Accepted Manuscript Rutin ameliorates cyclophosphamide induced oxidative stress and inflammation in Wistar rats: Role of NFκB/MAPK pathway Sana Nafees, Summya Rashid, Nemat Ali, Syed Kazim Hasan, Sarwat Sultana PII: DOI: Reference:

S0009-2797(15)00090-3 http://dx.doi.org/10.1016/j.cbi.2015.02.021 CBI 7297

To appear in:

Chemico-Biological Interactions

Received Date: Revised Date: Accepted Date:

15 July 2014 28 January 2015 26 February 2015

Please cite this article as: S. Nafees, S. Rashid, N. Ali, S. Kazim Hasan, S. Sultana, Rutin ameliorates cyclophosphamide induced oxidative stress and inflammation in Wistar rats: Role of NFκB/MAPK pathway, Chemico-Biological Interactions (2015), doi: http://dx.doi.org/10.1016/j.cbi.2015.02.021

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Rutin ameliorates cyclophosphamide induced oxidative stress and inflammation in Wistar rats: Role of NFκB/MAPK pathway

Sana Nafees, Summya Rashid, Nemat Ali, Syed Kazim Hasan and Sarwat Sultana*

Molecular Carcinogenesis and Chemoprevention Division, Dept. of Medical Elementology and Toxicology, Faculty of Science, Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi 110062, India.

*Corresponding author, mailing address: Dr. Sarwat Sultana Molecular Carcinogenesis and Chemoprevention Division, Dept. of Medical Elementology and Toxicology, Faculty of Science, Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi 110062, India. E-mail address: [email protected] 1

Abstract Cyclophosphamide is a potent anticancer agent. However its clinical use is restricted because of its marked organ toxicity associated with increased oxidative stress and inflammation. The present study was designed to demonstrate the protective effects of rutin, a naturally occurring bioflavonoid against the hepatotoxicity induced by CP. Rats were subjected to oral pretreatment of rutin (50 and 100 mg/kg b.wt.) against hepatotoxicity induced by i.p. injection of CP (150 mg/kg b.wt.) and were sacrificed after 24h. Hepatoprotective effects of rutin were associated with upregulation of antioxidant enzyme activities and down regulation of serum toxicity markers. Rutin was able to down regulate the levels of inflammatory markers like TNF-α, IL-6 and expressions of p38-MAPK, NFκB, i-NOS and COX-2. Histopathological changes further confirmed the biochemical and immunohistochemical results showing that CP caused significant structural damage to liver which were reversed by pretreatment of rutin. Therefore, our study revealed that rutin may be a promising modulator in attenuating CP induced oxidative stress, inflammation and hepatotoxicity via targeting NFκB and MAPK pathway.

Key words: Cyclophosphamide, rutin, oxidative stress, inflammation, NFκB, MAPK.

2

Abbreviations BSA- bovine serum albumin CAT- catalase CDNB- 1-chloro 2, 4-dinitrobenzene COX-2- Cycloxygenase-2 CP- Cyclophosphamide DAB-3,3-diaminobenzidine DTNB- 5, 5′-dithio bis-[2-nitrobenzoic acid] EDTA- ethylene diamine tetra acetic acid GPx -glutathione peroxidase GR- glutathione reductase GSH- reduced glutathione GSSG- oxidized glutathione IL-6 -Interleukin-6 i-NOS- inducible nitric oxide synthase LPO-lipidperoxidation MAPKs- mitogen-activated protein kinases MDA- malondialdehyde NADPH- reduced nicotinamide adenine dinucleotide phosphate NFκB - Nuclear factor kappa B NO- Nitric oxide NOS- nitric oxide synthase ROS- reactive oxygen species TNF-α -Tumor Necrosis Factor XO- xanthine oxidase

3

1. Introduction Chemotherapeutic drugs are basically used for the treatment of an array of malignancies. However their therapeutic usage is inadequate because of their clinical side effects [1]. Cyclophosphamide [CP] is an anticancer drug which is extensively used in the treatment of multiple human malignancies and disorders like breast cancer, lung cancer, systemic lupus erythematosus, rheumatoid arthritis and multiple sclerosis [2]. On the contrary, CP usage is recurrently restricted due to its adverse side effects and toxicities including nephrotoxicity, hepatotoxicity, cardiotoxicity, immunotoxicity and mutagenicity [3]. Acrolein which is a toxic metabolite of CP is responsible for reactive oxygen species [ROS] production and augmentation of lipid peroxidation [4]. One possible mechanism behind hepatotoxity of CP may be oxidative stress as reported previously [5]. The anti-neoplastic effects of CP are due to phosphoramide mustard, whereas acrolein is correlated to its toxic side effects [6]. Acrolein is a powerful soft electrophile that causes hepatoxicity by inactivating proteins and depleting GSH which results in oxidative stress [7]. It has been reported that acrolein toxicity is mediated by increased oxidants, thereby generating oxidative injury[8], signifying that it is not only direct oxidant but it also generates oxidants by various other pathways [9]. Inflammation plays a key role in the regulation of oxidative stress [10]. ROS leads to the activation of an array of transcription factors like nuclear factor kappa B [NFκB], which results in release of proinflammatory cytokines like Tumor Necrosis Factor [TNF-α] and Interleukin-6 [IL-6] [11] which further activate cycloxygenase-2 [COX-2] [12]. It has been reported that in addition to inflammation various antineoplastic agents have also been shown to activate transcription factor NFκB [13]. NFκB is one of the most important transcription factor which helps in the regulation of genes involved in the process of inflammation, cell proliferation and survival [14]. COX-2 is one of the downstream targets of NFκB.

