Skip to main content
  • Research article
  • Open access
  • Published:

CCl4-induced hepatotoxicity: protective effect of rutin on p53, CYP2E1 and the antioxidative status in rat

Abstract

Background

Rutin is a polyphenolic natural flavonoid which possesses antioxidant and anticancer activity. In the present study the hepatoprotective effect of rutin was evaluated against carbon tetrachloride (CCl4)-induced liver injuries in rats.

Methods and materials

24 Sprague–Dawley male rats were equally divided into 4 groups for the assessment of hepatoprotective potential of rutin. Rats of group I (control) received only vehicles; 1 ml/kg bw of saline (0.85%) and olive oil (3 ml/kg) and had free access to food and water. Rats of group II, III and IV were treated with CCl4 (30% in olive oil, 3 ml/kg bw) via the intraperitoneal route twice a week for four weeks. The rutin at the doses of 50 and 70 mg/kg were administered intragastrically after 48 h of CCl4 treatment to group III and IV, respectively. Protective effect of rutin on serum enzyme level, lipid profile, activities of antioxidant enzymes and molecular markers were calculated in CCl4-induced hepatotoxicity in rat.

Results

Rutin showed significant protection with the depletion of alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), gamma glutamyl transpeptidase (γ-GT) in serum as was raised by the induction of CCl4. Concentration of serum triglycerides, total cholesterol and low density lipoproteins was increased while high-density lipoprotein was decreased with rutin in a dose dependent manner. Activity level of endogenous liver antioxidant enzymes; catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GSHpx), glutathione-S-transferase (GST) and glutathione reductase (GSR) and glutathione (GSH) contents were increased while lipid peroxidation (TBARS) was decreased dose dependently with rutin. Moreover, increase in DNA fragmentation and oxo8dG damages while decrease in p53 and CYP 2E1 expression induced with CCl4 was restored with the treatment of rutin.

Conclusion

From these results, it is suggested that rutin possesses hepatoprotective properties.

Peer Review reports

Background

Exposure to toxic chemicals, environmental pollutants and drugs can cause cellular injuries through metabolic activation of reactive oxygen species (ROS) [1]. Carbon tetrachloride (CCl4) has been used extensively to study hepatotoxicity in animal models by initiating lipid peroxidation, thereby causing injuries to kidney, heart, testis and brain [2–4], in addition to liver pathogenesis [5]. Liver is particularly susceptible to oxidative stress due to the direct release of CCl4 metabolites and cytokines, which propagate inflammatory response [6]. CCl4 is one of the xenobiotics that has been reported to induce acute and chronic tissue injuries [7, 8] through bioactivation of the phase I cytochrome P450 system to form reactive metabolic trichloromethyl radicals (·CCl3) and peroxy trichloromethyl radicals (·OOCCl3). These free radicals can covalently bind to macromolecules such as proteins, lipids and nucleic acids. The double allylic hydrogen bonds of polyunsaturated fatty acid (PUFA) are susceptible to abstraction by free radicals; CCl4 exposure induces an increase in lipoperoxide and free peroxide radical concentrations that are highly reactive and cause injury or necrosis [9, 10].

An increase in unsaturated fatty acid lipoperoxide and free peroxide radical concentrations [11, 12], can induce alterations in the cholesterol profile and decrease in hepatic antioxidant enzymes [13], in addition to induction of oxidative DNA damage including formation of DNA adducts, genetic mutations, strand breakage and chromosomal alterations [14]. These free radicals can cause depletion of CYP2E1 activity [15] and increase in oxo8dG concentration in tissues of experimental animals [16]. DNA fragmentation induces p53 gene expression, blocks cells in the G phase of the cell cycle, and gives additional time for DNA repair, while severe DNA damage triggers apoptosis [17]. It has been reported that CCl4 administration increases the silver-stained nucleolar organizer region, alters its size, morphology or spreading in the nucleus, which may be utilized as an indicator of genotoxicity, neoplasia and hyperplasia to complement other histological procedures [11].

