REVIEW PAPER
Effect of ionizing radiation on the male reproductive system
 
More details
Hide details
1
Diagnostic Techniques Unit, Faculty of Health Sciences, Medical University of Lublin, Poland
 
2
Department of Clinical Dietetics, Faculty of Health Sciences, Medical University of Lublin, Poland
 
3
Prof. W. Orłowski Independent Public Clinical Hospital / Medical Center for Postgraduate Education, Warsaw, Poland
 
4
Institute of Rural Health, Poland
 
 
Corresponding author
Artur Wdowiak   

Diagnostic Techniques Unit, Faculty of Health Sciences, Medical University of Lublin, Poland
 
 
Ann Agric Environ Med. 2019;26(2):210-216
 
KEYWORDS
TOPICS
ABSTRACT
Introduction and objective:
In the light of current data concerning the growing exposure to ionizing radiation (IR) originating from atrificial sources, especially from medical ones, and also related to occupational exposure, it is justifiable to systematize the state of knowledge concerning the effect of IR on the male reproductive system.

Brief description of the state of knowledge:
There is no basis for the application of the hypothesis of hormesis in the area of male reproductive health. Regarding the impact of IR on spermatogenesis, spermatogonia are less susceptible to the occurrence of DNA damage after exposition to IR, but are characterized by slower DNA repair compared to somatic cells. Damage to the genes after exposure to IR is possible at each stage of spermatogenesis; however, haploidal spermatids show the highest radiosensitivity in this respect. The genetic risk of the cells differentiating during spermatogenesis is limited to one cycle of spermatogenesis, whereas the genetic instability may persist for the whole period of life, and DNA damage induced by IR may be transmitted to future generations. The minimum dose causing detectable DNA damage was 30 Gy. While exceeding this dose, the number of single-strand DNA breaks increases. Among males exposed to IR, a decrease was observed in sperm motility and in the percentage of morphologically normal spermatozoa as well as in an intensification of vacuolization. The genetic material in the sperm of these males showed higher fragmentation and methylation of genomic DNA.

Conclusion:
In the context of the epidemiological situation concerning the prevalence of infertility, while assessing the health effects of exposure to IR from artificial, including medical sources, the reproductive risk should be considered.

 
REFERENCES (64)
1.
Polish National Atomic Energy Agency. President’s Annual Report. Warsaw, 2014. Available from: www.paa.gov.pl (access: 2019.01.24).
 
2.
Mettler FA, Thomadsen BR, Bhargavan M, Gilley BD, Gray JE, Lipoti JA, et al. Medical radiation exposure in the US in 2006: preliminary results. Health Phys. 2008; 95(5): 502–507. (https://doi.org/10.1097/01.HP....).
 
3.
Do KH. General Principles of Radiation Protection in Fields of Diagnostic Medical Exposure. J Korean Med Sci. 2016; 31(Suppl. 1): S6–9. https://doi.org/10.3346/jkms.2....
 
4.
Polish National Atomic Energy Agency. President’s Annual Report. Warsaw, 2017. Available from: www.paa.gov.pl (access: 2019.01.24).
 
5.
Møller AP, Mousseau TA. The effects of natural variation in background radioactivity on humans, animals and other organisms. Biol Rev Camb Philos Soc. 2013; 88(1): 226–254. https://doi.org/10.1111/j.1469....
 
6.
Winters BR, Walsh TJ. The epidemiology of male infertility. Urol Clin North Am. 2014; 41(1): 195–204. https://doi.org/10.1016/j.ucl.....
 
7.
International Atomic Energy Agency. International basic safety standards for protection against ionizing radiation. Vienna: 1996. Available from: www-pub.iaea.org (access: 2019.01.24).
 
8.
Krajewski P. [Biological effects of ionizing radiation]. Warsaw: Central Laboratory for Radiological Protection, Faculty of Physics, Warsaw University of Technology; 2009. Available from: www.if.pw.edu.pl (access: 2019.01.24). [In Polish].
 
9.
Calabrese EJ, Baldwin LA. Defining hormesis. Human Exp Toxicol. 2002; 21(2): 91–97.
 
10.
Yan Cheng C. Spermatogenesis: Biology and Clinical Implications. 1st ed. Boca Raton: CRC Press, 2018.
 
11.
Cordelli E, Eleuteri P, Grollino MG, Benassi B, Blandino G, Bartoleschi C, et al. Direct and delayed X-ray-induced DNA damage in male mouse germ cells. Environ Mol Mutagen. 2012; 53(6): 429–439. https://doi.org/10.1002/em.217....
 
12.
Haines GA, Hendry JH, Daniel CP, Morris ID. Germ cell and dose-dependent DNA damage measured by the comet assay in murine spermatozoa after testicular X-irradiation. Biol Reprod. 2002; 67(3): 854–861.
 