4

Uncontrolled expression of COX-2 has been reported in different premalignant and malignant phases [15]. Nitric oxide [NO] is a free radical which is synthesized by the enzyme nitric oxide synthase [NOS]. Among its three isoforms, inducible-nitric oxide synthase [i-NOS] is responsible for the production of elevated levels of NO. It is often considered to be the key executor of toxicity in oxidative stress [16]. The process of i-NOS expression involves different signal transduction pathways, including nuclear translocation of the transcription factor, NFκB [17]. The mitogen-activated protein kinases [MAPKs] are proline targeted serine-threonine kinases, which are transducers of environmental stimulus to the nucleus [18]. The p38MAPK is a member of the MAPK family which is activated by oxidative stress which leads to accumulation of ROS in cell [19]. In various studies it has been reported that the activation of p38-MAPK has been associated in regulating the expression of NFκB and i-NOS [20]. In order to prevent the toxic side effects of CP and its metabolites, augmentation of antioxidant defence enzymes with natural antioxidants is necessary. Therefore, there is a need for effective agents which protect the normal tissue and cells from the undesirable effects of toxicity induced by chemotherapy. Therefore amalgamation of treatment regimen with effective antioxidants might be the desirable approach to alleviate the toxicity induced by CP [21, 22]. Various studies have reported that plant extracts possessing antioxidant activity protect against hepatotoxicity induced by CP [23, 24]. The flavonoids belong to the family of phenolic compounds and possess biochemical and pharmacological properties like antibacterial, antiviral, anti-inflammatory, antiallergic, antithrombotic, antimutagenic and antineoplastic [25]. They affect the cell growth, differentiation and its function owing to their radical scavenging activity. Rutin is a glycone of quercetin having a flavonol structure. It is present in fruits and fruit peel, predominantly in citrus fruits like oranges, grapefruits, lemons and limes. It is also found in buckwheat seeds 5

[26]. It is a member of bioflavonoids with antioxidant, anti-inflammatory, antiallergenic, antiviral and anticarcinogenic properties [27]. It has been reported to scavenge superoxide radicals. It can chelate metal ions, such as ferrous cations, which are involved in the Fenton reaction, which generates ROS and prevents lipid peroxidation [28]. However to the best of our knowledge, there is no report about the effect of rutin against CP induced hepatic oxidative stress and anti-inflammatory activity. Therefore, the main objective of our study was to check the role of activation of NFκB and MAPK pathway, expression of p38-MAPK, NFκB, COX-2, i-NOS and proinflammatory cytokines like TNF-α and IL-6 in the pathogenesis of oxidative damage caused by CP in rat liver and to investigate the protective effect of rutin implicated in the hepatotoxic insult might be caused by acrolein, one of the metabolites of CP. 2. Materials and Methods 2.1. Chemicals Rutin, CP, GR, GSSG, GSH, DTNB, CDNB, BSA, NADPH etc. were obtained from Sigma– Aldrich, USA. All other chemicals and reagents were of the highest purity grade and commercially available. 2.2. Animals Eight weeks old male Wistar rats (150–200 g) were obtained from the Central Animal House Facility of Hamdard University, New Delhi and were housed in a ventilated room at 25 ± 5 0

C under a 12 h light/dark cycle. The animals were acclimatized for 1 week before the study

and had free access to standard laboratory feed (Hindustan Lever Ltd., Bombay, India) and water ad libitum. The study was approved by the Committee for the Purpose of Control and Supervision of Experimental Animals (CPCSEA) having Registration number and date of registration: (IAEC No: 173/ CPCSEA, 28th Jan 2000).

6

2.3. Treatment regimen Male Wistar rats (30) were divided into five groups containing 6 rats in each group. Group I served as a saline treated control. Group II served as positive control and administered intraperitoneal dose (150 mg/kg b.wt.) of CP freshly dissolved in normal saline. Rutin was orally administered at two doses, 50 and 100 mg/kg b.wt. to groups III and IV respectively for 20 consecutive days. Group V received only a high dose of rutin for 20 consecutive days. Group II, III, and IV were given CP (150 mg/kg b.wt.) on 20th day after rutin pretreatment. After 24 h of CP administration rats were sacrificed by cervical dislocation under mild anaesthesia using i.p. pentobarbital (50 mg/kg b.wt.) and blood was taken for various serological

parameters.

Liver

samples

were

taken

at

the

same

time

for

immunohistochemistry, various biochemical and histological parameters. There was 100% survival of animals in all the groups. 2.4. Post-mitochondrial supernatant preparation [PMS] Tissue processing and preparation of post-mitochondria supernatant (PMS) were done as described by S. Nafees et al. [29]. 2.5. Measurement of liver toxicity markers: serum aspartate aminotransferase (AST) and alanine aminotransferase [ALT] AST and ALT activities were determined by the method of Reitman and Frankel [30]. 2.6. Assay for Lactate dehydrogenase [LDH] activity Lactate dehydrogenase activity has been estimated in serum by the method described by Khan et al. [31]. 2.7. Assay for Reduced glutathione [GSH] estimation Reduced glutathione was determined by the method of Jollow et al. [32]. 2.8. Assay for Glutathione reductase [GR] activity Glutathione reductase activity was measured by the method of Carlberg and Mannervik [33].

7

2.9. Assay for Glutathione peroxidase [GPx] activity The activity of Glutathione peroxidase was calculated by the method of Mohandas et al. [34]. 2.10. Assay for catalase activity The catalase activity was assessed by the method of Claiborne [35]. 2.11. Assay for lipid peroxidation [LPO] The assay for lipid peroxidation was done following the method of Wright et al. [36]. 2.12. Assay for Xanthine oxidase [XO] activity Xanthine oxidase activity was done following the method of S. Nafees et al. [37].

2.13. Estimation of protein concentration The protein concentration in all samples was determined by the method of O.H. Lowry [38]. 2.14. Assay for Tumor Necrosis Factor Alpha [TNF-α] TNF-α levels were determined by rat TNF-α kit [eBioscience, Inc., San Diego., USA]. The method is based on enzyme-linked immuno-sorbent assay [ELISA]. Analysis was performed by Elisa Plate Reader [Benchmark plus, BioRad] according to the manufacturer’s instruction. 2.15. Assay for IL-6 IL-6 levels were determined by rat IL-6 kit [eBioscience, Inc., San Diego., USA]. The method is based on enzyme-linked immuno-sorbent assay [ELISA]. Analysis was performed by Elisa Plate Reader [Benchmark plus, BioRad] according to the manufacturer’s instruction. 2.16. Histopathological examination The livers were quickly removed after sacrifice of rats and were fixed in 10% neutral buffered formalin solution for histopathological processing. Sections were stained with hematoxyline and eosin before being observed under an Olympus microscope at 40x magnification. 2.17. Immunohistochemistry

