Mol Biol Rep DOI 10.1007/s11033-014-3292-5

Protective effect of ellagic acid against TCDD-induced renal oxidative stress: Modulation of CYP1A1 activity and antioxidant defense mechanisms Viswanadha Vijaya Padma • Palaniswamy Kalai Selvi Samadi Sravani



Received: 16 March 2013 / Accepted: 13 February 2014 Ó Springer Science+Business Media Dordrecht 2014

Abstract 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) belongs to toxicologically important class of poly halogenated aromatic hydrocarbons and produce wide variety of adverse effects in humans. The present study investigated the protective effect of ellagic acid, a natural polyphenolic compound against TCDD-induced nephrotoxicity in Wistar rats. TCDD-induced nephrotoxicity was reflected in marked changes in the histology of kidney, increase in levels of kidney markers (serum urea, serum creatinine) and lipid peroxides. A significant increase in activity of phase I enzyme CYP1A1 with concomitant decline in the activities of phase II enzymes [non-enzymic antioxidant and various enzymic antioxidants such as superoxide dismutase, catalase, glutathione peroxidase, glutathione-stransferase] was also observed. In addition, TCDD treated rats showed alterations in ATPase enzyme activities such as Na? K?-ATPase, Mg2? ATPase and Ca2? ATPase. Oral pre-treatment with ellagic acid prevented TCDD-induced alterations in levels of kidney markers. Ellagic acid pretreatment significantly counteracted TCDD-induced oxidative stress by decreasing CYP1A1 activity and enhancing the antioxidant status. Furthermore, ellagic acid restored TCDD-induced histopathological changes and alterations in ATPase enzyme activities. The results of the present study show that significant protective effect rendered by ellagic acid against TCDD-induced nephrotoxicity might be attributed to its antioxidant potential.

V. Vijaya Padma (&)  P. Kalai Selvi  S. Sravani Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore 641046, Tamil Nadu, India e-mail: [email protected]

Keywords ATPase  CYP1A1  Oxidative stress  Reactive oxygen species  TCDD Abbreviations AhR Aryl hydrocarbon receptor ARNT Aryl hydrocarbon receptor nuclear translocator CAT Catalase EA Ellagic acid GPx Glutathione peroxidase GSH Glutathione GST Glutathione-s-transferase ROS Reactive oxygen species SOD Superoxide dismutase TCDD 2,3,7,8-Tetrachlorodibenzo-p-dioxin XRE Xenobiotic responsive elements

Introduction 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is one of the most toxic and the representative compound of dioxins. TCDD exposure results in adverse effects such as wasting syndrome, dermal toxicity, immunotoxicity, hepatotoxicity, carcinogenicity, teratogenicity, reproductive disorders, endocrine disruption, neurotoxicity and numerous other biochemical alterations [1–3]. TCDD is unintentionally produced from various manufacturing industries and during incineration of municipal and industrial wastes. The released TCDD and their congeners, are found in the environment including air, food, and soil. The exposure of TCDD among the general population mainly occurs through dietary consumption of meat, dairy products and fish. Since, TCDD is lipophilic in nature it tends to bioaccumulate in the food

123

Mol Biol Rep

chain. The half-life of TCDD is estimated to be 5.8–11.3 years in humans [4]. Adverse health effects induced by TCDD are mediated through aryl hydrocarbon receptor (AhR) signaling mechanism [5, 6]. The AHR is a ubiquitous cytosolic protein, which gets activated upon binding to TCDD and translocates into nucleus and dimerizes with the aryl hydrocarbon receptor nuclear translocator (ARNT). The AhR/ARNT heterodimer binds to xenobiotic responsive elements (XREs) and activates transcription of numerous phase I and II xenobiotic-metabolizing genes. One of the most sensitive targets of this heterodimer is CYP1A1 gene [7]. Cytochrome P450 is a super family of microsomal hemoproteins involved in the process of chemically induced carcinogenesis involved in conversion of procarcinogens into ultimate carcinogens [8]. During such metabolic process it generates reactive oxygen species and initiates oxidative stress [9]. ROS mediates toxic responses by creating an atmosphere of oxidative stress by increased lipid peroxidation and decreased antioxidant status [10]. Ellagic acid (EA), a natural polyphenolic antioxidant found in high amounts in the fruits and seeds of raspberries, strawberries, blueberries, pomegranates, walnuts and other plant foods, is being hailed as a potent antioxidant, possessing broad chemo protective properties against a variety of different carcinogens [11]. EA has been found to possess cardioprotecive, hepatoprotective and nephroprotective properties against various conditions of oxidative stress [12– 15]. Recently Hseu et al. [16] have reported that Nrf-2 is believed to play a role in protective effect of EA against UVA induced oxidative stress and apoptosis. Previous reports have documented that EA exhibits better protection than vitamin E against TCDD- induced fetal growth redardation [17]. However TCDD-induced nephrotoxicity is sparsely reported in the literature and protection offered by antioxidants against these effects is not well understood. So the present study, aimed at studying TCDD-induced oxidative stress in kidney tissue and extent of protection offered by EA against these effects. Protection offered by EA was evaluated by determining kidney markers (serum urea and creatinine), antioxidant status (GSH, SOD, CAT, GST and GPx), lipid peroxidation levels, ATPase enzyme activities (Na? K? ATPase, Ca2? ATPase and Mg2? ATPase) and CYP1A1 activity.