13.
Haines GA, Hendry JH, Daniel CP, Morris ID. Increased levels of comet-detected spermatozoa DNA damage following in vivo isotopic- or X-irradiation of spermatogonia. Mutat Res. 2001; 495(1–2): 21–32.
 
14.
van der Meer Y, Huiskamp R, Davids JA, van der Tweel I, de Rooij DG. The sensitivity to X rays of mouse spermatogonia that are committed to differentiate and of differentiating spermatogonia. Radiat Res. 1992; 130(3): 296–302.
 
15.
Bastos H, Lassalle B, Chicheportiche A, Riou L, Testart J, Allemand I, et al. Flow cytometric characterization of viable meiotic and postmeiotic cells by Hoechst 33342 in mouse spermatogenesis. Cytometry A. 2005; 65(1): 40–49.
 
16.
Paris L, Cordelli E, Eleuteri P, Grollino MG, Pasquali E, Ranaldi R, et al. Kinetics of gamma-H2AX induction and removal in bone marrow and testicular cells of mice after X-ray irradiation. Mutagenesis. 2011; 26(4): 563–572. https://doi.org/10.1093/mutage....
 
17.
Rube CE, Zhang S, Miebach N, Fricke A, Rűbe C. Protecting the heritable genome: DNA damage response mechanisms in spermatogonial stem cells. DNA Repair. 2011; 10(2): 159–168. https://doi.org/10.1016/j.dnar....
 
18.
Zheng H, Olive PL. Influence of oxygen on radiation-induced DNA damage in testicular cells of C3H mice. Int J Radiat Biol. 1997; 71(3): 275–282.
 
19.
Zeng F, Baldwin DA, Schultz RM. Transcript profiling during preimplantation mouse development. Dev Biol. 2004; 272(2): 483–496.
 
20.
Ahmadi A, Ng SC. Developmental capacity of damaged spermatozoa. Hum Reprod. 1999; 14(9): 2279–2285. (https://doi.org/10.1093/humrep...).
 
21.
Schulte RT, Ohl DA, Sigman M, Smith GD. Sperm DNA damage in male infertility: etiologies, assays, and outcomes. J Assist Reprod Genet. 2010; 27(1): 3–12. https://doi.org/10.1007/s10815....
 
22.
Villani P, Fresegna AM, Ranaldi R, Eleuteri P, Paris L, Pacchierotti F, et al. X-ray induced DNA damage and repair in germ cells of PARP1 (-/-) male mice. Int J Mol Sci. 2013; 14(9): 18078–18092. https://doi.org/10.3390/ijms14....
 
23.
Ahmed EA, de Boer P, Philippens ME, Kal HB, de Rooij DG. Parp1-XRCC1 and the repair of DNA double strand breaks in mouse round spermatids. Mutat Res. 2010; 683(1–2): 84–90. https://doi.org/10.1016/j.mrfm....
 
24.
Ahmed EA, Scherthan H, De Rooij DG. DNA double strand break response and limited repair capacity in mouse elongated spermatids. Int J Mol Sci. 2015; 16(12): 29923–29935. https://doi.org/10.3390/ijms16....
 
25.
Ahmed EA, van der Vaart A, Barten A, Kal HB, Chen J, Lou Z, et al. Differences in DNA double strand breaks repair in male germ cell types: lessons learned from a differential expression of Mdc1 and 53BP1. DNA Repair. 2007; 6(9): 1243–1254.
 
26.
Pampfer S, Streffer C. Increased chromosome aberration levels in cells from mouse fetuses after zygote x-irradiation. Int J Radiat Biol. 1989; 55(1): 85–92.
 
27.
Ahmed EA, Sfeir A, Takai H, Scherthan H. Ku70 and non-homologous end joining protect testicular cells from DNA damage. J Cell Sci. 2013; 126(Pt 14): 3095–3104. https://doi.org/10.1242/jcs.12....
 
28.
Goedecke W, Eijpe M, Offenberg HH, van Aalderen M, Heyting C. Mre11 and Ku70 interact in somatic cells, but are differentially expressed in early meiosis. Nat Genet. 1999; 23(2): 194–198.
 
29.
Hamer G, Roepers-Gajadien HL, van Duyn-Goedhart A, Gademan IS, Kal HB, van Buul PP, et al. Function of DNA-protein kinase catalytic subunit during the early meiotic prophase without Ku70 and Ku86. Biol Reprod. 2003; 68(3): 717–721.
 
30.
Le Guen T, Ragu S, Guirouilh-Barbat J, Lopez BS. Role of the double-strand break repair pathway in the maintenance of genomic stability. Mol Cell Oncol. 2015; 2(1): e968020. https://doi.org/10.4161/237235....
 
