Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Int Immunopharmacol ; 65: 408-412, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30380516

ABSTRACT

Mastitis is the inflammation of the mammary glands caused by bacteria. It causes severe economic loss to dairy industry. Curcumin, a polyphenol obtained from turmeric, has considerable anti-inflammatory effect. Since it is rapidly eliminated from the body, its oral bioavailability is low. However, nanoformulation of curcumin significantly enhances its therapeutic efficiency by improving its oral bioavailability. We evaluated whether nanocurcumin could be more effective than normal curcumin against bovine Staphylococcus aureus mastitis in mouse model. Curcumin-loaded PLGA nanoparticles (CUR-NP) were prepared by solid-in-oil-in-water emulsion method. The mouse model of mastitis was induced by inoculation of a field strain of S. aureus (bovine mastitis isolate) on the 9th day of parturition through the duct of the mammary gland. CUR-NP and curcumin were given orally for 7 days (day 2 to day 8 of parturition) prior to S. aureus inoculation. We determined the levels of inflammatory cytokines and the mRNA expression of NF­κB. S. aureus infection increased the levels of tumor necrosis factor­α, interleukin­1ß and myeloperoxidase in mammary tissues and C-reactive protein in serum. Both CUR-NP and curcumin significantly attenuated the levels of these cytokines. However, comparatively, the ameliorative efficiency of CUR-NP was better than normal curcumin. S. aureus infection-induced NF­κB mRNA expression was significantly reduced to the healthy control level by CUR-NP. Our study demonstrates that the nanoformulation of curcumin can reduce pro-inflammatory mediators in S. aureus-infected mammary tissues by improving NF­κB signaling. Besides, compared to normal curcumin, this nanoformulation appears to be a better alternative against murine mastitis.


Subject(s)
Curcumin/therapeutic use , Mammary Glands, Animal/immunology , Nanoparticles/therapeutic use , Staphylococcal Infections/immunology , Staphylococcus aureus/physiology , Animals , C-Reactive Protein/metabolism , Cattle , Curcumin/chemistry , Disease Models, Animal , Female , Interleukin-1beta/metabolism , Mammary Glands, Animal/microbiology , Mammary Glands, Animal/pathology , Mastitis , Mice , NF-kappa B/metabolism , Nanoparticles/chemistry , Peroxidase/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
2.
Toxicol Ind Health ; 32(3): 410-21, 2016 Mar.
Article in English | MEDLINE | ID: mdl-24105067

ABSTRACT

Arsenic exposure through drinking water causes oxidative stress and tissue damage in the kidney and brain. Curcumin (CUR) is a good antioxidant with limited clinical application because of its hydrophobic nature and limited bioavailability, which can be overcome by the encapsulation of CUR with nanoparticles (NPs). The present study investigates the therapeutic efficacy of free CUR and NP-encapsulated CUR (CUR-NP) against sodium arsenite-induced renal and neuronal oxidative damage in rat. The CUR-NP prepared by emulsion technique and particle size ranged between 120 and 140 nm, with the mean particle size being 130.8 nm. Rats were divided into five groups (groups 1-5) with six animals in each group. Group 1 served as control. Group 2 rats were exposed to sodium arsenite (25 ppm) daily through drinking water for 42 days. Groups 3, 4, and 5 were treated with arsenic as in Group 2; however, these animals were also administered with empty NPs, CUR (100 mg/kg body weight), and CUR-NP (100 mg/kg), respectively, by oral gavage during the last 14 days of arsenic exposure. Arsenic exposure significantly increased serum urea nitrogen and creatinine levels. Arsenic increased lipid peroxidation (LPO), reduced glutathione content and the activities of superoxide dismutase, catalase, glutathione peroxidase, and glutathione reductase were depleted significantly in both kidney and brain. Treatment with free CUR and CUR-NP decreased the LPO and increased the enzymatic and nonenzymatic antioxidant system in kidney and brain. Histopathological examination showed that kidney and brain injury mediated by arsenic was ameliorated by treatment. However, the amelioration percentage indicates that CUR-NP had marked therapeutic effect on arsenic-induced oxidative damage in kidney and brain tissues.