Materials and methods Chemicals TCDD was a kind gift from Dr. Howard G. Shertzer, Professor, Director, Environmental Genetics and Molecular Toxicology Division, University of Cincinnati. Ellagic Acid (purity [99 %), 7-ethoxy resorufin, dicumarol, resorufin were purchased from Sigma Aldrich Private

123

Limited, Bangalore, India. All other chemicals were purchased from Hi Media Laboratories, Mumbai, India. Animals Male albino wistar rats weighing between 150 and 200 g were procured from Kerala Agricultural University Mannuthy, Kerala, India. The study protocol was approved from the Institutional Animal Ethics Committee constituted in accordance with the rules and guidelines of the Committee for the purpose of control and supervision of experiments and animals (CPCSEA), India. The rats were maintained in a controlled environment with standard temperature (28 ± 2 °C) and humidity with an alternating light and dark cycle. The animals were fed with commercially available pelleted rat chow (Sai Durga Private Limited, Bangalore, India) and water ad libitum. After a week of acclimatization, animals were randomly divided into control and test groups with six rats maintained in each group. Treatment schedule Rats were given intra peritoneal injection of TCDD (15 lg/kg body weight) dissolved in 0.15 ml of corn oil) and EA (10 mg/kg body weight) dissolved in 0.15 ml of 50 % DMSO was administered orally. Experimental procedure The animals were divided into six groups with 6 rats in each group. The dosing protocol of TCDD (15 lg/kg body weight) was chosen from the previous reports of Shertzer et al. [18], which showed that such a dosing schedule intensifies TCDD inducible oxidative stress response. EA dose (10 mg/kg body weight) was fixed based on the reports of Turk et al. [19] and our preliminary studies also confirmed the same. Groups

Concentration and volume of toxin/ antioxidant/vehicle administered

Treatment period

Sacrifice

Group I (control)





On 21st day

Group II (DMSO— vehicle for EA)

0.15 ml of 50 % DMSO administered/day

From 1st– 10th day

On 21st day

Group III (corn oil—vehicle for TCDD)

0.15 ml of corn oil administered/day

From 11th– 13th day

On 21st day

Group IV (EA)

10 mg of EA/kg body weight dissolved in 0.15 ml of 50 % DMSO per day by oral administration

From 1st– 10th day

On 21st day

Mol Biol Rep

Groups

Concentration and volume of toxin/ antioxidant/vehicle administered

Treatment period

Sacrifice

Group V (TCDD)

15 lg TCDD/kg body weight dissolved in 0.15 ml corn oil per day by intraperitoneal injection

From 11th– 13th day

On 21st day

10 mg of EA/kg body weight dissolved in 0.15 ml of 50 % DMSO per day by oral administration

From 1st– 10th day

15 lg TCDD/kg body weight dissolved in 0.15 ml corn oil per day by intraperitoneal injection

From 11th– 13th day

Group VI (pretreatment with EA followed by TCDD treatment)

On 21st day

After the treatment period the animals were fasted overnight, weighed and anesthetized by exposing to diethyl ether. Blood was collected from jugular vein and serum was separated and used for kidney marker enzyme assays. The kidney tissues were dissected out, washed in chilled physiological saline, patted dry and weighed. A small portion of the kidney tissue was stored in 10 % formalin for histopathological examination. The remaining tissue was stored at -80 °C for further analysis. About 100 mg kidney tissue was weighed and homogenized in chilled 0.1 M Tris–HCl buffer in Potter–Elvehjem Teflon homogenizer. The homogenates were centrifuged at 3,000 rpm for 15 min and supernatant was aliquoted and used for assessing oxidative stress parameters and ATPase enzyme activities. Serum kidney marker analysis Blood samples were allowed to clot at room temperature and then centrifuged at 1,200 g for 15 min. The clear serum was separated and used for biochemical assays. Renal function was assessed by measurement of serum urea [20] and serum creatinine levels [21]. Histopathology A small portion of kidney tissue from the experimental animals was fixed in 10 % neutral buffered formalin and processed by standard procedure for paraffin embedding

and serial sections of about 5 lm sizes were cut and were stained with hematoxylin and eosin (H and E) dyes and studied under light microscope. Assessment of oxidative stress status and ATPases activities Oxidative stress was measured by estimating the enzymic and non-enzymic antioxidants such as catalase (CAT) [22], superoxide dismutase (SOD) [23], glutathione-s-transferase (GST) [24], glutathione peroxidase (GPX) [25], reduced glutathione (GSH) [26] and thio barbituric acid reactive substances (TBARS) [27]. Membrane bound ATPase enzyme activities namely Na? K? ATPase [28], Ca2? ATPase [29] and Mg2? ATPase [30] were assayed in the tissue homogenate. Metabolic activation of TCDD by CYP1A1 Microsomes from kidney tissues were isolated by the method described by Walawalkar and Iyer, [31] and CYP1A1 activity (EROD) was determined by the method of Peters et al. [32]. Statistical analysis The data was analyzed using one-way analysis of variance (ANOVA) followed by Tukey’s multiple comparison test.

