Generic placeholder image

Current Clinical Pharmacology

Editor-in-Chief

ISSN (Print): 1574-8847
ISSN (Online): 2212-3938

Research Article

Protective Effect of Selenium-L-methionine on Radiation-induced Acute Pneumonitis and Lung Fibrosis in Rat

Author(s): Peyman Amini, Sedighe Kolivand, Hana Saffar, Saeed Rezapoor, Elahe Motevaseli*, Masoud Najafi*, Farzad Nouruzi, Dheyauldeen Shabeeb and Ahmed Eleojo Musa

Volume 14, Issue 2, 2019

Page: [157 - 164] Pages: 8

DOI: 10.2174/1574884714666181214101917

Abstract

Background: In this study, we aimed to detect the changes in the level of interleukin (IL)-4 and IL-13 cytokines and their downstream genes including interleukin-13 receptor subunit alpha-2 (IL13Ra2), interleukin-4 receptor subunit alpha-1 (IL4Ra1), dual oxidase 1 (DUOX1) and dual oxidase 2 (DUOX2). The protective effects of Selenium-L-methionine on radiation-induced histopathological damages and changes in the level of these cytokines and genes were detected.

Methods: Four groups of 20 rats (5 rats in each) namely, control; Selenium-L-methionine, radiation and radiation plus Selenium-L-methionine were used in this study. 4 mg/kg of Selenium-Lmethionine was administered 1 day before irradiation and five consecutive days after irradiation. Irradiation was done using a dose of 15 Gy 60Co gamma rays at 109 cGy/min. All rats were sacrificed 10 weeks after irradiation for detecting changes in IL-4 and IL-13 cytokines, the expressions of IL13Ra2, IL4Ra1, Duox1 and Duox2 and histopathological changes.

Results: The level of IL-4 but not IL-13 increased after irradiation. This was associated with increased expression of IL4Ra1, Duox1 and Duox2, in addition to changes in morphological properties. Selenium-L-methionine could attenuate all injury markers following lung irradiation.

Conclusion: Selenium-L-methionine can protect lung tissues against toxic effects of ionizing radiation. It is possible that the modulation of immune responses and redox interactions are involved in the radioprotective effect of this agent.

Keywords: Selenium-L-methionine, lung, radiation, IL-4. IL-13, radiotherapy, pneumonitis, interleukin.

