Publication Cover
Redox Report
Communications in Free Radical Research
Volume 28, 2023 - Issue 1
1,929
Views
4
CrossRef citations to date
0
Altmetric
Research Article

Zinc improves sexual performance and erectile function by preventing penile oxidative injury and upregulating circulating testosterone in lead-exposed rats

, , , , , , & ORCID Icon show all

References

  • Martin K, Hosam MS. Introductory chapter: introducing heavy metals. In: HE-DM Saleh, RF Aglan, editors. Heavy metals. IntechOpen; 2018. doi:10.5772/intechopen.74783
  • Arif TJ, Mudsser A, Kehkashan S, et al. Heavy metals and human health: mechanistic insight into toxicity and counter defense system of antioxidants. Int J Mol Sci. 2015;16:29592–29630.
  • Cheng SP. Heavy metals pollution in China: origion, pattern and control. Environ Sci Pollute Res Int. 2003;10:192–198.
  • Colborn T, vom Saal FS, Soto AM. Developmental effects of endocrine-disrupting chemicals in wildlife and humans. Environ Health Perspect. 1993;101:378–384.
  • Adegunlola JG, Afolabi OK, Akhigbe RE, et al. Lipid peroxidation in brain tissue following administration of low and high doses of arsenite and L-ascorbate in Wistar strain rats. Toxicol Int. 2012;19:47–50.
  • Ige SF, Akhigbe RE. Common onion (Allium cepa) extract reverses cadmium-induced organ toxicity and dyslipidaemia via redox alteration in rats. Pathophysiology. 2013;20:269–274.
  • Ige SF, Akhigbe RE, Adewale AA, et al. Effect of allium cepa (Onion) extract on cadmium–induced nephrotoxicity in rats. Kidney Res J. 2011a;1(1):41–47.
  • Ige SF, Akhigbe RE, Edeogho O, et al. Hepatoprotective activities of allium cepa in cadmium-treated rats. Int J Pharm Pharm Sci. 2011;3(5):60–63.
  • Ige SF, Akhigbe RE. The role of Allium cepa on aluminum-induced reproductive dysfunction in experimental male rat models. J Hum Reprod Sci. 2012b;5:200–205.
  • Ige SF, Olaleye SB, Akhigbe RE, et al. Testicular toxicity and sperm quality following cadmium exposure in rats: Ameliorative potentials of Allium cepa. J Hum Reprod Sci. 2012a;5:37–42.
  • Gupta RK, Archambeault DR, Yao HH-C. Genetic mouse models for female reproductive toxicology studies. Compr Toxicol. 2010;11:561–575.
  • Rhomberg LR, Seeley M, Verslycke T, et al. Environmental hormone disruptors. Encyclopedia Toxicol. 2014:378–380.
  • Vigeh M, Smith DR, Hsu PC. How does lead induce male infertility? Iran J Reprod Med. 2011;9:1–8.
  • Winder C. Lead, reproduction and development. NeuroToxicology. 1993;14:303–317.
  • Xuezhi J, Youxin L, Yilan W. Studies of lead exposure on reproductive system: A review of work in China. Biomed Environ Sci. 1992;5:266–275.
  • Pizent A, Tariba B, Živković T. Reproductive toxicity of metals in Men. Arch Ind Hyg Toxicol. 2012;63:35–46.
  • Gandhi J, Hernandez RJ, Chen A, et al. Impaired hypothalamic-pituitary-testicular axis activity, spermatogenesis, and sperm function promote infertility in males with lead poisoning. Zygote. 2017;25:103–110.
  • Apostoli P, Kiss P, Porru S, et al. Male reproductive toxicity of lead in animals and humans. ASCLEPIOS Study Group. Occup Environ Med. 1998;55:364–374.
  • Alexander BH, Checkoway H, Faustman EM, et al. Contrasting associations of blood and semen lead concentrations with semen quality among lead smelter workers. Am J Ind Med. 1998;34:464–469.
  • Bentaiba K, Belhocine M, Chugrani F, et al. Effectiveness of Withania frutescens root extract on testicular damage induced by lead acetate in adult albino rats. Reprod Toxicol. 2023;115:102–110.