Results Effect of EA on renal biomarkers and histopathological alterations induced by TCDD The biomarkers of renal function, serum urea and creatinine were estimated in the present study. The TCDD induced kidney toxicity was reflected in the significant increase (P \ 0.01) in levels of these two kidney markers when compared to control rats. Pre-treatment with EA followed by TCDD treatment significantly decreased (P \ 0.001) their levels in serum when compared to TCDD alone treated group. Rats treated with corn oil/DMSO/EA alone did not show any change in the serum kidney markers when compared to control rats (Fig. 1). Histological changes on TCDD treatment showed sclerosed glomerulus with lose of shape, shrinkage and increase in interstium (Fig. 2a, b) shows interstitial inflammation with dilated tubules. Histopathological examination of control (Fig. 3a) and rats which received DMSO (Fig. 3b)/corn oil (Fig. 3c)/EA (Fig. 3d) alone showed normal kidney tissue architecture with prominent shape of glomerules and tubules. Rats pre-treated with EA followed by TCDD

123

Mol Biol Rep

Fig. 1 Effect of ellagic acid and TCDD on renal markers a serum creatinine b serum urea. Group I control, Group II DMSO (vehicle for EA), Group III corn oil (vehicle for TCDD), Group IV EA, Group V TCDD, Group VI pre-treatment of EA followed by TCDD treatment. Each bar represents, mean ± standard error of mean (SEM) of each

group. **P \ 0.01, when compared to control. ###P \ 0.001, when compared to TCDD treated group. NS non significant when compared to control. (One way ANOVA followed by Tukey’s multiple comparison)

Fig. 2 Histopathological alterations induced by TCDD in kidney of wistar rats. a Sclerosed glomeruli with lose of shape, shrinkage and increase in interstium b interstitial inflammation, sclerosed glomeruli and dilated tubules. (H&E, magnification 9400)

Fig. 3 Histopathology of rats treated with ellagic acid and TCDD. a Control rats, b rats treated with DMSO (solvent control for ellagic acid), c rats treated with corn oil (solvent control for TCDD), d rats treated with ellagic acid (10 mg/kg body weight), showing typical

123

kidney morphology with normal glomeruli and tubules. e Rats pretreated with ellagic acid (10 mg/kg body weight) followed by TCDD (15 lg/kg body weight) shows normal glomeruli with no signs of sclerosis and inflammation. (H&E, magnification 9400)

treatment (Fig. 3e) showed normal kidney architecture which was comparable to that of control rats. Protective effect of EA on oxidative stress induced by TCDD

##

P \ 0.01,

###

P \ 0.001, when compared to TCDD treated group

The results of the present study showed a significant increase (P \ 0.001) in the levels of TBARS with concomitant decrease (P \ 0.001) in the levels of non- enzymic antioxidant (GSH) in rats treated with TCDD when compared to control. The activities of various antioxidant enzymes like SOD, CAT, GST and GPX were found to be significantly decreased (P \ 0.001) in TCDD treated rats when compared to control. Pre-treatment with EA followed by TCDD treatment significantly increased the antioxidant status with SOD, GPX (P \ 0.001) and GSH, GST, CAT (P \ 0.01) whereas the levels of TBARS significantly decreased (P \ 0.001) when compared to TCDD treated rats. Rats treated with EA/corn oil/DMSO showed nonsignificant levels when compared to control rats (Table 1).

*** P \ 0.001, when compared to control.

2.4 ± 0.01 5.4 ± 0.05 5.4 ± 0.04 5.0 ± 0.05 GST

5.1 ± 0.04

59 ± 1.12 GPX

Group I control, Group II DMSO (vehicle for ellagic acid), Group III corn oil (vehicle for TCDD), Group IV EA, Group V TCDD, Group VI pre-treatment of EA followed by TCDD treatment. Values are mean ± standard error of mean of each group. NS not significant when compared to control. (One way ANOVA followed by Tukey’s multiple comparison). Units are expressed as: LPO in nanomoles of TBA reactants/mg of protein, GSH in nanomoles of GSH/g of tissue, SOD, CAT, GST and GPx activities in Units/mg protein. SOD 1 U the amount of enzyme required to give 50 % inhibition of pyrogallol auto-oxidation. Catalase 1 U the amount of enzyme that consumes 1 nmol of H2O2/min, GST 1 U the amount of enzyme that conjugates 1 lmol CDNB/min, GPx 1 U the amount of enzyme that converts 1 l mole GSH to GSSG in the presence of H2O2/min

4.2 ± 0.02###

49 ± 0.96##

*** NS NS

0.030 ± 0.002## 0.050 ± 0.001 CAT

56 ± 1.28

59 ± 1.09

NS

58 ± 0.90

41 ± 1.65

***

0.010 ± 0.002 0.049 ± 0.002

NS NS

0.041 ± 0.003

98 ± 1.60### 119 ± 2.10 SOD

0.042 ± 0.001

NS

*** NS

32 ± 1.70 120 ± 2.10

45 ± 2.00 GSH

115 ± 1.90

NS

NS NS

111 ± 1.54

41 ± 1.30NS 42 ± 1.90NS

30.65 ± 1.50 32.00 ± 1.11 LPO

30.21 ± 1.17

NS

*** NS

37 ± 1.06## 22 ± 1.24*** 42 ± 1.12NS

39.20 ± 1.37### 31.45 ± 1.22

72.45 ± 1.45

***

Group V Group IV

NS NS

Group III Group II Group I Parameters

Table 1 Effect of ellagic acid on TCDD-induced oxidative stress and antioxidant status in experimental rats

NS

Group VI

Mol Biol Rep

Effect of EA and TCDD on ATPases enzyme activities The rats treated with TCDD showed a significant decrease in both Na?/K? ATPase (P \ 0.001) and Mg2? ATPase (P \ 0.01) activities and significant increase in the Ca2? ATPase (P \ 0.001) activity when compared to control rats. In rats pre-treated with EA followed by TCDD treatment showed a marked increase in Na?/K? ATPase (P \ 0.01) and Mg2? ATPase activities (P \ 0.01) with significant decrease (P \ 0.01) in Ca2? ATPases activity when compared to TCDD alone treated rats. Whereas rats which received EA/corn oil/DMSO alone did not show any significant change in ATPases activities when compared to control rats (Fig. 4). Inhibition of metabolic activation of TCDD by EA EROD activity describes the rate of the CYP1A mediated de ethylation of the substrate 7-ethoxyresorufin (7-ER) to form the product resorufin. The catalytic activity towards this substrate is an indication of the amount of enzyme present and is measured as the concentration of resorufin produced [33]. The results are expressed as percentage fold induction of EROD activity with control values considered as 100 %. In the present study, the activities of CYP1A1 were found to be significantly increased (P \ 0.01) in TCDD treated rats when compared to control rats. In rats pre-treated with EA followed by TCDD treatment there was a significant (P \ 0.001) decrease in activity when compared to TCDD treated rats. Rats which received EA/Corn oil/DMSO alone did not show significant change in the activity when compared to control rats (Fig. 5).