Subject(s)
Antioxidants/pharmacology , Arsenic/toxicity , Brain Diseases/chemically induced , Curcumin/pharmacology , Kidney Diseases/chemically induced , Nanoparticles/chemistry , Oxidative Stress/drug effects , Administration, Oral , Animals , Antioxidants/administration & dosage , Brain Chemistry/drug effects , Curcumin/administration & dosage , Kidney/chemistry , Kidney/drug effects , Male , Particle Size , Rats , Rats, Wistar
3.
Environ Toxicol ; 30(6): 628-37, 2015.
Article in English | MEDLINE | ID: mdl-24347089

ABSTRACT

We investigated the therapeutic effectiveness of the nanoparticle-encapsulated curcumin (CUR-NP) against sodium arsenite-induced hepatic oxidative damage in rats. The CUR-NP prepared by emulsion technique was spherical in shape with an encapsulation efficiency of 86.5%. The particle size ranged between 120 and 140 nm with the mean particle size being 130.8 nm. Rats were divided into five groups of six each. Group 1 served as control. Group 2 rats were exposed to sodium arsenite (25 ppm) daily through drinking water for 42 days. Groups 3, 4, and 5 were treated with arsenic as in group 2, however, they were administered, empty nanoparticles, curcumin (100 mg/kg bw) and CUR-NP (100 mg/kg bw), respectively, by oral gavage during the last 14 days of arsenic exposure. Arsenic increased the activities of serum alanine aminotransferase and aspartate aminotransferase and caused histological alterations in liver indicating hepatotoxicity. Arsenic increased lipid peroxidation, depleted reduced glutathione and decreased the activities of superoxide dismutase, catalase, glutathione peroxidase and glutathione reductase in liver. All these effects of arsenic were attenuated with both curcumin and CUR-NP. However, the magnitude of amelioration was more pronounced with CUR-NP. The results indicate that curcumin given in nano-encapsulated form caused better amelioration than free curcumin. © 2013 Wiley Periodicals, Inc. Environ Toxicol 30: 628-637, 2015.


Subject(s)
Arsenic Poisoning/prevention & control , Arsenic/toxicity , Chemical and Drug Induced Liver Injury/prevention & control , Curcumin/pharmacology , Alanine Transaminase/blood , Animals , Arsenites/toxicity , Aspartate Aminotransferases/blood , Catalase/metabolism , Chemical and Drug Induced Liver Injury/enzymology , Curcumin/administration & dosage , Curcumin/chemistry , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Lipid Peroxidation/drug effects , Male , Nanoparticles , Particle Size , Rats , Rats, Wistar , Sodium Compounds/toxicity , Superoxide Dismutase/metabolism
4.
Mol Biol Rep ; 41(11): 7413-22, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25078984

ABSTRACT

We explored whether nanoformulation of curcumin can cause better protective effect than free curcumin against arsenic-induced genotoxicity. Curcumin-loaded Poly(lactic-co-glycolic acid) nanoparticles (CUR-NP) were prepared by emulsion technique. The CUR-NP were water soluble and showed biphasic release pattern. Rats were divided into 5 groups of 6 each. Group I served as the control. Group II rats were exposed to sodium arsenite (25 ppm) daily through drinking water for 42 days. Groups III, IV and V were maintained as in Group II, however, they were also administered empty nanoparticle, curcumin (100 mg/kg bw) and CUR-NP (100 mg/kg bw), respectively, by oral gavage during the last 14 days of arsenic exposure. On the 43rd day, genotoxic effects were evaluated in bone marrow cells. Arsenic increased chromosomal aberrations, micronuclei formation and DNA damage. Both free curcumin and CUR-NP attenuated these arsenic-mediated genotoxic effects. However, the result suggests that nanoformulation have better protective effect than free curcumin at the same dose level.