  • Elsheikh NAH, Omer NA, Yi-Ru W, et al. Protective effect of betaine against lead-induced testicular toxicity in male mice. Andrologia. 2020;52:e13600.
  • Farmand F, Ehdaie A, Roberts CK, et al. Lead-induced dysregulation of superoxide dismutases, catalase, glutathione peroxidase, and guanylate cyclase. Environ Res. 2005;98:33–39.
  • Kasperczyk S, Kasperczyk A, Ostalowska A, et al. Activity of glutathione peroxidase, glutathione reductase, and lipid peroxidation in erythrocytes in workers exposed to lead. Biol Trace Elem Res. 2004;102:61–72.
  • Martinez-Haro M, Green AJ, Mateo R. Effects of lead exposure on oxidative stress biomarkers and plasma biochemistry in waterbirds in the field. Environ Res. 2011;111(4):530–538.
  • Wang L, Wang H, Hu M, et al. Oxidative stress and apoptotic changes in primary cultures of rat proximal tubular cells exposed to lead. Arch Toxicol. 2009;83:417–427.
  • Vaziri ND, Lin C-Y, Farmand F, et al. Superoxide dismutase, catalase, glutathione peroxidase and NADPH oxidase in lead-induced hypertension. Kidney Int. 2003;63:186–194.
  • Vaziri ND, Ding Y. Effect of lead on nitric oxide synthase expression in coronary endothelial cells. Role of superoxide. Hypertension. 2001;37(2I):223–226.
  • Fallah A, Mohammad-Hasani A, Colagar AH. Zinc is an essential element for male fertility: A review of Zn roles in men’s health, germination, sperm quality, and fertilization. J Reprod Infertil. 2018;19:69.
  • Jurowski K, Szewczyk B. Biological consequences of zinc deficiency in the pathomechanisms of selected diseases. J Biol Inorg Chem. 2014;19:1069–1079.
  • Roshanravan N, Alizadeh M. Effect of zinc supplementation on insulin resistance, energy and macronutrients intakes in pregnant women with impaired glucose tolerance. Iran J Public Health. 2015;44:211–217.
  • Chasapis CT, Loutsidou AC. Zinc and human health: An update. Arch Toxicol. 2012;86:521–534.
  • Ruz M, Carrasco F. Zinc as a potential coadjuvant in therapy for type 2 diabetes. Food Nutr Bull. 2013;34:215–221.
  • Foster M, Chu A. Zinc transporter gene expression and glycemic control in post-menopausal women with type 2 diabetes mellitus. J Trace Elem Med Biol. 2014;28:448–452.
  • Jansen J, Rosenkranz E. Disturbed zinc homeostasis in diabetic patients by in vitro and in vivo analysis of insulinomimetic activity of zinc. J Nutr Biochem. 2012;23:1458–1466.
  • Akhigbe RE, Hamed MA, Odetayo AF, et al. Zinc improves sexual and erectile function in HAART-treated rats via the upregulation of erectogenic enzymes and maintenance of redox balance. Aging Male. 2023;26(1):2205517.
  • Rafique M, Naheed K, Khalida P, et al. The effects of lead and zinc on the quality of semen of albino rats. J Coll Physicians Surg Pak. 2009;19:510–513.
  • Adeyemi DH, Odetayo AF, Hamed MA, et al. Impact of COVID 19 on erectile function. Aging Male. 2022;25(1):202–216.
  • Ajayi AF, Akhigbe RE. Assessment of sexual behaviour and fertility indices in male rabbits following chronic codeine use. Andrology. 2020a;8:509–515.
  • Hamed MA, Akhigbe RE, Aremu AO, et al. Zinc normalizes hepatic lipid handling via modulation of ADA/XO/UA pathway and caspase 3 signaling in highly active antiretroviral therapy-treated Wistar rats. Chem-Biol Interact. 2022a;368:110233.
  • Kelainy EG, Laila IM, Ibrahim SR. The effect of ferulic acid against lead-induced oxidative stress and DNA damage in kidney and testes of rats. Environ Sci Pollut Res. 2019;26(3):31675–31684.
  • Vukelic D, Djordjevic AB, Andelkovic M, et al. Derivation of benchmark doses for male reproductive toxicity in a subacute low-levl Pb exposure model in rats. Toxicol Lett. 2023;375:69–76.