123

Mol Biol Rep Fig. 4 Effect of EA on TCDDinduced changes in membrane bound ATPase enzyme activities. Group I control, Group II DMSO (vehicle for DMSO), Group III corn oil (vehicle for TCDD), Group IV EA, Group V TCDD, Group VI pre-treatment of EA followed by TCDD treatment. Each bar represents mean ± standard error of mean of each group. ***P \ 0.001, **P \ 0.01 when compared to control. ## P \ 0.01, when compared to TCDD treated group. NS non significant when compared to control. (One way ANOVA followed by Tukey’s multiple comparison)

Fig. 5 Effect of EA on TCDD-induced CYP1A1 activity. Group I control, Group II DMSO (vehicle for DMSO), Group III corn oil (vehicle for TCDD), Group IV EA, Group V TCDDm, Group VI pretreatment of EA followed by TCDD treatment. Each bar represents mean ± standard error of mean of each group. ***P \ 0.001, when compared to control. ###P \ 0.001, when compared to TCDD treated group. NS non significant when compared to control. (One way ANOVA followed by Tukey’s multiple comparison)

Discussion TCDD is classified as a group I carcinogen by the International Agency for Research on Cancer [34]. TCDD is resistant to degradation which allows it to bioaccumulate in food chain [35]. Since liver is the target organ for detoxification of environmental toxins, most of the studies have focused on TCDD-induced liver toxicity. Recent reports show that TCDD also causes adverse effects in various

123

extra hepatic tissues and sparse literature is available on TCDD induced nephrotoxicity and its protection by antioxidants. Lu et al. [36] has first demonstrated combined effect of TCDD and PCB congeners on kidney toxicity in rats. Ciftci et al. [37] have reported protective effect offered by quercetin and chrysin on TCDD induced renal damage by evaluating the antioxidant status. In present study the antioxidant ellagic acid was evaluated for its protective effect by assessing CYP1A1 activity (prime candidate involved in the generation of ROS), ATP ase enzyme activities and serum kidney markers in addition to oxidative stress parameters in TCDD-induced nephrotoxicity in male wistar rats. Phenolic antioxidants play a pivotal role in offering protection during various oxidative stress conditions. EA (gallic acid dimer), a naturally occurring polyphenolic compound possesses lipophilic and free radical scavenging property. EA exhibits antioxidant [11], anti-proliferative [38] and anti-mutagenic properties [39]. Studies on structure–function relationship of EA revealed that phenolic hydroxyl group and lactone are responsible for its protective action against various conditions of oxidative stress [40]. So, the present study evaluated the oxidative stress responses induced by TCDD in kidney of wistar rats and possible protective effect of EA on TCDD induced nephrotoxicity. Histopathological observation and levels of serum kidney markers reveal that TCDD causes severe damage to the kidney tissues. The rats treated with TCDD showed sclerosis in glomeruli with interstitial inflammation and dilation in distal tubules. The observed changes are consistent with earlier reports of Ciftci et al. [37] and Lu et al. [36]. Along with these changes the present study observed that

Mol Biol Rep

TCDD also induced inflammation and damages to the proximal and distal tubules. Pre-treatment with EA rendered significant protection against the histological changes induced by TCDD by maintaining the near normal architecture of glomerulus and tubules. The results of the present study corroborates with the earlier report of Atessahin et al. [15], where EA offered protective effect by reducing the histopathological alterations against cisplatininduced nephrotoxicity. Oxidative stress results when production of reactive oxidative species exceeds the capacity of cellular antioxidant defenses to remove these toxic species. Oxidative stress conditions induced by various xenobiotics result from upstream signaling mechanisms and in the case of TCDD, AhR play an important role in mediating the toxicity. Upon TCDD binding to AhR, it gets translocated to the nucleus where it associates with ARNT. The whole complex then acts as a transcription factor that binds to DNA promoter sequences termed xenobiotic responsive elements, thereby enhancing the transcription of array of genes [41]. Out of which induction of CYP1A1 by AhR is of toxicological relevance. TCDD acts as substrate for CYP1A1 and the formed metabolites potentially initiates toxic responses by increasing oxidative stress status [42, 43]. The ROS generated can directly react with cellular membrane phospholipid moieties due to the presence of their methylene group [44]. Polyunsaturated fatty acid oxidation yields short-lived lipid hydroperoxide molecules known to further perpetuate ROS production [45]. Lipid peroxidation and oxidative stress have been indicated as factors which lead to acute toxicity of TCDD [46, 47]. The cells exhibit various non- enzymic and enzymic antioxidants which act as natural defense mechanisms to counteract the oxidative stress conditions. Of which, reduced glutathione (GSH) is a non protein thiol which is actively present inside the cells and acts as first defense against oxidative attack. It participates in a variety of detoxification reactions, in addition to providing protection against oxidative damage by virtue of its thiol group. Peroxide level and thiol status is indicative of oxidative stress conditions, since increase in lipid peroxidation causes depletion in GSH levels [48]. So, in the present study these two levels were measured to analyze the extent of oxidative stress caused by TCDD. In the present study there was a significant decrease in total GSH and with concomitant increase in lipid peroxide levels in TCDD treated rats. Earlier report by Stohs, [49] also showed decreased thiol levels and increased lipid peroxide levels in mitochondria of TCDD treated rats. In rats pre-treated with EA followed by TCDD treatment there was a significant increase in GSH levels with concomitant decrease in lipid peroxide levels. Thus EA by its potent free radical scavenging property and glutathione sparing action might have

