ISSN 2415-3060 (print), ISSN 2522-4972 (online)
  • 38 of 45
Up
УЖМБС 2021, 6(6): 275–281
https://doi.org/10.26693/jmbs06.06.275
Biology

Cadmium Toxicity and Reproductive Function of Males

Ostrovska S. S. 1, Abramov S. V. 1, Pisarevska I. A. 1, Trushenko O. S. 1, Zherzhova T. A. 1, Pervysherst K. Y. 1, Stepura V. P. 1, Zakhariev A. V. 2
Abstract

The purpose of the review of foreign literature was to analyze current research on the effects of cadmium on male reproductive function. Results. According to the researcher data, at least 15–20% of cases of fertility decline in males fall on infertility. The etiology of this phenomenon in 50% of cases remains unknown, however, increasing environmental pollution contributes to a constant increase in male infertility. One of the most toxic pollutants is cadmium. Numerous animal model studies and human epidemiological studies indicate an adverse effect of cadmium on male fertility. Smoking is an important source of cadmium, which is absorbed into the human body. In vitro studies confirm the deleterious effects of cigarette smoke compounds on sperm motility and spermatozoon parameters. Depending on the concentration, nicotine suppresses the progressive motility of the spermatozoon parameters, starting from the lowest concentration used (1 ng/ml). Likewise, it decreases the percentage of viable spermatozoon parameters and increases the amount of spermatozoon parameters in late apoptosis with altered chromatin compactness or DNA fragmentation already after 3 hours of incubation. On average, the daily intake of cadmium in humans is 1.06 μg/kg body weight, the half-life of cadmium is more than 20-40 years, which causes its accumulation in the body. The testicles are the organ in which cadmium is stored in large quantities. Studies have shown that the testicles are extremely sensitive to cadmium because these organs are characterized by intense cellular activity, where vital spermatogenesis processes take place. Exposure to cadmium leads to reproductive tract abnormalities such as cryptorchidism and hypospadias, testicular cancer, subfertility or infertility, called testicular dysgenesis syndrome. In the genesis of the testicles during the embryonic and neonatal periods, Sertoli’s cells play a critical role, the development of which is influenced by cadmium. Exposure to cadmium (1-2 mg/kg, subcutaneously) in pregnant and lactating rats causes vacuolization of Sertoli’s cells and loss of cells in the epithelium of the seminiferous tubules in adult animals. Cadmium inhibits proliferation, induces apoptosis and DNA damage in immature Sertoli’s cells. Perinatal exposure to cadmium affects the development and function of fetal Leydig cells, which are endocrine cells in the testicle. In pregnant rats that received a single dose of cadmium (0.25, 0.5, and 1.0 mg/kg, intraperitoneally), synthesis of testosterone in the fetal tests was significantly reduced, while gene expression in cells was suppressed, and the androgen-dependent formation process was reduced. The mechanism by which cadmium mediates impaired male fertility is also associated with the production of reactive oxygen species in the testicles, which leads to oxidative stress that interferes with the development and functioning of the spermatozoon parameters. Exposure to cadmium, for both environmental and occupational reasons, can contribute to a decrease in the quality of human sperm, which confirms high toxicity of cadmium. Conclusion. Thus, in humans and other mammals, cadmium damages the male reproductive system, disrupts its structure, including the vascular system of the testicles, leads to DNA damage, inhibits functions of germ cells, leads to loss of sperm quality and quantity, sub-fertility or infertility

