DOI QR코드

DOI QR Code

ɴ-Acetylcysteine protects against diazinon-induced histopathological damage and apoptosis in renal tissue of rats

  • Gaiqin Dong (Department of Nephrology and Immunology, Children's Hospital of Soochow University) ;
  • Qingfeng Li (Department of Pediatrics, Affiliated Hospital of Yangzhou University) ;
  • Chun Yu (Department of Pediatrics, Affiliated Hospital of Yangzhou University) ;
  • Qing Wang (Department of Pediatrics, Affiliated Hospital of Yangzhou University) ;
  • Danhua Zuo (Department of Pediatrics, Affiliated Hospital of Yangzhou University) ;
  • Xiaozhong Li (Department of Nephrology and Immunology, Children's Hospital of Soochow University)
  • Received : 2023.08.29
  • Accepted : 2024.01.22
  • Published : 2024.04.15

Abstract

Diazinon (DZN) is a member of organophosphorus insecticides that has cytotoxic effects on different organs. ɴ-Acetyl cysteine (NAC) is a widely used antioxidant in clinical, in vivo and in vitro studies. We evaluated the protective role of NAC against DZN-induced toxicity in kidney tissue of Wistar rats. 30 male Wistar rats were divided into 5 groups of control, single dose of DZN, continuous dose of DZN, single doses of DZN + NAC and continuous doses of DZN + NAC. Kidney function test (blood urea nitrogen, creatinine and uric acid) was provided. Levels of malondialdehyde (MDA), total antioxidant capacity (TAC) and total sulfhydryl (T-SH) were determined in renal tissues. Renal cells apoptosis was detected using TUNEL assay. The mRNA expressions of apoptosis, oxidative stress and inflammatory mediators, including B-cell lymphoma-2 (Bcl2), Bcl-2-associated X protein (Bax), superoxide dismutase (SOD), catalase (CAT), Interleukin 10 (IL-10), Tumor necrosis factor-α (TNF-α), Caspase-3 and Caspase-8 were analyzed in kidney tissues using Real Time PCR method. Chronic exposure to DZN was associated with severe morphological changes in the kidney, as well as impairment of its function and decreased kidney weights. Continues treatment with DZN significantly decreased the percentage of renal apoptotic cells as compared to rats treated with continuous dose of DZN alone (17.69 ± 3.67% vs. 39.46% ± 2.44%; p < 0.001). Continuous exposure to DZN significantly decreased TAC and T-SH contents, as well as SOD and CAT expression, but increased MDA contents in the kidney tissues (p < 0.001). A significant increase was observed in mRNA expression of Bax, Caspase-3, Caspase-8, as well as TNF-α following exposure to DZN, but the expression of IL-10 and Bcl2 was significantly decreased. NAC can protect kidney tissue against DZN-induced toxicity by elevating antioxidants capacity, mitigating oxidative stress, inflammation and apoptosis.

Keywords

Acknowledgement

The authors of this manuscript wish to express their thanks and appreciation to Department of Nephrology and Immunology at Children's Hospital of Soochow University, and Department of Pediatrics at Affiliated Hospital of Yangzhou University, Yangzhou Jiangsu, China.