  • Akhigbe RE, Afolabi OA, Ajayi AF. L-Arginine reverses maternal and pre-pubertal codeine exposure-induced sexual dysfunction via upregulation of androgen receptor gene and NO/cGMP signaling. PLoS ONE. 2022;17(9):e0274411.
  • Ajayi AF, Akhigbe RE. Staging of the estrous cycle and induction of estrus in experimental rodents: an update. Fertil Res Pract. 2020b;6:5.
  • Akhigbe RE, Hamed MA, Odetayo AF. HAART and anti-Koch’s impair sexual competence, sperm quality and offspring quality when used singly and in combination in male Wistar rats. Andrologia. 2021a;53(2):e13951.
  • Hamed MA, Akhigbe TM, Akhigbe RE, et al. Glutamine restores testicular glutathione-dependent antioxidant defense and upregulates NO/cGMP signaling in sleep deprivation-induced reproductive dysfunction in rats. Biomed Pharmacother. 2022b;148:112765.
  • Oluwole DT, Akhigbe RE, Ajayi AF. Rohypnol-induced sexual dysfunction is via suppression of hypothalamic-pituitary-testicular axis: An experimental study in rats. Andrologia. 2020;53(2):e13931.
  • Zhang Z, Yu J, Xie J, et al. Improvement roles of zinc supplementation in low dose lead-induced testicular damage and glycolytic inhibition in mice. Toxicology. 2021;462:152933.
  • Akhigbe RE, Hamed MA. Co-administration of HAART and Antikoch triggers cardiometabolic dysfunction through an oxidative stress-mediated pathway. Lipids Health Dis. 2021;20:62.
  • Fossati P, Prencipe L, Berti G. Use of 3, 5-dichloro-2-hydroxybenzenesulfonic acid/4-aminophenazone chromogenic system in direct enzymic assay of uric acid in serum and urine. Clin Chem. 1980;26(/2):227–231.
  • Akhigbe R, Ajayi A. Testicular toxicity following chronic codeine administration is via oxidative DNA damage and up-regulation of NO/TNF-α and caspase 3 activities. Plos One. 2020;15(3):e0224052.
  • Beutler E, Duron O, Kelly BM. Improved method for the determination of blood glutathione. J Lab Clin Med. 1963;61:882–888.
  • Euler HV, Josephson K. Uber katalase. I Eur J Org Chem. 1972;452:158–181.
  • Fridovich I, Misra HP. The role of superoxide anion in the autooxidation of epinephrine and a simple assay for superoxide dismutase. J Biol Chem. 1972;247:3170–3175.
  • Rotruck JT, Pope AL, Ganther HE, et al. Selenium: biochemical role as a component of glutathione peroxidase. Science. 1973;179(4073):588–590.
  • Habig WH, Pabst MJ, Glutathione-S-transferases JW. The first enzymatic step in mercapturic acid formation. J Biol Chem. 1974;249(22):7130–7139.
  • Lizza EF, Rosen RC. Definition and classification of erectile dysfunction: report of the nomenclature committee of the international society of impotence research. Int J Impot Res. 1999;11:141.
  • Akhigbe RE, Hamed MA, Aremu AO. HAART exacerbates testicular damage and impaired spermatogenesis in anti-Koch-treated rats via dysregulation of lactate transport and glutathione content. Reprod Toxicol. 2021b;103:96–107.
  • Yakubu MT, Atoyebi AR. Brysocarpus coccineus (Schum & Thonn) root reinstates sexual competence and testicular function in paroxetine-induced sexual dysfunction in male Wistar rats. Andrologia. 2018;50(4):e12980.
  • Yakubu MT, Afolayan AJ. Effect of aqueous extract of Bulbine natalensis (Baker) stems on the sexual behaviour of male rats. Int J Androl. 2009;32:629–636.
  • Yakubu MT, Akanji MA. Effect of aqueous extract of Massularia acuminata stem on sexual behaviour of male Wistar rats. Evidence-Based Complement Alternate Med. 2011;2011. Article ID 738103.
  • Tajuddin A, Ahmad S, Latif A, et al. Effect of 50% ethanolic extract of Syzygium aromaticum (L.) Merr and Perry. (clove) On the sexual behaviour of normal male rats. BMC Complement Altern Med. 2003;3:6.