8

To examine the protective effects of rutin on markers of inflammation in the liver COX-2, iNOS, NFκB and p38-MAPK expression in the liver were assessed by immunohistochemical staining. Liver sections on polylysine coated slides obtained were fixed in neutral buffered formalin, and embedded in paraffin and were treated for COX-2, i-NOS, NFκB and p38MAPK antibodies for immunohistochemical analysis. The procedures were processed according to the manufacturer’s protocol recommended for the COX-2, i-NOS, NFκB and p38-MAPK immunohistochemistry with slight modifications. Following deparaffinization and rehydration, sections were irradiated in 0.1 mol/L sodium citrate buffer [pH 6.0] in a microwave oven [medium low temperature] for 20 min. Then the sections were exposed to 3% H2O2 for 10 min to bleach endogenous per-oxidases, followed by rinsing 3 times in Tris buffer [pH 7.4] for 10 min. Sections were selectively incubated under humid conditions using an anti-NFκB antibody [1:400;Thermo Fisher Scientific, USA], anti-COX-2 antibody [1:200; Santacruz Biotechnology, Inc., USA], anti-phospho-p38 [dilution 1:200, Santa Cruz] and anti-i-NOS antibody (1:200; Thermo Fisher Scientific, USA) for overnight at 4 °C. Next day, slides were washed 3 times in Tris buffer for 10 min each. The specificity of the antibodies was tested by omission of the primary antibodies and a positive control of rat tonsil tissue. After washing in Tris buffer (pH 7.4), tissues were visualized with 3,3’diaminobenzidine [DAB] and counterstained with hematoxyline. Finally, the sections were dehydrated in xylene, mounted with DPX and cover slipped. Slides prepared for each case were examined by light microscopy. Positive and negative controls were conducted in parallel with COX-2, i-NOS, NFκB and p38-MAPK stained sections. Staining of sections with commercially available antibodies served as the positive control. Negative controls included staining tissue sections with omission of the primary antibody. 2.18. Statistical analysis

9

The data from individual groups were presented as the mean ± SD. Differences between groups were analyzed using analysis of variance [ANOVA] followed by Tukey-Kramer multiple comparisons test and minimum criterion for statistical significance was set at p75%). According to staining intensity, sections were graded as follows: 0 (no staining), 1 (weak but detectable staining), 2 (distinct staining) or 3 (intense staining). Immunohistochemical staining scores were obtained by adding the diffuseness and intensity scores. All slides were examined by two independent observers who were unaware of the experimental protocol. The slides with discrepant evaluations were reevaluated, and a consensus was reached. Measurements were carried out using an Olympus BX51 (Hamburg, Germany) microscope at 40× magnification.

10

3. Results 3.1.

Rutin

pretreatment

modulates

aspartate

aminotransferase

[AST],

alanine

aminotransferase [ALT] and lactate dehydrogenase [LDH] activity AST, ALT and LDH were significantly elevated in CP-treated rats (Table 3). Prophylactic treatment of rutin at a higher dose prevented CP induced elevation in serum levels of these enzymes (##p < 0.01 and < 0.05,

##

p < 0.01 and

###

p < 0.01) as compared to untreated control. Significant reduction [#p

###

p < 0.001] in these marker enzymes was observed in the rutin

pretreatment groups and found to be effective in the reduction of level of these enzymes when compared to CP administered group. Only rutin treated group showed no significant difference compared with the control group.

Henceforth there was a significant recovery after rutin

supplementation [Table 1]. 3.2. Rutin pretreatment restored the hepatic reduced glutathione [GSH] level GSH level was depleted significantly [***p < 0.001] in CP treated group when compared with the control group. GSH level in the rutin pretreated groups increased significantly [##p < 0.01 and

###

p < 0.001] when compared to CP treated group in a dose dependent manner.

However rutin alone did not show any significant changes in GSH level compared to control group [Table 2]. 3.3. Rutin pretreatment ameliorates the activities of antioxidant enzymes A significant depletion in hepatic antioxidant enzymes viz., GPx, GR [Table 2] and CAT [Table 3] was observed in the CP treated group when compared to the control group [***p < 0.001]. Pretreatment with rutin was found to be significantly effective in restoring the activities of these antioxidant enzymes [#p < 0.05,

11

##

p < 0.01 and

###

p < 0.001] in a dose

dependent manner. There was no significant difference in the activity of these antioxidant enzymes between control and only rutin treated group. 3.4. Rutin pretreatment decreased XO activity and MDA formation A significant [**p < 0.01 and

***

p < 0.001] increase in the XO activity and MDA formation

was found in the CP administered group when compared with the control group. It has been observed that pretreatment with rutin leads to significant reduction in the elevated level of MDA and XO activity significantly [#p < 0.05, ##p < 0.01 and

###

p < 0.001] when compared

to CP treated group in a dose dependent manner. There was no significant difference in the activity of these antioxidant enzymes between control and only rutin treated group [Table 3]. 3.5. Cytokines analysis 3.5.1 Rutin inhibit the TNF-α and IL-6 production Levels of proinflammatory cytokine TNF-α and IL-6 were found elevated significantly in CP treated group as compared to control group [***p < 0.001]. Pretreatment with rutin significantly [#p < 0.05,

##

p < 0.01] decreased TNF-α and IL-6 levels. However no

significance difference was observed in the levels of TNF-α and IL-6 between control and only rutin pretreated group [Figure 1 & 2]. 3.6. Effect of rutin on histological changes The liver architecture of control group was normal, devoid of any signs of inflammatory changes. Nucleolus was clear, the central veins and hepatic lobule were clearly seen. In CP treated group, dilated and congested sinusoidal space with infiltration of lymphocytes and small portal space with inflammation were observed which are indicative of hepatocellular injury. Pretreatment with rutin reverted these changes in liver marked by less dilated and congested sinusoidal space with less inflammatory cell infiltration [Figure 3]. 3.7. Effect of rutin pretreatment on immunohistochemical analysis of i-NOS, COX-2 and P38-MAPK 12

The expressions of i-NOS, COX-2 and P38-MAPK are given in figure fig.4, fig.5 and fig.6. Staining intensity of i-NOS, COX-2 and P38-MAPK was remarkably high in CP treated group as compared to control group [***p < 0.001 and

