Abstract

Research Article

Zinc oxide nanoparticles attenuate the oxidative damage and disturbance in antioxidant defense system induced by cyclophosphamide in male albino rats

Azab Elsayed Azab*, Karema El M Shkal, Ahmed M Attia, Sabah G El-Banna and Rabia AM Yahya

Published: 30 April, 2020 | Volume 4 - Issue 1 | Pages: 001-008

Background: Cyclophosphamide is used for the treatment of malignant and non-malignant diseases, but, it induces oxidative damage and disturbance in the antioxidant defense system. Zinc oxide nanoparticles (ZnO NPs) are used in biomedical applications and consumer products. ZnO-NPs are protected cell membranes against oxidative damage, decrease free radicals and malondialdehyde (MDA) levels, and increase the antioxidant enzyme levels.

Objectives: The present aimed to evaluate the ameliorative effect of Zn-O nano-particles on oxidative damage and disturbance in the antioxidant defense system induced by cyclophosphamide in male albino rats.

Materials and Methods: 24 adult male albino rats were randomly divided into 4 groups (6 rats of each). Group I (Control group): Received 0.2 ml saline /day i.p. injection for 14 days (day by day), group II, (nZnO group): Received nZnO (5 mg/kg/day) b.w., intraperitoneally for 14 days, Group III (CP group): Received CP (20 mg/kg/day) b.w, day by day for 14 days by intraperitoneal injection, Group IV (CP + ZnO NPs group): Received nZnO group: Received nZnO (5 mg/kg/day) b.w., intraperitoneally for 14 days, plus CP (20 mg/kg/day) b.w., day by day for 14 days by intraperitoneal injection. After 24-hr from the last treatment, all animals were anesthetized using light ether. Blood, lungs, and liver samples were taken and prepared for biochemical measurements.

Results: Individual treatment of zinc oxide nanoparticles and CP induced liver cytochrome b5, cytochrome C reductase, and glutathione S-transferase (GST) compared to the control group, while CP increased P450. The combination of nZnO and CP prevents the elevation of cytochrome b5, P450, cytochrome C reductase, and GST compared with the CP treated group. Zinc oxide nanoparticles and CP increased liver thiobarbituric acid reactive substances (TBARS). The combination of nZnO and CP prevents the changes in TBARS concentrations compared with the CP. Injection of CP to rats reduced the activities of serum glutathione reductase (GR) and catalase (CAT) as compared with the control group. However, combination treatment of rats with nZnO and CP increased the activities of these enzymes compared with those treated with CP alone. Zinc oxide nanoparticles and CP increased serum and lung TBARS, while decreased glutathione (GSH) concentration compared to the control group, with more pronounced changes by CP. The combination of nZnO and CP prevents the changes in TBARS and GSH concentrations compared with the CP.

Conclusion: It can be concluded that CP induced oxidative stress and disturbance in the antioxidant defense system. Treatment of rats with zinc oxide nano-particles and CP together attenuated the oxidative damage and disturbance in the antioxidant defense system induced by CP. So, Patients treated with CP advised to take nZnO to prevent the side effects of chemotherapy. Further studies are necessary to evaluate the amelioration effect nZnO and other nano-particles against oxidative stress induced by CP in different doses and experimental models.

Read Full Article HTML DOI: 10.29328/journal.ibm.1001016 Cite this Article Read Full Article PDF

Keywords:

Cyclophosphamide; Zinc oxide nanoparticles; Attenuation; Ameliorative effect; Oxidative damage; Antioxidant defense system