Flavonoids are a large group of polyphenolic compounds that play an important role in detoxification of free radicals and are markedly found in fruits, vegetables and medicinal plants [18, 19]. Glycosidic flavonoids such as rutin are much more readily absorbed by humans than aglycones [20, 21]. Rutin possesses antitumor [22], anti-inflammatory [23] and antimutagenic potential [24], besides myocardial protection [25] and immunomodulating activities [26]. Therefore, the present study was designed to investigate the hepatoprotective effects of rutin against CCl4-induced oxidative stress and its role in alleviation of lipid peroxidation and restoration of p53 and CYP2E1 activity.

Methods

Drugs and chemicals

Reduced glutathione (GSH), oxidized glutathione (GSSG), glutathione reductase, gamma-glutamyl p-nitroanilide, glycylglycine, bovine serum albumin (BSA), 1,2-dithio-bis nitro benzoic acid (DTNB), 1-chloro-2,4-dinitrobenzene (CDNB), reduced nicotinamide adenine dinucleotide phosphate (NADPH), rutin, CCl4, flavine adenine dinucleotide (FAD), glucose-6-phosphate, Tween-20, 2,6-dichlorophenolindophenol, thiobarbituric acid (TBA), picric acid, sodium tungstate, sodium hydroxide, trichloroacetic acid (TCA) and perchloric acid (PCA) were purchased from Sigma Chemicals Co. USA.

Animals and treatment

Six week old, 24 Sprague–Dawley male rats (200–210 g) were provided by National Institute of Health Islamabad and were kept in ordinary cages at room temperature of 25±3°C with a 12 h dark/light cycles. They have free access to standard laboratory feed and water, according to the study protocol approved by Ethical Committee of Quaid-i-Azam University Islamabad for animal care and experimentation. To study the hepatoprotective effects of rutin, rats were equally divided into four groups (six rats). Animals of group I were treated with 1 ml/kg bw of saline (0.85%) intragastrically and olive oil (3 ml/kg bw) intraperitoneally twice a week for four weeks. Rats of group II, III and IV were treated with CCl4 (30% in olive oil) at a dose of 3 ml/kg bw intraperitoneally twice a week for four weeks. Animals of group II received only CCl4 treatment. However, animals of group III and IV received rutin at a dose of 50 and 70 mg/kg bw intragastrically, respectively, in addition to CCl4 treatment, twice a week for four weeks.

After 24 h of the last treatment, all the animals were weighted, sacrificed, collected the blood while liver was removed, weighted and perfuse in ice-cold saline solution. Liver samples were treated with liquid nitrogen and stored at −70 °C for further studies.

Assessment of hepatotoxicity

Liver marker enzymes (alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), gamma glutamyl transpeptidase (γ-GT), lipid profile [total cholesterol, low-density lipoprotein (LDL), high-density lipoprotein (HDL) and triglyceride] were estimated by using standard AMP diagnostic kits (Stattogger Strasse 31b 8045 Graz, Austria). CYP 2E1 (cat no: E90988Ra, Uscn Life Science Inc.), oxo8dG and p53 (PUMA ELISA Kit cat no: E91909Ra) concentration was determined with ELISA kit.

Assessment of oxidative stress

For determination of oxidative stress liver tissue was homogenized in 10 volumes of 100 mmol KH2PO4 buffer containing 1 mmol EDTA (pH 7.4) and centrifuged at 12,000 × g for 30 min at 4 °C. The supernatant was collected and used for the determination of protein and enzymatic studies as described below. Protein concentration was determined by using crystalline BSA as standard. CAT and SOD activities are determined with protocol of [27, 28] while phase II metabolizing enzyme, including glutathione-S-transferase (GST), glutathione reductase (GSR), glutathione peroxidase (GSH-Px), reduced glutathione (GSH) [29–32] and thiobarbituric acid reactive substances (TBARS) contents, respectively [33].

