Changes in semen characteristics and sex hormones of rats treated with iron oxide nanoparticles, silver nanoparticles and their mixture

Authors

  • Ibrahim Younus Ahmed Ministry of Health, Iraq.
  • Ibrahim Yousef Mokhtar Department of Environmental Studies, Institute of Graduate Studies and Research, Alexandria University, Egypt.
  • Abdel-NabiKamel Maher Department of Biochemistry, Medical Research Institute, Alexandria University, Egypt.
  • Alrawi Rakhad Grand Canyon University and Arizona College. Arizona, USA.
  • Mohammed Abdulrahman Jubran Center of Research and Educational Studies, Ministry of Education, Iraq.

DOI:

https://doi.org/10.30574/gscbps.2020.12.2.0272

Keywords:

Iron oxide nanoparticles, Silver nanoparticles, Male rats, Semen characteristics, Sex hormones.

Abstract

Few studies have demonstrated the toxicity of iron oxide nanoparticles (Fe2O3NPs), and silver nanoparticles (AgNPs), and their combination on reproductive performance.Therefore, the present study aimed to investigate the reproductive toxicity of iron oxide nanoparticles, silver nanoparticles and their combination in male rats. Forty Wistar male rats were divided into 4 equal groups. Group 1 served as control, group 2 was administered orally with Fe2O3NPs (5 mg/kg BW; >50 nm), group 3 was treated intraperitoneally with AgNPs (50 mg/kg BW; >100 nm) and group 4 was administered with the mixture of Fe2O3NPs with AgNPs. Animals were treated with doses every day for 79 days. The results revealed that semen parameters (sperm count, abnormal sperm and sperm motility) of adult male rats treated with Fe2O3NPs, AgNPs and their combination showed significant (P<0.05) reduction in sperm motility and sperm count, and significant (P<0.05) induction in abnormal sperm compared to the control group. Results also showed significant (P<0.05) decrease in the levels of testosterone and TSH, and significant (P<0.05) increase in FSH, LH, T3 and T4 levels in rats treated with Fe2O3NPs, AgNPs and their combination compared to the control group. The specific activity of testicular 17β-hydroxysteroid dehydrogenase (17β-HSD) was significantly decreased, while 17-ketosteroid reductase (17-KSR) was significantly increased in animals treated with Fe2O3NPs, AgNPs and their combination compared to the control group.  It was concluded that rats exposed to Fe2O3NPs, AgNPs or their combination caused testes inflammation and sex hormones imbalance that affect reproductive performance.

Metrics

Metrics Loading ...

References

Ema M, Kobayashi N, Naya M, Hanai S andNakanishi J. (2010). Reproductive and developmental toxicity studies of manufacturednanomaterials. Reprod. Toxicol, 30, 343-352.

Medina C, Santos-Martinez MJ, Radomski A, Corrigan OI and Radomski MW. (2007). Nanoparticles: pharmacological and toxicological significance. Br J. Pharmacol, 150, 552-558.

Kulthong K, Srisung S, Boonpavanitchakul K, Kangwansupamonkon W and Maniratanachote R. (2010). Determination of silver nanoparticle release from antibacterial fabrics into artificial sweat. Part Fiber Toxicol, 7, 2-9.

Chen X and Schluesener HJ. (2008). Nanosilver: a nanoproduct in medical application. Toxicol. Let, 176, 1-12.

Ji JH, Jung JH, Kim SS, Yoon JU, Park JD, Choi BS, Chung YH, Kwon IH, Jeong J, Han BS, Shin JH, Sung JH, Song KS and Yu IJ. (2007). Twenty-eight-day inhalation toxicity study of silver nanoparticles in Sprague-Dawley rats. Inhal. Toxicol, 19, 857-871.

Luoma PV. (2008). Cytochrome P450 and gene activation-from pharmacology to cholesterol elimination and regression of atherosclerosis. European. J. Clin. Pharmacol, 64, 841-850.

Braydich-Stolle L, Hussain S, Schlager JJ and Hofmann MC. (2005). In vitro Cytotoxicity of Nanoparticles in Mammalian Germline Stem Cells. Toxicol. Sci, 88, 412–419.

Hussain SM, Hess KL, Gearhart JM, Geiss KT and Schlager JJ. (2005). In vitro Toxicity of Nano particles in BRL 3A Rat Liver Cells. Toxicol. In Vitro, 19, 975-983.

Arora S, Jain J, Rajwade JM and Paknikar KM. (2008). Cellular Responses Induced by Silver Nanoparticles: In Vitro Studies. Toxicol. Let, 179, 93-100.

Krause W. (1995). Computer-assisted semen analysis systems: comparison with routine evaluation and prognostic value in male fertility and assisted reproduction. Human Reprod, 10, 60-66.

Dunson DB, Winberg CR, Perreault SD and Chapin RE. (1999).Summarizing the motion of self-propelled cells: applications to sperm motility, Biometrics, 55, 537-543.

Wells ME and Awa OA. (1970). New technique for assessing acrosomalcharacteristics of spermatozoa. J. Dairy Sci, 53, 227-232.

Zirkin BR and Chen H. (2000). Regulation of Leydig cell steroidogenic function during aging. Biol. Reprod, 63, 977-981.

Sakuma Y. (2009). Gonadal steroid action and brain sex differentiation in the rat. J. Neuroendocrinol, 21, 410-414.