**

p < 0.01]. Pretreatment with rutin

caused a noticeable reduction in the staining intensity of i-NOS, COX-2 and p-38 dose dependently [#p < 0.05, ##p < 0.01 and ###p < 0.001]. 3.8. Effect of rutin pretreatment on expression of NFκB The expression of NFκB protein is given in fig. 7. The immunohistochemical study revealed that in CP treated group intense and positive stained nuclei were seen as compared to the control group [**p < 0.01]. Rutin pretreatment reduced nuclear staining of NFκB dose dependently [#p < 0.05 and ##p < 0.01]. 4. Discussion Therapeutic effectiveness with anticancer drugs like CP is limited due to its evident toxicity on several target organs in addition to the risk of secondary tumors in patients [40]. Therefore there is an urgent need for potent chemopreventive agents which can diminish toxicity and effectively make the possible use of CP so that there will be improvement in the quality of life of the patients undergoing CP therapy. Findings of our study reveal that rutin exhibits preventive potential against CP-induced oxidative and inflammatory events. Biochemical, molecular and histopathological data demonstrates that alleviated effect of rutin is accredited to its antioxidative and antinflammatory potential. It has been evaluated that antioxidants show protection in the normal cells against the toxicity induced by CP [41, 42]. It has been reported that treatment with CP induces hepatotoxicity due to which there is elevation in the levels of serum marker enzymes [AST, ALT and LDH] which depicts hepatic damage [43]. Similarly in our study CP enhanced serum toxicity markers significantly and pretreatment with rutin showed protection against CP induced hepatotoxicity as shown by reduced level of serum enzymes which were compared to that of control group. It has been 13

evaluated that CP induced hepatic injury which led to the induction of oxidative stress due to excessive formation of ROS which caused lipid peroxidation [LPO] [44, 45]. Our results showed increased LPO in the liver of CP treated rats. Pretreatment with rutin prevented CP induced lipid peroxidation which may possibly be associated with the free radical scavenging activity of rutin, suppressing oxidative stress. GSH plays a crucial role in shielding cells against oxidative insult. CP treatment causes depletion of GSH level which might be attributed to the direct conjugation of acrolein with GSH, thus reducing its cellular level and leads to induction of oxidative stress [46]. Our study demonstrated that the mechanism of hepatoprotection by rutin against CP induced toxicity and hepatic damage is associated with the inhibition of oxidative stress by preventing GSH depletion which is in concurrence with the previous findings [47]. Pretreatment with rutin augmented the levels of GSH which may be because of enhancement of GSH synthesizing enzyme activities. Cellular antioxidant defence enzymes like CAT, GPx and GR help in detoxification of the free radicals and following oxidative stress. Dismutation of the superoxide anion leads to the formation of H2O2 and O2 which is further detoxified to water by CAT and GPx. During this reaction, GSH is oxidized to GSSG which is reduced back to GSH via GR. As a result, the activity of these antioxidant enzymes is essential for protection against oxidative stress. Hepatotoxicity induced by CP is attributed to oxidative stress caused by reduction of the antioxidant enzymes [48]. In our study activity of antioxidant enzymes like CAT, GPx and GR in the hepatic tissue were significantly decreased by CP treatment indicating marked oxidative stress. Our results demonstrated that the antioxidant status in the liver was markedly diminished after CP administration. Previous studies demonstrate that natural antioxidants protect against CP induced hepatic injury [49, 17]. In our study we have demonstrated that rutin showed protection against CP induced hepatotoxicity by preventing

14

the generation of oxidative stress and increasing the hepatic antioxidant defense armoury involving GSH-dependent enzymes. Cytokines are low molecular weight proteins which are secreted primarily by the activation of lymphocytes and macrophages and are responsible for the maintenance of homeostasis by lymphoid, inflammatory and hematopoietic cells respectively [50]. TNF-α is a key element for the prevention of inflammatory toxicity induced by chemical compounds [51]. It has been reported in the previous studies that the levels of TNF-α and IL-6 were increased in the rats administered with CP [52]. In our study rutin reduced the levels of TNF-α and IL-6 which is in agreement with the findings of the previous studies [53]. In several inflammatory diseases, NFκB pathway is a therapeutic target because it plays an essential role in the activation of transcriptional factors like TNF-α, IL-6, COX-2 and i-NOS [54]. Dysregulation of NFκB has been associated with different pathological conditions which includes cancer and other inflammatory diseases [55]. In our study we have demonstrated that rutin plays potential role in attenuating ROS generated and TNF-α induced inflammation through downregulation of p38-MAPK, NFκB, COX-2 and i-NOS. ROS are thought to be involved in the activation of NFκB [56]. Various antioxidants are known to exhibit an inhibitory action on the activation of NFκB by modulating the redox status of the cell [57, 58]. In line with this, TNF-α induced ROS generation may possibly be associated with the stimulation of NFκB pathway [56], while inhibition of NFκB signalling could be due to the significant free radical scavenging properties of rutin. Our results suggest the significance of intracellular ROS levels, for rutin-inhibited ROS induced NFκB activation in hepatic tissues. To prevent inflammatory diseases it is an effective approach to target COX-2 and i-NOS [59]. A number of proinflammatory cytokines such as TNF-α and IL-6 are well-known to participate in stimulating i-NOS and COX-2 [60, 61]. Our results demonstrated that rutin 15

remarkably inhibited the activation of TNF- α, IL-6, NFκB, COX-2 and i-NOS induced by CP may be correlated with the inactivation of these proinflammatory cytokines. It has been reported that ROS, proinflammatory cytokines further stimulates redox sensitive transcription factors by activating mitogen-activated protein kinase (MAPK) signalling pathways [62, 63]. p38-MAP kinase, one of the most widely studied cell signalling cascade, plays an important role in inflammation via regulation of activation of COX-2 and NFκB in response to oxidative stress [64]. p38-MAPK pathway regulates production of proinflammatory cytokines which play critical role in inflammation through transcriptional activation of transcription factors like NFκB strongly supports its role in inflammation [65]. Our results demonstrated that there was highest expression of p38, NFκB in CP treated group as compared to control group. However rutin significantly suppressed the activation of p38- MAPK and NFκB which supports the anti-inflammatory effect of rutin. Histopathological studies showed that CP caused destruction of the hepatic tissue which was supported by the swelling and reduction of sinusoidal spaces in liver tissues. This may be due to potency in metabolite of CP which caused destruction of the membrane. The pathological changes due to the CP induced toxicity are correlated with distorted enzyme activities [66] and upregulation of proteins. In our study rutin pretreatment showed a protective effect on liver morphology. 5. Conclusion Our study demonstrated that rutin alleviated ROS induced oxidative stress, inflammation via targeting p38-MAPK, NFκB, COX-2, i-NOS and TNF-α, IL-6 which may be due to the acrolein, metabolite of CP. More research is needed to explain the down-regulation pathways of the modulatory effects of rutin against CP induced hepatotoxicity. In conclusion, rutisn attenuates CP induced hepatic toxicity in Wistar rats and may be clinically useful in the form

16

of a combinational therapy after further confirmatory studies both at pre clinical and clinical levels.

