Sunteți pe pagina 1din 15

Our reference: PATPHY 726

P-authorquery-v9

AUTHOR QUERY FORM


Journal: PATPHY Please e-mail or fax your responses and any corrections to: E-mail: corrections.esch@elsevier.thomsondigital.com Article Number: 726 Dear Author, Please check your proof carefully and mark all corrections at the appropriate place in the proof (e.g., by using on-screen annotation in the PDF le) or compile them in a separate list. Note: if you opt to annotate the le with software other than Adobe Reader then please also highlight the appropriate place in the PDF le. To ensure fast publication of your paper please return your corrections within 48 hours. For correction or revision of any artwork, please consult http://www.elsevier.com/artworkinstructions. Any queries or remarks that have arisen during the processing of your manuscript are listed below and highlighted by ags in the proof. Click on the Q link to go to the location in the proof. Location in article Q1 Q2 Query / Remark: click on the Q link to go Please insert your reply or correction at the corresponding line in the proof Please conrm that given names and surnames have been identied correctly. Fig. 2 will appear in black and white in print and in color on the web. Based on this, the respective gure captions have been updated. Please check, and correct if necessary. Fax: +353 6170 9272

Thank you for your assistance.

PATPHY 726 114

ARTICLE IN PRESS

Pathophysiology xxx (2012) xxxxxx

Traditional extract of Pithecellobium dulce fruits protects mice against CCl4 induced renal oxidative impairments and necrotic cell death
Q1

3 4

Pabitra Bikash Pal, Sankhadeep Pal, Prasenjit Manna, Parames C. Sil


Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, West Bengal, India Received 4 April 2011; accepted 17 February 2012

5 6 7 8 9 10 11 12 13 14 15 16 17 18

Abstract The present study has been carried out to investigate the role of the aqueous extract of the fruits of Pithecellobium dulce (AEPD) against carbon tetrachloride (CCl4 ) induced renal oxidative injury in mice. HPLC analysis shows that AEPD contains phenolics, avonoids and saponins as the major active components. Creatinine and blood urea nitrogen (BUN) levels were assayed to determine renal protective action of AEPD in CCl4 -induced renal pathophysiology. Its antioxidant activity was determined by measuring radical scavenging activity, antioxidant enzymes activities, GSH content, protein carbonylation and lipid peroxidation. In addition, FACS analysis, DNA fragmentation and histological studies were carried out to determine its effect in CCl4 induced renal oxidative injury and cell death. CCl4 exposure increased the intracellular reactive oxygen species production, decreased intracellular antioxidant defence, reduced mitochondrial membrane potential, attenuated the intracellular ATP content and caused renal cell death mainly via the necrotic pathway as revealed by DNA fragmentation analysis. Treatment with AEPD both prior and post to the toxin exposure protected the organ from CCl4 induced oxidative insult. Histological studies also support our results. Combining, results suggest that the protective role of AEPD against CCl4 induced renal oxidative impairments is probably due to the antioxidative properties present in its active constituents. 2012 Published by Elsevier Ireland Ltd.
Keywords: Carbon tetrachloride; Oxidative impairment and kidney dysfunction; Necrosis; Pithecellobium dulce fruit extract; Antioxidants; Renal-protection; Male mice

19 20 21

22

1. Introduction Carbon tetrachloride (CCl4 ), an environmental toxin, is enormously used in chemical industry as an organic solvent. It is also used in medicine as a vermifuge in the treatment of hookworm disease. It is exposed to the whole body by
Abbreviations: AEPD, aqueous extract of the fruits of Pithecellobium dulce; BSA, bovine serum albumin; BUN, blood urea nitrogen; CCl4 , carbon tetrachloride; CDNB, 1-chloro-2,4-dinitrobenzene; DPPH, 2,2-diphenyl-1-picrylhydrazyl; DTNB, 5,5 -dithiobis(2-nitrobenzoic acid) [(Ellmans reagent)]; EDTA, ethylene diamine tetraacetic acid; GSH, glutathione; GSSG, glutathione disulde; H2 O2 , hydrogen peroxide; MDA, malonaldehyde; NEM, N-ethylmaleimide; NADH, nicotinamide adenine dinucleotide reduced disodium salt; NBT, nitro blue tetrazolium chloride; PMT, phenazine methosulphate; ROS, reactive oxygen species; NaN3 , sodium azide; TBA, thiobarbituric acid; TBARS, thiobarbituric acid reactive substances; TCA, trichloroacetic acid. Corresponding author. Tel.: +91 33 25693243; fax: +91 33 2355 3886. E-mail addresses: parames@bosemain.boseinst.ac.in, parames 95@yahoo.co.in (P.C. Sil). 0928-4680/$ see front matter 2012 Published by Elsevier Ireland Ltd. doi:10.1016/j.pathophys.2012.02.001

23 24 25 26

inhalation, ingestion or absorption through the skin. Its exposure results poisoning in many cases although liver, brain, kidney, muscle, fat and blood are found to be seriously affected. Long time administration of CCl4 caused brosis, cirrhosis and hepatic carcinoma. CCl4 toxicity results from its bioactivation to the trichloromethyl free radical by cytochrome P450 isozymes [1]. The trichloromethyl radical reacts with oxygen to form the highly toxic reactive trichloromethyl peroxy radical, a reactive oxygen species (ROS). Free radical induced lipid peroxidation believed to be one of the major causes of cell membrane damage leading to a number of pathological situations. Recent reports show that in addition to hepatic disorders, CCl4 also causes renal disorders by generating free radicals. Reports from Perez et al. [2], Ogeturk et al. [3] and Churchill et al. [4] suggest that acute and chronic renal injuries occur due to the exposure of this solvent. Many herbs and medicinal plants in India are rich in natural sources of antioxidants. These medicinal plants [e.g.

27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45

Please cite this article in press as: P.B. Pal, et al., Traditional extract of Pithecellobium dulce fruits protects mice against CCl4 induced renal oxidative impairments and necrotic cell death, Pathophysiology (2012), doi:10.1016/j.pathophys.2012.02.001

PATPHY 726 114

ARTICLE IN PRESS
P.B. Pal et al. / Pathophysiology xxx (2012) xxxxxx

2
46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73

Andrographis paniculata [5,6], Phylanthus niruri [712], Cajanus indicus [1318], Silybum marianum [19], Terminalia arjuna [2023], etc.] are used for the treatment of renal, hepatic and other organ disorders. Pithecellobium dulce is a well known medicinal plant, generally used for fencing and tanning, fodder for feed and pods food. In this particular study, protective role of the aqueous extract of the fruits of P. dulce was evaluated against CCl4 -induced renal pathophysiology using murine model. In our present study, free radical scavenging activity of the extract was determined from its 2,2-diphenyl-1picrylhydrazyl (DPPH), hydroxyl (OH) and superoxide (O2 ) quenching ability. Renal oxidantantioxidant status was determined by measuring the levels of (a) lipid peroxidation and protein carbonylation; (b) activities of the antioxidant enzymes superoxide dismutase (SOD), catalase (CAT), glutathione-S-transferase (GST), glutathione reductase (GR) and (c) the levels of cellular metabolites in the kidney tissue of the experimental mice. The mode of cell death in kidney tissue has been evaluated by DNA fragmentation analysis. The involvement of mitochondrial event and the intracellular ATP level has also been investigated in our study. In addition, histological studies were carried out to assess the ultrastructural changes in murine kidney. The results of the present study could clarify the role of AEPD in the protection of CCl4 -induced nephrotoxicity and may shed light on a possible solution to the unavoidable ways of its exposures in humans.

2.3. Chemicals and other reagents 2,2-Diphenyl-1-picryl hydrazyl (DPPH), Bradford reagent, bovine serum albumin (BSA), and protein estimation kit were purchased from SigmaAldrich Chemical Company (St. Louis, MO, USA). 1-Chloro-2,4-dinitrobenzene (CDNB), carbontetrachloride (CCl4 ), disodium hydrogen phosphate (Na2 HPO4 ), sodium dihydrogen phosphate (NaH2 PO4 ), 5,5 -dithiobis(2-nitrobenzoic acid) [DTNB, (Ellmans reagent)], glacial acetic acid, hydrogen peroxide (H2 O2 ), ethylene diamine tetra acetic acid (EDTA), phenazine methosulphate (PMT), nitro blue tetrazolium (NBT), sodium pyrophosphate, potassium dihydrogen phosphate (KH2 PO4 ), trichloro acetic acid (TCA), thiobarbituric acid (TBA), reduced glutathione (GSH) were bought from Sisco research laboratory, India.

