Logo-ajmb
Submitted: 19 Feb 2014
Revised: 07 Mar 2014
Accepted: 13 May 2014
First published online: 25 Sep 2014
EndNote EndNote

(Enw Format - Win & Mac)

BibTeX BibTeX

(Bib Format - Win & Mac)

Bookends Bookends

(Ris Format - Mac only)

EasyBib EasyBib

(Ris Format - Win & Mac)

Medlars Medlars

(Txt Format - Win & Mac)

Mendeley Web Mendeley Web
Mendeley Mendeley

(Ris Format - Win & Mac)

Papers Papers

(Ris Format - Win & Mac)

ProCite ProCite

(Ris Format - Win & Mac)

Reference Manager Reference Manager

(Ris Format - Win only)

Refworks Refworks

(Refworks Format - Win & Mac)

Zotero Zotero

(Ris Format - FireFox Plugin)

Abstract View: 1538
PDF Download: 818
Full Text View: 1066
Avicenna J Med Biochem. 2014;2(1): e18321.
doi:10.17795/ajmb-18321

Research Article

Hepatoprotective Effects of Vitamin E Against Malathion-Induced Mitochondrial Dysfunction in Rat Liver

Akram Ranjbar 1 * , Fariba Mohsenzadeh 2, Maryam Baeeri 3

1 Department of Toxicology and Pharmacology, Hamadan University of Medical Sciences, Hamadan, IR Iran
2 Department of Biology, Bu-Ali Sina University, Hamedan, IR Iran
3 Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, IR Iran
*Corresponding author: Akram Ranjbar, Department of Toxicology and Pharmacology, Hamadan University of Medical Sciences, Hamadan, IR Iran. Tel/Fax: +98-8118380031, E-mail: Email: akranjbar1389@yahoo.ccom

Abstract

 

Background: Malathion is an insecticide of the grouping of organophosphate pesticides (OPs), which shows strong insecticidal effects. In addition, vitamin E reacting to cell membrane site may prevent OP-induced oxidative injury.

Objectives: The aim of this study was to examine the protective function of vitamin E on toxicity of malathion, by measuring the activities of liver and liver mitochondrial superoxide dismutase (SOD), catalase (CAT),lipid peroxidation (LPO),and glutathione peroxidase (GPx) in rats.

Materials and Methods: The mitochondrial viability was determined in liver. ‎Effective doses of malathion(200 mg/kg/day) and vitamin E (alpha-tocopherylacetate [AT]; 15 mg/kg/day) were administered alone or in combination for 14 days. At the end of the experiment, the liver tissue and liver mitochondria of the animals were harvested and examined.

Results: In liver tissue, the activity of LPO and CAT was higher in the malathion group in comparison to controls. AT reduced malathion-induced LPO, SOD, CAT, and GPx in rat liver. Coadministration of AT with malathion improved LPO, SOD, and CAT levels in liver as well as CAT and GPx in liver mitochondria. Malathion-induced mitochondria toxicity was recovered by AT.

Conclusions: In conclusion, AT measurement can be beneficial for the safety or recovery of malathion-induced toxic injury in liver tissue and liver mitochondria.

Keywords: Malathion; Mitochondria, Liver; Oxidative Stress; Rats; Vitamin E

1. Background

 

 

Organophosphorus (OPs) pesticides are a major as well as the most diverse group of insecticides. The broad implication of OPs in public health and agricultural programs accompanies potentially dangerous impacts on humans, animals, plants, and environment (water, air, soil, and food) and causes acute and chronic poisoning (1, 2). OPs decrease the function of carboxylic ester hydrolases such as chymotrypsin, acetylcholinesterase (AChE), plasma or butyrylcholinesterase (BuChE), plasma and hepatic carboxylesterase (aliesterases), paraoxonases (PON1) and other esterases within the body (2-4). Malathion (O,O-dimethyl-S-[1,2-dicarboxyethyl] ethyl phosphorodithioate) is one of the main widely used Ops pesticides in agriculture and public health programs (5-7). Recently, it has been revealed that OPs produce oxidative stress in different tissues through the producing reactive oxygen species (ROS) (8, 9). Although ROS are part of normal oxidative metabolism, when produced in overload, they cause tissue injury including lipid peroxidation, DNA damage, and enzyme inactivation (10, 11). Moreover, oxidative stress is a process related to xenobiotic contact and different levels of environmental pollution (12). Hence, the mitochondrial respiratory chain serves as a main source of ROS derived from the disproportionate superoxide anions. Even though the presence of different antioxidants factors, e.g. ubiquinone and vitamin E, and antioxidant enzymes, and the mitochondria is the most powerful intracellular source of ROS; according to a hypothesis, the steady state concentration of O.-2 in the mitochondrial matrix is about five-to ten-fold higher than that in the cytosol or nucleus. Consequently, mitochondria might also be a main target for the damaging function of ROS. The contact of diverse macromolecules with ROS may impair the role of these organelles and may directly influence cell viability and lead to prompt cell death (13, 14). In biological systems, vitamin E is a key antioxidant acting as a potent chain-breaking mediator during the scavenging of peroxyl radicals (15, 16). Moreover vitamin E decreases the chain reaction of lipid peroxidation in membranes. Hence, a lot of studies have been done to find out the defending properties of vitamin E in specific biological models of injury (17, 18).