Graphical Abstract
[1]
Rezaeyan A, Fardid R, Haddadi GH, et al. Evaluating radioprotective effect of hes-peridin on acute radiation damage in the lung tissue of rats. J Biomed Phys Eng 2016; 6(3): 165-74.
[PMID: 27853724]
[2]
Mahmood J, Jelveh S, Calveley V, Zaidi A, Doctrow SR, Hill RP. Mitigation of lung injury after accidental exposure to radiation. Radiat Res 2011; 176(6): 770-80.
[http://dx.doi.org/10.1667/RR2562.1] [PMID: 22013884]
[3]
Farhood B, Mortezaee K, Goradel NH, et al. Curcumin as an anti-inflammatory agent: Implications to radiotherapy and chemotherapy. J Cell Physiol 2019; 234(5): 5728-40.
[http://dx.doi.org/10.1002/jcp.27442] [PMID: 30317564]
[4]
Keywan Mortezaee NHG. Peyman Amini, Dheyauldeen Shabeeb, Ahmed Eleojo Musa, Masoud Najafi, NADPH oxi-dase as a target for modulation of radiation response; implica-tions to carcinogenesis and radiotherapy. Curr Mol Pharmacol 2018; 12(1)
[http://dx.doi.org/10.2174/1874467211666181010154709]
[5]
Yahyapour R, Salajegheh A, Safari A, et al. Radiation-induced Non-targeted Effect and Carcinogenesis; Implications in Clinical Radiotherapy. J Biomed Phys Eng 2018; 8(4): 435-46.
[http://dx.doi.org/10.31661/jbpe.v0i0.713] [PMID: 30568933]
[6]
Di Maggio FM, Minafra L, Forte GI, et al. Portrait of inflammatory response to ionizing radiation treatment. J Inflamm (Lond) 2015; 12: 14.
[http://dx.doi.org/10.1186/s12950-015-0058-3] [PMID: 25705130]
[7]
Yahyapour R, Motevaseli E, Rezaeyan A, et al. Reduction-oxidation (redox) system in radiation-induced normal tissue injury: molecular mechanisms and implications in radiation therapeutics. Clin Transl Oncol 2018; 20(8): 975-88.
[http://dx.doi.org/10.1007/s12094-017-1828-6] [PMID: 29318449]
[8]
Farhood B, Goradel NH, Mortezaee K, et al. Intercellular communications-redox interactions in radiation toxicity; potential targets for radiation mitigation. J Cell Commun Signal 2019; 13(1): 3-16.
[http://dx.doi.org/10.1007/s12079-018-0473-3] [PMID: 29911259]
[9]
Frey B, Hehlgans S, Rödel F, Gaipl US. Modulation of inflammation by low and high doses of ionizing radiation: Implications for benign and malign diseases. Cancer Lett 2015; 368(2): 230-7.
[http://dx.doi.org/10.1016/j.canlet.2015.04.010] [PMID: 25888451]
[10]
Abratt RP, Morgan GW. Lung toxicity following chest irradiation in patients with lung cancer. Lung Cancer 2002; 35(2): 103-9.
[http://dx.doi.org/10.1016/S0169-5002(01)00334-8] [PMID: 11804681]
[11]
Jakubzick C, Kunkel SL, Puri RK, Hogaboam CM. Therapeutic targeting of IL-4- and IL-13-responsive cells in pulmonary fibrosis. Immunol Res 2004; 30(3): 339-49.
[http://dx.doi.org/10.1385/IR:30:3:339] [PMID: 15531774]
[12]
Mahmood J, Jelveh S, Zaidi A, Doctrow SR, Medhora M, Hill RP. Targeting the Renin-angiotensin system combined with an antioxidant is highly effective in mitigating radiation-induced lung damage. Int J Radiat Oncol Biol Phys 2014; 89(4): 722-8.
[http://dx.doi.org/10.1016/j.ijrobp.2014.03.048] [PMID: 24867538]
[13]
Mahmood J, Jelveh S, Zaidi A, Doctrow SR, Hill RP. Mitigation of radiation-induced lung injury with EUK-207 and genistein: effects in adolescent rats. Radiat Res 2013; 179(2): 125-34.
[http://dx.doi.org/10.1667/RR2954.1] [PMID: 23237541]
[14]
Lambeth JD. NOX enzymes and the biology of reactive oxygen. Nat Rev Immunol 2004; 4(3): 181-9.
[http://dx.doi.org/10.1038/nri1312] [PMID: 15039755]
