Selenium supplementation in the form of selenium nanoparticles and selenite sodium improves mature male mice reproductive performances

Document Type : Original Article

Authors

1 Department of Clinical Pathology and Internal Medicine, Faculty of Veterinary Medicine, Urmia University, Urmia, West Azerbaijan, Iran

2 Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, West Azerbaijan, Iran

3 Ph.D. Student of Histology, Faculty of Veterinary Medicine, Urmia University, Urmia, West Azerbaijan, Iran

Abstract

Objective(s): The current study was conducted to examine the possible protective and retentive effects of one-week intra-peritoneal (IP) administration of selenium nanoparticles (Se-NPs), compared to its bulk counterpart, selenite sodium (Ss), after one complete cycle of spermatogenesis in mature male mice.
Materials and Methods: Thirty adult male mice were divided into 3 groups. Control group was administrated phosphate-buffered saline (IP) and the other groups received Ss (0.50 mg kg-1) and Se-NPs (0.50 mg kg-1) for seven successive days. Then, the animals were monitored for 28 days and finally sacrificed and tissue and blood samples were taken. Histopathological features, sperm quality, in vitro fertilization (IVF) capability and selenium (Se) content in testicular tissue were analyzed. Antioxidant enzyme activities including catalase, glutathione peroxidase, and superoxide dismutase as well as total antioxidant capacity and malondialdehyde levels were assessed in blood and the tissue samples.
Results: Remarkable differences were found in sperm characteristics, histopathological features and oxidative stress biomarkers between control and treatment groups. Moreover, IVF evaluation and tissue Se concentration examination weren’t similar for Se-NPs and Ss.
Conclusion: Conclusively, Se-treated groups had more antioxidant capacity than the control group, but sperm quality and histopathological features revealed that Se-NPs might possess more antioxidative and retentive potential compared to Ss in one spermatogenesis cycle.