2. Objectives

 

 

This study aimed to examine antioxidant effects of vitamin E on malathion-induced oxidative stress in rat liver homogenate and mitochondria.

3. Materials and Methods

 

 

3.1. Reagents and Chemicals

Trichloroacetic acid (TCA), tetraethoxypropane (MDA), 2-thiobarbituric acid (TBA), n-butanol,malathion, sucrose, ethylenediaminetetraacetic acid (EDTA), Trishydrochloride acid, nitrobluetetrazolium (NBT), xanthine, xanthine oxidase, hydrogen peroxide (H2O2), Coomassie blue, bovine serum albumin (BSA), 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), were used in this study. All the chemicals were obtained from the Sigma Chemical Company (St. Louis, MO, USA).

3.2. Animals and Treatments

The male Wistar rats (180-250 g) were obtained from the animal colony of the Pastor Institute, Iran. Animals were maintained under standard conditions with the temperature of 22℃ ± 1℃, humidity of 45%, and 12:12 hours light/dark cycle. Animals were randomly allocated to four groups of five animals and treated for two weeks. The groups were as follows: control group, malathion group, alpha-tocopherol acetate (AT) group, and malathion-AT group. Malathion was administered intraperitoneally (200 mg/kg/day) alone or in combination with subcutaneous AT (15 mg/kg/day). One group of animals received normal saline only and was considered as the control group. At the end of the treatment (24 hours after receiving the last treatment dose), animals were sacrificed; then, liver tissue was extracted and stored in liquid nitrogen.

3.3. Isolation of Liver Mitochondria

The liver tissue was distincted with small cutters in a cold mannitol solution containing 0.225-mol D-mannitol, 75-mmol sucrose, and 0.2-mmol EDTA. The minced liver (30 g) was gently homogenized in a glass homogenizer with a Teflon pestle and then centrifuged at 700 ×g for ten minutes at 4°C to remove nuclei, unbroken cells, and other non-subcellular tissues. The supernatants were centrifuged at 7000 g for 20 minutes. These second supernatants were taken as the crude microsomal part and the pale loose upper layer of sediments, which was rich in swollen or broken mitochondria, lysosomes, and some microsomes, was washed away. The dark packed lower layer (heavy mitochondrial fraction) was resuspended in the mannitol solution and centrifuged twice at 7000 g for 20 minutes. Then weighty mitochondrial sediments were floated in Tris solution buffer (pH, 7.4) (19, 20).

3.4. Assessmentof Copper-Zinc superoxide dismutase Activity

Copper-zinc SOD (Cu/Zn SOD) was measured indirectly by assessing the reaction between SOD and the molecule NBT. The optical density (OD) changes at 560 nm over a five-minute period indicated the reduction of NBT by superoxide. The activity of total SOD was determined as the quantity of protein necessary for half-maximal inhibition of the NBT reaction (21).

3.5. Assessment of Glutathione Peroxidase Activity

In this method, the sample was added to a solution containing, glutathione (2 mmol), glutathione reductase (0.15 U/mL), sodium azide (0.4 mmol), reduced form of nicotinamide adenine dinucleotide phosphate (NADPH, 0.12 mmol), Tris (50 mmol, pH ¼ 7.5), EDTA (5 mmol), and dithiothreitol (DTT) buffer (1 mmol). The reaction was initiated by adding H2O2 and the diminish in absorbance at 340 nm/min over ten minutes was calculated (22).

3.6. Assessmentof Catalaseactivity

The activity of CAT in the samples was measured by assessing the absorbance decreasing at 240 nm in a reaction medium containing H2O2 (10 mmol) and sodium phosphate buffer (50 mmol, pH 7.0). One unit of the enzyme was reported as 1 mol H2O2 as substrate consumed per minute, and the specific activity was reported as units per milligram protein (23).