95

96 97 98 99 100 101 102 103 104 105 106 107 108 109

2.4. Preparation of fruit extract of P. dulce After collection, the fruits were cut into small pieces and they were homogenized in 50 mM phosphate buffer, pH 7.2 at 4 C. The homogenized mixture was centrifuged at 12,000 g for 30 min to get rid of unwanted debris and lyophilized. The freeze-dried material was weighed, dissolved in the same phosphate buffer and used in different experiments needed for this study.

110

111 112 113 114 115 116 117

2.5. Qualitative analysis of AEPD


74

118

2. Materials and methods 2.1. Plant P. dulce (PD), one of the important traditional medicinal plants in India, belongs to the family of Fabaceae. In this study, the fruits of PD were collected in the summer session of the current year from the local market, Kolkata, India. After collection, one voucher specimen of this fruit was deposited in the Central National Herbarium (CNH), Botanical Survey of India (BSI), Howrah, West Bengal, India. 2.2. Experimental animals All experiments in this study were performed on Swiss albino mice (male, body weight 20 2 g, purchased from M/S Gosh Enterprises, Kolkata, India). The animals were acclimatized under laboratory conditions for two weeks prior to the experiments. They were maintained under standard conditions of temperature (23 2 C) and humidity (50 10%) with an alternating 12 h light/dark cycles. The animals had free access to tap water and fed standard pellet diet (Agro Corporation Private Ltd., Bangalore, India). All the experiments with animals were carried out according to the Guidelines of the Institutional Animal Ethical Committee.

75

76 77 78 79 80 81 82

83

84 85 86 87 88 89 90 91 92 93 94

Phytochemical analysis of AEPD was carried out to investigate the presence of active ingredients such as phenolics, avonoids, and saponins (Figs. 1 and 2). About 0.5 g of crude powder was heated with 5 ml of ethyl alcohol over a steam bath for 3 min and then few drops of neutral ferric chloride solution were added. Appearance of violet colouration conrmed the presence of phenolic compounds in the extract [24]. A portion of crude powder was heated with 10 ml of ethyl acetate over a steam bath for 3 min. The mixture was ltered and 4 ml of the ltrate was shaken with 1 ml of dilute ammonia solution. Appearance of yellow colouration conrmed the presence of avonoids in the extract [25]. To nd out the presence of saponins, small amount (0.5 g) of crude powder was shaken with water in a test tube and it was warmed in a water bath. Persistent froth formation indicated the presence of saponins in the extract [26].

119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135

2.6. Quantitative analysis of AEPD 2.6.1. Total phenolic compounds estimation The content of total phenolic compounds of the extract was determined according to McDonalds method using FolinCiocalteau reagent (gallic acid as a standard) [27].

136

137 138 139 140

Please cite this article in press as: P.B. Pal, et al., Traditional extract of Pithecellobium dulce fruits protects mice against CCl4 induced renal oxidative impairments and necrotic cell death, Pathophysiology (2012), doi:10.1016/j.pathophys.2012.02.001

PATPHY 726 114

ARTICLE IN PRESS
P.B. Pal et al. / Pathophysiology xxx (2012) xxxxxx 3
144 145 146 147

whole solution was ltered through Whatman lter paper no. 42 (125 m). The ltrate was transferred into a crucible, evaporated into dryness and weighed to a constant weight [28].

2.6.3. Determination of saponin content Crude powder (20 g) was put into a conical ask and 100 ml of 20% aqueous ethanol was added. The samples were heated over a hot water bath for 4 h with continuous stirring at about 55 C. The mixture was ltered and the residue was extracted with another 200 ml of 20% ethanol. The extract was reduced to 40 ml over water bath at about 90 C. The concentrate was transferred into 250 ml separating funnel, 20 ml of diethyl ether was added and the mixture was shaken vigorously. The aqueous layer was recovered while the ether layer was discarded. The purication process was repeated. 60 ml of n-butanol was then added. The combined n-butanol extracts were washed twice with 10 ml of 5% aqueous sodium chloride. The remaining solution was heated in a water bath. After evaporation, the samples were dried in the oven to constant weight and the saponin content was calculated [29].

148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164

2.7. HPLC analysis of AEPD HPLC analysis of AEPD was carried out using a C18 column (8 nm 10 cm). The column was eluted with a mobile phase 70:30 methanol/water, ow rate 1 ml/min. UV detection was done at 254 nm. A standard curve was prepared using the mixture of standard compounds, such as gallic acid (standard for phenolic compounds), quercetinn (standard for avonoid compounds) and digitonin (standard for saponin compounds).

165

166 167 168 169 170 171 172 173

Fig. 1. Upper panel: reverse phase HPLC analysis of AEPD. Lower panel: reverse phase HPLC analysis of a mixture of pure compounds gallic acid (peak I), quercetin (peak II) and digitonin (peak III).
141 142 143

2.6.2. Flavonoid content estimation Crude powder (10 g) was extracted repeatedly with 100 ml of 80% aqueous methanol at room temperature. The

Fig. 2. (A) DPPH radical scavenging activity of AEPD in cell free system. Each measurement was made six times. Data represent the averages SD of 6 separate experiments. * the optimum dose of AEPD at which it shows its maximum DPPH radical scavenging activity. (B) Hydroxyl (black square) and Q2 superoxide radical (red circle) scavenging activities of AEPD in cell free system. Each measurement was made six times. Data represent the averages SD of 6 separate experiments. * the optimum dose of AEPD at which it shows its maximum hydroxyl and superoxide radicals scavenging activities. (For interpretation of the references to colour in this gure legend, the reader is referred to the web version of the article.)

Please cite this article in press as: P.B. Pal, et al., Traditional extract of Pithecellobium dulce fruits protects mice against CCl4 induced renal oxidative impairments and necrotic cell death, Pathophysiology (2012), doi:10.1016/j.pathophys.2012.02.001

PATPHY 726 114

ARTICLE IN PRESS
P.B. Pal et al. / Pathophysiology xxx (2012) xxxxxx

4
174 175

2.8. Determination of free radical scavenging activity of AEPD in cell free system 2.8.1. Determination of DPPH radical quenching activity The free radical scavenging activity of AEPD was measured by the DPPH radical quenching method [30]. Various concentrations of extract were added to DPPH in methanol (125 M, 2 ml) solution and the nal volume was by water to 4 ml. The solution was shaken and incubated at 37 C for 30 min in dark. The decrease in absorbance of DPPH was measured at 517 nm. Percent inhibition was calculated by comparing the absorbance values of control and the extract. Percent inhibition was calculated by comparing the absorbance values of control and the sample. Percentage inhibition = A 1 A2 100 A1

176 177 178 179 180 181 182 183 184 185 186 187

treatment (at a dose of 1 ml/kg body weight for 2 days, orally) respectively. To determine the time dependent effects of AEPD, mice were divided into nine groups each consisting of six animals. First two groups were served as normal control (received only water as vehicle) and toxin control (received CCl4 at a dose of 1 ml/kg body weight for 2 days, orally) respectively. Other seven groups of animals were treated with AEPD orally at a dose of 200 mg/kg body weight, once daily for 1, 3, 5, 7, 10, 12 and 14 days prior to CCl4 intoxication (at a dose of 1 ml/kg body weight for 2 days, orally). After 24 h to the administration of CCl4 all mice were sacriced. The CAT activities were measured in the kidney tissue homogenates of all the experimental mice. 2.10. Experimental mice groups and treatment The animals were divided into seven groups each consisted of six mice and they were treated as follows. Group 1 normal control (received only water as vehicle). Group 2 animals were administered with AEPD at a dose of 200 mg/kg body weight for 7 days, orally. Group 3 toxin control; animals were administered with CCl4 orally at a dose of 1 ml/kg body weight in liquid parafn (1:1, v/v) for 2 days. Group 4 animals were administered with AEPD at a dose of 200 mg/kg body weight for 7 days, orally prior to CCl4 administration (at a dose of 1 ml/kg body weight for 2 days). Group 5 animals were administered with AEPD at a dose of 200 mg/kg body weight for 7 days, orally after CCl4 administration (at a dose of 1 ml/kg body weight for 2 days). Group 6 animals were administered with vitamin C at a dose of 100 mg/kg body weight for 7 days, orally, prior to CCl4 administration (at a dose of 1 ml/kg body weight for 2 days). Group 7 animals were administered with CCl4 at a dose of 1 ml/kg body weight for 2 days and received normal diet for next 7 days. This group was included just to check the natural healing of the animals after toxin treatment. The in vivo experimental protocol has been summarized in Fig. 3. 2.11. Kidney tissue homogenate preparation

