Melatonin Attenuates Upregulation of Duox1 and Duox2 and Protects against Lung Injury following Chest Irradiation in Rats

Akbar Aliasgharzadeh, Bagher Farhood, Peyman Amini, Hana Saffar, Elahe Motevaseli, Saeed Rezapoor, Farzad Nouruzi, D Heyauldeen Shabeeb, Ahmed Eleojo Musa, Mehran Mohseni, Habiballah Moradi, Masoud Najafi, Akbar Aliasgharzadeh, Bagher Farhood, Peyman Amini, Hana Saffar, Elahe Motevaseli, Saeed Rezapoor, Farzad Nouruzi, D Heyauldeen Shabeeb, Ahmed Eleojo Musa, Mehran Mohseni, Habiballah Moradi, Masoud Najafi

Abstract

Objective: The Lung is one of the most radiosensitive organs of the body. The infiltration of macrophages and lymphocytes into the lung is mediated via the stimulation of T-helper 2 cytokines such as IL-4 and IL-13, which play a key role in the development of fibrosis. It is likely that these cytokines induce chronic oxidative damage and inflammation through the upregulation of Duox1 and Duox2, which can increase the risk of late effects of ionizing radiation (IR) such as fibrosis and carcinogenesis. In the present study, we aimed to evaluate the possible increase of IL-4 and IL-13 levels, as well as their downstream genes such as IL4ra1, IL13ra2, Duox1, and Duox2.

Materials and methods: In this experimental animal study, male rats were divided into 4 groups: i. Control, ii. Melatonintreated, iii. Radiation, and iv. Melatonin (100 mg/kg) plus radiation. Rats were irradiated with 15 Gy 60Co gamma rays and then sacrificed after 67 days. The expressions of IL4ra1, IL13ra2, Duox1, and Duox2, as well as the levels of IL-4 and IL-13, were evaluated. The histopathological changes such as the infiltration of inflammatory cells, edema, and fibrosis were also examined. Moreover, the protective effect of melatonin on these parameters was also determined.

Results: Results showed a 1.5-fold increase in the level of IL-4, a 5-fold increase in the expression of IL4ra1, and a 3-fold increase in the expressions of Duox1 and Duox2. However, results showed no change for IL-13 and no detectable expression of IL13ra2. This was associated with increased infiltration of macrophages, lymphocytes, and mast cells. Melatonin treatment before irradiation completely reversed these changes.

Conclusion: This study has shown the upregulation of IL-4-IL4ra1-Duox2 signaling pathway following lung irradiation. It is possible that melatonin protects against IR-induced lung injury via the downregulation of this pathway and attenuation of inflammatory cells infiltration.

Keywords: Duox1; Duox2; Lung; Melatonin; Radiation.

Conflict of interest statement

There is no conflict of interest in this study.

Copyright© by Royan Institute. All rights reserved.

Figures

Fig.1
Fig.1
Results of changes in the levels of IL-4 and IL-13 following irradiation with gamma rays and treatment with melatonin (MLT). A. IL-4 and B. IL- 13. a; Significant compared to control and b; Significant compared to radiation (Rad), ANOVA Tukey’s HSD post hoc, P

Fig.2

The expression of IL4ra1, Duox1, and…

Fig.2

The expression of IL4ra1, Duox1, and Duox2 following irradiation or melatonin treatment before irradiation…

Fig.2
The expression of IL4ra1, Duox1, and Duox2 following irradiation or melatonin treatment before irradiation in lung tissues of rats. A.IL4ra1, B.Duox1, and C.Duox2. a; Significant compared to control and b; Significant compared to radiation (Rad), ANOVA followed by Tukey’s HSD post hoc, P<0.05.

Fig.3

Histopathological investigation of the protective effect…

Fig.3

Histopathological investigation of the protective effect of melatonin on radiation-induced lung injury. Control and…

Fig.3
Histopathological investigation of the protective effect of melatonin on radiation-induced lung injury. Control and melatonin groups: no infiltration of macrophages and lymphocytes, as well as normal vascular and alveolar thickening, radiation: severe infiltration of macrophages and lymphocytes, as well as vascular thickening, while alveolar thickening mildly changed. A. Control; B. Melatonin, C. Radiation, D. Radiation+Melatonin (H&E staining ×100).

Fig.4

Results of trichrome staining showed a…

Fig.4

Results of trichrome staining showed a mild collagen deposition, while treatment with melatonin completely…

Fig.4
Results of trichrome staining showed a mild collagen deposition, while treatment with melatonin completely reversed collagen deposition. A. Control, B. Melatonin, C. Radiation, and D. Radiation+Melatonin (Masson’s Trichrome staining ×100).