Kjeld JM, Kuku SF, Harsoulis P, Fraser TR, Puah CM and Joplin GF. (1976). Infusions of hFSH and hLH in normal men. II. Serum testosterone response to infusedhLH and hFSH.,ActaEndocrinol, 81, 234-242.

Closset, J., Hennen, G., 1988. Biopotency of highly purified FSH and LH on the testis of immature hypophysectomized rats. J. Endocrinol., 119: 1-542.

Teerds KJJ, Rommerts RFG, De Rooij DG, Stocco DM, Colenbrander B, Gwensing J and Hennen G. (1989). Effects of pure FSH and LH preparations on the number and function of Leydig cells in immature hypophysectomized rats. J. Endocrinol, 120, 97-106.

Katryna B and Anita PP. (1980). Purification of rat testicular microsomal 17-ketosteroid reductase. Evidence that 17-ketosteroid reductase and 17-bhydroxysteroid dehydrogenase are distinct enzymes. J. Biol. Chem, 255, 5552-5559.

Bogovich K and Payne AH. (1980). Purification of rat testicular microsomal 17β- hydroxysteroid dehydrogenase are distinct enzymes. J. Biol. Chem, 255, 5552-5559.

Hedayati M, Yazdanparast R and Azizi F. (2001). Determination of human tumor necrosis factor α by a highly sensitive enzyme immunoassay. Biochem. Biophys. Res. Comm, 289, 295-298.

Hupp TR, Lane DP and Ball KL. (2000). Strategies for manipulating the p53 pathway in the treatment of human cancer. Biochem. J, 352, 1-17.

Renard P, Ernest I, Houbion A, Art M, Le Calvez H, Raes M and Remacle J. (2001). Development of a sensitive multi-well colorimetric assay for active NF kappa B. Nucleic Acids Res, 29, 1-21.

Gottifredi V and Prives C. (2001). Getting p53 out of the nucleus. Science, 292, 1851-1852.

Yang A, Kaghad M, Caput D and McKeon F. (2002). On the shoulders of giants: p63, p73 and the rise of p53. Trends Genet, 18, 90-95.

Yoshida H, Kuwauchi Y, Jinschek JR, Sun K, Kohyama M, Shimada S, Haruta M and Takeda S. (2012). Gas Molecules Interacting with Supported Nanoparticulate Catalysts at Reaction Conditions. Sci. Magazine, 335, 317-319.

Hughes GA. (2005). Nano structure-Mediated drug delivery, Nanomedicine, 1, 22-30.

Manin OI, Nikolaev VA, Kolomiitsev AA and LebedenkoIIu. (2007). Comparative toxicological evaluation of domestic golden alloys for soldering. Stomatologiia (Mosk), 86, 64-67.

Carlson C, Hussain SM and Schr AM. (20080. Unique cellular interaction of nanoparticles: size dependent generation of reactive oxygen species. J. Phys. Chem. B, 112, 13608-13619.

Yoshida Y, Itoh N, Saito Y, Hayakawa M and Niki E. (2004). Application of water-soluble radical initiator, 2, 2′-azobis-[2-(2-imidazolin-2-yl) propane] dihydrochloride, to a study of oxidative stress. Free Radical res, 38, 375-384.

De-Lamirande E and Gagnon C. (1992). Reactive oxygen species and human spermatozoa. J. andrology, 13, 379-386.

Aziz N, Saleh RA, Sharma RK, Lewis-Jones I, Esfandiari N, Thomas AJ and Agarwal A. (2004). Novel association between sperm reactive oxygen species production, sperm morphological defects, and the sperm deformity index. Fertil. Steril, 81, 349-354.

Nasri S, Rezai-Zarchi S, Kerishchi P and Sadeghi S. (2015). The Effect of Iron Oxide Nanoparticles on Sperm Numbers and Mobility in Male Mice. Zahedan J. Res. Med. Sci, 17, 29-31.

Stocco DM. (2000). The role of the StAR protein in steroidogenesis: challenges for the future. J. Endocrinol, 164, 247-253.

Buzzard JJ, Morrison JR, O’Bryan MK, Song Q and Wreford NG. (2000). Developmental expression of thyroid hormone receptorsin the rat testis. Biol. Reprod, 62, 664-669.

Chandra AK, Sengupta P, Goswami H and Sarkar M. (2013). Effects of dietary magnesium on testicular histol- ogy, steroidogenesis, spermatogenesis and oxidative stress markers in adult rats. Indian J. Exp. Biol, 51, 37-47.

Mendis-Handagama SM and Ariyaratne HB. (2001). Differentiation of the adult Leydig cell population in the postnatal testis. Biol. Reprod, 65, 660-671.

Yoshida M, Kitani T, Takenaka A, Kudoh K, Katsuda SI and Taya K. (2002). Lack of effects of oxolinic acid on spermatogenesis in young male adult and aged Wistar rats. Food. Chem. Toxicol, 40, 1815–1825.

Downloads

Published

2022-02-04

How to Cite

Ahmed , I. Y., Mokhtar , I. Y., Maher , A.-N., Rakhad , A., & Jubran , M. A. (2022). Changes in semen characteristics and sex hormones of rats treated with iron oxide nanoparticles, silver nanoparticles and their mixture. GSC Biological and Pharmaceutical Sciences, 12(2), 229–237. https://doi.org/10.30574/gscbps.2020.12.2.0272

Issue

Section

Original Article