31.
Kiliańska ZM, Żołnierczyk J, Węsierska-Gądek J. [Biological activity of the polymerase poly(ADP-ribose)polymerase-1]. Post Hig Med Dosw. 2010; 64: 344–363. [In Polish].
 
32.
West A, Lähdetie J. X-irradiation-induced changes in the progression of type B spermatogonia and preleptotene spermatocytes. Mol Reprod Dev. 2001; 58(1): 78–87.
 
33.
Zheng H, Olive PL. Influence of oxygen on radiation-induced DNA damage in testicular cells of C3H mice. Int J Radiat Biol. 1997; 71(3): 275–28.
 
34.
Chandley AC, Kofman-Alfaro S. Unscheduled DNA synthesis in human germ cells following UV irradiation. Exp Cell Res. 1971; 69(1): 45–48.
 
35.
Baudat F, Manova K, Yuen JP, Jasin M, Keeney S. Chromosome synapsis defects and sexually dimorphic meiotic progression in mice lacking Spo11. Mol Cell. 2000; 6(5): 989–998.
 
36.
Hamer G, Roepers-Gajadien HL, van Duyn-Goedhart A, Gademan IS, Kal HB, van Buul PP, et al. DNA double-strand breaks and gamma-H2AX signaling in the testis. Biol Reprod. 2003; 68(3): 628–634.
 
37.
Mahadevaiah SK, Turner JM, Baudat F, Rogakou EP, de Boer P, Blanco-Rodriguez J, et al. Recombinational DNA double-strand breaks in mice precede synapsis. Nat Genet. 2001; 27(3): 271–76.
 
38.
Romanienko PJ, Camerini-Otero RD. The mouse Spo11 gene is required for meiotic chromosome synapsis. Mol Cell. 2000; 6(5): 975–987.
 
39.
Beaumont HM. The effects of acute x-irradiation on primordial germ-cells in the female rat. Int J Radia Biol Relat Stud Phys Chem Med. 1966; 10(1): 17–28.
 
40.
Ahmed EA, Philippens ME, Kal HB, de Rooij DG, de Boer P. Genetic probing of homologous recombination and non-homologous end joining during meiotic prophase in irradiated mouse spermatocytes. Mutat Res. 2010; 688(1–2): 12–18. https://doi.org/10.1016/j.mrfm....
 
41.
Chicheportiche A, Bernardino-Sgherri J, De Massy B, Dutrillaux B. Characterization of Spo11-dependent and independent phospho-H2AX foci during meiotic prophase I in the male mouse. J Cell Sci. 2007; 120(Pt 10): 1733–1742. https://doi.org/10.1242/jcs.00....
 
42.
Allen JW, De Weese GK, Gibson JB, Poorman PA, Moses MJ. Synaptonemal complex damage as a measure of chemical mutagen effects on mammalian germ cells. Mutat Res. 1987; 190(1): 19–24.
 
43.
Baker SM, Plug AW, Prolla TA, Bronner CE, Harris AC, Yao X, et al. Involvement of mouse Mlh1 in DNA mismatch repair and meiotic crossing over. Nat Genet. 1996; 13(3): 336–342. https://doi.org/10.1038/ng0796....
 
44.
Jacquet P, de Saint-Georges L, Vankerkom J, Baugnet-Mahieu L. Embryonic death, dwarfism and fetal malformations after irradiation of embryos at the zygote stage: studies on two mouse strains. Mutat Res. 1995; 332(1–2): 73–87.
 
45.
Haines GA, Hendry JH, Daniel CP, Morris ID. Increased levels of comet-detected spermatozoa DNA damage following in vivo isotopic- or X-irradiation of spermatogonia. Mutat Res. 2001; 495(1–2): 21–3.
 
46.
Little MP, Goodhead DT, Bridges BA, Bouffler SD. Evidence relevant to untargeted and transgenerational effects in the offspring of irradiated parents. Mutat Res. 2013; 753(1): 50–67. https://doi.org/10.1016/j.mrre....
 
47.
Mikamo K, Kamiguchi Y, Tateno H. Spontaneous and in vitro radiation-induced chromosome aberrations in human spermatozoa: applications of a new method. In: Mendelsohn ML, Albertini RJ, editors. Mutation and the environment. Part b: Metabolism, testing methods and chromosomes. Wiley-Liss, New York; 1990. p. 447–456.
 
48.
Latini G, Dipaola L, Mantovani A, Picano E. Reproductive effects of low-to-moderate medical radiation exposure. Curr Med Chem. 2012; 19(36): 6171–6177.
 
49.
Fernández JL, Vázquez-Gundín F, Delgado A, Goyanes VJ, Ramiro-Díaz J, de la Torre J, et al. DNA breakage detection-FISH (DBD-FISH) in human spermatozoa: technical variants evidence different structural features. Mutat Res. 2000; 453(1): 77–82.
 