attenuated the oxidative stress status invoked by TCDD. The free radical scavenging property of EA leads to the inhibition of LPO and 8-OGDG formation both in vitro and in vivo [50, 51]. Protection offered by EA by decreasing the levels of lipid peroxides on fetotoxicity induced by TCDD in C57BL/67 mice [17] and increasing the levels of GSH in N-nitrosodiethylamine induced lung tumorigenesis in mice [52] has been reported earlier which supports the present findings. Apart from GSH, various antioxidant enzymes are involved to overcome the conditions of oxidative stress. Optimum levels and proper balance among these enzymes may be critical for the prevention of cellular oxidative stress. In the present study, a significant decrease in the activities of the antioxidant enzymes (SOD, GSH, GPx, and CAT) reflects TCDD-induced oxidative stress in renal tissue. The observed decline in antioxidant status after TCDD treatment is in line with the previous reports of Jin et al. [53] who has demonstrated significant decrease in the activities of antioxidant enzymes in reproductive system of mouse treated with TCDD. In addition, a dose-dependent suppression of SOD activity in cerebral cortex and hippocampus regions following TCDD treatment has been reported earlier by Hassoun et al. [54]. It could be said that, TCDD-induced decrease in activities of antioxidant enzymes might be caused by the expense of these enzymes in overcoming the oxidative stress conditions. In the present study, pre-treatment of EA followed by TCDD treatment significantly improved the antioxidant status. EA has been shown to offer protection by its free radical scavenging action and improved antioxidant status through Nrf-2 expression [11, 16]. The observed results on increase in antioxidant status and nephroprotective action of EA might be due to the above mentioned properties of EA. In addition, the ability of EA to render protective effect by enhancing the antioxidant mechanism against various oxidative conditions, support the present study findings. Das et al. [55] has shown protection offered by EA through increasing GST activity in liver and lungs of BALB/c mice against oxidative stress induced by benzo[a]pyrene. Protective effect of EA by improving the antioxidant status against selenite induced cataractogenesis [56] and cisplatin induced injuries to sperm [19] has been reported earlier. The membrane-bound enzymes such as Na?, K?-ATPase, Mg2?-ATPase and Ca2?-ATPase are responsible for the transport of sodium/potassium, magnesium and calcium ions across the cell membranes at the expense of ATP hydrolysis [57]. It is important to evaluate membrane bound ATPases enzyme activity during stress conditions, since changes in the activities of these enzymes affect the normal functioning of any organ. It is well documented that TCDD-induced Ca2? signaling plays a major role in TCDD-induced toxic

123

Mol Biol Rep

events [58–60]. TCDD-dependent increase in intracellular calcium levels activates phospholipase A2, which in turn mobilizes arachidonic acid from cell membrane. The released arachidonic acid acts as a substrate for COX-2 and initiates eicosanoid synthesis. TCDD induced early and sustained increase in intracellular free calcium and subsequent formation of eicosanoids has been reported by Puga et al. [59]. Studies have documented that eicosanoids, especially arachidonic acid metabolites have profound inhibitory action on Na?, K?-ATPase and Mg2?-ATPase activities [61]. The results of the present study reveal a significant decrease in Na?, K?-ATPase and Mg2?-ATPase activities during TCDD treatment when compared to control rats. Decline in Na?, K?-ATPase activity after TCDD treatment has been reported in liver tissues [62] and renal medullary cells [63]. However the Schwartzman et al. [63] has showed that arachidonic acid metabolite formed from cytochrome P450 played a major role in inhibition of in Na?, K?-ATPase activity. But it was interesting to note that there was a significant increase in Ca2?-ATPase activity in the present study on treatment with TCDD. It can be explained that an early and sustained increase in Ca2? levels might have increased the activity of Ca2?-ATPase as a cells adaptive mechanism to overcome Ca2? overloading and prevent subsequent calcium induced toxic events. From the results of the present study and available literature it can be said that TCDD-induced alterations in ATPase enzyme activities may be due the disturbances in Ca2? homeostasis and subsequent formation of eicosanoids. While EA pretreatment significantly restored the ATPase enzyme activities to values close to that of control rats. The ability of EA to maintain Ca2? homeostasis might be attributed to restore ATPase enzyme activities. To this end, Ou et al. [64] have reported that EA maintained Ca2? homeostasis in endothelial cells and thereby prevented oxidized low-density lipoprotein-induced apoptosis. An unpublished data from our laboratory also supports the fact that EA is involved in maintaining Ca2? homeostasis. Our laboratory has been involved in identifying the molecular mechanism of EA in offering cytoprotection against TCDD-induced toxic events by in vitro studies. From the results, we observed that EA significantly maintained the Ca2? homeostasis and prevented TCDD-induced oxidative stress in HepG2 cells. The above results indicate the protective nature of EA against TCDD-induced nephrotoxicity by enhancing antioxidant status and preventing oxidative stress. Since TCDD-induced sustained oxidative stress and other stress response is primarily mediated through TCDD-induced CYP1A1 activation and subsequent imbalance in the redox status [65, 66], we next determined whether EA had any effect on suppressing the activity of CYP1A1. Inhibition of CYP1A1 by natural compounds is an attractive strategy, as these compounds could prevent oxidative stress and subsequent