DNA damages

Hepatic DNA damages (fragmentation % by DPA assay), DNA ladder assay [34] and number of NORs per cell [35] were determined.

Statistical analysis

To determine the treatment effects, one-way analysis of variance was carried by computer software SPSS 13.0. Level of significance among the various treatments was determined by LSD at 0.05% and 0.01% level of probability.

Results

Body weight, liver weight

Treatment of CCl4 caused significant reduction (P< 0.01) in body weight while increased the absolute liver and relative liver weight comparatively to control group was significantly (P< 0.01) restored with 50 mg/kg bw and 70 mg/kg bw treatment of rutin (Table 1).

Table 1 Effect of Rutin on absolute liver weight, relative liver weight, and % increase in body weight

Lipids profile

Administration of CCl4 increased triglycerides, total cholesterol, LDL while decreased the HDL as shown in Table 2. Concentration of HDL was significantly (P<0.01) increased by rutin whereas concentration of triglycerides, total cholesterol and LDL was appreciably (P< 0.01) augmented to compensate the CCl4-induced toxicity.

Table 2 Effect of Rutin on Lipids profile

Genotoxicity studies

Exposure of CCl4 elicited the hepatic DNA damages (% fragmentation) and number of AgNORs/cell. Percent serum level of oxo8dG was increased whereas the % activity level of p53 and CYP 2E1 was decreased in hepatic samples of rat. Treatment of rats with 50 mg/kg bw and 70 mg/kg bw of rutin restored the level of these markers (Table 3). DNA ladder assay showed conformity to the DNA fragmentation assay (Figure 1).

Table 3 Effect of Rutin on Genotoxicity studies
Figure 1
figure 1

Protective effects of rutin on DNA; Lane 1–4 (non treated control), 5–8 (CCl 4 treated rats), 9,10 (CCl 4 +50 mg/kg b. w. rutin), 11,12 (CCl 4 +70 mg/kg b.w. rutin).

Indices of hepatotoxicity

Administration of CCl4 markedly increased (P<0.01) the activity of liver serum marker enzymes such as AST, ALT, ALP and γ-GT as compared with the control group. Elevations in the secretion of these enzymes were significantly decreased (P<0.01) by 50 mg/kg bw and 70 mg/kg bw of rutin as compared to the CCl4 group are shown in Table 4.

Table 4 Effect of Rutin on Indices of hepatotoxicity

Assessment of oxidative stress

CCl4 treatment in rats significantly decreased (P< 0.01) the activity of CAT, SOD, GST, GSH-Px, GSR, GSH while increased TBARS contents in liver samples. The increase of lipid peroxidation caused; reduction in the activities of antioxidant enzymes and glutathione (GSH) contents were markedly attenuated (P< 0.01) by administration of 50 mg/kg bw and 70 mg/kg bw of rutin in intoxicated rats (Table 5).

Table 5 Effect of Rutin on assessment of oxidative stress

Discussion

The fields of dietary modification and chemoprevention show considerable effective approaches against oxidative stress and are the focus of research these days [36]. Various studies have shown that several mutagens and carcinogens cause generation of oxygen-free radicals, which play a major role in the emergence of cancer and other health disturbances [37, 38]. The present study revealed that CCl4-induction in rats remarkably increased the level of ALT, AST, ALP and γ-GT. CCl4 causes acute hepatocyte injuries, altered membrane integrity and as a result enzymes in hepatocytes leak out [39]. However, after treatment with rutin, the pathological increases in ALT, AST, ALP and γ-GT were significantly restored. These results indicate that rutin has the ability to protect against CCl4-induced hepatocyte injury, which is in agreement with a previous study [40] that reported the protective consequence of polyphenolic compounds against CCl4-induced liver cirrhosis. Importantly, the increased serum concentrations of triglycerides, total cholesterol and LDL, and the decreased level of HDL, were restored to normal values with rutin co-treatment. This may be explained on the basis that rutin has a strong ability to chelate multivalent metal ions, especially zinc, calcium and iron. Indeed, its ability to chelate minerals has been reported to have some protective effects, such as decreasing iron mediated free radical formation and lowering serum cholesterol, triglycerides and lipid peroxides in experimental animals [41]. Similar findings were reported in another study [42] that investigated the hepatoprotective effects of plant bioactive compounds against CCl4-induced hepatic injury in rats.