References

  1. Dollery A. Cyclophosphamide. In: Dollery C., Editor. Therapeutic drugs. Edinburgh: Churchill Livingstone; 1999. 349-353.
  2. Nafees S, Rashid S, Ali N, Hasan SK, Sultana S. Rutin ameliorates cyclophosphamide-induced oxidative stress and inflammation in Wistar rats: Role of NFκB/MAPK pathway. Chemico-Biological Interactions. 2015; 231: 98-107. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/25753322
  3. Lindley CM, Hamilton G, McCune JS, Faucette S, Shord SS, et al. The effect of cyclophosphamide with and without dexamethasone on cytochrome P450 3A4 and 2B6 in human hepatocytes. Drug Metabol Dispo, 2002; 30: 814-821. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/12065440
  4. Pass GJ, Carrie D, Boylan M, Lorimore S, Wright E, et al. Role of hepatic cytochrome P450s in the pharmacokinetics and toxicity of cyclophosphamide: stidies with the hepatic cytochrome P450 reductase null mouse. Canc Res. 2005; 65: 4211-4217. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/15899812
  5. Seis H. Oxidative stress: introductory remarks. H Seis (Ed). Oxidative Stress, Academic Press, London. 1985; 1-8.
  6. Sachidanandam 1K, Fagan SC, Ergul A. Oxidative stress and cardiovascular disease: antioxidants and unresolved issues. Cardiovasc Drug Rev. 2005; 23: 115-132. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/16007229
  7. Borchi E, Bargelli V, Stillitano F, Giordano C, Sebastiani M, et al. Enhanced ROS production by NADPH oxidase is correlated to changes in antioxidant enzyme activity in human heart failure. Biochim Biophys Acta, 2010; 1802: 331-338. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/19892017
  8. Schacter LP, DelVillano BC, Gordon EM, Klein BL. Red cell superoxide dismutase and sickle cell anemia symptom severity. Am J Hematol. 1985; 19: 137-144. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/4003385
  9. Scott MD, Eaton JW, Kuypers FA, Chiu DT, Lubin BH. Enhancement of erythrocyte superoxide dismutase activity: effects on cellular oxidant defence. Blood. 1989; 74: 2542-2549. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/2553167
  10. Kao MPC, Ang DSC, Pall A, Struthers AD. Oxidative stress in renal dysfunction: mechanisms, clinical sequelae and therapeutic options. J Human Hypertension, 2010; 24: 1-8. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/19727125
  11. Jones N, Ray B, Ranjit KT, Manna AC. Antibacterial activity of ZnO nanoparticle suspensions on a broad spectrum of microorganisms. FEMS Microbiol Lett. 2008; 279: 71-76. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/18081843
  12. Wang SQ, Tooley IR. Photoprotection in the era of nanotechnology. Semin Cutan Med Surg. 2011; 30: 210-213. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/22123418
  13. Khalaf AA, Hassanen EI, Azouz RA, Zaki AR, Ibrahim MA, et al. Ameliorative effect of zinc oxide nanoparticles against dermal toxicity induced by lead oxide in rats. Int J Nanomed. 2019; 14: 7729-7741. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/31806958
  14. Hussein SA, EL-Senosi YA, El-Dawy K, Baz HA. Protective effect of zinc oxide nanoparticles on oxidative stress in experimental-induced diabetes in rats. Benha Veter Med J. 2014; 27: 405-414.
  15. Afifi M, abdelazim AM. Ameliorative effect of zinc oxide and silver nanoparticles on antioxidant system in the brain of diabetic rats. Asian Pac J Trop Biomed. 2015; 5: 874-877.
  16. Aitken RJ, Roman SD. Antioxidant systems and oxidative stress in the testes. Oxid Med Cell Longev. 2008; 1: 15-24. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/19794904
  17. El-Maddawy ZK, abd El Naby WSH. Protective effects of zinc oxide nanoparticles against doxorubicin induced testicular toxicity and DNA damage in male rats. Toxicol Res. 2019; 8: 654-662. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/31588342
  18. Raajshreer K, Durairaj B. Evaluation of the antityrosinase and antioxidant potential of zinc oxide nanoparticles synthesized from the brown seaweed –Turbinaria conoides. Int. J Appl Pharm. 2017; 9: 116-120.
  19. Dawei AI, Zhisheng W, Angu Z. Protective effects of nano-ZnO on the primary culture mice intestinal epithelial cells in in vitro against oxidative injury. Int J Nanotechnol App. 2009; 3: 1-6.