123

induction of various toxic events. The present study demonstrates that EA pre-treatment significantly decreased TCDD-induced CYP1A1 activity. The reduction in CYP1A1 activity might be either due to direct binding of EA to CYP1A1 or it could be due to its AhR antagonism. Further studies are required to understand the exact molecular mechanism of EA mediated CYP1A1 inhibition. In conclusion, the present study demonstrates that TCDDinduced nephrotoxicity is mediated through sustained oxidative stress with altered ATPase enzyme activities. Our data provide clear evidence that over all protective effect offered by EA against TCDD-induced nephrotoxicity is mediated through suppressing CYP1A1 activity and enhancing the antioxidant mechanism. Thus the present study highlights that consumption of ellagic acid helps in protection against environmental toxins which primarily mediate their toxic effects through oxidative stress. Conflict of interest The authors declare that there are no conflict of interest.

References 1. Gasiewicz TA (1997) Exposure to dioxin and dioxin-like compounds as a potential factor in developmental disabilities. Ment Retard Dev Disabil Res Rev 3:230–238 2. Peterson RE, Theobald HM, Kimmel GL (1993) Developmental and reproductive toxicity of dioxins and related compounds: cross-species comparisons. Crit Rev Toxicol 23:283–335 3. Pohjanvirta R, Tuomisto J (1994) Short-term toxicity of 2,3,7,8tetrachlorodibenzo-p-dioxin in laboratory animals: effects, mechanisms, and animal models. Pharmacol Rev 46:483–549 4. Olson JR (1994) Pharmacokinetics of dioxin and related chemicals. In: Schecter A (ed) Dioxins and health. Plenum Press, New York, pp 163–167 5. Fujii-Kuriyama Y, Mimura J (2005) Molecular mechanisms of AhR functions in the regulation of cytochrome P450 genes. Biochem Biophys Res Commun 338:311–317 6. Schmidt JV, Bradfield CA (1996) Ah receptor signaling pathways. Annu Rev Cell Dev Biol 12:55–89 7. Nebert DW, Roe AL, Dieter MZ, Solis WA, Yang Y, Dalton TP (2000) Role of the aromatic hydrocarbon receptor and [Ah] genebattery in the oxidative stress response, cell cycle control, and apoptosis. Biochem Pharmacol 59:65–85 8. Castell JV, Donato MT, Gomez-Lechon MJ (2005) Metabolism and bioactivation of toxicants in the lung. The in vitro cellular approach. Exp Toxicol Pathol 57:189–204 9. Park JY, Shigenaga MK, Ames BN (1996) Induction of cytochromeP4501A1 by 2,3,7,8-tetrachlorodibenzo-p-dioxin or indolo(3,2-b)-carbazole is associated with oxidative DNA damage. Proc Natl Acad Sci 93:2322–2327 10. Mates JM, Perez-Gomez C, Nunez de Castro I (1999) Antioxidant enzymes and human diseases. Clin Biochem 32:595–603 11. Priyadarsini KI, Khopde SM, Kumar SS, Mohan H (2002) Free radical studies of ellagic acid, a natural phenolic antioxidant. J Agric Food Chem 50:2200–2206 12. Kannan MM, Quine SD (2011) Ellagic acid ameliorates isoproterenol induced oxidative stress: evidence from electrocardiological, biochemical and histological study. Eur J Pharmacol 659:45–52