ROS formed during the biotransformation process of CCl4 are more reactive and toxic than the parental compound. Biotransformation of CCl4 occurs in the endoplasmic reticulum and the isoenzyme implicated in this process is CYP2E1 [43, 44]. Our results showed that the active free radical/intermediate of CCl4 caused a reduction in CYP2E1, which was markedly restored by rutin treatment. Our results showed conformity with previous investigations, which demonstrated that the polyphenolic natural product is responsible for its protective, effects [45, 46].

Results of the present study revealed that exposure of rats to CCl4 resulted in depletion of antioxidant activities. In consonance with our results, Szymonik-Lesiuk et al.[1] reported that CCl4 intoxication leads to changes in antioxidant enzymes and reactive intermediates involved in the bioactivation of CCl4 that may truss to those enzymes to prevent their inactivation. Furthermore, our results correspond with [11, 12], and are in agreement with an investigation following CCl4 intoxication [47].

Glutathione provides a first line of defense and scavenges free radical oxygen species (ROS). The decreased concentration of GSH in liver may be due to NADPH reduction or GSH utilization in the exclusion of peroxides [48]. GSH-dependent enzymes offer a second line of protection as they primarily detoxify noxious byproducts generated by ROS and help to avert dissemination of free radicals [49]. GSH-Px detoxifies peroxides by reacting with GSH and converting it into GSSG, which is reduced to GSH by GSR [50]. Our study revealed that CCl4 treatment in rats markedly changed the activity of antioxidant enzymes, which was reverted by the co-administration of rutin. Thiobarbituric acid reactive substances (TBARS), the final metabolites of peroxidized polyunsaturated fatty acids, are considered as a late biomarker of oxidative stress [51]. In our experiments, major decrease in lipid peroxidation and consequent reduction in TBARS were obtained by treatment with rutin. The increment in lipid peroxidation, as assessed by the elevated levels of TBARS following CCl4 administration, has been well documented [12]. Data of the present study indicated that lipid peroxidation induced by oxidative stress caused DNA damage. TBARS react with the DNA strand to form the M1G adduct, the mutagenic pirimedopurinone adduct of deoxyguanosine [52]. Administration of rutin markedly reduced the DNA damage, which is in close agreement with a previous study [11]. This level of DNA damage decreases the expression of p53 and blocks cells in the G phase of the cell cycle, which gives the cells additional time to repair the DNA damage. However, severe DNA damage may elicit apoptosis [53]. The data revealed that CCl4-induction caused marked reduction in p53. This result may be explained on the basis that CCl4 acts as a tumor promoter through increasing the intracellular concentration of ROS necrosis/regeneration and cell proliferation and/or may be due to mutation of p53. Our results regarding p53 are in agreement with previous studies [54, 55].

Conclusion

These results demonstrate that administration of rutin may be useful in the treatment and prevention of hepatic stress.

References

  1. Szymonik-Lesiuk SM, Czechowska M, Stryjecka-Zimmer M, Somka A, Madro K, Celiski M, Wielosz M: Catalase, superoxide dismutase, and glutathion peroxidase activities in various rat tissues after carbon tetrachloride intoxication. J. Hepatobil. Pancr. Surg. 2003, 10: 309-315. 10.1007/s00534-002-0824-5.

    Article  Google Scholar 

  2. Tirkey NG, Kaur G, Vij K, Chopra K: Hesperidin, a citrus bioflavonoid, decreases the oxidative stress produced by carbon tetrachloride in rat liver and kidney. BMC Pharmacol. 2005, 5: 15-21. 10.1186/1471-2210-5-15.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Preethi KC, Kuttan R: Hepato and reno protective action of Calendula officinalis L. flower extract. Indian J. Exp. Biol. 2009, 47: 163-168.

