Radio-protective response on the environmental pollutant induced oxidative stress

Abstract

The purpose of this study was to investigate the protective effect of low dose radiation on TCE induced oxidative damage in rats. The oxidative damage of both liver and kidney was assessed by serum alkaline phosphatase (ALP), Gamma Glutamyl Trans-Peptidase (GGTP), Alanine and Aspartate Amino Transferase (ALT & AST) activities in addition concentrations of cholesterol, high density lipoprotein-cholesterol (HDL-c), low density lipoprotein-cholesterol (LDL-c), Triacyglycerols (TGs), urea and creatinine were analyzed. Liver lipid peroxidation (MDA), Nitric Oxide (NO), reduced glutathione (GSH) levels, and activities of both Super-Oxide Dismutase (SOD) and Glutathione Peroxidase (GSH-Px) were measured. Results: TCE administration increase serum ALP, GGTP, ALT, AST activities, cholesterol, triacylglycerols, LDL-c, urea and creatinine concentrations, besides liver MDA and NO, whereas it decreased SOD, GSH-Px activities, GSH level in liver, HDL-c in serum. Low dose of gamma rays (0.5 Gy) exposure significantly improved lipid peroxidation and oxidative injury induced by TCE. Conclusion: The study indicates that treatment with low dose of gamma rays ameliorate harmful effects induced by TCE taking in consideration the effect of gamma radiation as a stimulant of radical detoxification.

Share and Cite:

Gharib, O. , Ellatif, U. , Abdellah, N. and Mohammad, M. (2012) Radio-protective response on the environmental pollutant induced oxidative stress. Advances in Bioscience and Biotechnology, 3, 989-996. doi: 10.4236/abb.2012.327122.

Conflicts of Interest

The authors declare no conflicts of interest.