223 224 225 226 227 228 229 230 231 232 233 234 235 236

237

188

238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264

189 190

A1 is the absorbance of the blank and A2 is the absorbance in the presence of AEPD. 2.8.2. Determination of hydroxyl radical scavenging activity The hydroxyl radical scavenging activity of the extract has been measured following the method of Nash [31]. In vitro hydroxyl radicals were generated by Fe3+ /ascorbic acid system. The detection of hydroxyl radicals was carried out by measuring the amount of formaldehyde produced from the oxidation of dimethyl sulfoxide (DMSO). The production of formaldehyde was detected spectrophotometrically at 412 nm. 2.8.3. Determination of superoxide radical scavenging activity The superoxide radical scavenging activity of the extract was measured following the method of Siddhuraju and Becker [32]. Compositions of reaction mixture are 0.1 M phosphate buffer, pH 7.4, 150 M nitroblue tetrazolium (NBT), 60 M phenazine methosulphate (PMT), 468 M NADH and various concentrations of the AEPD. Then the mixture was incubated in the dark for 10 min at 25 C and the absorbance was measured at 560 nm. Results were expressed as percentage inhibition of the superoxide radicals. 2.9. Determination of dose and time dependent effect of AEPD To nd out the dose of AEPD necessary for optimum protection against CCl4 induced renal-toxicity mice were divided into ten groups each consisting of six mice in each. First two groups were served as normal control (received only water as vehicle) and toxin control (received CCl4 at a dose of 1 ml/kg body weight for 2 days, orally) respectively. Other eight groups of animals were treated with AEPD orally at a dose of 25 mg, 50 mg, 75 mg, 100 mg, 150 mg, 200 mg, 250 mg and 300 mg/kg body weight for 7 days prior to CCl4

191 192 193 194 195 196 197 198 199 200

201 202 203 204 205 206 207 208 209 210 211

212 213

265

214 215 216 217 218 219 220 221 222

About 150 mg of kidney tissue was homogenized in 10 volume of 100 mM KH2 PO4 buffer containing 1 mM EDTA, pH 7.4 and centrifuged at 12,000 g for 30 min at 4 C from each mouse of separated groups. The supernatant was collected and used for following experiments as described below. Protein concentration of the supernatant was measured according to the method of Bradford [33] using crystalline BSA as standard.

266 267 268 269 270 271 272 273

Please cite this article in press as: P.B. Pal, et al., Traditional extract of Pithecellobium dulce fruits protects mice against CCl4 induced renal oxidative impairments and necrotic cell death, Pathophysiology (2012), doi:10.1016/j.pathophys.2012.02.001

PATPHY 726 114

ARTICLE IN PRESS
P.B. Pal et al. / Pathophysiology xxx (2012) xxxxxx 5

Fig. 3. Schematic diagram of in vivo experimental protocol.


274 275

2.12. Assessment of the levels of blood urea nitrogen (BUN) and creatinine Blood samples collected from puncturing rat heart were kept overnight to clot and then centrifuged at 3000 g for 10 min. Creatinine and BUN levels were estimated by using standard kits (Span diagnostic Ltd., India). 2.13. Lipid peroxidation and protein carbonylation estimation The extent of lipid peroxidation in terms of malondialdehyde (MDA) formation was measured according to the method of Esterbauer and Cheeseman [34]. Sample containing 1 mg protein was mixed with 1 ml TCA (20%), 2 ml TBA (0.67%) and heated for 1 h at 100 C. After cooling, the precipitate was removed by centrifugation. The absorbance of the sample was measured at 535 nm using a blank containing all the reagents except the sample. MDA content of the sample was calculated using the extinction co-efcient of MDA, which is 1.56 105 M1 cm1 . Protein carbonyl contents were determined according to the methods of Uchida and Stadtman [35]. The sample was treated with an equal volume of 0.1% (w/v) 2,4-DNPH in 2 N HCl and incubated for 1 h at room temperature and then treated with 20% TCA. After centrifugation, the precipitate was extracted three times with EtOH/EtOAc and dissolved in 8 M guanidine hydrochloride in 133 mM Tris solution

276 277 278 279

containing 13 mM EDTA. The absorbance was recorded at 365 nm. The results were expressed as nmol of DNPH incorporated/mg protein based on the molar extinction coefcient of 22,000 M1 cm1 for aliphatic hydrazones. 2.14. Determination of the activities of antioxidant enzymes The activities of antioxidant enzymes, superoxide dismutase (SOD), catalase (CAT), glutathione-S-transferase (GST) and glutathione reductase (GR) were measured in all experimental sets following the method of Sinha et al. [36]. Briey, in case of SOD assay tissue homogenates were centrifuged at 600 g for 8 min at 4 C. Supernatants were decanted and re-centrifuged at 5500 g for 15 min to form mitochondrial pellets. The supernatant was kept as cytosolic fraction. Five micrograms protein of the cytosolic fraction was mixed with sodium pyrophosphate buffer, PMT and NBT. The reaction was started by the addition of NADH. Then the reaction mixture was incubated at 30 C for 90 s and stopped by the addition of 1 ml of glacial acetic acid. The absorbance of the chromogen formed was measured at 560 nm. One unit of SOD activity is dened as the enzyme concentration required to inhibit chromogen production by 50% in 1 min under the assay condition. CAT activity was determined by following the decomposition of H2 O2 (7.5 mM) at 240 nm for 10 min and it was monitored spectrophotometrically. One unit of CAT activity

299 300 301 302

303 304

280 281

305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324

282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298

Please cite this article in press as: P.B. Pal, et al., Traditional extract of Pithecellobium dulce fruits protects mice against CCl4 induced renal oxidative impairments and necrotic cell death, Pathophysiology (2012), doi:10.1016/j.pathophys.2012.02.001

PATPHY 726 114

ARTICLE IN PRESS
P.B. Pal et al. / Pathophysiology xxx (2012) xxxxxx

6
325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341

is dened as the amount of enzyme, which reduces 1 mol of H2 O2 per minute. GST activity was assayed based on the conjugation reaction with glutathione in the rst step of mercapturic acid synthesis. The reaction mixture contains supernatant 25 g protein sample, KH2 PO4 buffer, EDTA, CDNB and GSH. The reaction was carried out at 37 C and monitored spectrophotometrically at 340 nm for 5 min. One unit of GST activity is 1 mol product formation per minute. GR activity was determined spectrophotometrically by monitoring the absorbance at 412 nm for 3 min at 24 C. Reaction mixture contained 1 mM EDTA, 0.3 mM DTNB, 2 mM NADPH and 20 mM GSSG. The enzyme activity was calculated using molar extinction coefcient of 13,600 M1 cm1 . One unit of enzyme activity is dened as the amount of enzyme, which catalyzes the oxidation of 1 mol NADPH per minute. 2.15. Estimation of the levels of cellular metabolites 2.15.1. Determination of GSH content Intracellular GSH level was measured according to the method of Ellman [37]. 720 L of sample was double diluted and 5% TCA was added to precipitate the protein content. After centrifugation (at 10,000 g for 5 min) the supernatant was taken, DTNB solution (Ellmans reagent) was added to it and the absorbance was measured at 412 nm. A standard curve was drawn using different known concentrations of GSH solution. With the help of this standard curve, GSH contents were calculated. 2.15.2. Determination of GSSG content Glutathione disulde (GSSG) was assessed by measuring its level using a kit from Calbiochem, USA following the method of Finck et al. [38]. Briey, kidney tissue samples were homogenized in ice cold m-phosphoric acid (100 g/L, Fluka). This treatment resulted in the precipitation of the proteins. After allowing the mixture to stand for 5 min at room temperature, homogenates were centrifuged (10,000 g; 10 min at 4 C). Supernatants were frozen at 80 C until further analysis. Prior to the estimation of GSSG, neutralization was achieved by adding 5 L of triethanolamine (4 mmol/L, Sigma Chemicals) to a 100 L supernatant. For GSSG assay, the thiol scavenging reagent 1-methyl-2-vinylpyridinium triuoromethanesulfonate (M2VP) was used to mask GSH very rapidly. 10 L of M2VP was added to 100 L of neutralized supernatant. Then 100 L of the recycling reagent, which contained NADPH (0.30 mM), DTNB (0.225 mM), and GR (1.6 units/mL) in a 100 mM phosphate/1 mM EDTA buffer (pH 7.4), was added. The change of absorbance was monitored at 412 nm for 3 min with a spectrophotometer. Standards and test samples were run in triplicate for each assay and the measurements were repeated three times. The reaction rate and calibration curves were used to calculate concentrations of GSSG.