    CAS  PubMed  Google Scholar 

  4. Khan RA, Khan MR, Sahreen S: Evaluation of Launaea procumbens use in renal disorders: A rat model. J. Ethanopharmacol. 2010, 128: 452-461. 10.1016/j.jep.2010.01.026.

    Article  Google Scholar 

  5. Murugesan GS, Sathishkumar M, Jayabalan R, Binupriya AR, Swaminathan K, Yun SEZ: Hepatoprotective and curative properties of Kombucha tea against carbon tetrachloride-induced toxicity. J Microbiol Biotechnol. 2009, 19: 397-402. 10.4014/jmb.0806.374.

    Article  CAS  PubMed  Google Scholar 

  6. Werner M, Costa MJ, Mitchell LG, Nayar R: Nephrotoxicity of xenobiotics. Review Clin. Chim. Acta. 1995, 237: 107-154. 10.1016/0009-8981(95)06068-O.

    Article  CAS  PubMed  Google Scholar 

  7. Ogeturk M, Kus I, Colakoglu N, Zararsiz I, Ilhan N, Sarsilmaz M: Caffeic acid phenethyl ester protects kidneys against carbon tetrachloride toxicity in ratsJ. Ethnopharmacol. 2005, 97: 273-280. 10.1016/j.jep.2004.11.019.

    Article  CAS  Google Scholar 

  8. Jaramillo-Juarez F, Rodriguez-Vazquez ML, Rincon-Sanchez AR, Consolacion M, Martinez GG, Ortiz J, Reyes JL: Caffeic acid phenethyl ester against carbon tetrachloride toxicity in rats. Ann Hepatol. 2008, 7: 331-338.

    CAS  PubMed  Google Scholar 

  9. Weber LW, Boll M, Stampfl M: Hepatotoxicity and mechanism of action of haloalkanes: carbon tetrachloride as a toxicological model. Crit. Revw. Toxicol. 2003, 33: 105-136. 10.1080/713611034.

    Article  CAS  Google Scholar 

  10. Miyazaki T, Bouscarel B, Ikegami T, Honda A, Matsuzaki Y: The protective effect of taurine against hepatic damage in a model of liver disease and hepatic stellate cells. Adv. Exp. Med. Biol. 2009, 643: 293-303. 10.1007/978-0-387-75681-3_30.

    Article  CAS  PubMed  Google Scholar 

  11. Khan MR, Rizvi W, Khan GN, Khan RA, Shaheen S: Carbon tetrachloride induced nephrotoxicity in rat: Protective role of Digera muricata. J Ethnopharmacol. 2009, 122: 91-99. 10.1016/j.jep.2008.12.006.

    Article  CAS  PubMed  Google Scholar 

  12. Khan RA, Daud A: Protective effects of Digera muricata (L.) Mart. on testis against oxidative stress of carbon tetrachloride in rat. Food Chem. Toxicol. 2009, 47: 1393-1399. 10.1016/j.fct.2009.03.020.

    Article  CAS  PubMed  Google Scholar 

  13. Adewole SO, Salako AA, Doherty OW, Naicker T: Effect of melatonin on carbon tetrachloride-induced kidney injury in Wistar rats. African J. Biomed. Res. 2007, 10: 153-164.

    Google Scholar 

  14. Jia X, Han C, Chen J: Effect of tea on preneoplastic lesions and cell cycle regulators in rat liver. Cancer Epidemiol. Biomark. Preven. 2002, 11: 1663-1667.

    CAS  Google Scholar 

  15. Fahmy SR, Hamdi SAH, Abdel-Salam HA: Curative effect of dietary freshwater and marine crustacean extracts on carbon tetrachloride-induced nephrotoxicity. Australian J. Basic Appl. Sci. 2009, 3: 2118-2129.