Fig.5

Infiltration of mast cells following irradiation…

Fig.5

Infiltration of mast cells following irradiation of lung tissues in rats. The administration of…

Fig.5
Infiltration of mast cells following irradiation of lung tissues in rats. The administration of melatonin before irradiation could not significantly attenuate mast cell infiltration. A. Control, B. Melatonin, C. Radiation, and D. Radiation+Melatonin (Giemsa staining ×100).
Fig.2
Fig.2
The expression of IL4ra1, Duox1, and Duox2 following irradiation or melatonin treatment before irradiation in lung tissues of rats. A.IL4ra1, B.Duox1, and C.Duox2. a; Significant compared to control and b; Significant compared to radiation (Rad), ANOVA followed by Tukey’s HSD post hoc, P<0.05.
Fig.3
Fig.3
Histopathological investigation of the protective effect of melatonin on radiation-induced lung injury. Control and melatonin groups: no infiltration of macrophages and lymphocytes, as well as normal vascular and alveolar thickening, radiation: severe infiltration of macrophages and lymphocytes, as well as vascular thickening, while alveolar thickening mildly changed. A. Control; B. Melatonin, C. Radiation, D. Radiation+Melatonin (H&E staining ×100).
Fig.4
Fig.4
Results of trichrome staining showed a mild collagen deposition, while treatment with melatonin completely reversed collagen deposition. A. Control, B. Melatonin, C. Radiation, and D. Radiation+Melatonin (Masson’s Trichrome staining ×100).
Fig.5
Fig.5
Infiltration of mast cells following irradiation of lung tissues in rats. The administration of melatonin before irradiation could not significantly attenuate mast cell infiltration. A. Control, B. Melatonin, C. Radiation, and D. Radiation+Melatonin (Giemsa staining ×100).