2.15.3. Determination of total thiols content The total thiols (total sulfhydryl groups) content was measured according to the method of Sedlak and Lindsay [39] with some modications. About 50 L of sample was mixed with 0.6 ml of TrisEDTA buffer, 40 L of 10 mM DTNB in methanol. The nal volume was made up to 1 ml by adding methanol. The reaction mixture was incubated at room temperature for 20 min and the absorbance was measured at 417 nm. The content of total thiols was calculated using molar extinction coefcient of 13,600 M1 cm1 . 2.16. Measurement of ROS level ROS level was estimated by using 2,7-dichlorouorescein diacetate (DCFDA) as a probe following the method of LeBel and Bondy [40] as modied by Kim et al. DCF-DA diffuses through the cell membrane where it is enzymatically deacetylated by intracellular esterases to the more hydrophilic nonuorescent reduced dye dichlorouorescin. In the presence of reactive oxygen metabolites, nonuorescent DCFH rapidly oxidized to highly uorescent product DCF. For in vivo study, 100 L of tissue homogenates were incubated with the assay media (20 mM trisHCl, 130 mM KCl, 5 mM MgCl2 , 20 mM NaH2 PO4 , 30 mM glucose and 5 M DCFDA) at 37 C for 15 min. The formation of DCF was measured at the excitation wavelength of 488 nm and emission wavelength of 610 nm for 10 min by using uorescence spectrometer (HITACHI, Model No F4500) equipped with a FITC lter. 2.17. DNA fragmentation analysis The extent of DNA fragmentation (DNA ladder) has been assayed by the procedure as described by Sellins and Cohen [41]. DNA samples were isolated from the kidney tissue of the normal as well as experimental mice and they were analyzed by electrophoresing the DNA samples on agarose/ethydium bromide gel. 2.18. Assessment of mitochondrial membrane potential and integrity Reduction in mitochondrial membrane potential ( ) and hence the mitochondrial integrity plays an important role in CCl4 induced cell death. For the detection of mitochondrial , mitochondria were isolated from the renal tissue of experimental mice. Briey, renal tissue was minced, washed with ice-cold isolation buffer (10 ml TrisMOPS [0.1 M; pH 7.4], 20 ml sucrose [1 M], and 1 ml EGTATris buffer [0.1 M; pH 7.4]) and the buffer was discarded. 5 ml of fresh isolation buffer was then added, and the mixture was homogenized. The homogenates were centrifuged at 600 g at 4 C for 10 min, and then the supernatants were centrifuged at 7000 g at 4 C for 10 min, after which the supernatants were discarded. The pellets were washed once with isolation buffer, and the centrifugation steps were repeated twice.

377 378 379 380 381 382 383 384 385 386

387

388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403

342

343 344 345 346 347 348 349 350 351 352

404

353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376

405 406 407 408 409 410

411 412

413 414 415 416 417 418 419 420 421 422 423 424 425 426

Please cite this article in press as: P.B. Pal, et al., Traditional extract of Pithecellobium dulce fruits protects mice against CCl4 induced renal oxidative impairments and necrotic cell death, Pathophysiology (2012), doi:10.1016/j.pathophys.2012.02.001

PATPHY 726 114

ARTICLE IN PRESS
P.B. Pal et al. / Pathophysiology xxx (2012) xxxxxx 7 Table 1B Quantitative extract analysis of AEPD. Serial no. 1 2 3 Component Phenol content (mg/g dry weight material) Flavonoid content (mg/g dry weight material) Saponin content (mg/g dry weight material) Concentration 45.12 1.08 55.47 1.57 72.81 2.35

427 428 429 430 431 432 433 434 435 436

After the three-step centrifugation, the supernatants were discarded, and the pellets were suspended in 1 ml of isolation buffer and used for further analysis [42]. Analytic ow cytometric measurements for the membrane potential ( m ) of isolated mitochondria were performed using a FACScan ow cytometer with an argon laser excitation at 488 nm. Mitochondrial membrane potential ( m ) was estimated to the exclusion of rhodamine 123 from intact mitochondria. ROS stimulated changes of mitochondrial membrane potential will promote the uptake of rhodamine 123. 2.19. Assay of ATP level

II) and saponins (peak III). The presence of these compounds in the extract has been characterized by comparing the HPLC proles of the respective pure standard compounds. 3.2. Free radical scavenging activity of AEPD

465 466 467

437

468

438 439 440 441

For the intracellular ATP assay, renal homogenate was mixed with TCA (6.5%, w/v) and left on ice for 10 min. The TCA extract was removed and used immediately for analysis of ATP as described by Kalbheim and Koch [43]. 2.20. Histological studies Kidneys from the normal and experimental mice were xed in 10% buffered formalin and were processed for parafn sectioning. Sections of about 5 m thickness were stained with hematoxylin and eosin to evaluate under light microscope. 2.21. Statistical analysis All the values are expressed as means SD (n = 6). Significant differences between the groups were determined with SPSS 10.0 software (SPSS Inc., Chicago, IL, USA) for Windows using one-way analysis of variance (ANOVA) and the group means were compared by StudentNewmanKeuls post hoc tests. A difference was considered signicant at the p < 0.05 level.

442

443 444 445 446 447

448

449 450 451 452 453 454 455

To begin with the evaluation of the antioxidant nature of AEPD, we looked for an effective radical scavenging cell free assay. So, we determined its radical scavenging power using the DPPH radical. The experimental results on the radical scavenging effect of AEPD have been shown in Fig. 2A. The plot shows that with increase in concentration of AEPD the color of the DPPH radical vanishes rapidly at 517 nm. It has been observed that AEPD showed maximum inhibition when it was incubated at a concentration of 20 mg/ml with DPPH solution. Hence, it can be anticipated that the active principle(s) present in AEPD possesses potent free radical scavenging activity. Hydroxyl (OH) and superoxide (O2 ) radicals scavenging activity of AEPD has been investigated in cell free system and Fig. 2B represents those results. It has been observed that at a dose of 20 mg/ml, AEPD can effectively scavenge OH and O2 radicals. 3.3. Dose and time dependent protective action of AEPD In the present study, we used CAT assay to determine the optimum dose and time necessary for AEPD to protect mouse kidney against CCl4 -induced renal oxidative damages. The experimental results have been represented in Fig. 4A and B. CCl4 exposure decreased the CAT activity. AEPD administration increased the CAT activity linearly up to a dose of 200 mg/kg body weight, when applied for 7 days prior to CCl4 intoxication. This dose and time of AEPD treatment were used for the subsequent experiments. 3.4. Kidneybody weight ratio CCl4 intoxication usually reduces the weight of different organs in pathophysiological situations. We, therefore, checked whether the kidney weight of the experimental animals was affected by CCl4 exposure and if that happened whether AEPD could protect it. Table 2A shows that CCl4 toxicity lowers kidney weight to body weight ratio and that could be prevented by the treatment with AEPD prior to toxin exposure. Post treatment with AEPD also showed similar result as the pre treated groups. It is evident from the data

469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485

486

487 488 489 490 491 492 493 494 495

456

3. Results 3.1. Phytochemical screening of AEPD for active components Qualitative analysis of AEPD (Table 1A) revealed the presence of phenolics, avonoids, and saponins as the major constituents in the extract. Quantitative estimation of the chemical constituents in the studied medicinal plant is summarized in Table 1B. HPLC prole of AEPD extract (Fig. 1) also shows the presence phenolics (peak I), avonoids (peak
Table 1A Qualitative extract analysis of AEPD. Serial no. 1 2 3 Component Phenolics Flavonoids Saponins Abundance + + +

457 458

459 460 461 462 463 464

496

497 498 499 500 501 502 503 504 505

+ sign indicates the presence of the compounds in the AEPD.