3.7. Measurement of ‎Lipid Peroxidation

In this method, the lipid peroxidation (LPO) product in the tissues was diluted by TBA reagent in acid heating reaction. Then, the samples were washed by adding 1.5 mL of 20% (w/v) TCA was added to 250 μL of samples and centrifuged in 3000 g for ten minutes. The precipitation was dissolved in sulfuric acid and 1.5 mL of the solution was added to 1.5 mL of 0.2% (w/v) TBA. Subsequent to incubation, 2 mL of n-butanol was added and the solution was centrifuged, cooled, and measured at 532 nm. Finally, the calibration curve of tetramethoxypropane standard solutions was used to verify the concentrations of TBA + MDA adducts in samples (24).

3.8. Total Protein

In this study, the protein content was measured using Bradford method. Strenuous Coomassie blue (G250) was diluted in 250 μL distilled water and then 750 μL of this diluted dye was added to 50 μL of the sample. The mixture was incubated at room temperature for ten minutes and an absorbance was measured at 595 nm by a spectrophotometer. Finally, a standard curve was constructed by using bovine serum albumin (BSA) as the standard (0.25 and 1 mg/mL) (25).

3.9. Assessment of‎ Mitochondria Toxicity

This assay is a quantitative colorimetric way to determine cell viability. It utilizes the yellow tetrazolium salt (MTT), which is metabolized by mitochondrial dehydrogenase enzyme from viable cells to yield a purple formazan reaction result at wavelength of 570 nm. Finally, the percentage of mitochondrial viability of each sample was measured (26).

3.10. Statistical Analysis

Data were presented as mean ± SEM of at least three independent experiments with SPSS Version 16 (SPSS, Chicago, IL, USA). The one-way ANOVA, followed by a Tukey’s post hoc test, was used to compare multiple groups. The significance level was set to 0.05 (P < 0.05) for all comparisons.

4. Results

 

 

4.1. In Liver Tissue

Malathion caused a significant raise in LPO when compared to the controls (P < 0.05). AT caused a significant decrease in LPO when compared to the malathion group (P < 0.05). AT caused a significant decrease in SOD activity in comparison to the malathion group (P < 0.05). In addition, coadministration of AT with malathion significantly reduced malathion-induced SOD activity (P < 0.05). Therefore, coadministration of AT with malathion significantly reduced malathion induced LPO (P < 0.05). In comparison to the malathion group, AT caused a significant decrease in GPx activity (P < 0.05). Malathion caused a significant increase in CAT activity in contrast to the control group (P < 0.05) while AT caused a significant decrease in CAT activity in comparison to the malathion group (P < 0.05), (Table 1).

Table 1.
Oxidative Stress Biomarkers in Rat Liver and Liver Mitochondria a,b
4.2. Liver Mitochondria

Malathion and AT did not cause significant change on LPO in comparison to the other groups; however, malathion and AT did not cause significant change on SOD activity in contrast to the other groups. AT significantly reduced malathion-induced GPx activity (P < 0.05). Moreover, coadministration of AT with malathion significantly reduced malathion-induced CAT activity (P < 0.05). Administration of AT significantly decreased toxicity of the cells in comparison to the malathion group (P < 0.05). In addition, coadministration of AT with malathion significantly decreased malathion-induced mitochondria toxicity (P < 0.05), (Table 1).

5. Discussion

 

 