Mol Biol Rep 13. Kannan MM, Quine SD (2013) Ellagic acid inhibits cardiac arrhythmias, hypertrophy and hyperlipidaemia during myocardial infarction in rats. Metabolism 62:52–61 14. Yuce A, Atessahin A, Ceribasi AO, Aksakal M (2007) Ellagic acid prevents cisplatin-induced oxidative stress in liver and heart tissue of rats. Basic Clin Pharmacol Toxicol 101:345–349 15. Atessahin A, Ceribasi AO, Yuce A, Bulmus O, Cikim G (2007) Role of ellagic acid against cisplatin-induced nephrotoxicity and oxidative stress in rats. Basic Clin Pharmacol Toxicol 100:121–126 16. Hseu YC, Chou CW, Senthil Kumar KJ, Fu KT, Wang HM, Hsu LS, Kuo YH, Wu CR, Chen SC, Yang HL (2012) Ellagic acid protects human keratinocyte (HaCaT) cells against UVA-induced oxidative stress and apoptosis through the upregulation of the HO-1 and Nrf-2 antioxidant genes. Food Chem Toxicol 50:1245–1255 17. Hassoun EA, Walter AC, Alsharif NZ, Stohs SJ (1997) Modulation of TCDD-induced fetotoxicity and oxidative stress in embryonic and placental tissues of C57BL/6 J mice by vitamin E succinate and ellagic acid. Toxicology 124:27–37 18. Shertzer HG, Nebert DW, Puga A, Ary M, Sonntag D, Dixon K, Robinson LJ, Cianciolo E, Dalton TP (1998) Dioxin causes a sustained oxidative stress response in the mouse. Biochem Biophys Res Commun 253:44–48 19. Turk G, Atessahin A, Sonmez M, Ceribasi AO, Yuce A (2008) Improvement of cisplatin-induced injuries to sperm quality, the oxidant-antioxidant system, and the histologic structure of the rat testis by ellagic acid. Fertil Steril 89:1474–1481 20. Natelson S, Scott ML, Beffa C (1951) A rapid method for the estimation of urea in biologic fluids. Am J Clin Pathol 21:275–281 21. Brod J, Sirota JH (1948) The renal clearance of endogenous ‘‘creatinine’’ in man. J Clin Invest 27:645–654 22. Aebi (1974) Catalase. In: Bergmeyer (ed) Methods of enzymatic analysis. Academic Press, New York, pp 673–684 23. Marklund S, Marklund G (1974) Involvement of the superoxide anion radical in the autoxidation of pyrogallol and a convenient assay for superoxide dismutase. Eur J Biochem 47:469–474 24. Habig WH, Pabst MJ, Jakoby WB (1974) Glutathione-s-transferases. The first enzymatic step in mercapturic acid formation. J Biol Chem 249:7130–7139 25. Rotruck JT, Pope AL, Ganther HE, Swanson AB, Hafeman DG, Hoekstra WG (1973) Selenium: biochemical role as a component of glutathione peroxidase. Science 179:588–590 26. Moron MS, Depierre JW, Mannervik B (1979) Levels of glutathione, glutathione reductase and glutathione-s-transferase activities in rat lung and liver. Biochim Biophys Acta 582:67–78 27. Ohkawa H, Ohishi N, Yagi K (1979) Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem 95:351–358 28. Bonting (1970) Sodium-potassium activated adenosine triphosphatase and cation transport. In: Bittar EE (ed) Membranes and ion transport. Wiley, London, pp 257–263 29. Hjerte´n S, Pan H (1983) Purification and characterization of two forms of a low-affinity Ca2?-ATPase from erythrocyte membranes. Biochim Biophys Acta 728:281–288 30. Ohnishi T, Suzuki T, Suzuki Y, Ozawa K (1982) A comparative study of plasma membrane Mg2?-ATPase activities in normal, regenerating and malignant cells. Biochim Biophys Acta 684:67–74 31. Walawalkar PS, Iyer KR (2006) Isolation and catalytic competence of different animal liver microsomal fractions prepared by calcium-aggregation method. Indian J Pharm Sci 68:262–265 32. Peters AK, van Londen K, Bergman A, Bohonowych J, Denison MS, van den Berg M, Sanderson JT (2004) Effects of polybrominated diphenyl ethers on basal and TCDD-induced ethoxyresorufin activity and cytochrome P450-1A1 expression in MCF7, HepG2, and H4IIE cells. Toxicol Sci 82:488–496 33. Kennedy SW, Jones SP (1994) Simultaneous measurement of cytochrome P4501A catalytic activity and total protein

34.

35.

36.

37.

38.

39.

40.

41.

42.

43.

44.

45. 46.

47.

48.

49. 50.

concentration with a fluorescence plate reader. Anal Biochem 222:217–223 IARC (1997) Polychlorinated dibenzo-para-dioxins and polychlorinated dibenzofurans. IARC Monogr Eval Carcinog Risks Hum 69:1–631 Van den Berg M, De Jongh J, Poiger H, Olson JR (1994) The toxicokinetics and metabolism of polychlorinated dibenzo-p-dioxins (PCDDs) and dibenzofurans (PCDFs) and their relevance for toxicity. Crit Rev Toxicol 24:1–74 Lu CF, Wang YM, Peng SQ, Zou LB, Tan DH, Liu G, Fu Z, Wang QX, Zhao J (2009) Combined effects of repeated administration of 2,3,7,8-tetrachlorodibenzo-p-dioxin and polychlorinated biphenyls on kidneys of male rats. Arch Environ Contam Toxicol 57:767–776 Ciftci O, Ozdemir I, Vardi N, Beytur A, Oguz F (2011) Ameliorating effects of quercetin and chrysin on 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced nephrotoxicity in rats. Toxicol Ind Health 28:947–954 Whitley AC, Stoner GD, Darby MV, Walle T (2003) Intestinal epithelial cell accumulation of the cancer preventive polyphenol ellagic acid-extensive binding to protein and DNA. Biochem Pharmacol 66:907–915 Loarca-Pina G, Kuzmicky PA, de Mejia EG, Kado NY (1998) Inhibitory effects of ellagic acid on the direct-acting mutagenicity of aflatoxin B1 in the Salmonella microsuspension assay. Mutat Res 398:183–187 Barch DH, Rundhaugen LM, Stoner GD, Pillay NS, Rosche WA (1996) Structure-function relationships of the dietary anticarcinogen ellagic acid. Carcinogenesis 17:265–269 Nebert DW, Duffy JJ (1997) How knockout mouse lines will be used to study the role of drug-metabolizing enzymes and their receptors during reproduction and development, and in environmental toxicity, cancer, and oxidative stress. Biochem Pharmacol 53:249–254 Spink DC, Katz BH, Hussain MM, Spink BC, Wu SJ, Liu N, Pause R, Kaminsky LS (2002) Induction of CYP1A1 and CYP1B1 in T-47D human breast cancer cells by benzo[a]pyrene is diminished by arsenite. Drug Metab Dispos 30:262–269 Androutsopoulos VP, Tsatsakis AM, Spandidos DA (2009) Cytochrome P450 CYP1A1: wider roles in cancer progression and prevention. BMC Cancer 9:187 Valko M, Rhodes CJ, Moncol J, Izakovic M, Mazur M (2006) Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chem Biol Interact 160:1–40 Marnett LJ (2000) Oxyradicals and DNA damage. Carcinogenesis 21:361–370 Pohjanvirta R, Kulju T, Morselt AF, Tuominen R, Juvonen R, Rozman K, Mannisto P, Collan Y, Sainio EL, Tuomisto J (1989) Target tissue morphology and serum biochemistry following 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure in a TCDD-susceptible and a TCDD-resistant rat strain. Fundam Appl Toxicol 12:698–712 Slezak BP, Hatch GE, DeVito MJ, Diliberto JJ, Slade R, Crissman K, Hassoun E, Birnbaum LS (2000) Oxidative stress in female B6C3F1 mice following acute and subchronic exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Toxicol Sci 54:390–398 Ganesan B, Buddhan S, Anandan R, Sivakumar R, AnbinEzhilan R (2010) Antioxidant defense of betaine against isoprenalineinduced myocardial infarction in rats. Mol Biol Rep 37:1319–1327 Stohs SJ (1990) Oxidative stress induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Free Radic Biol Med 9:79–90 Cozzi R, Ricordy R, Bartolini F, Ramadori L, Perticone P, De Salvia R (1995) Taurine and ellagic acid: two differently acting natural antioxidants. Environ Mol Mutagen 26:248–254