Keywords

Main Subjects


1. Schwarz K, Foltz CM. Selenium as an integral part of factor 3 against dietary necrotic liver degeneration. J Am Chem Soc 1957; 79:3292-3293.
2. Drake E. Cancer chemoprevention: selenium as a prooxidant, not an antioxidant. Med Hypotheses 2006; 67:318-322.
3. Kieliszek M, Błażejak S. Selenium: significance, and outlook for supplementation. Nutrition 2013; 29:713-718.
4. Driscoll DM, Copeland PR. Mechanism and regulation of selenoprotein synthesis Annu Rev Nutr 2003; 23:17-40.
5. Miyamoto Y, Koh YH, Park YS, Fujiwara N, Sakiyama H, Misonou Y, et al Oxidative stress caused by inactivation of glutathione peroxidase and adaptive responses. J Biol Chem 2003; 384:567-574.
6. Imai H, Nakagawa Y. Biological significance of phospholipid hydroperoxide glutathione peroxidase (PHGPx, GPx4) in mammalian cells. Free Radic Biol Med 2003; 34:145-169.
7. Becker K, Gromer S, Schirmer RH, Müller S. Thioredoxin reductase as a pathophysiological factor and drug target. FEBS J 2000; 267:6118-6125.
8. Rayman MP. Selenium and human health. The Lancet 2012; 379:1256-1268.
9. Lei C, Niu X, Wei J, Zhu J, Zhu Y. Interaction of glutathione peroxidase-1 and selenium in endemic dilated cardiomyopathy. Clin Chim Acta 2009; 399:102-108.
10. Reeves M, Hoffmann P. The human selenoproteome: recent insights into functions and regulation. Cell Mol Life Sci 2009; 66:2457-2478.
11. Schiavon R, Guidi G, Biasioli S, De Fanti E, Targa L. Plasma glutathione peroxidase activity as an index of renal function. Clin Chem Lab Med 1994; 32:759-766.
12. Burk RF, Hill KE. Selenoprotein P—expression, functions, and roles in mammals. Biochim Biophys Acta 2009; 1790:1441-1447.
13. Schomburg L, Köhrle J. On the importance of selenium and iodine metabolism for thyroid hormone biosynthesis and human health. Mol Nutr Food Res 2008; 52:1235-1246.
14. Aitken RJ, Baker MA. Oxidative stress and male reproductive biology. Reprod Fertil Dev 2004; 16:581-588.
15. Burk RF, Hill KE, Motley AK. Selenoprotein metabolism and function: evidence for more than one function for selenoprotein P. J. Nutr 2003; 133:1517S-1520S.
16. Moslemi MK, Tavanbakhsh S. Selenium–vitamin E supplementation in infertile men: effects on semen parameters and pregnancy rate. Int J Gen Med 2011; 4:99.
17. Wallace E, Calvin HI, Cooper GW. Progressive defects observed in mouse sperm during the course of three generations of selenium deficiency. Gamete Res 1983; 7:377-387.
18. Wu A, Oldfield J, Shull L, Cheeke P. Specific effect of selenium deficiency on rat sperm. Biol Reprod 1979; 20:793-798.
19. Ursini F, Heim S, Kiess M, Maiorino M, Roveri A, Wissing J, et al Dual function of the selenoprotein PHGPx during sperm maturation. Science 1999; 285:1393-1396.
20. Liu C, Chen Y, Zhang J, Huang M, Su Q, Lu Z, et al. Preliminary studies on influence of selenium deficiency to the developments of genital organs and spermatogenesis of infancy boars. J Anim Sci 1982; 2:000.
21. Brennan KM, Pierce JL, Cantor AH, Pescatore AJ, Xiao R, Power RF. Source of selenium supplementation influences testis selenium content and gene expression profiles in Single Comb White Leghorn roosters. Biol Trace Elem Res 2012; 145:330-337.
22. Foresta C, Flohé L, Garolla A, Roveri A, Ursini F, Maiorino M. Male fertility is linked to the selenoprotein phospholipid hydroperoxide glutathione peroxidase. Biol Reprod 2002; 67:967-971.
23. Olson GE, Winfrey VP, NagDas SK, Hill KE, Burk RF. Selenoprotein P is required for mouse sperm development. Biol Reprod 2005; 73:201-211.
24. Shalini S, Bansal M. Dietary selenium deficiency as well as excess supplementation induces multiple defects in mouse epididymal spermatozoa: understanding the role of selenium in male fertility. Int J Androl 2008; 31:438-449.
25. Yang H, Jia X. Safety evaluation of Se-methylselenocysteine as nutritional selenium supplement: Acute toxicity, genotoxicity and subchronic toxicity. Regul Toxicol Pharmacol 2014; 70:720-727.
26. Ip C, Ganther HE. Activity of methylated forms of selenium in cancer prevention. Br J Cancer 1990; 50:1206-1211.
27. Maier KJ, Knight AW. Ecotoxicology of selenium in freshwater systems. Rev Environ Contam Toxicol 1994; 31-48.
28. Zhang J-S, Gao X-Y, Zhang L-D, Bao Y-P. Biological effects of a nano red elemental selenium. Biofactors 2001; 15:27-38.
29. Zhang J, Wang X, Xu T. Elemental selenium at nano size (Nano-Se) as a potential chemopreventive agent with reduced risk of selenium toxicity: comparison with se-methylselenocysteine in mice. Toxicol Sci 2008; 101:22-31.
30. Abd-Allah S, Hashem KS. Selenium nanoparticles increase the testicular antioxidant activity and spermatogenesis in male rats as compared to ordinary selenium. Int J Adv Res 2015; 3:792-802.
31. Shi L-g, Yang R-j, Yue W-b, Xun W-j, Zhang C-x, Ren Y-s, et al Effect of elemental nano-selenium on semen quality, glutathione peroxidase activity, and testis ultrastructure in male Boer goats. Anim Reprod Sci 2010; 118:248-254.
32. Reeves PG, Nielsen FH, Fahey Jr GC. AIN-93 purified diets for laboratory rodents: final report of the American Institute of Nutrition ad hoc writing committee on the reformulation of the AIN-76A rodent diet. J Nutr 1993; 123:1939-1951.