50.
Santiso R, Tamayo M, Gosálvez J, Johnston S, Mariño A, Fernández C, et al. DNA fragmentation dynamics allows the assessment of cryptic sperm damage in human: evaluation of exposure to ionizing radiation, hyperthermia, acidic pH and nitric oxide. Mutat Res. 2012; 734(1–2): 41–49. https://doi.org/10.1016/j.mrfm....
 
51.
Kumar D, Salian SR, Kalthur G, Uppangala S, Kumari S, Challapalli S, et al. Semen abnormalities, sperm DNA damage and global hypermethyla-tion in health workers occupationally exposed to ionizing radiation. PLoS One. 2013; 8(7): e69927. https://doi.org/10.1371/journa....
 
52.
Zhou DD, Hao JL, Guo KM, Lu CW, Liu XD. Sperm quality and DNA damage in men from Jilin Province, China, who are occupationally exposed to ionizing radiation. Genet Mol Res. 2016; 15(1): 1–7. https://doi.org/10.4238/gmr.15....
 
53.
Evdokimov VV, Erasova VI, Demin AI, Dubinina EB, Liubchenko PN. State of the reproductive system of men who participated in the cleaning-up of aftereffects of the Chernobyl AES accident. Med Tr Prom Ekol. 1993; 3–4: 25–26.
 
54.
Fischbein A, Zabludovsky N, Eltes F, Grischenko V, Bartoov B. Ultramorphological sperm characteristics in the risk assessment of health effects after radiation exposure among salvage workers in Chernobyl. Environ Health Perspect. 1997; 105 (Suppl 6): 1445–1449. https://doi.org/10.1289/ehp.97....
 
55.
Goncharov NP, Katsiya GV, Kolesnikova GS, Dobracheva GA, Todua TN, Vax VV, et al. Endocrine and reproductive health status of men who had experienced short-term radiation exposure at Chernobyl. Int J Androl. 1998; 21(5): 271–276.
 
56.
Premi S, Srivastava J, Chandy SP, Ali S. AZFc somatic microdeletions and copy number polymorphism of the DAZ genes in human males exposed to natural background radiation. Hum Genet. 2007; 121(3–4): 337–346. https://doi.org/10.1007/s00439....
 
57.
Premi S, Srivastava J, Chandy SP, Ahmad J, Ali S. Tandem duplication and copy number polymorphism of the SRY gene in patients with sex chromosome anomalies and males exposed to natural background radiation. Mol Hum Reprod. 2006; 12(2): 113–121. https://doi.org/10.1093/molehr....
 
58.
Premi S, Srivastava J, Chandy SP, Ali S. Unique signatures of natural background radiation on human Y chromosomes from Kerala, India. PLoS One. 2009; 4(2): e4541. https://doi.org/10.1371/journa....
 
59.
Pathak D, Premi S, Srivastava J, Chandy SP, Ali S. Genomic instability of the DYZ1 repeat in patients with Y chromosome anomalies and males exposed to natural background radiation. DNA Res. 2006; 13(3): 103–109. https://doi.org/10.1093/dnares....
 
60.
Kumar D, Salian SR, Kalthur G, Uppangala S, Kumari S, Challapalli S, et al. Association between sperm DNA integrity and seminal plasma antioxidant levels in health workers occupationally exposed to ionizing radiation. Environ Res. 2014; 132: 297–304. https://doi.org/10.1016/j.envr....
 
61.
Kłucinski P, Wójcik A, Grabowska-Bochenek R, Gmiński J, Mazur B, Hrycek A, et al. Erythrocyte antioxidant parameters in workers occupationally exposed to low levels of ionizing radiation. Ann Agric Environ Med. 2008; 15(1): 9–12.
 
62.
Alvarez R, Tusell L, Miró R, Genescà A, Caballín MR, Ribas M, et al. Dose-response relationship for the induction of chromosomal abnormalities in gamma-irradiated human spermatozoa. Environ Mol Mutagen. 1997; 29(4): 357–366.
 
63.
Saiyad Musthafa M, Jawahar Ali A, Mohamed Ahadhu Shareef TH, Vijayakumar S, Iyanar K, Thangaraj K. Ionizing radiation effects on sex steroid hormone levels in serum and milt of freshwater fish Oreochromis mossambicus. Ecotoxicol Environ Saf. 2014; 101: 103–106. https://doi.org/10.1016/j.ecoe....
 
64.
Verçosa P, Falcão PH, Furtado AF. Effects of gamma irradiation on the reproductive capacity and the longevity of Panstrongylus megistus (Hemiptera: Reduviidae). Mem Inst Oswaldo Cruz. 1993; 88(2): 195–201.
 
eISSN:1898-2263
ISSN:1232-1966
Journals System - logo
Scroll to top