The main result of the present study was that malathion induced oxidative damages and mitochondrial dysfunction in rat liver. These results clearly indicate that malathion increased liver oxidative damage in rat by prompting of LPO, CAT, GPx, and SOD activities. Furthermore, AT was able to attenuate malathion-induced changes in almost all of the tested parameters. In addition, AT decreased the mitochondria toxicity in liver.The key function of mitochondria is producing energy, by oxidative phosphorylation and in the form of ATP, for cellular processes (27). This study reported the direct indications of damage in the liver mitochondria induced by malathion insecticide. It seems that the LPO byproducts induced a state of oxidative stress in mitochondria. Moreover, oxidative stress has been observed in some instinctive errors of metabolism (28), diseases (29-31), and pesticides poisoning (8, 9, 32, 33). In addition, both in vitro and in vivo studies showed that malathion toxicity is mediated by increased oxidants and oxidative injury (34), suggesting that malathion acts not only as a direct oxidant, but also as an initiator of oxidants (35). Latest studies have shown that the effects of acute and chronic exposure to malathion in oxidative stress induction is through the production of the ROS in the blood (36), liver (5), pancreas mitochondria (37), brain (38), and brain mitochondria (39) of rats. The results of previous studies showed that malathion poisoning results in oxidative stress condition, which is established by producing ROS, rise in cellular lipid peroxidation (LPO), and lastly, increase in level or activity of compensatory enzymatic and nonenzymatic antioxidant pathways (40). However, it has been revealed that malathion-induced oxidative stress modulates SOD and CAT activity in the liver (41). Our present results showed that malathion administration in rats caused significant induction in the activity of antioxidant enzymes, LPO, and increased mitochondria toxicity in the liver. In addition, malathion changed the antioxidant enzymes such as SOD, CAT, and GPx following subchronic exposure in animals (5, 42). Some studies have reported hepatotoxic effects of malathion exposure in both humans (43) and experimental animals (5, 42, 44); however, it has been confirmed that malathion-induced oxidative stress is due to the inactivation of mitochondrial respiratory complexes (39, 45). Our present results showed that malathion administration in rats caused significant mitochondrial toxicity.

Some studies have shown that mitochondria contains the highest concentration of AT (46), which accelerates ATP resynthesis in the ischemic heart tissue due to reperfusion (47). AT mostly acts as a chain-breaking antioxidant and radical scavenger, defending cell membrane against oxidative injury (48). Moreover, AT regulates production of ROS, maintains oxidative phosphorylation in the mitochondria, and accelerates damages of high-energy metabolites (49, 50). Finally, as evidenced by MTT assay, the present data verified that malathion-induced mitochondrial dysfunction could be modified by AT.

In conclusion, we reported that malathion induced a clear state of oxidative stress in liver that was in some way directly involved in the damage of hepatic mitochondria. In addition, AT was able to prevent the malathion-induced damage. These results proposed a possible mechanism to treat the oxidative injury caused by the OPs poisoning, in which the antioxidants such as AT may recover cell from injury. The present results showed that AT ‎could restore oxidant/antioxidant equilibrium in liver tissue and liver mitochondria and was also ‎capable of improving malathion-induced changes.

Footnotes

Implication for health policy/practice/research/medical education: This study proposed vitamin E as a new approach to the treatment of the oxidative damage caused by ‎the organophosphorus poisoning, in which the antioxidants might prevent cellular damage. ‎Results of the present study revealed that alpha-tocopheryl acetate ‎normalizes oxidant/antioxidant balance in liver ‎and therefore liver mitochondria is ‎able to recover malathion-induced changes.‎