References

  1. Larkin DJ, Tjeerdema RS (2000) Fate and effects of diazinon. Rev Environ Contam Toxicol 166:49-82
  2. Jokanovic M, Kosanovic M (2010) Neurotoxic effects in patients poisoned with organophosphorus pesticides. Environ Toxicol Pharmacol 29:195-201. https://doi.org/10.1016/j.etap.2010.01.006
  3. Oksay T et al (2013) N-acetyl cysteine attenuates diazinon exposure-induced oxidative stress in rat testis. Andrologia 45:171-177. https://doi.org/10.1111/j.1439-0272.2012.01329.x
  4. El-Shenawy NS et al (2010) Amelioratory effect of vitamin E on organophosphorus insecticide diazinon-induced oxidative stress in mice liver. Pestic Biochem Physiol 96:101-107. https://doi.org/10.1016/j.pestbp.2009.09.008
  5. Harchegani AB et al (2018) Mechanisms of diazinon effects on impaired spermatogenesis and male infertility. Toxicol Ind Health 34:653-664. https://doi.org/10.1177/0748233718778665
  6. Ahmadi-Naji R, Heidarian E, Ghatreh-Samani K (2017) Evaluation of the effects of the hydroalcoholic extract of Terminalia chebula fruits on diazinon-induced liver toxicity and oxidative stress in rats. Avicenna J Phytomed 7:454
  7. Moallem SA et al (2014) Effect of aqueous extract of Crocus sativus L. (saffron) stigma against subacute effect of diazinon on specific biomarkers in rats. Toxicol Ind health 30:141-146. https://doi.org/10.1177/0748233712452609
  8. Tsitsimpikou C et al (2013) Histopathological lesions, oxidative stress and genotoxic effects in liver and kidneys following long term exposure of rabbits to diazinon and propoxur. Toxicology 307:109-114. https://doi.org/10.1016/j.tox.2012.11.002
  9. Colovic MB et al (2015) In vitro evaluation of neurotoxicity potential and oxidative stress responses of diazinon and its degradation products in rat brain synaptosomes. Toxicol Lett 233:29-37. https://doi.org/10.1016/j.toxlet.2015.01.003
  10. Beydilli H et al (2015) Evaluation of the protective effect of silibinin against diazinon induced hepatotoxicity and free-radical damage in rat liver. Iran Red Crescent Med J 17:e25310. https://doi.org/10.5812/ircmj.17(4)2015.25310
  11. Razavi BM et al (2013) Protective effect of crocin on diazinon induced cardiotoxicity in rats in subchronic exposure. Chem Biol Interact 203:547-555. https://doi.org/10.1016/j.cbi.2013.03.010
  12. Aluigi M, Guida C, Falugi C (2010) Apoptosis as a specific biomarker of diazinon toxicity in NTera2-D1 cells. Chem Biol Interact 187:299-303. https://doi.org/10.1016/j.cbi.2010.03.031
  13. Shiri M et al (2016) Blockage of both the extrinsic and intrinsic pathways of diazinon-induced apoptosis in PaTu cells by magnesium oxide and selenium nanoparticles. Int J Nanomed 11:6239. https://doi.org/10.2147/IJN.S119680
  14. Gomez-Lechon MJ et al (2002) Sensitive markers used to identify compounds that trigger apoptosis in cultured hepatocytes. Toxicol Sci 65:299-308. https://doi.org/10.1093/toxsci/65.2.299
  15. Liu X et al (1996) Induction of apoptotic program in cell-free extracts: requirement for dATP and cytochrome c. Cell 86:147-157. https://doi.org/10.1073/pnas.95.16.956
  16. Jaafarzadeh M et al (2022) Protecting effects of N-acetyl cysteine supplementation against lead and cadmium-induced brain toxicity in rat models. Biol Trace Elem Res 200:4395-4403. https://doi.org/10.1007/s12011-021-03034-0
  17. Elmore S (2007) Apoptosis: a review of programmed cell death. Toxicol Pathol 35:495-516. https://doi.org/10.1080/01926230701320337
  18. Caroppi P et al (2009) Apoptosis and human diseases: mitochondrion damage and lethal role of released cytochrome C as proapoptotic protein. Curr Med Chem 16:4058-4065. https://doi.org/10.2174/092986709789378206
  19. Slattery J et al (2015) Clinical trials of ɴ-acetylcysteine in psychiatry and neurology: a systematic review. Neurosci Biobehav Rev 55:294-321. https://doi.org/10.1016/j.neubiorev.2015.04.015
  20. Wong KK et al (2023) The effects of ɴ-acetylcysteine on lung alveolar epithelial cells infected with respiratory syncytial virus. Malays J Pathol 45:43-50
  21. Shah MD, Iqbal M (2010) Diazinon-induced oxidative stress and renal dysfunction in rats. Food Chem Toxicol 48:3345-3353. https://doi.org/10.1016/j.fct.2010.09.003
  22. de Faria Guimaraes LP et al (2014) N-acetyl-cysteine is associated to renal function improvement in patients with nephropathic cystinosis. Pediatr Nephrol 29:1097-1102. https://doi.org/10.1007/s00467-013-2705-3
  23. Ahmadi F et al (2017) Effectiveness of N-acetylcysteine for preserving residual renal function in patients undergoing maintenance hemodialysis: multicenter randomized clinical trial. Clin Exp Nephrol 21:342-349. https://doi.org/10.1007/s10157-016-1277-5
  24. Radomska-Lesniewska DM, Skopinski P (2012) Review paper ɴ-acetylcysteine as an anti-oxidant and anti-inflammatory drug and its some clinical applications. Cent Eur J Immunol 37:57-66