Subject(s)
Arsenites/toxicity , Curcumin/pharmacology , DNA Damage/drug effects , Nanoparticles/therapeutic use , Sodium Compounds/toxicity , Analysis of Variance , Animals , Bone Marrow Cells/metabolism , Chromosome Aberrations/drug effects , Comet Assay , Curcumin/chemistry , Curcumin/therapeutic use , Lactic Acid/chemistry , Lactic Acid/therapeutic use , Micronuclei, Chromosome-Defective/drug effects , Molecular Structure , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Polyglycolic Acid/therapeutic use , Polylactic Acid-Polyglycolic Acid Copolymer , Rats
5.
Environ Toxicol Pharmacol ; 38(2): 397-405, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25128769

ABSTRACT

We assessed whether repeated arsenic exposure can decrease paracetamol-mediated antinociception by modulating serotonergic and endocannabinoid pathways. Rats were preexposed to elemental arsenic (4ppm) as sodium arsenite through drinking water for 28 days. Next day paracetamol's (400mg/kg, oral) antinociceptive activity was assessed through formalin-induced nociception. Serotonin content and gene expression of 5-HT1A, 5-HT2A and CB1 receptors were evaluated in brainstem and frontal cortex. Arsenic decreased paracetamol-mediated analgesia. Paracetamol, but not arsenic, increased serotonin content in these regions. Arsenic attenuated paracetamol-mediated increase in serotonin level. Paracetamol did not alter 5-HT1A expression, but caused down-regulation of 5-HT2A and up-regulation of CB1 receptors. Arsenic down-regulated these receptors. However, paracetamol-mediated down-regulation of 5-HT2A was more pronounced. Arsenic did not modify paracetamol's effect on 5-HT1A expression, but reduced paracetamol-mediated down-regulation of 5-HT2A and reversed up-regulation of CB1 receptors. Results suggest arsenic reduced paracetamol-induced analgesia possibly by interfering with pronociceptive 5-HT2A and antinociceptive CB1 receptors.


Subject(s)
Acetaminophen/administration & dosage , Analgesics/administration & dosage , Arsenites/toxicity , Drinking Water/chemistry , Receptor, Cannabinoid, CB1/metabolism , Receptors, Serotonin/metabolism , Sodium Compounds/toxicity , Water Pollutants, Chemical/toxicity , Acetaminophen/pharmacology , Analgesics/pharmacology , Animals , Brain Stem/metabolism , Drug Interactions , Frontal Lobe/metabolism , Gene Expression Regulation/drug effects , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/genetics , Receptors, Serotonin/genetics
6.
Toxicol Appl Pharmacol ; 280(1): 107-16, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25058445

ABSTRACT

We evaluated whether atorvastatin, an extensively prescribed statin for reducing the risks of cardiovascular diseases, can reduce the risk of arsenic-induced vascular dysfunction and inflammation in rats and whether the modulation could be linked to improvement in vascular NO signaling. Rats were exposed to sodium arsenite (100ppm) through drinking water for 90 consecutive days. Atorvastatin (10mg/kg bw, orally) was administered once daily during the last 30days of arsenic exposure. On the 91(st) day, blood was collected for measuring serum C-reactive protein. Thoracic aorta was isolated for assessing reactivity to phenylephrine, sodium nitroprusside and acetylcholine; evaluating eNOS and iNOS mRNA expression and measuring NO production, while abdominal aorta was used for ELISA of cytokines, chemokine and vascular cell adhesion molecules. Histopathology was done in aortic arches. Arsenic did not alter phenylephrine-elicited contraction. Atorvastatin inhibited Emax of phenylephrine, but it augmented the contractile response in aortic rings from arsenic-exposed animals. Sodium nitroprusside-induced relaxation was not altered with any treatment. However, arsenic reduced acetylcholine-induced relaxation and affected aortic eNOS at the levels of mRNA expression, protein concentration, phosphorylation and NO production. Further, it increased aortic iNOS mRNA expression, iNOS-derived NO synthesis, production of pro-inflammatory mediators (IL-1ß, IL-6, MCP-1, VCAM, sICAM) and serum C-reactive protein and aortic vasculopathic lesions. Atorvastatin attenuated these arsenic-mediated functional, biochemical and structural alterations. Results show that atorvastatin has the potential to ameliorate arsenic-induced vascular dysfunction and inflammation by restoring endothelial function with improvement in NO signaling and attenuating production of pro-inflammatory mediators and cell adhesion molecules.