References

  • 1. Ecobichon DJ. Pesticide use in developing countries. Toxicology. 2001;160(1-3):27-33. [PubMed]
  • 2. Cavari Y, Landau D, Sofer S, Leibson T, Lazar I. Organophosphate poisoning-induced acute renal failure. Pediatr Emerg Care. 2013;29(5):646-7. [DOI] [PubMed]
  • 3. Lee P, Tai DY. Clinical features of patients with acute organophosphate poisoning requiring intensive care. Intensive Care Med. 2001;27(4):694-9. [PubMed]
  • 4. Bunyan PJ, Jennings DM, Taylor A. Organophosphorus poisoning. Properties of avian esterases. J Agric Food Chem. 1968;16(2):326-31. [DOI]
  • 5. Akhgari M, Abdollahi M, Kebryaeezadeh A, Hosseini R, Sabzevari O. Biochemical evidence for free radical-induced lipid peroxidation as a mechanism for subchronic toxicity of malathion in blood and liver of rats. Hum Exp Toxicol. 2003;22(4):205-11. [PubMed]
  • 6. Aldridge WN, Miles JW, Mount DL, Verschoyle RD. The toxicological properties of impurities in malathion. Arch Toxicol. 1979;42(2):95-106. [PubMed]
  • 7. Murphy SD. Malathion inhibition of esterases as a determinant of malathion toxicity. J Pharmacol Exp Ther. 1967;156(2):352-65. [PubMed]
  • 8. Ranjbar A, Pasalar P, Abdollahi M. Induction of oxidative stress and acetylcholinesterase inhibition in organophosphorous pesticide manufacturing workers. Hum Exp Toxicol. 2002;21(4):179-82. [PubMed]
  • 9. Ranjbar A, Solhi H, Mashayekhi FJ, Susanabdi A, Rezaie A, Abdollahi M. Oxidative stress in acute human poisoning with organophosphorus insecticides; a case control study. Environ Toxicol Pharmacol. 2005;20(1):88-91. [DOI] [PubMed]
  • 10. Aruoma OI. Free radicals, oxidative stress, and antioxidants in human health and disease. J Am Oil Chem Soc. 1998;75(2):199-212. [DOI]
  • 11. Halliwell B. Free radicals and other reactive species in disease. Wiley Online Library. 2005. [DOI]
  • 12. Di Giulio RT. Indices of oxidative stress as biomarkers for environmental contamination. ASTM Special Technical Publication. 1991;(1124):15-31.
  • 13. Ott M, Gogvadze V, Orrenius S, Zhivotovsky B. Mitochondria, oxidative stress and cell death. Apoptosis. 2007;12(5):913-22. [DOI] [PubMed]
  • 14. Sastre J, Pallardo FV, Vina J. Mitochondrial oxidative stress plays a key role in aging and apoptosis. IUBMB Life. 2000;49(5):427-35. [DOI] [PubMed]
  • 15. Ingold KU, Webb AC, Witter D, Burton GW, Metcalfe TA, Muller DP. Vitamin E remains the major lipid-soluble, chain-breaking antioxidant in human plasma even in individuals suffering severe vitamin E deficiency. Arch Biochem Biophys. 1987;259(1):224-5. [PubMed]
  • 16. Bowry VW, Ingold KU, Stocker R. Vitamin E in human low-density lipoprotein. When and how this antioxidant becomes a pro-oxidant. Biochem J. 1992;288 ( Pt 2):341-4. [PubMed]
  • 17. Suleiman SA, Ali ME, Zaki ZM, el-Malik EM, Nasr MA. Lipid peroxidation and human sperm motility: protective role of vitamin E. J Androl. 1996;17(5):530-7. [PubMed]
  • 18. Meagher EA, Barry OP, Lawson JA, Rokach J, FitzGerald GA. Effects of vitamin E on lipid peroxidation in healthy persons. JAMA. 2001;285(9):1178-82. [PubMed]
  • 19. Ramasarma T. Generation of H2O2 in biomembranes. Biochim Biophys Acta. 1982;694(1):69-93.
  • 20. Ghazi-Khansari M, Mohammadi-Bardbori A, Hosseini MJ. Using Janus green B to study paraquat toxicity in rat liver mitochondria: role of ACE inhibitors (thiol and nonthiol ACEi). Ann N Y Acad Sci. 2006;1090:98-107. [DOI] [PubMed]
  • 21. Spitz DR, Oberley LW. An assay for superoxide dismutase activity in mammalian tissue homogenates. Anal Biochem. 1989;179(1):8-18. [PubMed]
  • 22. Mills GC. Glutathione peroxidase and the destruction of hydrogen peroxide in animal tissues. Arch Biochem Biophys. 1960;86:1-5. [PubMed]
  • 23. Aebi H. Catalase in vitro. Methods Enzymol. 1984;105:121-6. [PubMed]
  • 24. Ernster L, Nordenbrand K, Orrenius S, Das ML. Microsomal lipid peroxidation. Hoppe Seylers Z Physiol Chem. 1968;349(11):1604-5. [PubMed]
  • 25. Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 1976;72:248-54. [PubMed]
  • 26. Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods. 1983;65(1-2):55-63. [PubMed]
  • 27. Balaban RS, Nemoto S, Finkel T. Mitochondria, oxidants, and aging. Cell. 2005;120(4):483-95. [DOI] [PubMed]