  20. Badkoobeh P, Parivar K, Kalantar SM, Hosseini SD, Salabat A. Effect of nano-zinc oxide on doxorubicin- induced oxidative stress and sperm disorders in adult male Wistar rats. Iran J Reprod Med. 2013; 11: 355-364. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/24639766
  21. Fahmey MA, Hassan NHA, El-Fiky SA, Elalfy HG. A mixture of honey bee products ameliorates the genotoxic side effects of cyclophosphamide. Asian Pacific J Trop Dis. 2015; 5: 638-644.
  22. Moron MS, Depierre JW, Mannervik B. Levels of glutathione, glutathione reductase and glutathione S-transferase activities in rat lung and liver. Biochim. Biophys. Acta. 1979; 582: 67-78. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/760819
  23. Goldberg DM, Spooner RJ. In HV Bergmeyer (Ed). Methods of enzymatic analysis (3rd ed). 1983; 258-265.
  24. Góth L. A simple method for determination of serum catalase activity and revision of reference range. Clinica Chimica Acta. 1991; 196: 143-151. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/2029780
  25. Slater TF. Sawyer BC. The stimulatory effects of carbon tetrachloride and other halogeno-alkanes on peroxidative reactions in rats. Biochem J. 1971; 123: 805-814. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/4399399
  26. Omura T, Sato R. The carbon monoxide binding pigment of liver microsomes. 1-Evidence for its hematoprotein nature. J Biol Chem. 1964; 239: 1370-2378.
  27. Williams CH, Kamin H. Microsomal triphosphopyridine nucleotide-cytochrome-C reductase of liver. J Biol Chem. 1962; 237: 587-595.
  28. Habig W, Pabst M, Jakoby W. Glutathione-S-transferases. The first enzymatic step in mercapturic acid formation. J Biol Chem. 1974; 249: 7130-139. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/4436300
  29. Howell DC. Fundamental statistics for the behavioral sciences (3rd ed). Duxbury press. An imprint of Wads Worth publishing company Belmont. California. 1995; 163-166.
  30. De Oliveira CA, Gremano PM. Aflatoxins current concepts on mechanisms of toxicity and their involvement in the etiology of hepatocellular carcinoma. Rev. Saude Publica. 1997; 31: 417-424. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/9595771
  31. Verma RS, Mehta A, Srivastava N. Effect of phenobarbitone on cytochrome P450 activity and chlorpyrifos and 3, 5, 6-trichloropyridinol levels in liver and serum in rat. Indian J Biochem Biophys. 2005; 42: 254-257. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/23923552
  32. Goel A, Dani V, Dhawan D. Zinc mediates normalization of hepatic drug metabolizing enzymes in chlorpyrifos-induced toxicity. Toxicol Lett. 2007; 169: 26-33. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/17194553
  33. Sharma Y, Bashir S, Irshad M, Nag TC, Dogra TD. Dimethoate-induced effects on antioxidant status of liver and brain of rats following subchronic exposure. Toxicol. 2005; 215: 173-181. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/16112789
  34. Sivapiriya V, Jayanthisakthisekaran, Venkatraman S. Effects of dimethoate (O,O-dimethyl S-methyl carbamoyl methyl phosphorodithioate) and Ethanol in antioxidant status of liver and kidney of experimental mice. Pest Biochem Physiol. 2006; 85: 115-121.
  35. Vermilion JL, Ballou DP, Massey V, Coon MJ. Separate roles of FMN and FAD in catalysis by liver microsomal NADPH cytochrome P450 reductase. J Biol Chem. 1981; 256: 266-277. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/6778861
  36. Delvi RR. Alterations in hepatic phase I and phase II biotransformation enzymes by garlic oil in rats. Toxicol Lett. 1992; 60: 299-305. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/1595088
  37. Sheweita S, abd El-Gaber M, Bastawy M. Carbon tetrachloride changes the activity of cytochrome P450 system in the liver of male rats: role of antioxidants. Toxicol. 2001; 169: 83-92. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/11718950
  38. Patel JM, Block ER. Cyclophosphamide-induced depression of the antioxidant defense mechanisms of the lungs. Exp Lung Res. 1985; 8: 153-165. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/4029094
  39. Venkatesan N, Chandrakasan G. Modulation of cyclophosphamide induced early lung injury by curcumin, an anti-inflammatory antioxidant. Mol Cell Biochem. 1995; 142: 79-87. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/7753045