Subject(s)
Arsenic/toxicity , Endothelium, Vascular/drug effects , Heptanoic Acids/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Inflammation Mediators/physiology , Nitric Oxide/physiology , Pyrroles/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Atorvastatin , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Male , Organ Culture Techniques , Random Allocation , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology
7.
Environ Toxicol Pharmacol ; 37(1): 438-47, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24448467

ABSTRACT

We examined whether subacute arsenic exposure can reduce paracetamol-mediated antipyretic activity by affecting COX pathway and cannabinoid CB1 receptor regulation. Rats were preexposed to elemental arsenic (4 ppm) as sodium arsenite through drinking water for 28 days. Next day pyrexia was induced with lipopolysaccharide and paracetamol's (200 mg/kg, oral) antipyretic activity was assessed. The activities of COX-1 and COX-2, the levels of PGE2, TNF-α and IL-1ß and expression of CB1 receptors were assessed in brain. Arsenic inhibited paracetamol-mediated antipyretic activity. COX-1 activity was not affected by any treatments. Paracetamol decreased COX-2 activity, levels of PGE2, TNF-α and IL-1ß and caused up-regulation of CB1 receptors. Arsenic caused opposite effects on these parameters. In the arsenic-preexposed rats, paracetamol-mediated effects were attenuated, while CB1 receptor up-regulation was reversed to down-regulation. Results suggest that elevated COX-2 activity and reduced CB1 expression could be involved in the arsenic-mediated attenuation of the antipyretic activity of paracetamol.


Subject(s)
Acetaminophen/therapeutic use , Antipyretics/therapeutic use , Arsenic/pharmacology , Cyclooxygenase 2 Inhibitors/therapeutic use , Water Pollutants, Chemical/pharmacology , Acetaminophen/pharmacology , Animals , Antipyretics/pharmacology , Brain/drug effects , Brain/metabolism , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/metabolism , Drug Interactions , Fever/chemically induced , Fever/drug therapy , Fever/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides , Male , Membrane Proteins/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
Environ Toxicol ; 29(2): 187-98, 2014 Feb.
Article in English | MEDLINE | ID: mdl-22120977

ABSTRACT

We evaluated whether the commonly used analgesic-antipyretic drug acetaminophen can modify the arsenic-induced hepatic oxidative stress and also whether withdrawal of acetaminophen administration during the course of long-term arsenic exposure can increase susceptibility of liver to arsenic toxicity. Acetaminophen was co-administered orally to rats for 3 days following 28 days of arsenic pre-exposure (Phase-I) and thereafter, acetaminophen was withdrawn, but arsenic exposure was continued for another 28 days (Phase-II). Arsenic increased lipid peroxidation and reactive oxygen species (ROS) generation, depleted glutathione (GSH), and decreased superoxide dismutase (SOD), catalase, glutathione peroxidase (GPx), and glutathione reductase (GR) activities. Acetaminophen caused exacerbation of arsenic-mediated lipid peroxidation and ROS generation and further enhancement of serum alanine aminotransferase and aspartate aminotransferase activities. In Phase-I, acetaminophen caused further GSH depletion and reduction in SOD, catalase, GPx and GR activities, but in Phase-II, only GPx and GR activities were more affected. Arsenic did not alter basal and inducible nitric oxide synthase (iNOS)-mediated NO production, but decreased constitutive NOS (cNOS)-mediated NO release. Arsenic reduced expression of endothelial NOS (eNOS) and iNOS genes. Acetaminophen up-regulated eNOS and iNOS expression and NO production in Phase-I, but reversed these effects in Phase-II. Results reveal that acetaminophen increased the risk of arsenic-mediated hepatic oxidative damage. Withdrawal of acetaminophen administration also increased susceptibility of liver to hepatotoxicity. Both ROS and NO appeared to mediate lipid peroxidation in Phase-I, whereas only ROS appeared responsible for peroxidative damage in Phase-II.