Conflict of interest statement The authors declare that there are no conflicts of interest. Acknowledgment The authors are thankful to the University Grants commission (UGC), New Delhi, India for providing the funds to carry out this work. References [1] S. Rashid, N. Ali, S. Nafees et al. Mitigation of 5-Fluorouracil induced renal toxicity by

chrysin via targeting oxidative stress and apoptosis in wistar rats. Food Chem Toxicol. 66 (2014) 185-93. [2] M. Ayman, I. Mahmoud, E. Omnia et al. Amelioration of Cyclophosphamide-Induced Hepatotoxicity by the Brown Seaweed Turbenaria ornate. International Journal of Clinical Toxicology 1 (2013) 9-17. [3] S. R. Somnath, C. Pramita, B. Sudin. Intervention in cyclophosphamide induced oxidative stress and DNA damage by a flavonyl-thiazolidinedione based organo selenocyanate and evaluation of its efficacy during adjuvant therapy in tumor bearing mice. European Journal of Medicinal Chemistry. 73 (2014) 195-209. [4] F. Liu, X. L .Li, T. Lin et al. The cyclophosphamide metabolite, acrolein, induces cytoskeleton changes and oxidative stress in sertoli cells. Mol. Biol. (2012) Rep. 39, 493– 500.

17

[5] E. Selvakumar, C Prahalathan, Y. Mythili et al. Mitigation of oxidative stress in cyclophosphamide challenged hepatic tissue by DL-alpha-lipoic acid. Mol Cell Biochem. 272 (2005) 179-85. [6] Sung-Hwan Kim, In-Chul Lee, Jeong-Hyeon Lim et al. Protective effects of pine bark extract on developmental toxicity of cyclophosphamide in rats. Food and Chemical Toxicology 50 (2012) 109–115. [7] Lijuan Sun, Cheng Luo, Jiangang Long et al. Acrolein is a mitochondrial toxin: Effects on respiratory function and enzyme activities in isolated rat liver mitochondria. Mitochondrion 6 (2006) 136–142. [8] J. Luo, J., Shi. Acrolein induces axolemmal disruption, oxidative stress and mitochondrial impairment in spinal cord tissue. Neurochem. (2004) Int. 44, 475–486. [9] J R. Adams, J.D., Klaidman. Acrolein-induced oxygen radical formation. Free Radic. Biol. Med. 15 (1993) 187–193. [10] S. Reuter, S.C Gupta, M. M Chaturvedi et al. Oxidative stress, inflammation and cancer: How are they linked. Free Radicals in Biology and Medicine, 49 (2010) 1603–1616. [11] T. Luedde, R. F. Schwabe. NF-κB in the liver-linking injury, fibrosis and hepatocellular carcinoma. Nature Reviews. Gastroenterology and Hepatology 8 (2011) 108–118. [12] Y. J. Surh, K. S. Chun, H. H. Cha et al. Molecular mechanisms underlying chemopreventive activities of anti-inflammatory phytochemicals: Down-regulation of COX-2 and iNOS through suppression of NF-κB activation. Mutation Research. 480–481 (2001) 243–268.

18

[13] R. Simone, C. G. Subash, M. C. Madan et al. Oxidative stress, inflammation, and cancer: How are they linked. Free Radical Biology & Medicine 49 (2010) 1603–1616. [14] M. Karin, M .Delhase. The I kappa B kinase (IKK) and NF-kappa B: key elements of proinflammatory signalling. Seminars in Immunology. 12 (2000) 85–98. [15] C. S. Williams, M. Mann, R. N. Du Bois. The role of cyclooxygenases in inflammation, cancer, and development. Oncogene .18 (1999) 7908-7916. [16] O. I.Yusuf, O. Emin, C. Mustafa et al. Protective effect of curcumin in cisplatin-induced oxidative injury in rat testis: mitogen-activated protein kinase and nuclear factor-kappa B signaling pathways. Human Reproduction. 24 (2009) 1717-1725. [17] E. Gilad, H. R. Wong, B. Zingarelli et al. Melatonin inhibits expression of the inducible isoform of nitric oxide synthase in murine macrophages: role of inhibition of NF kappa B activation. FASEB J. 112 (1998) 685–693. [18] J. J. Haddad. Mitogen-activated protein kinases and the evolution of Alzheimer’s: a revolutionary neurogenetic axis for therapeutic intervention. Prog Neurobiol. 73 (2004) 359– 377. [19] E. Herlaar, Z. Brown. p38 MAPK signalling cascades in inflammatory disease. Mol Med Today. 5 (1995) 439–447. [20] V. G. Gutierrez, F. S. Maldonado, G. A. Ontiveros et al. Role of p38 in nitric oxide synthase and cyclooxygenase expression and nitric oxide and PGE2 synthesis in human gingival fibroblasts stimulated with lipopolysaccharides. Life Sci. 77 (2005) 60-73.