Please cite this article in press as: P.B. Pal, et al., Traditional extract of Pithecellobium dulce fruits protects mice against CCl4 induced renal oxidative impairments and necrotic cell death, Pathophysiology (2012), doi:10.1016/j.pathophys.2012.02.001

PATPHY 726 114

ARTICLE IN PRESS
P.B. Pal et al. / Pathophysiology xxx (2012) xxxxxx

Fig. 4. (A) Dose dependent effect of AEPD on CAT activity against CCl4 induced renal-toxicity. Cont: CAT activity in normal mice, CCl4 : CAT activity in CCl4 treated mice, AEPD-25, AEPD-50, AEPD-75, AEPD-100, AEPD-150, AEPD-200, AEPD-250, AEPD-300: CAT activity in AEPD treated mice for 7 days at a dose of 25, 50, 75, 100, 150, 200, 250 and 300 mg/kg body weight prior to CCl4 intoxication. Data are mean SD, for 6 animals per group and were analyzed by one-way ANOVA, with StudentNewmanKeuls post hoc tests. Differences were attributed at p < 0.05, and homogeneous subgroups share common superscripted letters. (B) Time dependent effect of AEPD on CAT activity against CCl4 induced renal-toxicity. Cont: CAT activity in normal mice, CCl4 : CAT activity in CCl4 treated mice, AEPD-1, AEPD-3, AEPD-5, AEPD-7, AEPD-10, AEPD-12 and AEPD-14: CAT activity in AEPD treated mice at a dose of 200 mg/kg body weight prior to CCl4 intoxication for 1, 3, 5, 7, 10, 12 and 14 days respectively. Data are mean SD, for 6 animals per group and were analyzed by one-way ANOVA, with StudentNewmanKeuls post hoc tests. Differences were attributed at p < 0.05, and homogeneous subgroups share common superscripted letters.
506 507

that CCl4 -induced renal pathophysiology could be protected by AEPD. 3.5. Effect of AEPD on BUN and creatinine levels In our study we found that CCl4 exposure did not alter the levels of serum markers; BUN and creatinine related to renal dysfunction (Table 2A) and treatment with AEPD also did not show any alteration. 3.6. Lipid peroxidation and protein carbonylation inhibition In oxidative stress related organ pathophysiology, lipid peroxidation and protein carbonylation are considered to be

508

509 510 511 512

the two important parameters. In our study, lipid peroxidation has been measured by estimating the concentration of MDA (lipid peroxidation end product). Table 2B represents the changes in MDA and protein carbonylation in the kidney tissue of the normal and experimental animals. CCl4 intoxication increased the levels of MDA and protein carbonylation. However, both pre and post treatment with AEPD decreased the levels of lipid peroxidation as well as protein carbonylation suggesting the anti-oxidant nature of AEPD in CCl4 -induced renal dysfunction. 3.7. Protective role of AEPD against CCl4 induced ROS production ROS plays a major role in CCl4 induced kidney dysfunction and cellular death of kidney. In order to assess

517 518 519 520 521 522 523 524 525 526 527 528

513 514

515 516

529 530

Table 2A Kidneybody weight ratios and the activities of the serum markers related to renal dysfunction in male mice. Animal groups Ratio of the kidney weight to the body weight (%) 1.11 1.05 0.62 0.95 0.99 0.94 0.65 0.04a 0.05a 0.06b 0.03a 0.04a 0.05a 0.06b Activities of the serum markers BUNa Normal AEPD CCl4 AEPD + CCl4 CCl4 + AEPD VitC + CCl4 Recovery 60.64 55.47 70.98 62.45 65.57 64.43 69.95 3.24a 2.62a 3.27a 4.52a 5.52a 4.82a 10.57a Creatininea 0.56 0.58 0.62 0.56 0.60 0.57 0.63 0.02a 0.02a 0.03a 0.02a 0.04a 0.06a 0.04a

Data are means SD, for 6 mice per group and were analyzed by one-way ANOVA, with StudentNewmanKeuls post hoc tests. Differences were attributed at p < 0.05, and homogeneous subgroups share common letters (a, b). a mg/dL.

Please cite this article in press as: P.B. Pal, et al., Traditional extract of Pithecellobium dulce fruits protects mice against CCl4 induced renal oxidative impairments and necrotic cell death, Pathophysiology (2012), doi:10.1016/j.pathophys.2012.02.001

PATPHY 726 114

ARTICLE IN PRESS
P.B. Pal et al. / Pathophysiology xxx (2012) xxxxxx 9

Table 2B Levels of the parameters as the index of lipid peroxidation (MDA) and protein carbonylation (PC) in the kidney tissue of the control and experimental mice. Animal groups Normal AEPD CCl4 AEPD + CCl4 CCl4 + AEPD VitC + CCl4 Recovery MDAa 4.72 5.23 10.42 7.41 7.85 7.13 9.34 0.25a 0.27a 0.52b 0.36c 0.41c 0.34c 0.45b PCa 11.5 10.92 27.89 13.35 14.01 12.89 25.54 0.57a 0.53a 1.43b 0.64a 0.71a 0.66a 1.28b

Data are means SD, for 6 animals per group and were analyzed by oneway ANOVA, with StudentNewmanKeuls post hoc tests. Differences were attributed at p < 0.05, and homogeneous subgroups share common letters (a, b). a nmol/mg protein.

531 532 533 534 535 536 537 538 539 540 541

the changes in the production of intracellular ROS under oxidative stress, kidneys were isolated from the experimental animals and studied the ROS production assay using DCFDA by uorescence spectrophotometer. Fig. 5 shows the effect of CCl4 on the intracellular ROS production and its reversal by the treatment with AEPD. Here, we observed that CCl4 administration caused increased production of intracellular ROS. Treatment with AEPD both prior and post to toxin administration decreased the intracellular ROS level compared to toxin control suggesting the intracellular ROS scavenging nature of AEPD.

Fig. 6. DNA fragmentation on agarose/ethydium bromide gel. DNA isolated from experimental kidney tissues was loaded onto 1% (w/v) agarose gels. Lane 1: Marker (1 kb DNA ladder); Lane 2: DNA isolated from normal kidney tissue; Lane 3: DNA isolated from AEPD treated kidney samples; Lane 4: DNA isolated from CCl4 intoxicated kidney; Lane 5: DNA isolated from AEPD pre-treated kidney samples; Lane 6: DNA isolated from kidney sample of the AEPD post-treated mouse and Lane 7: DNA isolated from kidney tissue of the mouse of recovery group.

3.8. Antioxidant enzyme activities Antioxidant enzymes are considered to be the rst line of cellular defence that prevents cellular ingredients from oxidative damage. Among them SOD and CAT act as important enzymes in the elimination of ROS. In order to remove excess free radicals from the system, GST utilizes GSH during their course of reactions. Due to decrease in GSH content for toxicity simultaneously decreased the activities of GST with a concomitant decrease in the activity of GSH regenerating enzyme, GR. We, therefore, determined the activities of these enzymes and presented the data in Table 2C. A signicant reduction in the activities of all antioxidant enzymes have been observed in the kidney tissue of the CCl4 intoxicated experimental animals. Results suggest that both pre as well as post treatment with AEPD could protect the rst line of renal dysfunction in oxidative damage induced by CCl4 .

542

543 544 545 546 547 548 549 550 551 552 553 554 555 556 557

Fig. 5. The intracellular ROS production was detected by DCF-DA method. Cont: ROS level in normal animals, AEPD: ROS level in the animals treated with AEPD only; CCl4 : ROS level in the CCl4 intoxicated animals; AEPD + CCl4 : ROS level in the animals treated with AEPD prior to CCl4 intoxication; CCl4 + AEPD: ROS level in the animals treated with AEPD post to CCl4 intoxication and Recovery: ROS level in the animals of recovery group. Data are means SD, for 6 animals per group and were analyzed by one-way ANOVA, with StudentNewmanKeuls post hoc tests. Differences were attributed at p < 0.05, and homogeneous subgroups share common superscripted letters.

3.9. Effect on cellular metabolites Table 2D depicts the effects of AEPD and CCl4 on the levels of the cellular metabolites in the kidney tissue of the experimental animals. CCl4 decreased the levels of total thiols as well as GSH and increased the level of GSSG. Administration of AEPD could ameliorate the alteration in the thiol parameters in the CCl4 intoxicated animals.