Subject(s)
Acetaminophen/toxicity , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Arsenic/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Water Pollutants, Chemical/toxicity , Animals , Catalase/metabolism , Chemical and Drug Induced Liver Injury/etiology , Drug Synergism , Gene Expression Regulation/drug effects , Glutathione/metabolism , Glutathione/pharmacology , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Lipid Peroxidation/drug effects , Male , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/genetics , Oxidation-Reduction , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
9.
Altern Lab Anim ; 41(4): P42-4, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24168138
10.
Int Immunopharmacol ; 17(1): 65-70, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23747587

ABSTRACT

We evaluated whether the nanoformulation of curcumin could be more effective than free curcumin against arsenic-induced immune dysfunction in rats. Curcumin was encapsulated in polylactic-co-glycolic acid (PLGA). Nanocurcumin (CUR-NP) exhibited a spherical shape with the mean particle size of 130.8 nm. Rats were randomly divided into five groups of six each. Group I was kept as the control. In Group II, rats were exposed to sodium arsenite (25 ppm) daily through drinking water for 42 days. Groups III, IV and V were treated with arsenic as in Group II, however, they were administered with nanoparticle, curcumin (100 mg/kg bw) and CUR-NP (100 mg/kg bw), respectively, by oral gavage during the last 14 days of arsenic exposure. At term, serum and spleen were collected. Immune dysfunction was evaluated by assessing cellular and humoral immunities. Arsenic significantly decreased the splenic lymphocyte proliferation in response to the antigen -- Keyhole Limpet Hemocyanin (KLH) and mitogen -- concanavalin-A. Arsenic reduced both the delayed type hypersensitivity response and secondary antibody (IgG) response to KLH. It also reduced the lipopolysaccharide-stimulated nitric oxide production in splenic lymphocytes. Free curcumin and CUR-NP treatment significantly attenuated these arsenic-mediated effects. However, the magnitude of the effects indicates that CUR-NP has better ameliorative potential than free curcumin at the equivalent dose level.


Subject(s)
Arsenic/toxicity , Curcumin/pharmacology , Nanoparticles/chemistry , Animals , Curcumin/chemistry , Lactic Acid/chemistry , Male , Nitric Oxide , Nitrites , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Random Allocation , Rats , Rats, Wistar
11.
Environ Toxicol Pharmacol ; 33(2): 267-76, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22236721

ABSTRACT

We evaluated the modulatory role of the groundwater contaminant arsenic on the pharmacodynamic responses of the nonsteroidal analgesic-antipyretic drug ketoprofen and the major pro-inflammatory mediators linked to the mechanism of ketoprofen's therapeutic effects. Rats were pre-exposed to sodium arsenite (0.4, 4 and 40 ppm) through drinking water for 28 days. The pharmacological effects of orally administered ketoprofen (5 mg/kg) were evaluated the following day. Pain, inflammation and pyretic responses were, respectively, assessed through formalin-induced nociception, carrageenan-induced inflammation and lipopolysaccharide-induced pyrexia. Arsenic inhibited ketoprofen's analgesic, anti-inflammatory and antipyretic effects. Further, arsenic enhanced cyclooxygenase-1 and cyclooxygenase-2 activities and tumor necrosis factor-α, interleukin-1ß and prostaglandin-E(2) production in hind paw muscle. These results suggest a functional antagonism of ketoprofen by arsenic. This may relate to arsenic-mediated local release of tumor necrosis factor-α and interleukin-1ß, which causes cyclooxygenase induction and consequent prostaglandin-E(2) release. In conclusion, subacute exposure to environmentally relevant concentrations of arsenic through drinking water may aggravate pain, inflammation and pyrexia and thereby, may reduce the therapeutic efficacy of ketoprofen.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arsenites/toxicity , Drinking Water/chemistry , Fever/prevention & control , Inflammation/prevention & control , Ketoprofen/pharmacology , Pain/prevention & control , Sodium Compounds/toxicity , Water Pollutants, Chemical/toxicity , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Arsenites/administration & dosage , Carrageenan , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Fever/chemically induced , Fever/metabolism , Formaldehyde , Hindlimb , Inflammation/chemically induced , Inflammation/metabolism , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Ketoprofen/administration & dosage , Lipopolysaccharides , Male , Membrane Proteins/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Pain/chemically induced , Pain/metabolism , Rats , Rats, Wistar , Sodium Compounds/administration & dosage , Time Factors , Tumor Necrosis Factor-alpha/metabolism , Water Pollutants, Chemical/administration & dosage
12.
Exp Toxicol Pathol ; 64(5): 487-93, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21130633

