Potential of Zingiber officinale Extract against Chlorpyrifos Toxicity in Caprine Testicular Cells

Jump To References Section

Authors

  • Reproductive Physiology Laboratory, Department of Zoology, Kurukshetra University, Kurukshetra – 136119, Haryana ,IN
  • Reproductive Physiology Laboratory, Department of Zoology, Kurukshetra University, Kurukshetra – 136119, Haryana ,IN
  • Department of Biochemistry, Sri Venkateshvara College, University of Delhi, Delhi – 110021 ,IN
  • Department of Library and Information Science, Kurukshetra University, Kurukshetra – 136119, Haryana ,IN
  • Reproductive Physiology Laboratory, Department of Zoology, Kurukshetra University, Kurukshetra – 136119, Haryana ,IN

DOI:

https://doi.org/10.18311/ti/2022/v29i4/29728

Keywords:

Apoptosis, Chlorpyrifos, Testicular Cells, Zingiber officinale.
Toxicology

Abstract

Chlorpyrifos, an organophosphate insecticide, is currently under studies due to its reproductive toxicity in non-targeted organisms. The present study on its efficacy as reproductive toxicant, revealed its effect on testicular cells of caprines. CPF was found to decrease cell viability in testicular cells, induced histo-architectural alterations like loss of tubular interactions and empty luminal spaces in testis; initiated DNA damage associated apoptosis as revealed by MTT, Histology and EB/AO fluorescence assay respectively, at both the doses (10 and 50 μM). CPF mediated toxicity induced oxidative stress in testicular cells as revealed by dose- and time-dependent increase in lipid peroxidation and decline in ferric reducing antioxidant power. However, supplementation of Zingiber officinale (ginger) extract reduced the CPF mediated decrease in cell viability, histological defects, incidence of apoptosis, and generated oxidative stress parameters; thus, proving its potential as a therapeutic agent against CPF, preventing testicular cells toxicity mediated reproductive disorders.

Downloads

Download data is not yet available.

Published

2023-02-07

How to Cite

Saraf, P., Panchal, H., Sharma, K., Sharma, S., & Bhardwaj, J. K. (2023). Potential of <i>Zingiber officinale</i> Extract against Chlorpyrifos Toxicity in Caprine Testicular Cells. Toxicology International, 29(4), 465–473. https://doi.org/10.18311/ti/2022/v29i4/29728
Received 2022-03-05
Accepted 2022-05-06
Published 2023-02-07

 

References

Anderson HR, Vinggaard AM, Rasmussen TH, Gjermandsen IM, Bonefeld-Jørgensen EC. Effects of currently used pesticides in assays for estrogenicity, androgenicity and aromatase activity in vitro. Toxicol Appl Pharmacol. 2002; 179:1–12. PMid: 11884232. https://doi.org/10.1006/taap.2001.9347

Bhardwaj JK, Saraf P. Granulosa cell apoptosis by impairing antioxidant defense system and cellular integrity in caprine antral follicles post malathion exposure. Environ Toxicol. 2016; 31(12):1944–54. PMid: 26420608. https://doi.org/10.1002/tox.22195

Bustos-Obregon EL, Gonzalez-Hormazabal P. Effect of a single dose of malathion on spermatogenesis in mice. Asian J Androl. 2003; 5(2):105–17.

Bhardwaj JK, Saraf P, Kumari P, Mittal M, Kumar V. N-Acetyl-cysteine mediated inhibition of spermatogonial cells apoptosis against malathion exposure in testicular tissue. J Biochem Mol Toxicol, 2018a; 32(4):e22046. PMid: 29457669. https://doi.org/10.1002/jbt.22046

Pina-Guzman B, Solis-Heredia MJ, Quintanilla-Vega B. Diazinon alters sperm chromatin structure in mice by phosphorylating nuclear protamines. Toxicol Appl Pharmacol. 2005; 15:189–98. PMid: 15629194. https://doi.org/10.1016/j.taap.2004.06.028

El-Mazoudy RH, Attia AA. Endocrine-disrupting and cytotoxic potential of anticholinesterase insecticide, diazinon in reproductive toxicity of male mice. J Hazard Mate. 2012; 210:111–20. PMid: 22284168. https://doi.org/10.1016/j.jhazmat.2011.12.073

Farag AT, Radwan MH, Sorour F, Okazy AE, Agamy S, Khaliek El-Sebae A. Chlorpyrifos induced reproductive toxicity in male mice. Reprod Toxicol. 2010; 29(1):80–5. PMid: 19850121. https://doi.org/10.1016/j.reprotox.2009.10.003