References

[1] Macklis, R.M. and Bresford, B. (1991) Radiation hormesis. Journal of Nuclear Medicine, 32, 350-359.
[2] Feinendegen, L.F. (2005) Evidence for beneficial low level radiation effects and radiation hormesis. British Journal of Radiology, 78, 3-7. doi:10.1259/bjr/63353075
[3] Bhattacharjee, D. (1996) Role of radio-adaptation on radiation-induced thymic lymphoma in mice. Mutation Research, 358, 231-235. doi:10.1016/S0027-5107(96)00125-X
[4] De Azzam, E.I., Toledo, S.M., Raaphorst, G.P. and Mitchel, R.E.J. (1996) Low dose ionizing radiation decreases the frequency of neoplastic transformation to a level below the spontaneous rate in C3H 10T 1/2 cells. Radiation Research, 146, 369-373. doi:10.2307/3579298
[5] Yonezawa, M., Misonoh, J. and Hosokawa, Y. (1996) Two types of X-ray induced radio-resistance in mice, presence of 4 dose ranges with distinct biological effects. Mutation Research, 358, 237-243. doi:10.1016/S0027-5107(96)00126-1
[6] Manno, M., Bertazzon, A., Burlina, L. and Galzigna, L. (1985) Interaction of low doses of ionizing radiation and carbon tetrachloride on liver superoxide dismutase and glutathione peroxidase in mice. Enzyme, 34, 107-112.
[7] Yamaoka, K., Kojima, S., Takahashi, M., Nomura, T. and Iriyama, K. (1998) Change of glutathione peroxidase synthesis along with that of super oxide dismutase synthesis in mice spleens after low-dose X-ray irradiation. Biochimica et Biophysica Acta, 1381, 265-270. doi:10.1016/S0304-4165(98)00021-X
[8] Kojima, S., Matuski, O. and Nourma, T. (1997) Effect of small doses of gamma ray on glutathione synthesis in mouse. International Conference on low doses of ionizing radiation Biology Effect and regulatory Control, Sevillr, 17-21.
[9] Nomura, T. and Yamaoka, K. (1999) Low-dose γ-ray irradiation reduces oxidative damage induced by CCl4 in mouse liver. Free Radical Biology & Medicine, 27, 1324-1333.
[10] Kojima, S., Matsumori, S., Ishida, H. and Yamaoka, K. (2000) Possible role of elevation of glutathione in the acquisition of enhanced proliferation of mouse splenocytes exposed to small dose gammarays. International Journal of Radiation Biology, 76, 1641-1647. doi:10.1080/09553000050201136
[11] Agency for toxic substance and disease registry (ATSDR) (2003) Managing hazardous materials incidents Vol. III. Medical management guidelines for acute chemical exposures: Trichloroethylene (TCE). US Department of Health and Human Services, Public Health Service, Atlanta.
[12] Boyes, W.K., Bushnell, P.J., Crofton, K.M., Evans, M. and Simmons, J.E. (2000) Neurotoxic and pharmacokinetic response to trichloroethylene as a function of exposure scenario. Environmental Health Perspectives, 108, 317-322. doi:10.1289/ehp.00108s2317
[13] Lash, L.H. and Parker, J.C. (2001) Hepatic and renal toxicities associated with trichloroethylene. Pharmacological Reviews, 53, 177-208.
[14] Bull, R.J. (2000) Mode of action of liver tumor induction by trichloroethylene and its metabolites, trichloroacetate and dichloroacetate. Environmental Health Perspectives, 108, 241-259. doi:10.1289/ehp.00108s2241
[15] Gharib, O.A. (2009) Effects of Kombucha on oxidative stress induced nephrotoxicity in rats. Chinese Medicine, 4, 23. doi:10.1186/1749-8546-4-23
[16] Chen, S.J., Wang, J.L., Chen, J.H. and Huang, R.N. (2002) Possible involvement of glutathione and P53 in trichloroethylene and percholoroethylene. Induced lipid peroxidetion and apoptosis in human lung cancer cells. Free Radical Biology & Medicine, 33, 464-472.
[17] Torasson, M., Clark, J., Dankovic, D., Mathias, P., Skaggs, S., Walker, C. and Werren, D. (1999) Oxidative stress and DNA damage in Fischer rats following acute exposure to trichloroethylene or perchloroethylene. Toxicology, 138, 43. doi:10.1016/S0300-483X(99)00083-9
[18] Ellis, G., Belfield, A. and Goldberg, D.M. (1971) Colorimetric determination of acid phosphatase activity using adenosine 3’-monophosphate as substrate. Journal of Clinical Pathology, 24, 493-500. doi:10.1136/jcp.24.6.493
[19] Szasz, G., Weimann, G., Stahler, O., Wahefedl, A.W. and Persijn, J.P. (1974) New substances for measuring gamma-glutamyl transpeptidase activity. Journal of Clinical Chemistry & Clinical Biochemistry, 12, 228-232.
[20] Retiman, S. and Frankel, S. (1957) A colorimetric method for the determination of serum glutamic oxalacetic and glutamic pyruvic transaminases. American Journal of Clinical Pathology, 28, 56-63.
[21] Castelli, W.P. (1986) Triglyceride tissue: A view from Framingham. American Heart Journal, 112, 432-437. doi:10.1016/0002-8703(86)90296-6
[22] Lopes, V.M.F, Stone, P., Ellis, S.and Colwell, J.A. (1977) Cholesterol determination in high density lipoproteins separated by three different methods. Clinical Chemistry, 23, 882-884.
[23] Meiattini, F., Prencipe, L., Bardelli, F., Giannini, G. and Tarli, P. (1978) The 4-hydroxybenzoate/4-amino phenazone chromogentic system used in the enzymic determination of serum cholesterol. Clinical Chemistry, 24, 2161-2165.
[24] Bucolo, G. and David, H. (1973) Quantitative determination of serum triglyceride by the use of enzymes. Clinical Chemistry, 19, 476-482.
[25] Patton, C.J. and Crouch, S.R. (1977) Spectrophotometric and kinetics investigation of the Berthelot reaction for the determination of ammonia. Analytical Chemistry, 49, 464-469. doi:10.1021/ac50011a034
[26] Folin, O.Z. (1943) Micro Method for the determination of non-protein nitrogen, in laboratory manual of biological chemistry with supplement. 5th Ed, Appleton-Century Co. Inc., New York, 271.
[27] Yoshioka, T., Kawada, K., Shimada, T.and Movi, M. (1979) Lipid peroxidation in maternal and cord blood and protective mechanism against activated oxygen toxicity in the blood. American Journal of Obstetrics & Gynecology, 135, 372-376.
