Effect of melatonin on male offspring testis and sperm parameters in BALB/c mice after exposing their mother to METHamphetamine during pregnancy and lactation

Document Type : Original Article

Authors

1 Department of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran

2 Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran

3 Applied Biomedical Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran

4 Medical Genetics and Molecular Medicine Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran

Abstract

Objective(s): Methamphetamine (METH) is a psychostimulant that has harmful effects on all organs,  the nervous system, cardiovascular system, and reproductive system. Since many METH consumers are young people of reproductive age, it poses a risk to the next generation of METH consumers. METH can pass through the placenta and is also secreted into breast milk. Melatonin (MLT) is the primary hormone of the pineal gland that regulates the circadian cycle, and it is also an antioxidant that can mitigate the effects of toxic substances. This study aims to investigate the protective effect of melatonin against the detrimental effects that METH has on the reproductive system of male newborns, whose mothers consumed METH during pregnancy and lactation.
Materials and Methods: In the current study, 30 female adult balb/c mice were divided into three groups: control group, vehicle group that received normal saline, and the experimental group that received 5 mg/kg METH intraperitoneally during gestation and lactation. After lactation, the male offspring of each group were randomly divided into two subgroups, one of which received 10 mg/kg melatonin intragastrically for 21 days (corresponding to the lactation period of the mice) (METH-MLT) and the other did not (METH -D.W). After treatment, the mice were sacrificed and testicular tissue and epididymis were obtained for the following tests.
Results: The diameter of seminiferous tubules, SOD activity, total Thiol groups concentration, catalase activity, sperm count, and PCNA and CCND gene expression were significantly increased in the METH-MLT group compared with the METH-DW. Apoptotic cells and MDA level ameliorated in the METH-MLT group compared with METH-D.W, and testicular weight had no notable change.
Conclusion: The current study represents that consumption of METH during pregnancy and lactation can have adverse effects on the histological and biochemical factors of testis and sperm parameters of male newborns,  which can be mitigated by taking melatonin after the end of the breastfeeding period.

