010) Proteomic analysis of radiation-induced changes in rat lung: Modulation by the superoxide dismutase mimetic MnTE-2-PyP(5+). Int J Radiat Oncol Biol Phys 78:547–554

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Yang H, Asaad N, Held KD (2005) Medium-mediated intercellular communication is involved in bystander responses of X-ray-irradiated normal human fibroblasts. Oncogene 24:2096–2103

    Article  CAS  PubMed  Google Scholar 

  • Yang H, Hreggvidsdottir HS, Palmblad K, Wang H, Ochani M, Li J, Lu B, Chavan S, Rosas-Ballina M, Al-Abed Y, Akira S, Bierhaus A, Erlandsson-Harris H, Andersson U, Tracey KJ (2010) A critical cysteine is required for HMGB1 binding to Toll-like receptor 4 and activation of macrophage cytokine release. Proc Natl Acad Sci U S A 107:11942–11947

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Yang, H., Wang, H., Chavan, S. S. & Andersson, U. 2015. High Mobility Group Box Protein 1 (HMGB1): The Prototypical Endogenous Danger Molecule. Molecular Medicine, 21, S6-S12.

  • Yoshida T, Goto S, Kawakatsu M, Urata Y, Li T-s (2012) Mitochondrial dysfunction, a probable cause of persistent oxidative stress after exposure to ionizing radiation. Free Radic Res 46:147–153

    Article  CAS  PubMed  Google Scholar 

  • Zhang M, Kleber S, Rohrich M, Timke C, Han N, Tuettenberg J, Martin-Villalba A, Debus J, Peschke P, Wirkner U, Lahn M, Huber PE (2011) Blockade of TGF-beta signaling by the TGFbetaR-I kinase inhibitor LY2109761 enhances radiation response and prolongs survival in glioblastoma. Cancer Res 71:7155–7167

    Article  CAS  PubMed  Google Scholar 

  • Zhang H, Wang Y-a, Meng A, Yan H, Wang X, Niu J, Li J, Wang H (2013) Inhibiting TGFβ1 has a protective effect on mouse bone marrow suppression following ionizing radiation exposure in vitro. J Radiat Res 54:630–636

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Zhao Y, de Toledo SM, Hu G, Hei TK, Azzam EI (2014) Connexins and cyclooxygenase-2 crosstalk in the expression of radiation-induced bystander effects. Br J Cancer 111:125–131

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • 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 U S A 102:14641–14646

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Zhou H, Ivanov VN, Lien Y-C, Davidson M, Hei TK (2008a) Mitochondrial Function and NF-κB Mediated Signaling in Radiation-Induced Bystander Effects. Cancer Res 68:2233–2240

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Zhou H, Ivanov VN, Lien Y-C, Davidson M, Hei TK (2008b) Mitochondrial function and nuclear factor-κB–mediated signaling in radiation-induced bystander effects. Cancer Res 68:2233–2240

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Zhou R, Yazdi AS, Menu P, Tschopp J (2011) A role for mitochondria in NLRP3 inflammasome activation. Nature 469:221–225

    Article  CAS  PubMed  Google Scholar 

  • Zorov DB, Juhaszova M, Sollott SJ (2006) Mitochondrial ROS-induced ROS release: an update and review. Biochim Biophys Acta 1757:509–517

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Masoud Najafi.

Ethics declarations

Conflict of interest

No

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Farhood, B., Goradel, N.H., Mortezaee, K. et al. Intercellular communications-redox interactions in radiation toxicity; potential targets for radiation mitigation. J. Cell Commun. Signal. 13, 3–16 (2019). https://doi.org/10.1007/s12079-018-0473-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12079-018-0473-3

Keywords

Navigation