123

Mol Biol Rep 51. Laranjinha J, Vierira O, Almeida L, Madeira V (1996) Inhibition of metmyoglobin/H2O2-dependent low density lipoprotein lipid peroxidation by naturally occurring phenolic acids. Biochem Pharmacol 51:395–402 52. Khanduja KL, Gandhi RK, Pathania V, Syal N (1999) Prevention of N-nitrosodiethylamine-induced lung tumorigenesis by ellagic acid and quercetin in mice. Food Chem Toxicol 37:313–318 53. Jin MH, Ko HK, Hong CH, Han SW (2008) In utero exposure to 2,3,7,8-Tetrachlorodibenzo-p-dioxin affects the development of reproductive system in mouse. Yonsei Med J 49:843–850 54. Hassoun EA, Li F, Abushaban A, Stohs SJ (2001) Production of superoxide anion, lipid peroxidation and DNA damage in the hepatic and brain tissues of rats after subchronic exposure to mixtures of TCDD and its congeners. J Appl Toxicol 21:211–219 55. Das M, Bickers DR, Mukhtar H (1985) Effect of ellagic acid on hepatic and pulmonary xenobiotic metabolism in mice: studies on the mechanism of its anticarcinogenic action. Carcinogenesis 6:1409–1413 56. Sakthivel M, Elanchezhian R, Ramesh E, Isai M, Jesudasan CN, Thomas PA, Geraldine P (2008) Prevention of selenite-induced cataractogenesis in Wistar rats by the polyphenol, ellagic acid. Exp Eye Res 86:251–259 57. Balasubramaniyan V, Nalini N (2006) Leptin alters brain adenosine triphosphatase activity in ethanol-mediated neurotoxicity in mice. Singap Med J 47:864–868 58. Canga L, Paroli L, Blanck TJ, Silver RB, Rifkind AB (1993) 2,3,7,8-tetrachlorodibenzo-p-dioxin increases cardiac myocyte intracellular calcium and progressively impairs ventricular contractile responses to isoproterenol and to calcium in chick embryo hearts. Mol Pharmacol 44:1142–1151 59. Puga A, Hoffer A, Zhou S, Bohm JM, Leikauf GD, Shertzer HG (1997) Sustained increase in intracellular free calcium and

123

60.

61. 62.

63.

64.

65.

66.

activation of cyclooxygenase-2 expression in mouse hepatoma cells treated with dioxin. Biochem Pharmacol 54:1287–1296 Sul D, Kim HS, Cho EK, Lee M, Kim HS, Jung WW, Hwang KW, Park SY (2009) 2,3,7,8-TCDD neurotoxicity in neuroblastoma cells is caused by increased oxidative stress, intracellular calcium levels, and tau phosphorylation. Toxicology 255:65–71 Therien AG, Blostein R (2000) Mechanisms of sodium pump regulation. Am J Physiol Cell Physiol 279:C541–C566 Peterson RE, Madhukar BV, Yang KH, Matsumura F (1979) Depression of adenosine triphosphatase activities in isolated liver surface membranes of 2,3,7,8-tetrachlorodibenzo-p-dioxin-treated rats: correlation with effects on ouabain biliary excretion and bile flow. J Pharmacol Exp Ther 210:275–282 Schwartzman M, Ferreri NR, Carroll MA, Songu-Mize E, McGiff JC (1985) Renal cytochrome P450-related arachidonate metabolite inhibits (Na? K?)ATPase. Nature 314:620–622 Ou HC, Lee WJ, Lee SD, Huang CY, Chiu TH, Tsai KL, Hsu WC, Sheu WH (2010) Ellagic acid protects endothelial cells from oxidized low-density lipoprotein-induced apoptosis by modulating the PI3 K/Akt/eNOS pathway. Toxicol Appl Pharmacol 248:134–143 Huang P, Rannug A, Ahlbom E, Hakansson H, Ceccatelli S (2000) Effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin on the expression of cytochrome P450 1A1, the aryl hydrocarbon receptor, and the aryl hydrocarbon receptor nuclear translocator in rat brain and pituitary. Toxicol Appl Pharmacol 169:159–167 Toborek M, Barger SW, Mattson M, Espandiari P, Robertson LW, Hennig B (1995) Exposure to polychlorinated biphenyls cause endothelial cell dysfunction. J Biochem Toxicol 10:219–226

Protective effect of ellagic acid against TCDD-induced renal oxidative stress: modulation of CYP1A1 activity and antioxidant defense mechanisms.

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) belongs to toxicologically important class of poly halogenated aromatic hydrocarbons and produce wide varie...
935KB Sizes 0 Downloads 3 Views