[15]
Yoshihara A, Hara T, Kawashima A, et al. Regulation of dual oxidase expression and H2O2 production by thyroglobulin. Thyroid 2012; 22(10): 1054-62.
[http://dx.doi.org/10.1089/thy.2012.0003] [PMID: 22874065]
[16]
Groves AM, Johnston CJ, Misra RS, Williams JP, Finkelstein JN. Effects of IL-4 on pulmonary fibrosis and the accumulation and phenotype of macrophage subpopulations following thoracic irradiation. Int J Radiat Biol 2016; 92(12): 754-65.
[http://dx.doi.org/10.1080/09553002.2016.1222094] [PMID: 27539247]
[17]
Ameziane-El-Hassani R, Talbot M, de Souza Dos Santos MC, et al. NADPH oxidase DUOX1 promotes long-term persistence of oxidative stress after an exposure to irradiation. Proc Natl Acad Sci USA 2015; 112(16): 5051-6.
[http://dx.doi.org/10.1073/pnas.1420707112] [PMID: 25848056]
[18]
Eskalli Z, Achouri Y, Hahn S, et al. Overexpression of interleukin-4 in the thyroid of transgenic mice upregulates the expression of Duox1 and the anion transporter pendrin. Thyroid 2016; 26(10): 1499-512.
[http://dx.doi.org/10.1089/thy.2016.0106] [PMID: 27599561]
[19]
Azmoonfar R, Amini P, Saffar H, et al. Metformin protects against radiation-induced pneumonitis and fibrosis and attenuates upregulation of dual oxidase genes expression. Adv Pharm Bull 2018; 8(4): 697-704.
[20]
Yahyapour R, Amini P, Saffar H, et al. Metformin Protects Against Radiation-Induced Heart Injury and Attenuates the Up-regulation of Du-al Oxidase Genes Following Rat’s Chest Irradiation. nt J Mol Cell Med 2018; 7(3): 0.
[21]
Stỳblo M, Walton FS, Harmon AW, Sheridan PA, Beck MA. Activation of superoxide dismutase in selenium-deficient mice infected with influenza virus. J Trace Elem Med Biol 2007; 21(1): 52-62.
[http://dx.doi.org/10.1016/j.jtemb.2006.11.001] [PMID: 17317526]
[22]
Bourdon E, Loreau N, Lagrost L, Blache D. Differential effects of cysteine and methionine residues in the antioxidant activity of human serum albumin. Free Radic Res 2005; 39(1): 15-20.
[http://dx.doi.org/10.1080/10715760400024935] [PMID: 15875807]
[23]
Sieber F, Muir SA, Cohen EP, et al. Dietary selenium for the mitigation of radiation injury: effects of selenium dose escalation and timing of supplementation. Radiat Res 2011; 176(3): 366-74.
[http://dx.doi.org/10.1667/RR2456.1] [PMID: 21867430]
[24]
Sieber F, Muir SA, Cohen EP, et al. High-dose selenium for the mitigation of radiation injury: a pilot study in a rat model. Radiat Res 2009; 171(3): 368-73.
[http://dx.doi.org/10.1667/0033-7587-171.3.368] [PMID: 19267564]
[25]
Weiss JF, Srinivasan V, Kumar KS, Landauer MR. Radioprotection by metals: selenium. Adv Space Res 1992; 12(2-3): 223-31.
[http://dx.doi.org/10.1016/0273-1177(92)90112-B] [PMID: 11537012]
[26]
Szabo S, Ghosh SN, Fish BL, et al. Cellular inflammatory infiltrate in pneumonitis induced by a single moderate dose of thoracic x radiation in rats. Radiat Res 2010; 173(4): 545-56.
[http://dx.doi.org/10.1667/RR1753.1] [PMID: 20334527]
[27]
Medhora M, Haworth S, Liu Y, et al. Biomarkers for Radiation Pneumonitis Using Noninvasive Molecular Imaging. J Nucl Med 2016; 57(8): 1296-301.
[http://dx.doi.org/10.2967/jnumed.115.160291] [PMID: 27033892]
[28]
Mahmood J, Jelveh S, Calveley V, Zaidi A, Doctrow SR, Hill RP. Mitigation of lung injury after accidental exposure to radiation. Radiat Res 2011; 176(6): 770-80.
[http://dx.doi.org/10.1667/RR2562.1] [PMID: 22013884]