558

559 560 561 562 563 564

Please cite this article in press as: P.B. Pal, et al., Traditional extract of Pithecellobium dulce fruits protects mice against CCl4 induced renal oxidative impairments and necrotic cell death, Pathophysiology (2012), doi:10.1016/j.pathophys.2012.02.001

PATPHY 726 114

ARTICLE IN PRESS
P.B. Pal et al. / Pathophysiology xxx (2012) xxxxxx

10

Table 2C Effect of CCl4 and AEPD on the activities of the antioxidant enzymes in kidneys of the normal and experimental mice. Animal groups Normal AEPD CCl4 AEPD + CCl4 CCl4 + AEPD VitC + CCl4 Recovery SODa 71.17 68.79 25.15 63.12 55.23 65.71 26.07 3.45a 2.81a 2.62b 3.08a 3.62a 3.05a 2.67b CATb 55.12 53.57 20.41 50.45 45.17 49.73 22.96 2.44a 2.12a 0.85b 1.92a 1.75a 2.15a 1.01b GSTb 3.47 2.12 0.85 1.95 1.75 2.05 0.91 0.21a 0.17a 0.09b 0.15a 0.12a 0.14a 0.11b GRc 17.45 16.35 5.25 14.33 12.18 15.15 6.89 0.95a 0.81a 0.35b 0.77a 0.69a 0.81a 0.42b

Data are means SD, for 6 mice per group and were analyzed by one-way ANOVA, with StudentNewmanKeuls post hoc tests. Differences were attributed at p < 0.05, and homogeneous subgroups share common letters (a, b). a Unit/mg protein. b mol/min/mg protein. c nmol/min/mg protein.
565

3.10. Effect on DNA damage Fig. 6 represents the results carried out to evaluate whether AEPD protects CCl4 induced renal DNA damage. DNA fragmentation was examined by agarose gel electrophoresis. A smear on agarose gel was observed in CCl4 treated group, indicating random DNA fragmentation which is a hallmark of cellular necrosis. AEPD treatment was found to be effective in preventing the toxin induced smear formation suggesting its protective power for the prevention of kidney cells from CCl4 -induced DNA damage and necrotic death.

566 567 568 569 570 571 572 573 574

575 576

3.11. Effect on mitochondrial membrane potential and ATP level Oxidative stress-induced cell death is directly related to mitochondrial dysfunction. Disruption of mitochondrial membrane potential and loss of ATP production cause cell death. Therefore, we assessed the mitochondrial membrane potential (Fig. 7) and the ATP level (Fig. 8). It has been observed that CCl4 exposure reduced the mitochondrial membrane potential as well as intracellular ATP level. AEPD administration however found to be effective in preventing this event.

577 578 579 580 581 582 583 584 585

Table 2D Status of the thiol based antioxidants in the kidney tissue of the CCl4 and AEPD treated mice. Animal groups Normal AEPD CCl4 AEPD + CCl4 CCl4 + AEPD VitC + CCl4 Recovery GSHa 7.89 7.87 2.88 6.63 6.55 6.67 2.89 0.38a 0.39a 0.142b 0.33a 0.31a 0.33a 0.143b GSSGa 0.27 0.26 0.87 0.59 0.51 0.57 0.86 0.014a 0.016a 0.042b 0.028a 0.024a 0.027a 0.042b Total thiolsa 125.55 115.74 59.97 104.21 96.33 103.98 59.91 6.26a 5.77a 2.99b 5.21a 4.87a 5.19a 2.98b Fig. 7. Mitochondrial membrane potentials ( ) were measured in the kidney tissue of normal and experimental animals. Upper panel represents the histogram analysis and lower panel represents the corresponding levels as percentage over control. Cont: in normal animals, AEPD: in the animals treated with AEPD only; CCl4 : in the CCl4 intoxicated animals; in the animals treated with AEPD prior to CCl4 intoxiAEPD + CCl4 : in the animals treated with AEPD post to CCl4 cation; CCl4 + AEPD: intoxication and Recovery: in the animals of recovery group. Data are mean SD, for 6 animals per group and were analyzed by one-way ANOVA, with StudentNewmanKeuls post hoc tests. Differences were attributed at p < 0.05, and homogeneous subgroups share common superscripted letters.

Data are means SD, for 6 mice per group and were analyzed by oneway ANOVA, with StudentNewmanKeuls post hoc tests. Differences were attributed at p < 0.05, and homogeneous subgroups share common letters (a, b). a nmol/mg protein.

Please cite this article in press as: P.B. Pal, et al., Traditional extract of Pithecellobium dulce fruits protects mice against CCl4 induced renal oxidative impairments and necrotic cell death, Pathophysiology (2012), doi:10.1016/j.pathophys.2012.02.001

PATPHY 726 114

ARTICLE IN PRESS
P.B. Pal et al. / Pathophysiology xxx (2012) xxxxxx 11
589 590 591 592 593 594 595

in Fig. 9. CCl4 exposed animals showed extensive tubular damage by swollen and necrotic epithelial cells. Treatment with AEPD both prior to and after the toxin exposure showed a considerable improvement in kidney morphology. Results of the histological assessment support the outcome of the earlier studies by exhibiting CCl4 -induced necrosis in the kidney tissue and its protection by AEPD.

4. Discussion We observed that CCl4 administration caused murine renal dysfunction and cell death by disturbing the status of antioxidant enzyme activities, lipid peroxidation, protein carbonylation followed by the reduction in mitochondrial membrane potential, loss in, unprogrammed cell death, necrosis. This impairment of renal function and cell death could, however, be protected by the treatment of AEPD prior and post to CCl4 intoxication. As the rst step of dening a substance to be antioxidant, we usually determine its radical scavenging activity. DPPH radical scavenging assay is considered to be a convenient tool in cell free system for this purpose. We found that incubation of AEPD with increased concentrations and DPPH radical at a xed concentration diminished the absorbance at 517 nm (Fig. 2A). This experiment shows the ability of the extract to scavenge DPPH radicals. In addition, the hydroxyl as well as superoxide radicals scavenging activity of AEPD were also investigated in cell free systems and found that it could effectively scavenge these radicals (Fig. 2B). We

596

597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615

Fig. 8. ATP levels were measured in the kidney tissue of normal and experimental animals. Cont: level in normal animals, AEPD: level in the animals treated with AEPD only; CCl4 : level in the CCl4 intoxicated animals; AEPD + CCl4 : level in the animals treated with AEPD prior to CCl4 intoxication; CCl4 + AEPD: level in the animals treated with AEPD post to CCl4 intoxication and Recovery: level in the animals of recovery group. Data are mean SD, for 6 animals per group and were analyzed by one-way ANOVA, with StudentNewmanKeuls post hoc tests. Differences were attributed at p < 0.05, and homogeneous subgroups share common superscripted letters.
586

3.12. Histological assessment Histological assessments of different kidney segments of the normal and experimental animals have been presented

587 588

Fig. 9. Haematoxylin and eosin stained kidney section of experimental mice. Cont: kidney section from normal mice (100); AEPD: kidney section of the mice treated with AEPD only (100); CCl4 : mice administered with CCl4 (100); AEPD + CCl4 : kidney section from the AEPD pre treated mice (100); CCl4 + AEPD: kidney section from the AEPD post treated mice (100) and Recovery: kidney section from the mice of recovery group (100).

Please cite this article in press as: P.B. Pal, et al., Traditional extract of Pithecellobium dulce fruits protects mice against CCl4 induced renal oxidative impairments and necrotic cell death, Pathophysiology (2012), doi:10.1016/j.pathophys.2012.02.001

PATPHY 726 114

ARTICLE IN PRESS
P.B. Pal et al. / Pathophysiology xxx (2012) xxxxxx

12

Fig. 10. Schematic diagram of the antioxidative role AEPD and its protective role against CCl4 induced renal pathophysiology.