    CAS  Google Scholar 

  16. Sai K, Tyson CA, Thomas DW, Dabbs JE, Hasegawa R, Kurokawa Y: Oxidative DNA damage induced by potassium bromate in isolated rat renal proximal tubules and renal nuclei. Cancer Lett. 1994, 87: 1-7. 10.1016/0304-3835(94)90402-2.

    Article  CAS  PubMed  Google Scholar 

  17. Khan RA: Protective effect of Launaea procumbens (L.) on lungs against CCl4 induced toxicity in rat. BMC Compl Alter Med. 2012, 12: 133-10.1186/1472-6882-12-133.

    Article  Google Scholar 

  18. Potter JD: Cancer prevention: epidemiology and experiment. Cancer Lett. 1997, 114: 7-9. 10.1016/S0304-3835(97)04615-6.

    Article  CAS  PubMed  Google Scholar 

  19. Rice-Evans C, Packer L: Flavonoids in health and diseases. 1998, New York: Marcel Decker Inc., 483-522.

    Google Scholar 

  20. Hollman PC, Katan MB: Absorption, metabolism and bioavailability of flavonoids. Flavonoids in Health and Disease. Edited by: Rice-Evans C, Packer L. 1998, New York: Marcel Decker, 483-522.

    Google Scholar 

  21. Nakamura Y, Ishimitsu S, Tonogai Y: Effects of quercetin and rutin on serum and hepatic lipid concentrations, fecal steroid excretion and serum antioxidant properties. J. Health Sci. 2000, 46: 229-240. 10.1248/jhs.46.229.

    Article  CAS  Google Scholar 

  22. Deschner EE, Ruperto J, Wong G, Newmark HL: Quercetin and rutin as inhibitors of azoxymethanol-induced colonic neoplasia. Carcinogenesis. 1991, 12: 1193-1196. 10.1093/carcin/12.7.1193.

    Article  CAS  PubMed  Google Scholar 

  23. Aleksandrov PN, Speranskaia TV, Bobkov Iu G, Zagorevskii VA, Zykov DA: Effect of rutin and esculamine on models of aseptic inflammation. Farmakol. Toksikol. 1986, 49: 84-86.

    CAS  PubMed  Google Scholar 

  24. Bear WL, Teel RW: Effects of citrus flavonoids on the mutagenicity of heterocyclic amines and on cytochrome P450 1A2 activity. Anticancer Res. 2000, 20: 3609-3614.

    CAS  PubMed  Google Scholar 

  25. Pozin VM, Skuratovskaia SG, Pocheptsova GA: Changes in the vascular wall and ischemic damages to the myocardium in reversible episodes of heart muscle ischemia. Fiziologic. Zhur. 1996, 42: 10-16.

    CAS  Google Scholar 

  26. Chen S, Gong J, Liu F, Mohammed U: Naturally occurring polyphenolic antioxidants modulate IgE-mediated mast cell activation. Immunology. 2000, 100: 471-480. 10.1046/j.1365-2567.2000.00045.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Chance B, Maehly AC: Assay of catalase and peroxidases. Methods Enzymol. 1955, 11: 764-775.

    Article  Google Scholar 

  28. Kakkar P, Das B, Viswanathan PN: A modified spectrophotometric assay of superoxide dismutase. Indian J. Biochem. Biophys. 1984, 21: 130-132.

    CAS  PubMed  Google Scholar 

  29. Habig WH, Pabst MJ, Jakoby WB: Glutathione-S-transferases: the first enzymatic step in mercapturic acid formation. J Biol Chem. 1974, 249: 7130-7139.

    CAS  PubMed  Google Scholar 

  30. Carlberg I, Mannervik EB: Glutathione level in rat brain. J Biol Chem. 1975, 250: 4475-4480.

    Google Scholar 

  31. Mohandas J, Marshal JJ, Duggin GG, Horvath JS, Tiller DJ: Differential distribution of glutathione and glutathione-related enzymes in rabbit kidney. Possible implications in analgesic nephropathy. Biochem Pharmacol. 1984, 33: 1801-1807. 10.1016/0006-2952(84)90353-8.