[28] Moshage, H., Kok, B., Huizenga, J.R. and Jansen, P.L.M. (1995) Nitrie and nitrate determination in plasma a critical evaluation. Clinical Chemistry, 41, 892-896.
[29] Ellman, G.C. (1959) Tissue sulfhydryl groups. Archives of Biochemistry and Biophysics, 82, 70-77. doi:10.1016/0003-9861(59)90090-6
[30] Marklund, S. and Marklund, G. (1974) Involvement of the super oxide anion radical in the antioxidation of pyrogallol and a convenient assay for superoxide dismutase. European Journal of Biochemistry, 47, 469-474. doi:10.1111/j.1432-1033.1974.tb03714.x
[31] Gross, R.T., Bracci, R., Rudolph, N., Schroeder, E. and Kochen, J.A. (1967) Hydrogen peroxide toxicity and detoxification in the erythrocytes of newborn infants. Blood, 29, 481-493.
[32] IAEA (1980) Elemental analysis of biological materials. Current problem and techniques with special reference to trace elements. International Atomic Energy Agency, IAEA, Vienna. Technical Reports Series, 379.
[33] Kingstone, H.M. and Jassie, L. (1988) Introduction to microwave sample preparation. Theory and practice. American Chemical Socity Professional Reference Book, Washington DC, 263.
[34] Gregus, Z. and Klaassen, C.D. (1986) Disposition of metals in rats: A comparative study of fecal urinary and billary excretion and tissue distribution of eighteen metals. Toxicology and Applied Pharmacology, 85, 24-38. doi:10.1016/0041-008X(86)90384-4
[35] Ramaiah, S.K., Apte, U.M. and Mehenadale, HM. (2001) Cytochrome P450 induction increases thioacetamide liver injury in dietrestricted rats. Drug Metabolism and Disposition, 29, 1088-1095.
[36] Anand, S.S., Mumtaz, M.M. and Mehendale, H.M. (2005) Dose dependent liver regeneration in chloroform trichloroethylene and allyl alcohol ternary mixture hepatotoxicity in rats. Archives of Toxicology, 79, 671-682. doi:10.1007/s00204-005-0675-3
[37] Pratt, D.S. and Kaplan, M.M. (2002) Evaluation of liver function. In: Braunwald, E., et al., Eds., Harrison’s Principles of Internal Medicine, McGraw-Hill, New York, 1711-1715.
[38] Lawerence, B.P., Will, Y., Reed, D.J. and Kerkvliet, N.I. (2000) Gamma-glutamyl transpeptidase knockout mice as a model for understanding the consequence of diminished glutathione on T cell dependent immune response. European Journal of Immunology, 30, 1902-1910. doi:10.1002/1521-4141(200007)30:7<1902::AID-IMMU1902>3.0.CO;2-A
[39] Moffatt, P., Plaa, G.L. and Denizeau, F. (1996) Rat hepatocytes with elevated metallothionein expression are resistant to N-methyl-N#-nitro-Nnitrosoguanidine cytotoxicity. Toxicology and Applied Pharmacology, 136, 200-207. doi:10.1006/taap.1996.0025
[40] Chisolm, G.M., Hazen, S.L., Fox, P.L. and Cathcart, M.K. (1999) The oxidation of lipoprotein by monocytes macrophagaes Biochemical and Biological mechanism. The Journal of Biological Chemistry, 274, 5959-5962. doi:10.1074/jbc.274.37.25959
[41] Browning, J.D. and Horton, J.D. (2004) Molecular mediators of hepatic steatosis and liver injury. The Journal of Clinical Investigation, 114, 147-152.
[42] Ohshima, H. and Bartsch, H. (1994) Chronic infection and inflammatory processes as cancer risk factors: Possible role of nitric oxide in carcinogenesis. Mutation Research, 305, 253-264. doi:10.1016/0027-5107(94)90245-3
[43] Goel, S.K., Rao, G.S., Pandya, K.P. and Shanker, R. (1992) Trichloroethylene toxicity in mice: A biochemical, hematological and pathological assessment. Indian Journal of Experimental Biology, 30, 402-406.
[44] Khan, S., Priyamvada, S., Khan, S.A., Khan, W., Farooq, N., Khan, F., et al. (2009) Effect of trichloroethylene (TCE) toxicity on the enzymes of carbohydrate metabolism, brush border membrane and oxidative stress in kidney and other rat tissues. Food and Chemical Toxicology, 47, 1562-1568. doi:10.1016/j.fct.2009.04.002
[45] Mostafa, S.A. (1998) Effect of allyl as glutathione depleting agent on carbohydrate metabolism in rats. Journal of Egyptian German Society of Zoology, 26, 13-34.
[46] Fletcher, L.M., Roberts, F.D., Irving, M.G., Powell, L.W. and Halliday, J.W. (1989) Effects of iron loading on free radical scavenging enzymes and lipid peroxidation in rat liver. Gastroenterology, 97, 1011-1018.
[47] Sadani, G. and Nadkarni, G.D. (1994) Role of antioxidant enzyme defense in sparing rat hepatocytes from toxicity of ricin at low dose. Indian Journal of Experimental Biology, 32, 354-355.
[48] Kumar, P., Prasad, A.K., Maji, B.K., Mani, U. and Dutta, K.K. (2001) Hepatotoxic alterations induced by inhalation of trichloroethylene (TCE) in rats. Biomedical and Environmental Sciences, 14, 325-332.
[49] Hultberg, M. and Hultberg, B. (2005) Glutathione turnover in human cell lines in the presence of agents with glutathione influencing potential with and without acivicin inhibition of gamma-glutamyl transferase peptidase. Biochimica Biophysica Acta, 1726, 42-47. doi:10.1016/j.bbagen.2005.08.007
[50] Dashti, H.M., Al-Sayer, H., Behbehani, A., Madda, J. and Christenson, J.T. (1992) Liver cirrhosis induced by carbon tetrachloride and the effect of superoxide dismutase and xanthine oxidase inhibitor treatment. Journal of the Royal College of Surgeons of Edinburgh, 37, 23-28.
[51] Keen, C.L., Ensunsa, J.L., Watson, M.H., Baly, D.L., Donovan, S.M., et al. (1999) Nutritional aspects of manganese for experimental studies. Neurotoxicology, 20, 213-223.
[52] Pathak, C.M., Avti, P.K., Kumar, S., Khanduja, K.L. and Sharma, S.C. (2007) Whole body exposure to low dose gamma radiation promotes kidney antioxidant status in Balble/c-mice. Journal of Radiation Research, 48, 113-120. doi:10.1269/jrr.06063

Copyright © 2024 by authors and Scientific Research Publishing Inc.

Creative Commons License

This work and the related PDF file are licensed under a Creative Commons Attribution 4.0 International License.