616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642

then determined its effect on intracellular ROS production by using 2,7-dichlorouorescein diacetate (DCFDA) as a probe. Results showed that CCl4 intoxication increased the rate of DCF formation, an indicator of intracellular ROS production, in the renal tissue of the experimental animals. Treatment with AEPD, both pre and post to the toxin administration, could, however, prevent the increased production of ROS and maintain the intracellular antioxidant power close to normal. Results of these studies clearly established AEPD as a potent radical scavenger. It is, therefore, likely to believe that AEPD might alter the toxic effects of any toxicant-induced excessive free radicals by quenching those harmful species. It has been well established that exposure to various organic compounds including a number of environmental pollutants and drugs can cause cellular damages through metabolic activation of those compounds to highly reactive substances such as reactive oxygen species (ROS). CCl4 is one of such extensively studied environmental toxicant. Reports from our laboratory and other investigators have already established that in addition to hepatic problems, the industrial solvent CCl4 also causes disorders in kidneys, lungs, testis and brain as well as in blood by generating free radicals [44]. The reactive metabolite trichloromethyl radical (CCl3 ) has been formed from the metabolic conversion of CCl4 by cytochrome P-450 [45]. As O2 tension rises, a greater fraction of CCl3 present in the system reacts very rapidly with O2 and many orders of magnitude more reactive

free radical, CCl3 OO is generated from CCl3 [46]. Although various enzymatic and non-enzymatic systems in the cell try to cope up with the ROS and other free radicals, a condition of oxidative stress establishes when the defense capacities inside the cells against ROS become insufcient [47]. In the present study, it has been observed that CCl4 caused a signicant decrease in antioxidant enzyme activities, depleted the GSH content and enhanced lipid peroxidation as well as protein carbonylation in kidney tissue although levels of the markers related to renal damages practically remained unaltered. The exact reason for this in CCl4 induced renal pathophysiology is not clearly known. One possible explanation is that, although the time of CCl4 exposure to the animals was sufcient to induce renal oxidative stress but not enough to cause sufcient damage for its dysfunction. Similar results have been reported by other investigators in CCl4 induced renal damage in rats [48]. AEPD, on the other hand, was found to be highly effective in this organ pathophysiology as its administration both prior and post to CCl4 intoxication could not only prevent the CCl4 induced increased levels of lipid peroxidation and protein carbonylation, but also protected the antioxidant machineries of the kidney as revealed from the enhanced levels of SOD, CAT GST and GR activities, increased level of GSH. Oxidative stress induced by oxygen-derived species can produce a multiplicity of modications in DNA including base and sugar lesions, strand breaks, DNA-protein

643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669

Please cite this article in press as: P.B. Pal, et al., Traditional extract of Pithecellobium dulce fruits protects mice against CCl4 induced renal oxidative impairments and necrotic cell death, Pathophysiology (2012), doi:10.1016/j.pathophys.2012.02.001

PATPHY 726 114

ARTICLE IN PRESS
P.B. Pal et al. / Pathophysiology xxx (2012) xxxxxx 13
723

670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720

cross-links and base-free sites. If left un-repaired, oxidative DNA damage can lead to detrimental biological consequences in organisms, including cell death, mutations and transformation of cells to malignant cells. So, in addition to excessive oxidative stress, DNA fragmentation acts as a mediator of cell death [49]. In the present study it has been observed that CCl4 intoxication caused random fragmentation of genomic DNA leading to the formation of a DNA smear on agarose gel (Fig. 6), suggesting that CCl4 induced renal cell damage and death occur via necrotic pathway. CCl4 induced increased production of ROS caused reduction in mitochondrial membrane potential and loss in intracellular ATP content. AEPD treatment could, however, prevent CCl4 -induced over production of ROS, degree of DNA fragmentation, reduction in mitochondrial membrane potential (Fig. 7) and loss in ATP content (Fig. 8). These results clearly showed the antioxidative and antinecrotic nature of AEPD in CCl4 -induced renal pathophysiology and dysfunction. The fruits of P. dulce contain about, 2.5% protein, 0.4% fat, 18% carbohydrate, 77.8% water and 1.3% ber as reported by the Indian Council of Scientic and Industrial Research (CSIR) [50]. The report also mentioned that the fruits are enriched with vital vitamins such as thiamine, riboavin, niacin and ascorbic acid and contain some essential minerals such as Ca, P, Fe, Na and K. Few essential amino acids like valine, lysine, phenylalanine, and tryptophan have also been found in this fruit. Alcoholic extraction yielded saponin, sterol glucoside, avone and lecithin. In the present study, we also veried the presence of phenolics, avonoids and saponins in the plant extract. The observed effects of the extract could be related to chemically dened compounds. Flavonoids show their antioxidative action through scavenging or chelating process. Phenolic content is also important because of the presence of hydroxyl groups possessing scavenging ability. It can, therefore, be speculated that the observed antioxidant effects of AEPD could be due to the presence of avonoids and phenolic contents. Histopathological examination clearly demonstrated that CCl4 administration caused a signicant abnormality in the renal morphology showing marked tubular damages. Complete loss of brush borders, extensive tubular casts and debris as well as tubular dilatations was also observed in the renal tissue of the experimental animal. Treatment of AEPD prevented any such CCl4 -induced alterations and kept the kidney histologically close to normal (Fig. 9). In conclusion, aqueous extract of the fruits of P. dulce protects kidneys against CCl4 induced oxidative damages in mice model and could be used as an effective protector in studies pathophysiology (Fig. 10). Future works would characterize the responsible active principle(s) present in it and elucidate its possible mode of action.

Acknowledgement The authors are grateful to Mr. Prasanta Pal for excellent technical assistance for the study.

724 725

References
[1] J.L. Raucy, J.C. Kraner, J.M. Lasker, Bioactivation of halogenated hydrocarbons by cytochrome P4502E1, Crit. Rev. Toxicol. 23 (1993) 120. [2] A.J. Perez, M. Courel, J. Sobrado, L. Gonzalez, Acute renal failure after topical application of carbon tetrachloride, Lancet 1 (1987) 515516. [3] M. Ogeturk, I. Kus, A. Kavakli, J. Oner, A. Kukner, M. Sarsilmaz, Reduction of carbon tetrachloride-induced nephropathy by melatonin administration, Cell Biochem. Funct. 23 (2005) 8592. [4] D.N. Churchill, A. Finn, M. Gault, Association between hydrocharbon exposure and glomerulonephritis, an araisal of the evidence, Nephron 33 (1983) 169172. [5] P.K. Singha, S. Roy, S. Dey, Protective activity of andrographolide and arabinogalactan proteins from Andrographis paniculata Nees. against ethanol-induced toxicity in mice, J. Ethnopharmacol. 111 (2007) 1321. [6] B. Kligler, C. Ulbricht, E. Basch, C.D. Kirkwood, Andrographis paniculata for the treatment of upper respiratory infection: a systematic review by the natural standard research collaboration, Explore (NY) 2 (2006) 2529. [7] R. Bhattacharjee, P.C. Sil, The protein fraction of Phyllanthus niruri plays a protective role against acetaminophen induced hepatic disorder via its antioxidant properties, Phytother. Res. 20 (2006) 595601. [8] R. Bhattacharjee, P.C. Sil, Protein isolate from the herb, Phyllanthus niruri L. (Euphorbiaceae), plays hepatoprotective role against carbon tetrachloride induced liver damage via its antioxidant properties, Food Chem. Toxicol. 45 (2007) 817826. [9] M. Chatterjee, K. Sarkar, P.C. Sil, Herbal (Phyllanthus niruri) protein isolate protects liver from nimesulide induced oxidative stress, Pathophysiology 13 (2006) 95102. [10] M. Chatterjee, P.C. Sil, Hepatoprotective effect of aqueous extract of Phyllanthus niruri on nimesulide-induced oxidative stress in vivo, Indian J. Biochem. Biophys. 43 (2006) 299305. [11] M.K. Sarkar, M. Kinter, B. Mazumder, P.C. Sil, Purication and characterization of a novel antioxidant protein molecule from Phyllanthus niruri, Food Chem. 114 (2009) 14051412. [12] M.K. Sarkar, P.C. Sil, Hepatocytes are protected by herb Phyllanthus niruri protein isolate against thioacetamide toxicity, Pathophysiology 14 (2007) 113120. [13] K. Sarkar, P.C. Sil, Attenuation of acetaminophen-induced hepatotoxicity in vivo and in vitro by a 43-kD protein isolated from the herb Cajanus indicus L., Toxicol. Mech. Methods 17 (2007) 305315. [14] K. Sarkar, A. Ghosh, M. Kinter, B. Mazumder, P.C. Sil, Purication and characterization of a 43 kD hepatoprotective protein from the herb Cajanus indicus L., Protein J. 25 (2006) 411421. [15] K. Sarkar, P.C. Sil, A 43 kDa protein from the herb Cajanus indicus L. protects thioacetamide induced cytotoxicity in hepatocytes, Toxicol. In Vitro 20 (2006) 634640. [16] A. Ghosh, P.C. Sil, Protection of acetaminophen induced mitochondrial dysfunctions and hepatic necrosis via Akt-NF-kappaB pathway: role of a novel plant protein, Chem. Biol. Interact. 177 (2009) 96106. [17] A. Ghosh, P.C. Sil, Anti-oxidative effect of a protein from Cajanus indicus L. against acetaminophen-induced hepato-nephro toxicity, J. Biochem. Mol. Biol. 39 (2007) 10391049. [18] A. Ghosh, K. Sarkar, P.C. Sil, Protective effect of a 43 kD protein from the leaves of the herb, Cajanus indicus L. on chloroform induced hepatic-disorder, J. Biochem. Mol. Biol. 39 (2006) 197207.