33. Hassanin KM, El-Kawi SHA, Hashem KS. The prospective protective effect of selenium nanoparticles against chromium-induced oxidative and cellular damage in rat thyroid. Int J Nanomedicine 2013; 8:1713-1720.
34. Oakberg EF. Duration of spermatogenesis in the mouse and timing of stages of the cycle of the seminiferous epithelium. Dev Dyn 1956; 99:507-516.
35. Babaei M, Najafi G, Jalali AS, Behfar M. Effects of unilateral Iatrogenic vas deferens trauma on fertility: An experimental in vitro fertilization mice model study. Bull Emerg Trauma 2015; 3:122.
36. Johnson M. Changes in the blood-testis barrier of the guinea-pig in relation to histological damage following iso-immunization with testis. Reprod Abstr Ser 1970; 22:119-127.
37. Cooper TG, Noonan E, Von Eckardstein S, Auger J, Baker HG, Behre HM, et al. World Health Organization reference values for human semen characteristics. Hum Reprod 2010; 16:231-245.
38. Nazarizadeh A, Asri-Rezaie S. Comparative study of antidiabetic activity and oxidative stress induced by zinc oxide nanoparticles and zinc sulfate in diabetic rats. AAPS Pharm Sci Tech 2015; 17:1-10.
39. Asri-Rezaei S, Dalir-Naghadeh B. Evaluation of antioxidant status and oxidative stress in cattle naturally infected with Theileria annulata. Vet Parasitol 2006; 142:179-186.
40. Aebi H. [13] Catalase in vitro. Methods Enzymol 1984; 105:121-126.
41. Buege JA, Aust SD. [30] Microsomal lipid peroxidation. Methods Enzymol 1978; 52:302-310.
42. Zhang Q, Chen L, Guo K, Zheng L, Liu B, Yu W, et al Effects of different selenium levels on gene expression of a subset of selenoproteins and antioxidative capacity in mice. Biol Trace Elem Res 2013; 154:255-261.
43. Asri-Rezaei S, Dalir-Naghadeh B, Nazarizadeh A, Noori-Sabzikar Z. Comparative study of cardio-protective effects of zinc oxide nanoparticles and zinc sulfate in streptozotocin-induced diabetic rats. J Trace Elem Med Biol 2017; 42:129-141.
44. El‐Sharawy M, Eid E, Darwish S, Abdel‐Razek I, Islam M, Kubota K, et al. Effect of organic and inorganic selenium supplementation on semen quality and blood enzymes in buffalo bulls. Animal Sci J 2017; 88:999-1005.
45. Working PK. Male reproductive toxicology: comparison of the human to animal models. Environ Health Perspect 1988; 77:37-44.
46. Högstrand K, Böhme J. Gene conversion of major histocompatibility complex genes in the mouse spermatogenesis is a premeiotic event. Mol Biol Cell 1997; 8:2511-2517.
47. Mangoli E, Talebi AR, Anvari M, Pourentezari M. Effects of experimentally-induced diabetes on sperm parameters and chromatin quality in mice. Int J Reprod Biomed 2013; 11:53-60.
48. Ramamurthy Ch, Sampath KS, Arunkumar P, Kumar MS, Sujatha V, Premkumar K, Thirunavukkarasu C. Green synthesis and characterization of selenium nanoparticles and its augmented cytotoxicity with doxorubicin on cancer cells. Bioprocess Biosyst Eng. 2013; 36:1131-1139.
49. He Y, Chen S, Liu Z, Cheng C, Li H, Wang M. Toxicity of selenium nanoparticles in male Sprague–Dawley rats at supranutritional and nonlethal levels. Life Sci 2014; 115:44-51.
50. Jang D-Y, Kim S-J, Jeong J-H, Nam SY, Kim J-S, Yun YW, et al. Protective effects of sodium selenite and selenium nanoparticles against experimental colon carcinogenesis in mice. Prev Vet Med 2016; 40:101-108.
51. Ehlig C, Hogue D, Allaway W, Hamm D. Fate of selenium from selenite or seleno-methionine, with or without vitamin E in lambs. J Nutr 1967; 92:121-126.
52. Lopez PL, Preston R, Pfander W. Whole-body retention, tissue distribution and excretion of selenium-75 after oral and intravenous administration in lambs fed varying selenium intakes. J Nutr 1969; 97:123-132.
53. Hu C, Li Y, Xiong L, Zhang H, Song J, Xia M. Comparative effects of nano elemental selenium and sodium selenite on selenium retention in broiler chickens. Anim Feed Sci Technol 2012; 177:204-210.
54. Sánchez-Gutiérrez M, García-Montalvo E, Izquierdo-Vega J, Del Razo L. Effect of dietary selenium deficiency on the in vitro fertilizing ability of mice spermatozoa. Cell Biol Toxicol 2008; 24:321-329.
55. Zhang X, Wu H, Wu D, Wang Y, Chang J, Zhai Z, Meng A, Liu P, Zhang L, Fan F. Toxicologic effects of gold nanoparticles in vivo by different administration routes. Int J Nanomedicine 2010; 5: 771–781.
56. Kaur P, Bansal MP. Effect of selenium-induced oxidative stress on the cell kinetics in testis and reproductive ability of male mice. Nutrition 2005; 21:351-357.
57. Nouri K, Walch K, Weghofer A, Imhof M, Egarter C, Ott J. The impact of a standardized oral multinutrient supplementation on embryo quality in in vitro fertilization/intracytoplasmic sperm injection: A prospective randomized trial. Gynecol  Obstet Invest 2016; 82:8-14.
58. Goel A, Dani V, Dhawan D. Protective effects of zinc on lipid peroxidation, antioxidant enzymes and hepatic histoarchitecture in chlorpyrifos-induced toxicity. Chem Biol Interact 2005; 156:131-140.
59. Brigelius-Flohé R, Wingler K, Müller C. Estimation of individual types of glutathione peroxidases. Methods Enzymol 2002; 347:101-12.