Keywords

Main Subjects


1. Vearrier D, Greenberg MI, Miller SN, Okaneku JT, Haggerty DA. Methamphetamine: History, pathophysiology, adverse health effects, current trends, and hazards associated with the clandestine manufacture of methamphetamine. Dis Mon 2012;58:38-89.
2. Chomchai C, Chomchai S. Global patterns of methamphetamine use. Curr Opin Psychiatry 2015;28:269-274.
3. Sabour M, Khoradmehr A, Kalantar SM, Danafar AH, Omidi M, Halvaei I, et al. Administration of high dose of methamphetamine has detrimental effects on sperm parameters and DNA integrity in mice. Int J Reprod Biomed 2017;15:161-168.
4. Nudmamud‐Thanoi S, Thanoi S. Methamphetamine induces abnormal sperm morphology, low sperm concentration and apoptosis in the testis of male rats. Andrologia 2011;43:278-282.
5. Saberi A, Sepehri G, Safi Z, Razavi B, Jahandari F, Divsalar K, et al. Effects of methamphetamine on testes histopathology and spermatogenesis indices of adult male rats. Addict Health 2017;9:199-205.
6. Nudmamud-Thanoi S, Sueudom W, Tangsrisakda N, Thanoi S. Changes of sperm quality and hormone receptors in the rat testis after exposure to methamphetamine. Drug Chem Toxicol 2016;39:432-438.
7. Yamamoto Y, Yamamoto K, Hayase T, Abiru H, Shiota K, Mori C. Methamphetamine induces apoptosis in seminiferous tubules in male mice testis. Toxicol Appl Pharmacol 2002;178:155-160.
8. Krasnova IN, Cadet JL. Methamphetamine toxicity and messengers of death. Brain Res Rev 2009;60:379-407.
9. Alavi SH, Taghavi MM, Moallem SA. Evaluation of effects of methamphetamine repeated dosing on proliferation and apoptosis of rat germ cells. Syst Biol Reprod Med 2008;54:85-91.
10. Dinger J, Hinner P, Reichert J, Rüdiger M. Methamphetamine consumption during pregnancy-effects on child health. Pharmacopsychiatry 2017;50:107-113.
11. Chomchai C, Chomchai S, Kitsommart R. Transfer of methamphetamine (MA) into breast milk and urine of postpartum women who smoked MA tablets during pregnancy: Implications for initiation of breastfeeding. J Hum Lact 2016;32:333-339.
12. Baei F, Rajabzadeh A, Bagheri J, Jalayeri Z, Ebrahimzadeh-Bideskan A. Effect of methamphetamine exposure during pregnancy and lactation on polysialic acid-neural cell adhesion molecule expression in rat’s offspring hippocampus. Metab Brain Dis 2017;32:991-1002.
13. Tordjman S, Chokron S, Delorme R, Charrier A, Bellissant E, Jaafari N, et al. Melatonin: Pharmacology, functions and therapeutic benefits. Curr neuropharmacol 2017;15:434-343.
14. Li R, Luo X, Li L, Peng Q, Yang Y, Zhao L, et al. The protective effects of melatonin against oxidative stress and inflammation induced by acute cadmium exposure in mice testis. Biol Trace Elem Res 2016;170:152-164.
15. Li C, Zhou X. Melatonin and male reproduction. Clin Chim Acta 2015;446:175-80.
16. Wang L, Qu G, Dong X, Huang K, Kumar M, Ji L, et al. Long-term effects of methamphetamine exposure in adolescent mice on the future ovarian reserve in adulthood. Toxicol Lett 2016;242:1-8.
17. Mei Q, Diao L, Xu Jm, Liu Xc, Jin J. A protective effect of melatonin on intestinal permeability is induced by diclofenac via regulation of mitochondrial function in mice. Acta Pharmacol Sin 2011;32:495-502.
18. Karimi S, Hejazian SH, Alikhani V, Hosseini M. The effects of tamoxifen on spatial and nonspatial learning and memory impairments induced by scopolamine and the brain tissues oxidative damage in ovariectomized rats. Adv Biomed Res 2015;4:196-203.
19. Kei S. Serum lipid peroxide in cerebrovascular disorders determined by a new colorimetric method. Clin Chim Acta 1978;90:37-43.
20. Asgharzadeh F, Bargi R, Beheshti F, Hosseini M, Farzadnia M, Khazaei M. Thymoquinone prevents myocardial and perivascular fibrosis induced by chronic lipopolysaccharide exposure in male rats:-thymoquinone and cardiac fibrosis. J Pharmacopuncture 2018;21:284-293.
21. Gaharwar US, Meena R, Rajamani P. Iron oxide nanoparticles induced cytotoxicity, oxidative stress and DNA damage in lymphocytes. J Appl Toxicol 2017;37:1232-1244.
22. Khoshbakht S, Motejaded F, Karimi S, Jalilvand N, Ebrahimzadeh-Bideskan A. Protective effects of selenium on electromagnetic field-induced apoptosis, aromatase P450 activity, and leptin receptor expression in rat testis. Iran J Basic Med Sci 2021;24:322-330.
23. Jalilvand N, Hosseini M, Beheshti F, Ebrahimzadeh-Bideskan A. Protective effect of PPARγ agonist pioglitazone, on testicular tissue and sperm parameters in hypothyroid rats. Toxin Rev 2019;40:267-276.
24. Park Y-S, Lee S-H, Choi HW, Lee HS, Lee JS, Seo JT. Abnormal human sperm parameters contribute to sperm DNA fragmentation in men with varicocele. World J Mens Health 2018;36:239-247.
25. Boustan A, Mosaffa F, Jahangiri R, Heidarian-Miri H, Dahmardeh-Ghalehno A, Jamialahmadi K. Role of SALL4 and Nodal in the prognosis and tamoxifen resistance of estrogen receptor-positive breast cancer. Mol Biol Res Commun 2021;10:109-119.
26. Alamolhodaei NS, Rashidpour H, Gharaee ME, Behravan J, Mosaffa F. Overexpression of ABCC2 and NF-Κβ/p65 with reduction in cisplatin and 4OH-tamoxifen sensitivity in MCF-7 breast cancer cells: The influence of TNF-α. Pharm Sci 2020;26:150-158.
27. Hardeland R, Pandi-Perumal SR, Cardinali DP. Melatonin. Int J Biochem Cell Biol 2006;38:313-316.
28. Açikgöz O, Gönenç S, Kayatekin BM, Uysal N, Pekçetin Ç, Şemin İ, et al. Methamphetamine causes lipid peroxidation and an increase in superoxide dismutase activity in the rat striatum. Brain Res 1998;813:200-202.
29. Hotchkiss RS, Strasser A, McDunn JE, Swanson PE. Cell death. N Engl J Med 2009;361:1570-1583.
30. Finaud J, Lac G, Filaire E. Oxidative stress. Sport Med 2006;36:327-358.
31. Espino J, Bejarano I, Redondo PC, Rosado JA, Barriga C, Reiter RJ, et al. Melatonin reduces apoptosis induced by calcium signaling in human leukocytes: Evidence for the involvement of mitochondria and bax activation. J Membr Biol 2010;233:105-118.
32. Kumaresan A, Das Gupta M, Datta TK, Morrell JM. Sperm DNA integrity and male fertility in farm animals: A review. Front Vet Sci 2020;7:321-335.
33. O’Brien J, Zini A. Sperm DNA integrity and male infertility. Urology 2005;65:16-22.
34. Heidari-Rarani M, Noori A, Ghodousi A. Effects of methamphetamine on pituitary gonadal axis and spermatogenesis in mature male rats. Zahedan J Res Med Sci 2014;16:37-42.
35. Golias C, Charalabopoulos A, Charalabopoulos K. Cell proliferation and cell cycle control: A mini review. Int J Clin Pract 2004;58:1134-1141.
36. Whitfield ML, George LK, Grant GD, Perou CM. Common markers of proliferation. Nat Rev Cancer 2006;6:99-106.
37. Strzalka W, Ziemienowicz A. Proliferating cell nuclear antigen (PCNA): A key factor in DNA replication and cell cycle regulation. Ann Bot 2011;107:1127-1140.
38. Ekthuwapranee K, Sotthibundhu A, Govitrapong P. Melatonin attenuates methamphetamine‐induced inhibition of proliferation of adult rat hippocampal progenitor cells in vitro. J Pineal Res 2015;58:418-428.
39. Zwirska-Korczala K, Jochem J, Adamczyk-Sowa M, Sowa P, Polaniak R, Birkner E, et al. Influence of melatonin on cell proliferation, antioxidative enzyme activities and lipid peroxidation in 3T3-L1 preadipocytes-an in vitro study. J Physiol Pharmacol 2005;56:91-99.
40. Osatd-Rahimi N, Saburi E, Karimi S, Boustan A, Ebrahimzadehbideskan A. The therapeutic effect of melatonin on female offspring ovarian reserve and quality in BALB/c mice after exposing their mother to methamphetamine during pregnancy and lactation. Iran J Basic Med Sci 2023;26:208-215.