    Article  CAS  PubMed  Google Scholar 

  32. Jollow DJ, Mitchell JR, Zampaglione N, Gillete JR: Bromobenzene induced liver necrosis. Protective role of glutathione and evidence for 3, 4-bromobenzene oxide as a hepatotoxic metabolite. Pharmacol. 1974, 11: 151-169. 10.1159/000136485.

    Article  CAS  Google Scholar 

  33. Iqbal M, Sharma SD, Zadeh HR, Hasan N, Abdulla M, Athar M: Glutathione metabolizing enzymes and oxidative stress in ferric nitrilotriacetate (Fe-NTA) mediated hepatic injury. Redox Report. 1996, 2: 385-391.

    CAS  Google Scholar 

  34. Wu B, Ootani A, Iwakiri R, Sakata Y, Fujise T, Amemori S, Yokoyama F, Tsunada S, Fujimoto K: T cell deficiency leads to liver carcinogenesis in Azoxymethane-treated rats. Exp. Biol. Med. 2005, 231: 91-98.

    Google Scholar 

  35. Trere D, Zilbering A, Dittus D, Kim P, Ginsberg PC, Daskal I: AgNOR quantity in needle biopsy specimens of prostatic adenocarcinomas: correlation with proliferation state, Gleason score, clinical stage, and DNA content. Clin Mol Pathol. 1996, 49: 209-213. 10.1136/mp.49.4.M209.

    Article  Google Scholar 

  36. Aruoma OI: Methodological consideration for characterizing potential antioxidant action of bioactive components in plants foods. Mutat Res. 2003, 523–524: 9-20.

    Article  PubMed  Google Scholar 

  37. Sun Y: Free radicals, antioxidant enzymes and carcinogenesis. Free Radic Biol Med. 1990, 8: 583-599. 10.1016/0891-5849(90)90156-D.

    Article  CAS  PubMed  Google Scholar 

  38. Aleynick SI, Leo MA, Ma X, Aleynick MK: Polyenoyl phasphatidylcholine prevents CCl4 induced lipid peroxidation while C.S. it attenuates liver fibrosis. J Hepatol. 1997, 27: 554-561. 10.1016/S0168-8278(97)80361-3.

    Article  Google Scholar 

  39. Cheng HL, Hu YY, Wang RP, Liu C, Liu P, Zhu DY: Protective actions of salvianolic acid on hepatocyte injured by peroxidation in vitro. World J Gastroenterol. 2000, 6: 402-404.

    Article  Google Scholar 

  40. Xiao-hui H, Liang-qi C, Xi-ling C, Kai S, Yun-jian L, Long-juan Z: Polyphenol epigallocatechin-3-gallate inhibits oxidative damage and preventive effects on carbon tetrachloride–induced hepatic fibrosis. Nutr. Biochem. 2007, 3: 511-515.

    Google Scholar 

  41. Zhou JR, Erdman JWJ, Zhou JR, Erdman JWJ: Phytic acid in health and disease. Crit. Rev. Food Sci. Nutr. 1995, 35: 495-508. 10.1080/10408399509527712.

    Article  CAS  PubMed  Google Scholar 

  42. Sreelatha S, Padma PR, Umadevi M: Protective effects of Coriandrum sativume xtracts on CCl4- induced hepatotoxicity in rats.Food Chem. Toxicol. 10.1016/j.fct.2009.12.022.

  43. Recknagel RO, Glande EAJ: Carbon tetrachloride hepatotoxicity: an example of lethal cleavage. Critical. Rev. Toxicol. 1973, 2: 263-297. 10.3109/10408447309082019.

    Article  Google Scholar 

  44. Al-Shabanah OA, Alam K, Nagi MN, Al-Rikabi AC, Al-Bekairi AM: Protective effect of aminoguanidine, a nitric oxide synthetase inhibiter against CCl4 induced hepatotoxicity in mice. Life Sci. 2000, 66: 265-270.