ABSTRACT

The aim of present study was to investigate the protective effect of curcumin on cypermethrin-induced changes in blood biochemical markers and tissue antioxidant enzyme in rats. Rats were divided into six groups of six each: group I used as control and II and III groups were used as vehicle control. While, groups IV, V and VI were orally treated with curcumin (100 mg/kg body weight), cypermethrin (25 mg/kg body weight) and cypermethrin plus curcumin, respectively for 28 days. Serum biochemical markers were measured in the serum, and the levels of lipid peroxidation and antioxidant enzyme activity were determined in the liver, kidney and brain. Cypermethrin administration caused elevated level of blood biochemical markers in serum and lipid peroxidation in liver, kidney and brain. While the activities of non-enzymatic and enzymatic antioxidants levels were decreased except superoxide dismutase in liver, kidney and brain tissues. The presence of curcumin with cypermethrin significantly decreased the blood biochemical markers and lipid peroxidation but significantly increased the reduced glutathione, catalase and glutathione peroxidase level and preserved the normal histological architecture of the liver, kidney and brain. Our results indicate that curcumin can be potent protective agent against cypermethrin-induced biochemical alterations and oxidative damage in rats.


Subject(s)
Antioxidants/pharmacology , Curcumin/pharmacology , Insecticides/toxicity , Oxidative Stress/drug effects , Pyrethrins/toxicity , Animals , Brain/drug effects , Kidney/drug effects , Lipid Peroxidation/drug effects , Liver/drug effects , Male , Rats , Rats, Wistar
13.
Food Chem Toxicol ; 49(4): 974-82, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21219961

ABSTRACT

We examined whether acetaminophen could alter renal oxidative stress induced by arsenic; also whether withdrawal of acetaminophen treatment can increase susceptibility of kidney to arsenic toxicity. Acetaminophen (400 and 1600 mg/kg) was co-administered orally to rats for 3 days after preexposure to arsenic (25 ppm) for 28 days (Phase-I) and thereafter, acetaminophen was withdrawn, but arsenic exposure was continued for another 28 days (Phase-II). Acetaminophen enhanced arsenic-induced lipid peroxidation, GSH depletion and ROS production and further decreased superoxide dismutase, catalase, glutathione peroxidase and glutathione reductase activities. Increased peroxidation did not alter kidney weight, but increased serum urea nitrogen and creatinine. Arsenic did not alter basal, iNOS-mediated NO production or iNOS expression. Arsenic decreased cNOS-mediated NO release and eNOS expression in Phase-II. Acetaminophen increased their expressions and NO production in Phase-I. In Phase-II, arsenic-mediated effects on NO remained mostly unaffected with acetaminophen. Results reveal that acetaminophen enhanced the risk of arsenic-mediated oxidative stress in kidney. Discontinuation of acetaminophen administration also increased the susceptibility of kidney to nephrotoxic effect of arsenic. It appeared ROS were primarily responsible for oxidative stress in both the phases. NO may have a minor role in Phase-I, but does not contribute to redox signaling mechanism in Phase-II.


Subject(s)
Acetaminophen/toxicity , Arsenic/toxicity , Kidney/drug effects , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Animals , Base Sequence , Biomarkers/blood , Catalase/metabolism , DNA Primers , Glutathione/metabolism , Kidney/enzymology , Kidney/metabolism , Lipid Peroxidation/drug effects , Male , Organ Size/drug effects , Oxidation-Reduction , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
14.
Environ Toxicol ; 26(3): 250-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-19950220