  • Akhigbe RE, Dutta S, Sengupta P, et al. Adropin in immune and energy balance: ‘a molecule of interest’ in male reproduction. Chem Biol Lett. 2021c;8(4):213–223.
  • Beckman JS, Koppenol WH. Nitric oxide, superoxide, and peroxynitrite: The good, the bad, and ugly. Am J Physiol. 1996;71:1424–1437.
  • Ademosun AO, Adebayo AA, Oboh G. Anogeissus leiocarpus attenuates paroxetine-induced erectile dysfunction in male rats via enhanced sexual behavior, nitric oxide level and antioxidant status. Biomed Pharmacother. 2019;111:1029–1035.
  • Oyeleye SI, Adefegha SA, Dada FA, et al. Effect of p-coumaric acid on the erectogenic enzyme activities and non-protein thiol level in the penile tissue of normal and doxorubicin-induced oxidative stress male rat. Andrologia. 2019;51(6):e13281.
  • da Silva CB, Wolkmer P, da Silva AS, et al. Cholinesterases as markers of the inflammatory process in rats infected with Leptospira interrogans serovar Icterohaemorrhagiae. J Med Microbiol. 2012;61(2):278–284.
  • Kenia PN, Clinton WR. Mechanisms in erectile function and dysfunction: an overview. In: K Nunes, editor. Erectile dysfunction – Disease-associated mechanisms and novel insights into therapy 2012; (Vol. 1). Zagreb, Croatia: In Tech. p. 3–22.
  • Akhigbe R, Ajayi A. The impact of reactive oxygen species in the development of cardiometabolic disorders: a review. Lipids Health Dis. 2021;20:23.
  • Li Z, Shen Y, Chen Y, et al. High uric acid inhibits cardiomyocyte viability through the ERK/P38 pathway via oxidative stress. Cell Physiol Biochem. 2018;45:156–1164.
  • Akhigbe RE. Discordant results in plant toxicity studies in Africa: attempt of standardization. In: K Victor, editor. Toxicological survey of African medicinal plants. Waltham, MA: Elsevier Inc.; 2014. p. 53–61.
  • Sayre LM, Smith MA, Perry G. Chemistry and biochemistry of oxidative stress in neurodegenerative disease. Curr Med Chem. 2001;8:721.
  • Mohamad NV, Wong SK, Wan Hasan WN, et al. The relationship between circulating testosterone and inflammatory cytokines in men. Aging Male. 2019;22(2):129–140.
  • Aitken RJ. COVID-19 and human spermatozoa— potential risks for infertility and sexual transmission? Andrology: 12859. 2021;9(1):48–52.
  • Dehghan SF, Mehrifar Y, Ardalan A. The relationship between exposure to lead-containing welding fumes and the levels of reproductive hormones. Ann Glob Health. 2019;85(1):125, 1–6.
  • Mokhtari M, Zoboori M. The effects of lead acetate on sexual behaviour and the level of testosterone in adult male rats. Int J Fertility Sterility. 2011;5:13–20.
  • Mulligan T, Schmitt B. Testosterone for erectile failure. J Gen Intern Med. 1993;8:517.
  • Beyer C, Gonzalez-Mariscal G. Effects of sex steroids on sensory and motor spinal mechanisms. Psychoneuroendocrinology. 1994;19:517.
  • Hull EM, Muschamp JW, Sato S. Dopamine and serotonin: influences on male sexual behavior. Physiol Behav. 2004;83(2):291.
  • Cortese MM, Suschek CV, Wetzel W, et al. Zinc protects endothelial cells from hydrogen peroxide via Nrf2-dependent stimulation of glutathione biosynthesis. Free Radical Biol Med. 2008;44:2002–2012.
  • Ha K-H, Chen Y, Cai J, et al. Increased Glutathione synthesis through an ARE-Nrf2–dependent pathway by Zinc in the RPE: implication for protection against Oxidative Stress. IOVS. 2006;47(6):2709–2715.
  • Li B, Cui W, Tan Y, et al. Zinc is essential for the transcription function of Nrf2 in human renal tubule cells in vitro and mouse kidney in vivo under the diabetic condition. J Cell Mol Med. 2014;18(5):895–906.