    Article  CAS  PubMed  Google Scholar 

  45. Janbaz KH, Gilani AH: Evaluation of protective potential of Artemisia maritima extract on acetaminophen and CCl4 induced liver damage. J Ethnopharmacol. 1955, 47: 43-47.

    Article  Google Scholar 

  46. Chandan BK, Sharma AK, Anand KK: Boerhaavia diffusa: a study of its hepatoprotective activity. J Ethnopharmacol. 1991, 31: 299-307. 10.1016/0378-8741(91)90015-6.

    Article  CAS  PubMed  Google Scholar 

  47. Manna P, Sinha M, Sil PC: Aqueous extract of Terminalia arjuna prevents carbon tetrachloride induced hepatic and renal disorders. BMC Compl. Altern. Med. 2007, 6: 33-37.

    Article  Google Scholar 

  48. Yadav P, Sarkar S, Bhatnagar D: Action of Capparis deciduas against alloxan-induced oxidative stress and diabetes in rat tissues. Pharmacol Res. 1997, 36: 221-228. 10.1006/phrs.1997.0222.

    Article  CAS  PubMed  Google Scholar 

  49. Gumieniczek A: Effects of repaglinide on oxidative stress in tissues of diabetic rabbits. Diab. Res. Clin. Pract. 2005, 68: 89-95. 10.1016/j.diabres.2004.09.018.

    Article  CAS  Google Scholar 

  50. Maritim AC, Sanders RA, Watkins JB: Effects of α-lipoic acid on biomarkers of oxidative stress in streptozotocin-induced diabetic rats. J Nutr Biochem. 2003, 14: 288-294. 10.1016/S0955-2863(03)00036-6.

    Article  CAS  PubMed  Google Scholar 

  51. Cheeseman KH: Mechanisms and effects of lipid peroxidation. Mol Aspects Med. 1993, 14: 191-197. 10.1016/0098-2997(93)90005-X.

    Article  CAS  PubMed  Google Scholar 

  52. Marnett JL: Oxyridicals and DNA damage. Carcinogenesis. 2000, 21: 61-70.

    Article  Google Scholar 

  53. Van Gijssel HE, Maassen CB, Mulder GJ, Meerman JH: P53 protein expression by hepatocarcinogens in the rat liver and its potential role in mitoinhibition of normal hepatocytes as a mechanism of hepatic tumor promotion. Carcinogenesis. 1997, 18: 1027-1031. 10.1093/carcin/18.5.1027.

    Article  CAS  PubMed  Google Scholar 

  54. Farazi PA, Zeisberg M, Glickman J, Zhang Y, Kalluri R, DePinh RA: Chronic bile duct injury associated with fibrotic matrix micro environment provokes cholangiocarcinoma in p53-deficient mice. Nutri. Biochem. 2006, 6: 6622-66277.

    Google Scholar 

  55. Kuo PL, Lin C, Kuo PL, Lin CC: Green tea constituent (−) - epigallocatechin-3-gallate inhibits Hep G2 cell. Proliferaration and induces apoptosis through p53-dependent and fas-mediated pathways. J Biomed Sci. 2003, 10: 219-27.

    CAS  PubMed  Google Scholar 

Pre-publication history

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Rahmat A Khan.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

RAK made a significant contribution to conception and design of the study, acquisition and analyses of data and drafting of the manuscript. MRK and SS made contribution in sample collection and design. All the authors read the revised manuscript and approved.

Rahmat A Khan, Muhammad R Khan and Sumaira Sahreen contributed equally to this work.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Khan, R.A., Khan, M.R. & Sahreen, S. CCl4-induced hepatotoxicity: protective effect of rutin on p53, CYP2E1 and the antioxidative status in rat. BMC Complement Altern Med 12, 178 (2012). https://doi.org/10.1186/1472-6882-12-178

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1472-6882-12-178

Keywords