Bhardwaj JK, Saraf P. N-acetyl Cysteine-Mediated effective attenuation of methoxychlor-induced granulosa cell apoptosis by counteracting reactive oxygen species generation in caprine ovary. Environ Toxicol. 2017; 32(1):156–66. PMid: 26635070. https://doi.org/10.1002/tox.22221

Bhardwaj JK, Kumar V, Saraf P, Kumara P, Mittal M. Current status and changing national scenario of goat population: A review. Agricul Rev. 2018b; 39(2):91–103. https://doi.org/10.18805/ag.-1752

Al-Gubory KH. Environmental pollutants and lifestyle factors induce oxidative stress and poor prenatal development. Reprod Biomed Online. 2014; 29:17–31. PMid: 24813750. https://doi.org/10.1016/j.rbmo.2014.03.002

Haneia IM, Hasan AA, Reda HE, Wessam MA, Mona IS. Effect of methomyl on fertility, embryotoxicity and physiological parameters in female rat. J Appl Pharm Sci. 2013; 3(12):109–19.

Bhardwaj JK, Mittal M, Saraf P, Kumari P. Pesticides induced oxidative stress and female infertility: a review. Toxin Reviews. 2018c; 39(3):1–13. https://doi.org/10.1080/15569543.2018.1474926

Bhardwaj JK, Saraf P. Malathion induced granulosa cell apoptosis in caprine antral follicles: An ultrastructural and flow cytometric analysis. Microsc Microanal. 2014; 20(6):1861–8. PMid: 25409908. https://doi.org/10.1017/S1431927614013452

Pearse, AGE. Histochemistry. Theoretical and applied, 1968; (2 vols), London: Churchill.

Buege JA, Aust SD. Microsomal lipid peroxidation. Meth Enzymol. 1978; 51:302–10. PMid: 672633. https://doi.org/10.1016/S0076-6879(78)52032-6

Benzie IFF, Strain JJ. Ferric Reducing Ability of Plasma (FRAP) as a measure of antioxidant power: The FRAP assay. Anal Biochem. 1996; 239:70–6. PMid: 8660627. https://doi.org/10.1006/abio.1996.0292

Ricci JE, Munoz-Pinedo C, Fitzgerald P, Bailly-Maitre B, Perkins GA, Yadava N, Scheffler IE, Ellisman MH, Green DR. Disruption of mitochondrial function during Apoptosis is mediated by caspase cleavage of the p75 subunit of Complex 1 of the electron transport chain. Cell. 2004; 117:773–86. PMid: 15186778. https://doi.org/10.1016/j.cell.2004.05.008

Tsai-Turton M, Luong BT, Tan Y, Luderer U. Cyclophosphamide-induced apoptosis in COV434 human granulosa cells involves oxidative stress and glutathione depletion. Toxicol Sci. 2007; 98(1):216–30. PMid: 17434952. https://doi.org/10.1093/toxsci/kfm087

Shi YQ, Wang Y, Song Y, Li HW, Liu CJ, Wu ZG, Yang K. P,p’-DDE induces testicular apoptosis in prepubertal rats via the Fas/FasL pathway. Toxicol Lett. 2010; 193:79–85. PMid: 20025943. https://doi.org/10.1016/j.toxlet.2009.12.008

Nafisi S, Saboury AK, Keramata N, Neaultc JF, Tajmir-Riahic HA. Stability and structural features of DNA intercalation with ethidium bromide, acridine orange and methylene blue. J Mol Struct. 2007; 827:35–43. https://doi.org/10.1016/j.molstruc.2006.05.004

Banerjee S, Banerjee S, Saraswat G, Bandyopadhyay SA, Kabir SN. Female reproductive aging is master-planned at the level of ovary. PLoS ONE. 2014; 9:e96210. PMid: 24788203 PMCid: PMC4008600. https://doi.org/10.1371/journal.pone.0096210

Li R, Wei M, Shao J. Effects of verapamil on the immediate-early gene expression of bone marrow mesenchymal stem cells stimulated by mechanical strain in vitro. Med Sci Monit Basic Res. 2013; 19:68–75. PMid: 23435320 PMCid: PMC3638684. https://doi.org/10.12659/MSMBR.883790

Liu K, Liu P, Liu R, Wu X. Dual AO/EB staining to detect apoptosis in Osteosarcoma cells compared with flow cytometry. Med Sci Monit Basic Res. 2015; 21:15–20. PMid: 25664686 PMCid: PMC4332266. https://doi.org/10.12659/MSMBR.893327