19

[21] K. Bhatia, F. Ahmad, H. Rashid et al. Protective effect of Sallylcysteine against cyclophosphamide-induced bladder hemorrhagic cystitis in mice. Food Chem.Toxicol. 46 (2008) 3368–3374. [22] A. S. Jalali, S. Hasanzadeh, H. Malekinejad. Achillea millefolium inflorescence aqueous extract ameliorates cyclophosphamide-induced toxicity in rat testis: stereological evidences. Chin J. Nat. Med. 10 (2012) 0247–0254. [23] K.B.H. Kumar, R. Kuttan. Chemoprotective activity of an extract of Phyllanthus amarus against cyclophosphamide induced toxicity in mice. Phytomedicine 12 (2005) 494–500. [24] P. T. Sudharsan, Y. Mythili, E. Selvakumar et al. Cardioprotective effect of pentacyclic triterpene, lupeol and its ester on cyclophosphamide induced oxidative stress. Hum. Exp. Toxicol. 24 (2005) 313–318. [25] V. Uivarosi, S. F. Barbuceanu, V. Aldea et al. Synthesis, spectral and thermal studies of new rutin vanadyl complexes. Molecules 15, 1578 Basel, Switzerland 2010. [26] K. Sattanathan, C. K. Dhanapal, R. Umarani et al. Beneficial health effects of rutin supplementation in patients with diabetes mellitus. Journal of Applied Pharmaceutical Science. 8 (2011) 227-231. [27] M. M. Khan, S. S. Raza, H. Javed et al. Rutin Protects Dopaminergic Neurons from Oxidative Stress in an Animal Model of Parkinson’s Disease. Neurotox Res. 22 (2012) 1–15. [28] I.B. Afanasev, A.I. Dorozhko, A.V. Brodskii et al. Chelating and free radical scavenging mechanisms of inhibitory action of rutin and quercetin in lipid peroxidation. Biochem Pharmacol. 38 (1989) 1763-1769.

20

[29] S. Nafees, S. T. Ahmad, A. Wani. Modulatory effects of gentisic acid against genotoxicity and hepatotoxicity induced by cyclophosphamide in Swiss albino mice. Journal of pharmacy and pharmacology. 64 (2012) 259-267. [30] S. Reitman, S. A. Frankel. Colorimetric method for the determination of serum oxaloacetate and glutamic pyruvate transaminases. Am J Clin Pathol. 28 (1957) 56-66. [31] R. Khan, A. Q. Khan, W. Qamar. Chrysin protects against cisplatin-induced colon toxicity via amelioration of oxidative stress and apoptosis: probable role of p38MAPK and p53. Toxicol Appl Pharmacol. 258 (2012) 315-329. [32] D. J. Jollow et al. Bromobenzene induced liver necrosis: protective role of glutathione and evidence for 3, 4-bromobezene oxide as the hepatotoxic metabolite. Pharmacol. 11 (1974) 151-169. [33] I. Carlberg, B. Mannervik. Glutathione reductase levels in rat brain. J Biol Chem. 250 (1975) 5475-5480. [34] J. Mohandas et al. Differential distribution of glutathione and glutathione related enzymes in rabbit kidney. Cancer Res. 44 (1984) 5086-5091.

[35] A. Claiborne Catalase activity. In: Greenwald RA, ed. CRC Handbook of Methods in Oxygen Radical Research. Boca Raton, FL: CRC Press. 283-284 1985. [36] J. R. Wright et al. Cytosolic factors that affect microsomal lipid peroxidation in lung and liver. Arch Biochem Biophys. 206 (1981) 296-304. [37] S. Nafees , N. Ali , S. Rashid et al. Chemopreventive Effect of Bauhinia Purpurea against Chemically Induced Hepatocarcinogenesis via Amelioration of Oxidative Damage, Cell Proliferation and Induction of Apoptosis in Wistar Rats. Toxicology international. 20 (2013) 117-125. 21

[38] O. H. Lowry, N. J. Rosebrough, A. L. Farr, et al. Protein measurement with the Folin phenol reagent. Journal of Biological Chemistry. 193 (1951) 265–275. [39] Arriazu, R., Pozuelo, J.M., Henriques-Gil, N., Perucho, T., Martin, R., Rodriguez, R., Santamaria, L., 2006. Immunohistochemical study of cell proliferation, Bcl-2, p53 and caspase-3 expression on preneoplastic changes induced by cadmium and zinc chloride in the ventral rat prostate. J. Histochem. Cytochem. 54, 981–536. [40] D. N. Tripathi, V. Jena Astaxanthin inhibits cytotoxic and genotoxic effects of cyclophosphamide in mice germ cells. Toxicology. 248 (2008) 96-103. [41] K. Sheeja, G. Kuttan. Ameliorating effects of Andrographis Paniculata extract against cyclophosphamide-induced toxicity in mice. Asian Pac. J. Cancer Prev. 7 (2006) 609-614. [42] D.N. Tripathi, G. B. Jena Astaxanthin intervention ameliorates cyclophosphamideinduced oxidative stress, DNA damage and early hepatocarcinogenesis in rat: role of Nrf2, p53, p38 and phase-II enzymes. Mutat. Res. 696 (2010) 69-80. [43] S. Senthilkumar, T. Devaki, M.B. Manohar et al. Effect of squalene on cyclophosphamide induced toxicity. Clin. Chim. Acta. 364 (2006) 335–342. [44] K. Bhatia, M. Kaur, F. Atif et al. Aqueous extract of Trigonella foenum-graecum L. ameliorates additive urotoxicity of buthionine sulfoximine and cyclophosphamide in mice. Food Chem. Toxicol. 44 (2006) 1744–1750. [45] S. Kouidhi, K. Rouissi, B. Hamrita et al. Therapeutic effects of aloe Vera plant extract against cyclophosphamide and buthionine sulfoximine induced toxicities in the bladder. Biochem. Pharmacol. 1 (2012) 1.

22

[46] Z. Yousefipour, K. Ranganna, M.A Newaz et al. Mechanism of acrolein-induced vascular toxicity. J. Physiol. Pharmacol. 56 (2005) 337-353. [47] M.M. Khan, A. Ahmad, T. Ishrat et al. Rutin protects the neural damage induced by transient focal ischemia in rats. Brain Res. 1292 (2009) 123-35. [48] N. S. Rajasekaran, H. Devaraj, S.N. Devaraj The effect of glutathione monoester (GME) on glutathione (GSH) depleted rat liver. J. Nutr. Biochem. 13 (2002) 302–306. [49] R. Haque, B. Bin-Hafeez, S. Parvez et al. Aqueous extract of walnut (Juglans regia L.) protects mice against cyclophosphamide induced biochemical toxicity. Hum. Exp. Toxicol. 22 (2003) 473–480. [50] R. J. Dunlop, C.W. Campbell. Cytokines and advanced cancer. J Pain Symp Manag. 20 (2000) 214-32. [51] Q. Ma, K. Kineer. Chemoprotection by phenolic antioxidants. J Biol Chem. 277 (2002) 2477-2484. [52] C.B.D. Anna., F. Francisco, F. M. Larissa et al. Protective effect of simvastatin in the cyclophosphamide-induced hemohrragic cystitis in Rats. Acta Cirurgica Brasileira. 25 (2010) 43-46. [53] A. Wani, A. Seth., S. Sultana. Rutin attenuates cisplatin induced renal inflammation and apoptosis by reducing NFkB, TNF-α and caspase-3 expression in wistar rats. Food Chem. Toxicol 49 (2011) 2013-2021. [54] E.O. Farombi, S. Shrotriya, Y. J. Surh. Kolaviron inhibits dimethyl nitrosamine induced liver injury by suppressing COX-2 and i-NOS expression via NF-kB and AP-1. Life Sci. 84 (2009) 149-155.