  40. Kaya H, Oral B, Ozguner F, Tahan V, Babar Y, et al. The effect of melatonin application on lipid peroxidation during cyclophosphamide therapy in female rats. Zentralbl Gynakol. 1999; 121: 499-502. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/10573825
  41. Lear L, Nation RL, Stupans I. Effects of cyclophosphamide and adriamycin on rat hepatic microsomal glucuronidation and lipid peroxidation. Biochem Pharmacol. 1992; 44: 747-753. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/1510722
  42. Mathew S, Kuttan G. Antioxidant activity of Tinospora cordifolia and its usefulness in the amelioration of cyclophosphamide induced toxicity. J Exp Clin Cancer Res. 1997; 16: 407-411. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/9505214
  43. Premkumar K, Pachiappan A, Abraham SK, Santhiya ST, Gopinath PM. Effect of Spirulina fusiformis on cyclophosphamide and mitomycin-C induced genotoxicity and oxidative stress in mice. Fitoterapia. 2001; 72: 906-911. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/11731115
  44. Halliwell B, Gutteridge. JM. Free radicals in biology and medicine, 2nd ed. Oxford: Clarendon Press. 1989.
  45. Haque R, Bin-Hafeez B, Parvez S, Pandey S, Sayeed I, et al. Aqueous extract of walnut (Juglans regia L.) protects mice against cyclophosphamide-induced biochemical toxicity. Hum Exp Toxicol. 2003; 22: 473-480. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/14580007
  46. Patel JM. Stimulation of cyclophosphamide-induced pulmonary microsomal lipid peroxidation by oxygen. Toxicol. 1987; 45: 79-91. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/3603576
  47. Selvakumar E, Prahalathan C, Mythili Y, Varalakshmi P. Mitigation of oxidative stress in cyclophosphamide-challenged hepatic tissue by DL-alpha-lipoic-acid. Mol Cell Biochem. 2005; 272: 179-185. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/16010986
  48. Adams JD, Klaidman LK. Acrolein induced oxygen radical formation. Free Radic Biol Med. 1993; 15: 187-193. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/8397144
  49. Uchida K. Current status of acrolein as a lipid product. Trends Cardiovasc Med. 1999; 9: 109-113. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/10639724
  50. Kehrer JP. Free radicals as mediators of tissue injury and disease. Crit Rev Toxicol. 1993; 23: 21-48. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/8471159
  51. Lin HM. Yen FL, Ng LT, Lin CC. Protective eff ects of Ligustrum lucidum fruit extract on acute butylated hydroxytoluene-induced oxidative stress in rats. J Ethnopharmacol. 2007; 111: 129-136.
  52. Parke DV, Sapota A. Chemical toxicity and reactive oxygen species. Int J Occup Med Env Health. 1996; 9: 331. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/9117192
  53. Durken M, Agbenu J, Finckh B, Hubner C, Pichlmeier U, et al. Deteriorating free radical-trapping capacity and antioxidant status in plasma during bone marrow transplantation. Bonne Marrow Transplant. 1995; 15: 7570. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/7670403
  54. Sharma V, Singh P, Pandey AK, Dhawan A. Induction of oxidative stress, DNA damage and apoptosis in mouse liver after sub-acute oral exposure to zinc oxide nanoparticles. Mutat Res. 2012; 745: 84-91. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/22198329
  55. Ali-Osman F. Quenching of DNA cross-link precursors of chloroethylnitrosoureas and attenuation of DNA interstrand cross-linking by glutathione. Cancer Res. 1989; 49: 5258-5260. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/2766294
  56. McDiarmid MA, Iype PT, Koldner K, Jacobnson KD, Strickland PT. Evidence for acrolein-modified DNA in peripheral blood leucocytes of cancer patients treated with cyclophosphamide. Mutat Res. 1991; 248: 93-99. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/2030715
  57. Touliatos JS, Neitzel L, Whitworth C, Rybak LP, Malafa M. Effect of cisplatin on the expression of glutathione-S-transferase in the cochlea of the rat. Eur Arch Otorhinolaryngol. 2000; 25: 6-9. PubMed: https://www.ncbi.nlm.nih.gov/pubmed/10664037

Figures:

Figure 1

Figure 1

Figure 1

Figure 2

Figure 1

Figure 3

Figure 1

Figure 4

Figure 1

Figure 5

Figure 1

Figure 6

Figure 1

Figure 7

Figure 1

Figure 8

Figure 1

Figure 9

Similar Articles

Recently Viewed

Read More

Most Viewed

Read More

Help ?