Escobar ML, Echeverria OM, Casasa AS, Garcia G, Aguilar SJ, Vazquez-Nin GH. Involvement of pro-apop-totic and pro-autophagic proteins in granulosa cell death. Cell Biol. 2013; 1(1):9–17. https://doi.org/10.11648/j.cb.20130101.12

Devine PJ, Perreault SD, Luderer U. Roles of reactive oxygen species and antioxidants in ovarian toxicity. Biol Reprod. 2012; 86(2):27. PMid: 22034525 PMCid: PMC3290661. https://doi.org/10.1095/biolreprod.111.095224

Das UN. Essential fatty acids, lipid peroxidation and apoptosis. Prostaglandins Leukot Essent Fatty Acids. 1999; 61:157–63. PMid: 10582655. https://doi.org/10.1054/plef.1999.0085

Mahfouz R, Sharma R, Thiyagarajan A, Kale V, Gupta S, Sabanegh E, Agarwal A. Semen characteristics and sperm DNA fragmentation in infertile men with low and high levels of seminal reactive oxygen species. Fertil Steril. 2010; 94:2141–6. PMid: 20117780. https://doi.org/10.1016/j.fertnstert.2009.12.030

Riaz H, Begum A, Raza SA, Khan ZM, Yousaf H, Iariq A. Antimicrobial property and phytochemical study of ginger found in local area of Punjab, Pakistan. Int Curr Pharm J. 2015; 4(7):405–9. https://doi.org/10.3329/icpj.v4i7.23591

De Lima, RMT, Dos Reis AC, de Menezes APM, Santos JVO, Filho JWGO, Ferreira JRO, de Alencar MVOB, da Mata AMOF, Khan IN, Islam A, Uddin SJ, Ali ES, Islam MT, Tripathi S, Mishra SK, Mubarak MS, Melo-Cavalcante AAC. Protective and therapeutic potential of ginger (Zingiber officinale) extract and [6]-gingerol in cancer: A comprehensive review. Phytother Res. 2018; 32(10):1885–907. PMid: 30009484. https://doi.org/10.1002/ptr.6134

Abarikwu SO. Protective effect of quercetin on atrazineinduced oxidative stress in the liver, kidney, brain, and heart of adult Wistar rats. Toxicol Int. 2014; 21:148–55. PMid: 25253923 PMCid: PMC4170555. https://doi.org/10.4103/0971-6580.139794

Jain J, Hasan W, Yadav RS, Jat D. Protective Effects of Quercetin against Rotenone Induced Histopathological and Biochemical Alteration in Testes of Mice. Toxicol Int. 2021; 28:57–65.

Bhardwaj JK, Kumar V, Saraf P, Panchal H, Rathee V, Sachdeva SN. Efficacy of N-acetyl-L-cysteine against glyphosate induced oxidative stress and apoptosis in testicular germ cells preventing infertility. J Biochem Mol Toxicol. 2021; e22979.

Bhardwaj JK, Kumar V, Panchal H, Sachdeva SN. Transmission electron microscopic analysis of glyphosate induced cytotoxicity and its attenuation by N-acetyl-L-cysteine in caprine testicular germ cells in vitro. Ultrastruct Pathol. 2021; 45(6):407–13. PMid: 34698588. https://doi.org/10.1080/01913123.2021.1993 400

Bhardwaj JK, Kumari P, Saraf P, Yadav AS. Antiapoptotic effects of vitamin C and E against Cypermethrin induced oxidative stress and spermatogonial germ cell apoptosis. J Biochem Mol Toxicol. 2018; 32(8):e22174. PMid: 29975445. https://doi.org/10.1002/jbt.22174

Kumar P, Duhan M, Kadyan K, Bhardwaj JK, Saraf P, Mittal M. Multicomponent synthesis of some molecular hybrid containing thiazolepyrazole as apoptosis inducer. Drug Res. 2018; 68:72–9. PMid: 28910831. https://doi.org/10.1055/s-0043-116947

Bhardwaj JK, Kumari P, Saraf P, Mittal M, Yadav AS. Male infertility owing to pesticide poisoning and anti-oxidant induced amelioration: A review. J Cell Tissue Res, 2017; 17(3):6307–14.

Phogat A, Singh J, Kumar V, Malik N. Toxicity of the acetamiprid insecticide for mammals: A review. Environ Chem Letters. 2022; 20:1453–78. https://doi.org/10.1007/s10311-021-01353-1

Sharma RK, Bhat RA, Goyal AK, Bhardwaj JK. Germ cells apoptosis during spermatogenesis in mammals. J Entom Zoo Stud. 2015; 3(3):506–15.