726

727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783

721

Conict of interest statement The authors have declared that no conict of interest exists.

722

Please cite this article in press as: P.B. Pal, et al., Traditional extract of Pithecellobium dulce fruits protects mice against CCl4 induced renal oxidative impairments and necrotic cell death, Pathophysiology (2012), doi:10.1016/j.pathophys.2012.02.001

PATPHY 726 114

ARTICLE IN PRESS
P.B. Pal et al. / Pathophysiology xxx (2012) xxxxxx [35] K. Uchida, E.R. Stadtman, Covalent attachment of 4-hydroxynonenal to glyceraldehydes-3-phosphate dehydrogenase, J. Biol. Chem. 268 (1993) 63886393. [36] M. Sinha, P. Manna, P.C. Sil, Taurine, a conditionally essential amino acid, ameliorates arsenic-induced cytotoxicity in murine hepatocytes, Toxicol. In Vitro 21 (2007) 14191428. [37] G.L. Ellman, Tissue sulphydryl group, Arch. Biochem. Biophys. 82 (1959) 7077. [38] B.N. Finck, X. Han, M. Courtois, F. Aimond, J.M. Nerbonne, A. Kovacs, R.W. Gross, D.P. Kelly, A critical role for PPARalpha-mediated lipotoxicity in the pathogenesis of diabetic cardiomyopathy: modulation by dietary fat content, Proc. Natl. Acad. Sci. 100 (2003) 12261231. [39] J. Sedlak, R.H. Lindsay, Estimation of total, protein-bound, and nonprotein sulfhydryl groups in tissue with Ellmans reagent, Anal. Biochem. 24/25 (1968) 192205. [40] C.P. LeBel, S.C. Bondy, Sensitive and rapid quantitation of oxygen reactive species formation in rat synaptosomes, Neurochem. Int. 17 (1990) 435440. [41] K.S. Sellins, J.J. Cohen, Gene induction by gamma-irradiation leads to DNA fragmentation in lymphocytes, J. Immunol. 139 (1987) 31993206. [42] I. Korichneva, J. Waka, U. Hammerling, Regulation of the cardiac mitochondrial membrane potential by retinoids, J. Pharmacol. Exp. Ther. 305 (2003) 426433. [43] D. Kalbheim, H. Koch, Methodologic studies of the quantitative microdetermination of ATP in biological materials with the rey enzyme system, Zeitschrift. fr klinische. Chemie. und klinische. Biochemie. 5 (1967) 299394. [44] F.F. Ahmad, D.L. Cowan, A.Y. Sun, Detection of free radical formation in various tissues after acute carbon tetrachloride administration in gerbil, Life Sci. 41 (1987) 24692475. [45] T. Noguchi, K.L. Fong, E.K. Lai, S.S. Alexander, M.M. King, L. Olson, J.L. Poyer, P.B. Mccay, Specicity of aphenobarbitalinduced cytochrome P450 for metabolism of carbontetrachloride to the trichloromethyl radical, Biochem. Pharmacol. 31 (1982) 615624. [46] J.E. Packer, T.F. Slater, R.L. Willson, Reactions of the carbon tetrachlonde-related peroxy free radical with aminoacids: pulse radiolysis evidence, Life Sci. 23 (1978) 26172620. [47] B. Halliwell, J.M.C. Gutteridge, Free Radicals in Biology and Medicine, Oxford University Press (2000) 148149. [48] N. Tirkey, S. Pilkhwal, A. Kuhad, K. Chopra, Hesperidin, a citrus bioavonoid, decreases the oxidative stress produced by carbon tetrachloride in rat liver and kidney, BMC Pharmacol. 5 (2005) 2. [49] A.S Burke, K. Redeker, R.C. Kurten, L.P. James, J.A. Hinson, Mechanisms of chloroform-induced hepatotoxicity: oxidative stress and mitochondrial permeability transition in freshly isolated mouse hepatocytes, J. Toxicol. Env. Heal. A 70 (2007) 19361945. [50] C.S.I.R. (Council of Scientic and Industrial Research), The Wealth of India. 11 vols, 19481976, New Delhi.
833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882

14
784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832

[19] H. Farghali, L. Kamenikova, S. Hynie, E. Kmonickova, Silymarin effects on intracellular calcium and cytotoxicity: a study in perfused rat hepatocytes after oxidative stress injury, Pharmacol. Res. 41 (2000) 231237. [20] J. Ghosh, J. Das, P. Manna, P.C. Sil, Arjunolic acid, a triterpenoid saponin, prevents acetaminophen (APAP) induced liver and hepatocytes injury via the inhibition of APAP bioactivation and JNK-mediated mitochondrial protection, Free Radic. Biol. Med. 48 (2010) 535553. [21] P. Manna, J. Ghosh, J. Das, P.C. Sil, Contribution of type 1 diabetes to rat liver dysfunction and cellular damage via activation of NOS, PARP, I B /NF- B, MAPKs and mitochondria dependent pathways: prophylactic role of arjunolic acid, Free Radic. Biol. Med. 48 (2010) 14651484. [22] P. Manna, M. Sinha, P. Pal, P.C. Sil, Arjunolic acid, a triterpenoid saponin, ameliorates arsenic-induced cyto-toxicity in hepatocytes, Chem. Biol. Interact. 170 (2007) 187200. [23] P. Manna, M. Sinha, P.C. Sil, Aqueous extract of Terminalia arjuna prevents carbon tetrachloride induced hepatic and renal disorders, BMC Compl. Altern. Med. 6 (2006) 33. [24] C. Chitravadivu, S. Manian, K. Kalaichelvi, Qualitative analysis of selected medicinal plants, Tamilnadu, India, Middle-East J. Sci. Res. 4 (2009) 144146. [25] H.O. Edeoga, D.E. Okwu, B.O. Baebie, Phytochemical constituents of some Nigerian Medicinal plants, African Biotech. 4 (2005) 685688. [26] L.D. Kapoor, A. Singh, S.L. Kapoor, S.N. Shrivastava, Survey of Indian medicinal plants for saponins, alkaloids and avonoids, Lloydia 32 (1969) 297302. [27] S. McDonald, P.D. Prenzler, M. Autolovich, K. Robards, Phenolic content and anti-oxidant activity of olive extract, Food Chem. 73 (2001) 7384. [28] B.A. Boham, R. Kocipal-Abyazan, Flavonoids and condensed tannins from leaves of Hawaiian vaccinium vaticulatum and V. calycinum, Pac. Sci. 48 (1974) 458463. [29] B.O. Obadoni, P.O. Ochuko, Phytochemical studies and comparative efcacy of crude extracts of some homeostatic plants in Edo and Delta States of Nigeria, Global J. Pure Appl. Sci. 8 (2001) 203208. [30] M.S. Blois, Antioxidant determination by use of a stable free radical, Nature 29 (1957) 11991200. [31] T. Nash, The colorimetric estimation of formaldehyde by means of the Hantzsch reaction, J. Biochem. 55 (1953) 416421. [32] P. Siddhuraju, K. Becker, Antioxidant properties of various solvent extracts of total phenolic constituents from three different agroclimatic origins of drumstick tree (Moringa oleifera Lam.) leaves, J. Agric. Food Chem. 51 (2003) 21442155. [33] M.M. Bradford, A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding, Anal. Biochem. 72 (1976) 248254. [34] H. Esterbauer, K.H. Cheeseman, Determination of aldehydic lipid peroxidation products: malonaldehyde and 4-hydroxynonenal, Methods Enzymol. 186 (1990) 407421.

Please cite this article in press as: P.B. Pal, et al., Traditional extract of Pithecellobium dulce fruits protects mice against CCl4 induced renal oxidative impairments and necrotic cell death, Pathophysiology (2012), doi:10.1016/j.pathophys.2012.02.001

S-ar putea să vă placă și