23

[55] D.O.Moon, M.O. Kim, S.H. Kang. Sulforaphane suppresses TNF-α mediated activation of NF-jB and induces apoptosis through activation of reactive oxygen species-dependent caspase-3. Cancer Lett. 274 (2009) 132-142. [56] G. Gloire, S. Legrand-Poels, J. Piette. NFkB activation by reactive oxygen species: fifteen years later. Biochem Pharmacol. 72 (2006) 1493-1505. [57] S. Martinez-Florez, B. Gutierrez-Fernandez, S .Sanchez-Campos et al. Quercetin attenuates NFkB activation and nitric oxide production in interleukin-1b-activated rat hepatocyte. J Nutr. 135 (2005) 1359-1365. [58] D. Trombetta, F. Cimino, M.T. Cristiani et al. In vitro protective effects of two extracts from bergamot peels on human endothelial cells exposed to tumor necrosis factor-alpha. J Agric Food Chem. 58 (2010) 8430-8436. [59] R. H. Liu, J. H. Hotchkiss. Potential genotoxicity of chronically elevated nitric oxide: a review. Mutat Res. 339 (1995) 73-89. [60] S. Ghosh, M.S. Hayden. New regulators of NFkB in inflammation. Nat Rev Immunol. 8 (2008) 837-848. [61] R. Medzhitov, T. Horng. Transcriptional control of the inflammatory response. Nat Rev Immunol. 9 (2009) 692-703. [62] M.E. Goetz, A. Luch. Reactive species: a cell damaging rout assisting to chemical carcinogenesis. Cancer Letters 266 (2008) 73-83. [63] D.X. Hou, K. Kai, J.J. Li et al. Anthocyanidins inhibit activator protein 1 activity and cell transforation: structureactivity relationship and molecular mechanisms. Carcinogenesis 25 (2004) 29-36.

24

[64] J.M. Kyriakis, J.Avruch. Mammalian mitogen-activated protein kinase signal transduction pathways activated by stress and inflammation. Physiological Reviews 81 (2001) 807-869. [65] S. Kumar, J. Boehm, J.C. Lee. p38 MAP kinases: key signalling molecules as therapeutic targets for inflammatory diseases. Nature Reviews Drug Discovery 2 (2003) 717726. [66] S. Subramanian, D. Thiruvengadam, M. Bhakthavatchalam et al. Effect of squalene on cyclophosphamide induced toxicity. Clinica Chimica Acta. 364 (2006) 335-342.

25

Figure Legends Fig.1: Results represent mean ± SE of six animals per group. Levels of proinflammatory cytokine

TNF-α was found elevated significantly in CP treated group when compared with the control group [***p < 0.001]. Pretreatment with rutin significantly [#p < 0.05,

##

p < 0.01] decreased

TNF-α level dose dependently. There is no significance difference in the level of TNF-α between control and only rutin treated group. Rutin; D1 = 50 mg/kg/b.wt.; D2 = 100 mg/kg/b.wt. Fig.2 Results represent mean ± SE of six animals per group. Levels of proinflammatory cytokine

IL-6 was found elevated significantly in CP treated group when compared with the control group [***p < 0.001]. Pretreatment with rutin significantly [#p < 0.05,

##

p < 0.01] decreased

IL-6 level dose dependently. There is no significance difference in the level of IL-6 between control and only rutin treated group. Rutin; D1 = 50 mg/kg/b.wt.; D2 = 100 mg/kg/b.wt.

Fig. 3 [A-E] Representative photomicrograph of histopathological examination of rat liver 40x [A]

The liver architecture of control group was normal, devoid of any signs of inflammatory changes. Nucleolus was clear, the central veins and hepatic lobule were clearly seen. [B] In CP treated group, dilated and congested sinusoidal space with infiltration of lymphocytes and small portal space with inflammation were observed which are indicative of hepatocellular injury as shown by arrows. [C & D] Pretreatment with rutin reverted these changes in liver marked by less dilated and congested sinusoidal space with less inflammatory cell infiltration as shown by arrows. [E] Liver showed normal histology almost similar to control. Rutin; D1 = 50

mg/kg/b.wt.; D2 = 100 mg/kg/b.wt. Fig. 4 [A-D] Representative photomicrograph of immunohistochemistry of i-NOS of rat liver 40 x

[A] Control group showing negligible staining. [B] Staining intensity of i-NOS, was remarkably high in CP treated group as compared to control group as shown by arrows. 26

[C&D] Pretreatment with rutin caused a noticeable reduction in the staining intensity of iNOS dose dependently as shown by arrows. Rutin; D1 = 50 mg/kg/b.wt.;

D2 = 100

mg/kg/b.wt. Fig. 4.1. Results represent mean±standard error of mean of 6 animals. Results obtained are significantly different from group I (**p < 0.01). Results obtained are significantly different from group II (##p < 0.01, and

###

p < 0.001). Rutin; D1 = 50 mg/kg/b.wt.; D2 = 100

mg/kg/b.wt. Fig. 5 [A-D] Representative photomicrograph of immunohistochemistry of COX-2 of rat liver 40 x.