Keywords: cadmium, toxicity, male reproductive system, male infertility

Full text: PDF (Ukr) 238K

References
  1. Choy JT, Eisenberg ML. Male infertile as a window to health. Fertil Steril. 2018;110(5):810-814. https://www.ncbi.nlm.nih.gov/pubmed/30316415. https://doi.org/10.1016/j.fertnstert.2018.08.015
  2. Gao Y, Mruk DD, Cheng CY. Sertoli cells are the target of environmental toxicants in the testis - a mechanistic and therapeutic insight. Expert Opin Ther Targets. 2015;19:1073-1090. https://www.ncbi.nlm.nih.gov/pubmed/25913180. PMCID: PMC4822540. https://doi.org/10.1517/14728222.2015.1039513
  3. Wirth JJ, Mijal RS. Adverse effects of low level heavy metal exposure on male reproductive function. Syst Biol Reprod Med. 2010;56:147-167. https://www.ncbi.nlm.nih.gov/pubmed/20377313. https://doi.org/10.3109/19396360903582216
  4. Levine H, Jоrgensen N, Martino-Andrade A, Mendiola J, Weksler-Derri D, Mindlis I, et al. Temporal trends in sperm count: a systematic review and meta-regression analysis. Hum Reprod Update. 2017;23:646-659. https://www.ncbi.nlm.nih.gov/pubmed/28981654. PMCID: PMC6455044. https://doi.org/10.1093/humupd/dmx022
  5. Condorelli RA, LaVignera S, Giacone F, Iacoviello L,Vicari E, Mongioi L, et al. In vitro effects of nicotine on sperm motility and bio-functional flow cytometry sperm parameters. Int J Immunopathol Pharmacol. 2013;26(3):739-746. https://www.ncbi.nlm.nih.gov/pubmed/24067470. https://doi.org/10.1177/039463201302600317
  6. Alamo A, Condorelli RA, Mongioì LM, Cannarella R, Giacone F, Calabrese V, et al. Environment and male fertility: effects of benzo-α-pyrene and resveratrol on human sperm function in vitro. J Clin Med. 2019;8(4):E561. https://www.ncbi.nlm.nih.gov/pubmed/31027257. PMCID: PMC6518055. https://doi.org/10.3390/jcm8040561
  7. Wan HT, Mruk DD, Wong CK, Cheng CY. The apical ES-BTB-BM functional axis is an emerging target for toxicant-induced infertility. Trends Mol Med. 2013;19:396-405. https://www.ncbi.nlm.nih.gov/pubmed/23643465. PMCID: PMC3699959. https://doi.org/10.1016/j.molmed.2013.03.006
  8. Skakkebaek NE, Rajpert-De Meyts E, Main KM. Testicular dysgenesis syndrome: an increasingly common developmental disorder with environmental aspects. Hum Reprod. 2001;16:972-978. https://www.ncbi.nlm.nih.gov/pubmed/11331648. https://doi.org/10.1093/humrep/16.5.972
  9. Loebenstein M, Thorup J, Cortes D, Clasen-Linde E, Hutson JM, Li R. Cryptorchidism, gonocyte development, and the risks of germ cell malignancy and infertility: a systematic review. J Pediatr Surg. 2019;S0022-3468(19):30450-30456. https://www.ncbi.nlm.nih.gov/pubmed/31327540. https://doi.org/10.1016/j.jpedsurg.2019.06.023
  10. Skinner MK, Bhandari RK, Haque MM, Nilsson EE. Environmentally induced epigenetic transgenerational inheritance of altered SRY genomic binding during gonadal sex determination. Environ Epigenet. 2015;1:dvv004. https://www.ncbi.nlm.nih.gov/pubmed/27175298. PMCID: PMC4862609. https://doi.org/10.1093/eep/dvv004
  11. Rebourcet D, Wu J, Cruickshanks L, Smith S.E, Milne L, Fernando A, Wallace RJ, et al. Sertoli cells modulate testicular vascular network development, structure, and function to influence circulating testosterone concentrations in adult male mice. Endocrinology. 2016;157:2479-2488. https://www.ncbi.nlm.nih.gov/pubmed/27145015. PMCID: PMC4891787. https://doi.org/10.1210/en.2016-1156
  12. Li X, Liu J, Wu S, Zheng W, Li H, Bao S, et al. In utero single low-dose exposure of cadmium induces rat fetal Leydig cell dysfunction. Chemosphere. 2018;194:57-66. https://www.ncbi.nlm.nih.gov/pubmed/29197250. https://doi.org/10.1016/j.chemosphere.2017.11.159
  13. Bekheet SH. Comparative effects of repeated administration of cadmium chloride during pregnancy and lactation and selenium protection against cadmium toxicity on some organs in immature rats' offsprings. Biol Trace Elem Res. 2011;144:1008-1023. https://www.ncbi.nlm.nih.gov/pubmed/21614561. https://doi.org/10.1007/s12011-011-9084-z
  14. Zhang J, Huang B, Hu G, Zhan X, Xie T, Li S, et al. Aldosterone blocks rat stem Leydig cell development in vitro. Front Endocrinol. 2018;9:4. https://www.ncbi.nlm.nih.gov/pubmed/29416526. PMCID: PMC5787991. https://doi.org/10.3389/fendo.2018.00004
  15. Yu X, Hong S, Faustman E.M. Cadmium-induced activation of stress signaling pathways, disruption of ubiquitin-dependent protein degradation and apoptosis in primary rat Sertoli cell-gonocyte cocultures. Toxicol Sci. 2008;104:385-396. https://www.ncbi.nlm.nih.gov/pubmed/18463101. PMCID: PMC2734295. https://doi.org/10.1093/toxsci/kfn087