  25. De Andrade KQ et al (2015) Oxidative stress and inflammation in hepatic diseases: therapeutic possibilities of ɴ-acetylcysteine. Int J Mol Sci 16:30269-30308. https://doi.org/10.3390/ijms161226225
  26. de Oliveira Filho LD et al (2015) Effect of N-acetylcysteine in hearts of rats submitted to controlled hemorrhagic shock. Rev Bras Cir Cardiovasc 30:173-181. https://doi.org/10.5935/1678-9741.20140097
  27. Esfahani M et al (2023) Resveratrol: a panacea compound for diazinon-induced renal toxicity. Toxin Reviews 42:40-50. https://doi.org/10.1080/15569543.2021.2008452
  28. Bradford N (1976) A rapid and sensitive method for the quantitation microgram quantities of a protein isolated from red cell membranes. Anal Biochem 72:e254. https://doi.org/10.1006/abio.1976.9999
  29. Benzie IF, Strain J (1999) [2] Ferric reducing/antioxidant power assay: direct measure of total antioxidant activity of biological fluids and modified version for simultaneous measurement of total antioxidant power and ascorbic acid concentration. Methods Enzymol 299:15-27. https://doi.org/10.1016/s0076-6879(99)99005-5
  30. Tietze F (1969) Enzymic method for quantitative determination of nanogram amounts of total and oxidized glutathione: applications to mammalian blood and other tissues. Anal Biochem 27:502-522. https://doi.org/10.1016/0003-2697(69)90064-5
  31. Rush T et al (2010) Mechanisms of chlorpyrifos and diazinon induced neurotoxicity in cortical culture. Neuroscience 166:899-906. https://doi.org/10.1016/j.neuroscience.2010.01.025
  32. Lari P et al (2015) Evaluation of diazinon-induced hepatotoxicity and protective effects of crocin. Toxicol Ind Health 31:367-376. https://doi.org/10.1177/0748233713475519
  33. Pourtaji A et al (2016) Proteomics screening of adenosine triphosphate-interacting proteins in the liver of diazinon-treated rats. Hum Exp Toxicol 35:1084-1092. https://doi.org/10.1177/0960327115619771
  34. Sastry KV, Malik PV (1982) Acute and chronic effects of diazinon on the activities of three dehydrogenases in the digestive system of a freshwater teleost fish Channa punctatus. Toxicol Lett 10:55-59. https://doi.org/10.1016/0378-4274(82)90267-3
  35. Fujioka K, Casida JE (2007) Glutathione S-transferase conjugation of organophosphorus pesticides yields S-phospho-, S-aryl-, and S-alkylglutathione derivatives. Chem Res Toxicol 20:1211-1217. https://doi.org/10.1021/tx700133c
  36. Lari P et al (2014) Alteration of protein profile in rat liver of animals exposed to subacute diazinon: a proteomic approach. Electrophoresis 35:1419-1427. https://doi.org/10.1002/elps.201300475
  37. Kim SY, Chun E, Lee KY (2011) Phospholipase A(2) of peroxiredoxin 6 has a critical role in tumor necrosis factor-induced apoptosis. Cell Death Differ 18:1573-1583. https://doi.org/10.1038/cdd.2011.21
  38. Pakzad M et al (2013) Sublethal exposures of diazinon alters glucose homostasis in Wistar rats: biochemical and molecular evidences of oxidative stress in adipose tissues. Pestic Biochem Physiol 105:57-61. https://doi.org/10.1016/j.pestbp.2012.11.008
  39. Boussabbeh M et al (2016) Diazinon, an organophosphate pesticide, induces oxidative stress and genotoxicity in cells deriving from large intestine. Environ Sci Pollut Res Int 23:2882-2889. https://doi.org/10.1007/s11356-015-5519-y
  40. Yaghubi Beklar S et al (2021) The hydroalcoholic extract of Zingiber officinale diminishes diazinon-induced hepatotoxicity by suppressing oxidative stress and apoptosis in rats. Biotech Histochem 96:269-275. https://doi.org/10.1080/10520295.2020.1794039
  41. Ogasawara N et al (2017) Modulation of immunological activity on macrophages induced by diazinon. Toxicology 379:22-30. https://doi.org/10.1016/j.tox.2017.01.014
  42. Hedayati A, Hassan Nataj Niazie E (2015) Hematological changes of silver carp (Hypophthalmichthys molitrix) in response to Diazinon pesticide. J Environ Health Sci Eng 13:52. https://doi.org/10.1186/s40201-015-0208-9
  43. Zeinali M et al (2017) Protective effects of chrysin on sub-acute diazinon-induced biochemical, hematological, histopathological alterations, and genotoxicity indices in male BALB/c mice. Drug Chem Toxicol 41:270-280. https://doi.org/10.1080/01480545.2017.1384834
  44. Finamor IA et al (2014) The protective effect of N-acetylcysteine on oxidative stress in the brain caused by the long-term intake of aspartame by rats. Neurochem Res 39:1681-1690. https://doi.org/10.1007/s11064-014-1360-9 
  45. Li J et al (2015) N-acetylcysteine relieves oxidative stress and protects hippocampus of rat from radiation-induced apoptosis by inhibiting caspase-3. Biomed Pharmacother 70:1-6. https://doi.org/10.1016/j.biopha.2014.12.029