[29]
Farhood B, Goradel NH, Mortezaee K, Khanlarkhani N, Najafi M, Sahebkar A. Melatonin and cancer: From the promotion of genomic stability to use in cancer treatment. J Cell Physiol 2019; 234(5): 5613-27.
[http://dx.doi.org/10.1002/jcp.27391] [PMID: 30238978]
[30]
Yahyapour R, Shabeeb D, Cheki M, et al. Radiation protection and mitigation by natural antioxidants and flavo-noids; implications to radiotherapy and radiation disasters. Curr Mol Pharmacol 2018; 11(4): 285-304.
[http://dx.doi.org/10.2174/1874467211666180619125653] [PMID: 29921213]
[31]
Sieber F, Muir SA, Cohen EP, et al. High-dose selenium for the mitigation of radiation injury: a pilot study in a rat model. Radiat Res 2009; 171(3): 368-73.
[http://dx.doi.org/10.1667/0033-7587-171.3.368] [PMID: 19267564]
[32]
Brown SL, Kolozsvary A, Liu J, Jenrow KA, Ryu S, Kim JH. Antioxidant diet supplementation starting 24 hours after exposure reduces radiation lethality. Radiat Res 2010; 173(4): 462-8.
[http://dx.doi.org/10.1667/RR1716.1] [PMID: 20334518]
[33]
Bagheri H, Rezapour S, Najafi M, et al. Protection against radia-tion-induced micronuclei in rat bone marrow erythrocytes by Curcumin and selenium L-methionine. Iran J Med Sci 2018.
[34]
Choi SH, Kim M, Lee HJ, Kim EH, Kim CH, Lee YJ. Effects of NOX1 on fibroblastic changes of endothelial cells in radiationinduced pulmonary fibrosis. Mol Med Rep 2016; 13(5): 4135-42.
[http://dx.doi.org/10.3892/mmr.2016.5090] [PMID: 27053172]
[35]
Pazhanisamy SK, Li H, Wang Y, Batinic-Haberle I, Zhou D. NADPH oxidase inhibition attenuates total body irradiation-induced haematopoietic genomic instability. Mutagenesis 2011; 26(3): 431-5.
[http://dx.doi.org/10.1093/mutage/ger001] [PMID: 21415439]
[36]
Wu Y, Doroshow JH. AACR Cancer Progress Report 2014.
[37]
Yahyapour R, Amini P, Rezapour S, et al. Radiation-induced inflammation and autoimmune diseases. Mil Med Res 2018; 5(1): 9.
[http://dx.doi.org/10.1186/s40779-018-0156-7] [PMID: 29554942]
[38]
Yahyapour R, Amini P, Rezapoor S, et al. Targeting of Inflammation for Radiation Protection and Mitigation. Curr Mol Pharmacol 2018; 11(3): 203-10.
[http://dx.doi.org/10.2174/1874467210666171108165641] [PMID: 29119941]
[39]
Raad H, Eskalli Z, Corvilain B, Miot F, De Deken X. Thyroid hydrogen peroxide production is enhanced by the Th2 cytokines, IL-4 and IL-13, through increased expression of the dual oxidase 2 and its maturation factor DUOXA2. Free Radic Biol Med 2013; 56: 216-25.
[http://dx.doi.org/10.1016/j.freeradbiomed.2012.09.003] [PMID: 23010498]
[40]
Hodny Z, Reinis M, Hubackova S, Vasicova P, Bartek J. Interferon gamma/NADPH oxidase defense system in immunity and cancer. OncoImmunology 2015; 5(2)e1080416
[http://dx.doi.org/10.1080/2162402X.2015.1080416] [PMID: 27057461]
[41]
Wu Y, Antony S, Juhasz A, et al. Upregulation and sustained activation of STAT1 are essential for interferon-gamma (IFN-gamma)- induced dual oxidase-2 (DUOX2) and dual oxidase A2 (DUOXA2) expression in human pancreatic cancer cell lines. J Biol Chem 2011; jbc. M110. 191031.
[42]
Schnabel R, Lubos E, Messow CM, et al. Selenium supplementation improves antioxidant capacity in vitro and in vivo in patients with coronary artery disease: The SElenium Therapy in Coronary Artery disease Patients (SETCAP) Study. Am Heart J 2008; 156(6): 1201-11.

© 2024 Bentham Science Publishers | Privacy Policy