References

    1. Yahyapour R, Shabeeb D, Cheki M, Musa AE, Farhood B, Rezaeyan A, et al. Radiation protection and mitigation by natural antioxidants and flavonoids; implications to radiotherapy and radiation disasters. Curr Mol Pharmacol. 2018 ahead of print.
    1. Mortezaee K, NH Goradel, P Amini, D Shabeeb, A E Musa, M Najafi. NADPH oxidase as a target for modulation of radiation response; implications to carcinogenesis and radiotherapy. Curr Mol Pharmacol. 2018 ahead of print.
    1. Abratt RP, Morgan GW. Lung toxicity following chest irradiation in patients with lung cancer. Lung Cancer. 2002;35(2):103–109.
    1. Ghafoori P, Marks LB, Vujaskovic Z, Kelsey CR. Radiation-induced lung injury.Assessment, management, and prevention. Oncology. 2008;22(1):37-47; discussion 52-53..
    1. Tsoutsou PG, Koukourakis MI. Radiation pneumonitis and fibrosis: mechanisms underlying its pathogenesis and implications for future research. Int J Radiat Oncol Biol Phys. 2006;66(5):1281–1293.
    1. Haddadi GH, Rezaeyan A, Mosleh-Shirazi MA, Hosseinzadeh M, Fardid R, Najafi M, et al. Hesperidin as radioprotector against radiation- induced lung damage in rat: a histopathological study. J Med Phys. 2017;42(1):25–32.
    1. Zhao W, Robbins ME. Inflammation and chronic oxidative stress in radiation-induced late normal tissue injury: therapeutic implications. Curr Med Chem. 2009;16(2):130–143.
    1. Bahig H, Filion E, Vu T, Chalaoui J, Lambert L, Roberge D, et al. Severe radiation pneumonitis after lung stereotactic ablative radiation therapy in patients with interstitial lung disease. Pract Radiat Oncol. 2016;6(5):367–374.
    1. Chen Y, Williams J, Ding I, Hernady E, Liu W, Smudzin T, et al. Radiation pneumonitis and early circulatory cytokine markers. Semin Radiat Oncol. 2002;12(1 Suppl 1):26–33.
    1. Di Maggio FM, Minafra L, Forte GI, Cammarata FP, Lio D, Messa C, et al. Portrait of inflammatory response to ionizing radiation treatment. J Inflamm (Lond) 2015;12:14–14.
    1. Rube CE, Uthe D, Schmid KW, Richter KD, Wessel J, Schuck A, et al. 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. 2000;47(4):1033–1042.
    1. 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):339–349.
    1. Wu Y, Doroshow JH. IL-4/IL-13 induce Duox2/DuoxA2 expression and reactive oxygen production in human pancreatic and colon cancer cells. Cancer Res. 2014;74(19 Suppl):5–9.
    1. Chung SI, Horton JA, Ramalingam TR, White AO, Chung EJ, Hudak KE, et al. IL-13 is a therapeutic target in radiation lung injury. Sci Rep. 2016;6:39714–39714.
    1. Cheki M, Yahyapour R, Farhood B, Rezaeyan A, Shabeeb D, Amini P, et al. COX-2 in radiotherapy: a potential target for radioprotection and radiosensitization. Curr Mol Pharmacol. 2018;11(3):173–183.
    1. Tripathi DN, Jena GB. Effect of melatonin on the expression of Nrf2 and NF-kappaB during cyclophosphamide-induced urinary bladder injury in rat. J Pineal Res. 2010;48(4):324–331.
    1. Miller SC, Pandi-Perumal SR, Esquifino AI, Cardinali DP, Maestroni GJ. The role of melatonin in immuno-enhancement: potential application in cancer. Int J Exp Pathol. 2006;87(2):81–87.
    1. Najafi M, Salehi E, Farhood B, Nashtaei MS, Hashemi Goradel N, Khanlarkhani N, et al. Adjuvant chemotherapy with melatonin for targeting human cancers: a review. J Cell Physiol. 2019 ahead of print.
    1. Farhood B, Goradel NH, Mortezaee K, Khanlarkhani N, Salehi E, Nashtaei MS, et al. Melatonin as an adjuvant in radiotherapy for radioprotection and radiosensitization. Clin Transl Oncol. 2018 ahead of print.
    1. 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. 2018 ahead of print.
    1. Ghosh SN, Zhang R, Fish BL, Semenenko VA, Li XA, Moulder JE, et al. Renin-Angiotensin system suppression mitigates experimental radiation pneumonitis. Int J Radiat Oncol Biol Phys. 2009;75(5):1528–1536.
    1. Pfaffl MW. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res. 2001;29(9):e45–e45.
    1. Eskalli Z, Achouri Y, Hahn S, Many MC, Craps J, Refetoff 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):1499–1512.
    1. Ameziane-El-Hassani R, Talbot M, de Souza Dos Santos MC, Al Ghuzlan A, Hartl D, Bidart JM, 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):5051–5056.
    1. Harper RW, Xu C, Eiserich JP, Chen Y, Kao CY, Thai P, et al. Differential regulation of dual NADPH oxidases/peroxidases, Duox1 and Duox2, by Th1 and Th2 cytokines in respiratory tract epithelium. FEBS Lett. 2005;579(21):4911–4917.
    1. 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):754–765.
    1. Hebestreit H, Biko J, Drozd V, Demidchik Y, Burkhardt A, Trusen A, et al. Pulmonary fibrosis in youth treated with radioiodine for juvenile thyroid cancer and lung metastases after chernobyl. Eur J Nucl Med Mol Imaging. 2011;38(9):1683–1690.
    1. Yahyapour R, Motevaseli E, Rezaeyan A, Abdollahi H, Farhood B, Cheki M, et al. Reduction-oxidation (redox) system in radiationinduced normal tissue injury: molecular mechanisms and implications in radiation therapeutics. Clin Transl Oncol. 2018;20(8):975–988.
    1. Yahyapour R, Motevaseli E, Rezaeyan A, Abdollahi H, Farhood B, Cheki M, et al. Mechanisms of radiation bystander and nontargeted effects: implications to radiation carcinogenesis and radiotherapy. Curr Radiopharm. 2018;11(1):34–45.
    1. Andersen LP, Werner MU, Rosenkilde MM, Harpsøe NG, Fuglsang H, Rosenberg J, et al. Pharmacokinetics of oral and intravenous melatonin in healthy volunteers. BMC Pharmacol Toxicol. 2016;17:8–8.
    1. Gooneratne NS, Edwards AY, Zhou C, Cuellar N, Grandner MA, Barrett JS. Melatonin pharmacokinetics following two different oral surge-sustained release doses in older adults. J Pineal Res. 2012;52(4):437–445.
    1. Jou MJ, Peng TI, Yu PZ, Jou SB, Reiter RJ, Chen JY, et al. Melatonin protects against common deletion of mitochondrial DNA‐augmented mitochondrial oxidative stress and apoptosis. J Pineal Res. 2007;43(4):389–403.
    1. Talib WH. Melatonin and cancer hallmarks. Molecules. 2018;23(3) pii: E518.
    1. Yahyapour R, Amini P, Rezapoor S, Rezaeyan A, Farhood B, Cheki M, et al. Targeting of inflammation for radiation protection and mitigation. Curr Mol Pharmacol. 2018;11(3):203–210.

Source: PubMed

3
Abonnieren