  16. Wu S, Yan M, Ge R, Cheng C.Y. Crosstalk between sertoli and germ cells in male fertility. Trends Mol Med. 2019;26:215-231. https://www.ncbi.nlm.nih.gov/pubmed/31727542. https://doi.org/10.1016/j.molmed.2019.09.006
  17. Wang Y, Ni C, Li X, Lin Z, Zhu Q, Li L, et al. Phthalate-induced fetal Leydig cell dysfunction mediates male reproductive tract anomalies. Front Pharmacol. 2019;10:1309. https://www.ncbi.nlm.nih.gov/pubmed/31780936. PMCID: PMC6851233. https://doi.org/10.3389/fphar.2019.01309
  18. Tian H, Chen S, Leng Y, Li T, Li Z, Chen H, et al. Exposure to cadmium during gestation and lactation affects development and function of Leydig cells in male offspring. Environ Toxicol. 2018;33:351-360. https://www.ncbi.nlm.nih.gov/pubmed/29214744. https://doi.org/10.1002/tox.22522
  19. Leite R, Peloso EF, Gadelha F R, Dolder M.A. Environmentally realistic doses of cadmium as a possible etiologic agent for idiopathic pathologies. Biol Trace Elem Res. 2015;168:133-140. https://www.ncbi.nlm.nih.gov/pubmed/25850543. https://doi.org/10.1007/s12011-015-0322-7
  20. Morielli T, O'flaherty C. Oxidative stress impairs function andincreasesredox protein modifications in human spermatozoa. Reproduction. 2015;149:113-123. https://www.ncbi.nlm.nih.gov/pubmed/25385721. PMCID: PMC5489333. https://doi.org/10.1530/REP-14-0240
  21. Mahmoudi R, Azizi A, Abedini S, Hemayatkhah-Jahromi V, Abidi H, Jafari-Barmak M. Green tea improves rat sperm quality and reduced cadmium chloride damage effect in spermatogenesis cycle. J Med Life. 2018;11:371-380. https://www.ncbi.nlm.nih.gov/pubmed/30894897. PMCID: PMC6418325. https://doi.org/10.25122/jml-2018-0005
  22. Pandya C, Pillai P, Nampoothiri LP, Bhatt N, Gupta S, Gupta S. Effect of lead and cadmium co-exposure on testicular steroid metabolism and antioxidant system of adult male rats. Andrologia. 2012;44(1):813-822. https://www.ncbi.nlm.nih.gov/pubmed/21933223. https://doi.org/10.1111/j.1439-0272.2010.01137.x
  23. Chen N, Su P, Wang M, Li YM. Ascorbic acid inhibits cadmium-induced disruption of the blood-testis barrier by regulating oxidative stress-mediated p38 MAPK pathways. Environ Sci Pollut Res. 2018;25:21713-21720. https://www.ncbi.nlm.nih.gov/pubmed/29790047. https://doi.org/10.1007/s11356-018-2138-4
  24. Zhang J, Cai Z, Ma C, Xiong J, Li H. Impacts of outdoor air pollution on human semen quality: a meta-analysis and systematic review. BioMed Res Int. 2020:1-9. https://www.ncbi.nlm.nih.gov/pubmed/32420369. PMCID: PMC7204269. https://doi.org/10.1155/2020/7528901
  25. Zhu Q, Li X, Ge R-S. Toxicological Effects of Cadmium on Mammalian. Front Genet. 2020;11:527. https://www.ncbi.nlm.nih.gov/pubmed/32528534. PMCID: PMC7265816. https://doi.org/10.3389/fgene.2020.00527
  26. Zhao LL, Ru YF, Liu M, Tang JN, Zheng JF, Wu B, et al. Reproductive effects of cadmium on sperm function and early embryonic development in vitro. PLoS ONE. 2017;12:e0186727. https://www.ncbi.nlm.nih.gov/pubmed/29095856. PMCID: PMC5667747. https://doi.org/10.1371/journal.pone.0186727
  27. Zhu H, Li K, Liang J, Zhang J, Wu Q. Changes in the levels of DNA methylation in testis and liver of SD rats neonatally exposed to 5-aza-2'-deoxycytidine and cadmium. J Appl Toxicol. 2011;31:484-495. https://www.ncbi.nlm.nih.gov/pubmed/21449042. https://doi.org/10.1002/jat.1673
  28. Nakayama SMM, Nakata H, Ikenaka Y, Yabe J, Oroszlany B, Yohannes Y. B, et al. One year exposure to Cd- and Pb-contaminated soil causes metal accumulation and alteration of global DNA methylation in rats. Environ Pollut. 2019;252:1267-1276. https://www.ncbi.nlm.nih.gov/pubmed/31252124. https://doi.org/10.1016/j.envpol.2019.05.038
  29. Ghosh K, Chatterjee B, Behera P, Kanade SR. The carcinogen cadmium elevates CpG-demethylation and enrichment of NFYA and E2F1 in the promoter of oncogenic PRMT5 and EZH2 methyltransferases resulting in their elevated expression in vitro. Chemosphere. 2019;242:125186. https://www.ncbi.nlm.nih.gov/pubmed/31675590. https://doi.org/10.1016/j.chemosphere.2019.125186
  30. Fiore M, Giacone F, Altomare M, Asero P, Ledda C, Romeo G. Exposure to multiple metals/metalloids and human semen quality: A cross-sectional study. Ecotoxicol Environ Saf. 2021;215(1):112165. https://www.ncbi.nlm.nih.gov/pubmed/33773149. https://doi.org/10.1016/j.ecoenv.2021.112165