[A] Control group showing negligible staining. [B] Staining intensity of COX-2 was remarkably high in CP treated group as compared to control group as shown by arrows. [C & D] Pretreatment with rutin caused a noticeable reduction in the staining intensity of COX-2 dose dependently as shown by arrows. Rutin; D1 = 50 mg/kg/b.wt.; D2 = 100 mg/kg/b.wt. Fig. 5.1. Results represent mean±standard error of mean of 6 animals. Results obtained are significantly different from group I (**p < 0.01). Results obtained are significantly different from group II (##p < 0.01 and

###

p < 0.001). Rutin; D1 = 50 mg/kg/b.wt.; D2 = 100

mg/kg/b.wt. Fig. 6 [A-D] Representative photomicrograph of immunohistochemistry of p38-MAPK of rat liver 40x. [A] Control group showing negligible staining [B] Staining intensity of p38-MAPK, was

remarkably high in CP treated group as compared to control group as shown by arrows. [C & D] Pretreatment with rutin caused a noticeable reduction in the staining intensity of p38MAPK dose dependently as shown by arrows. Rutin; D1 = 50 mg/kg/b.wt.; D2 = 100 mg/kg/b.wt. Fig.6.1. Results represent mean±standard error of mean of 6 animals. Results obtained are significantly different from group I (**p < 0.01). Results obtained are significantly different from group II (#p < 0.05 and ##p < 0.01). Rutin; D1 = 50 mg/kg/b.wt.; D2 = 100 mg/kg/b.wt. 27

Fig. 7 [A-D] Representative photomicrograph of immunohistochemistry of NFκB of rat liver 40x.

[A] Control group showing very less immunopositive cells. [B] Number of immunopositive cells was remarkably high in CP treated group as shown by arrows. [C&D] Pretreatment with rutin caused a noticeable reduction in the number of NFκB immunopositive cells dose dependently as shown by arrows. Rutin; D1 = 50 mg/kg/b.wt.; D2 = 100 mg/kg/b.wt. Fig.7.1. Results represent mean±standard error of mean of 6 animals. Results obtained are significantly different from group I (**p < 0.01). Results obtained are significantly different from group II (#p < 0.05 and ##p < 0.01). Rutin; D1 = 50 mg/kg/b.wt.; D2 = 100 mg/kg/b.wt.

28

Immunohistochemical Score

5

***

4.5 4

#

3.5 3 2.5 2

###

1.5 1 0.5 0

Group I

Group II

Group III

Group IV

Fig.5.1 Quantitative estimation of Cox-2

Immunohistochemical Score

6

**

5 4

## 3

###

2 1 0 Group I

Group II

Group III

Group IV

Fig.4.1 Quantitative estimation of i-NOS

Immunohistochemical Score

6

**

5 4

#

3

##

2 1 0 Group I

Group II

Group III

Group IV

Fig.6.1 Quantitative estimation of P38-MAPK

35

** 30 25 Labelling index %

#

20 15

##

10 5 0 Group I

Group II Group III Fig.7.1 Quantitative estimation of NFҡB

Group IV

Table.1 Results of pretreatment of Rutin on serum markers enzymes like AST, ALT and LDH on cyclophosphamide administration in liver of Wistar rats. Treatment regimen per group

AST(IU/L)

ALT(IU/L)

LDH (n mol NADH oxidised / min/ mg protein) 163.99 ± 40.70

Group I (control)

51.36 ± 0.08

49.01±0.25

Group II (only CP)

98.79 ± 0.39***

100.24±0.87***

507.84 ± 25.26 **

Group III (D1+CP)

71.85 ± 0.54 ###

70.13±0.30###

296.24 ± 40.70 #

Group IV (D2+CP)

53.07 ± 0.22 ###

52.89±0.33 ###

179.86 ± 3.34##

Group V (only D2)

52.98 ± 4.09

51.85±1.72

169.28 ± 12.06

. Results represent mean ± SE of six animals per group. Results obtained are significantly different from control group (**P < 0.01,

***

P < 0.001). Results obtained are significantly different from

cyclophosphamide administered group (#P < 0.05, ##P < 0.01 and b.wt.; D2 = 100mg/kg b.wt.

29

###

P < 0.001). Rutin; D1= 50mg/kg

Table.2 Results of pretreatment of Rutin on antioxidant enzymes like GSH, GR and GPx on cyclophosphamide administration in liver of Wistar rats. Treatment regimen per group

Group I (control)

GSH (n mol CDNB Conjugate formed /g tissue) 9.15 ± 0.18

GR (n mol NADPH Oxidized/min/ mg protein) 298.09 ±2.94

Group II (only CP)

2.07 ± 0.31 ***

107.03±2.58***

Group III (D1+CP)

5.50 ± 0.66 ##

205.45±5.64###

Group IV (D2+CP)

8.17 ± 0.16 ###

288.02±1.03 ###

Group V (only D2)

8.63 ± 0.12

291.57±0.43

GPX (n mol NADPH Oxidized/min/ mg protein) 265.28 ± 0.91 99.82 ± 0.97 *** 173.21 ± 7.06### 246.96 ± 13.85### 251.11 ± 1.38

Results represent mean ± SE of six animals per group. Results obtained are significantly different from control group (***P < 0.001). Results obtained are significantly different from cyclophosphamide treated group (##P < 0.01 and (###P < 0.001). RutinD1= 50mg/kg/b wt; D2 = 100mg/kg/b wt.

30

Table.3 Results of pretreatment of Rutin on antioxidant enzymes like catalase, XO and LPO on cyclophosphamide administration in liver of Wistar rats. Treatment regimen per group

Group I (control)

Catalase (nmol H2O2 consumed/min/mg protein) 29.28 ± 2.19

XO (µg uric acid formed/min/mg protein) 0.27 ± 0.07

LPO (n mol MDA formed / hr/ g tissue)

Group II (only CP)

10.94 ± 7.84***

0.86 ± 0.05 **

27.71 ± 1.46 ***

Group III (D1+CP)

20.40 ± 8.38#

0.65 ±0.02 #

19.55 ± 0.56 #

Group IV (D2 +CP)

26.14 ± 20.47 ##

0.31 ±0.031##

12.12 ± 1.36 ###

Group V (only D2)

27.08 ± 16.22

0.29 ± 0.05

11.10 ± 0.66

10.89 ± 0.45

Results represent mean ± SE of six animals per group. Results obtained are significantly different from control group (**P < 0.01 and #

***

P < 0.001). Results obtained are significantly different from

cyclophosphamide treated group ( P < 0.05,

##

P < 0.01) and (###P

MAPK pathway.

Cyclophosphamide is a potent anticancer agent. However its clinical use is restricted because of its marked organ toxicity associated with increased o...
1MB Sizes 0 Downloads 15 Views