  • 28. Ristoff E, Larsson A. Oxidative stress in inborn errors of metabolism: lessons from glutathione deficiency. J Inherit Metab Dis. 2002;25(3):223-6. [PubMed]
  • 29. Madamanchi NR, Vendrov A, Runge MS. Oxidative stress and vascular disease. Arterioscler Thromb Vasc Biol. 2005;25(1):29-38. [DOI] [PubMed]
  • 30. Christen Y. Oxidative stress and Alzheimer disease. Am J Clin Nutr. 2000;71(2):621-9.
  • 31. Jenner P. Oxidative stress in Parkinson's disease. Ann Neurol. 2003;53 Suppl 3:S26-36. discussion S36-8 [DOI] [PubMed]
  • 32. Banerjee BD, Seth V, Ahmed RS. Pesticide-induced oxidative stress: perspectives and trends. Rev Environ Health. 2001;16(1):1-40. [PubMed]
  • 33. Abdollahi M, Ranjbar A, Shadnia S, Nikfar S, Rezaie A. Pesticides and oxidative stress: a review. Med Sci Monit. 2004;10(6):RA141-7. [PubMed]
  • 34. Luo J, Shi R. Acrolein induces axolemmal disruption, oxidative stress, and mitochondrial impairment in spinal cord tissue. Neurochem Int. 2004;44(7):475-86. [PubMed]
  • 35. Adams JD, Jr., Klaidman LK. Acrolein-induced oxygen radical formation. Free Radic Biol Med. 1993;15(2):187-93. [PubMed]
  • 36. Abdollahi M, Mostafalou S, Pournourmohammadi S, Shadnia S. Oxidative stress and cholinesterase inhibition in saliva and plasma of rats following subchronic exposure to malathion. Comp Biochem Physiol C Toxicol Pharmacol. 2004;137(1):29-34. [DOI] [PubMed]
  • 37. Pournourmohammadi S, Ostad SN, Azizi E, Ghahremani MH, Farzami B, Minaie B, et al. Induction of insulin resistance by malathion: evidence for disrupted islets cells metabolism and mitochondrial dysfunction. Pestic Biochem Physiol. 2007;88(3):346-52. [DOI]
  • 38. Fortunato JJ, Feier G, Vitali AM, Petronilho FC, Dal-Pizzol F, Quevedo J. Malathion-induced oxidative stress in rat brain regions. Neurochem Res. 2006;31(5):671-8. [DOI] [PubMed]
  • 39. Ranjbar A, Ghahremani MH, Sharifzadeh M, Golestani A, Ghazi-Khansari M, Baeeri M, et al. Protection by pentoxifylline of malathion-induced toxic stress and mitochondrial damage in rat brain. Hum Exp Toxicol. 2010;29(10):851-64. [DOI] [PubMed]
  • 40. Mittler R. Oxidative stress, antioxidants and stress tolerance. Trends Plant Sci. 2002;7(9):405-10. [PubMed]
  • 41. John S, Kale M, Rathore N, Bhatnagar D. Protective effect of vitamin E in dimethoate and malathion induced oxidative stress in rat erythrocytes. J Nutr Biochem. 2001;12(9):500-4. [PubMed]
  • 42. Possamai FP, Fortunato JJ, Feier G, Agostinho FR, Quevedo J, Wilhelm Filho D, et al. Oxidative stress after acute and sub-chronic malathion intoxication in Wistar rats. Environ Toxicol Pharmacol. 2007;23(2):198-204. [DOI] [PubMed]
  • 43. El-Desoky G, Abdelreheem M, Al-Othman A, ALOthman Z, Mahmoud M, Yusuf K. Potential hepatoprotective effects of vitamin E and selenium on hepatotoxicity induced by malathion in rats. Afr J Pharm Pharmacol. 2012;6(11):806-13. [DOI]
  • 44. El-Gharieb MA, El-Masry TA, Emara AM, Hashem MA. Potential hepatoprotective effects of vitamin E and Nigella sativa oil on hepatotoxicity induced by chronic exposure to malathion in human and male albino rats. Toxicol Environ Chem . 2010;92(2):391-407. [DOI]
  • 45. Kalender S, Uzun FG, Durak D, Demir F, Kalender Y. Malathion-induced hepatotoxicity in rats: the effects of vitamins C and E. Food Chem Toxicol. 2010;48(2):633-8. [DOI] [PubMed]
  • 46. Karami-Mohajeri S, Abdollahi M. Mitochondrial dysfunction and organophosphorus compounds. Toxicol Appl Pharmacol. 2013;270(1):39-44. [DOI] [PubMed]
  • 47. Rafique R, Schapira AH, Coper JM. Mitochondrial respiratory chain dysfunction in ageing; influence of vitamin E deficiency. Free Radic Res. 2004;38(2):157-65. [PubMed]
  • 48. Janero DR. Therapeutic potential of vitamin E against myocardial ischemic-reperfusion injury. Free Radic Biol Med. 1991;10(5):315-24. [PubMed]
  • 49. Burton GW, Joyce A, Ingold KU. Is vitamin E the only lipid-soluble, chain-breaking antioxidant in human blood plasma and erythrocyte membranes? Arch Biochem Biophys. 1983;221(1):281-90. [PubMed]
  • 50. Cheng G, Zielonka J, McAllister DM, Mackinnon AC, Jr., Joseph J, Dwinell MB, et al. Mitochondria-targeted vitamin E analogs inhibit breast cancer cell energy metabolism and promote cell death. BMC Cancer. 2013;13:285. [DOI] [PubMed]