ABSTRACT

Acetaminophen (AP) is a widely used, cheap, and over-the-counter nonsteroidal anti-inflammatory drug. Its toxicity depends on the cytochrome P-450 (CYP)-mediated oxidation to the toxic metabolite N-acetyl-p-benzoquinoneimine. On the other hand, arsenic, a global groundwater and environmental contaminant of major public health concern, decreases hepatic CYP content and its dependent monoxygenase activities. We hypothesized that arsenic exposure would reduce the AP toxicity. Our aim was to evaluate the effects of repeated preexposure or coexposure to arsenic on the oxidative stress induced by a single or repeated oral administration of AP in rat kidney and its possible relationship with the effects of arsenic on certain antioxidants. Rats were exposed to arsenic through drinking water at 25 ppm for 28 days. The dosages of AP used for a single administration after arsenic preexposure for 28 days were 420 and 1000 mg kg(-1) , while for daily concurrent administration with arsenic for 28 days were 105 and 420 mg kg(-1) body weight. AP increased lipid peroxidation (LPO) in rat kidney where its acute administration caused more LPO than its subacute dosing. Repeated arsenic exposure differentially altered the AP-induced LPO. Arsenic preexposure antagonized LPO induced by the acute AP administration; in contrast, arsenic coexposure aggravated the repeated dose (AP)-mediated LPO. Arsenic-mediated alterations in renal sensitivity to LPO did not appear to be linked to the antioxidants such as reduced glutathione, superoxide dismutase, catalase, glutathione peroxidase, and glutathione reductase; nor could it be related to glutathione-S-transferase activity. The results indicated that repeated arsenic preexposure decreased susceptibility of rat kidney to acute AP-mediated oxidative stress; on the contrary, its coexposure rendered the rat kidney more vulnerable to oxidative stress induced by the repeated dosing of AP.


Subject(s)
Acetaminophen/toxicity , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Arsenic/toxicity , Environmental Pollutants/toxicity , Kidney/drug effects , Acetaminophen/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Antioxidants/metabolism , Arsenic/metabolism , Catalase/metabolism , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Environmental Pollutants/metabolism , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Glutathione Transferase/metabolism , Kidney/metabolism , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/metabolism , Male , Malondialdehyde/metabolism , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Oxidation-Reduction , Oxidative Stress/drug effects , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
15.
Environ Toxicol Pharmacol ; 30(3): 289-91, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21787662

ABSTRACT

Cypermethrin is a synthetic pyrethroid insecticide used worldwide in agriculture, home pest control, protection of foodstuff and disease vector control. The aim of the present study was to investigate the protective effect of curcumin on cypermethrin-induced genotoxicity in rats. Administration of cypermethrin (25mg/kg, p.o.) for 28 days resulted in significant increase in the frequency of micronuclei formation in bone marrow cells and DNA damage in blood cells. Curcumin (100mg/kg, p.o.) administration caused significant reduction in micronuclei formation and, marked reduction in DNA damage. The present study revealed that presence of curcumin could diminish cypermethrin-induced genotoxicity in rats.

16.
Food Chem Toxicol ; 48(2): 605-10, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19932728

ABSTRACT

We evaluated whether repeated arsenic preexposure can increase acetaminophen-induced hepatic oxidative stress. Rats were exposed to arsenic (25 ppm; rat equivalent concentration of maximum groundwater contamination level) via drinking water for 28 days. Next day, they were given single oral administration of acetaminophen (420 or 1000 mg/kg b.w.). Hepatotoxicity was evaluated by assessing serum biomarkers, cytochrome-P450 (CYP) content, CYP3A4- and CYP2E1-dependent enzymes, lipid peroxidation and antioxidants. Arsenic or acetaminophen increased serum ALT and AST activities and depleted CYP. Arsenic decreased, but acetaminophen increased CYP-dependent enzyme activities. These agents independently increased lipid peroxidation and decreased antioxidants. Arsenic did not alter the effects of acetaminophen on serum biomarkers, caused further CYP depletion and decreased acetaminophen-mediated induction of drug-metabolizing enzymes. Arsenic enhanced the lower dose of acetaminophen-mediated lipid peroxidation and glutathione depletion with no further alterations in enzymatic antioxidants. However, arsenic attenuated the higher dose-mediated lipid peroxidation and glutathione depletion with improvement in glutathione peroxidase and glutathione reductase activities, further decrease in catalase and no alterations in superoxide dismutase and glutathione-S-transferase activities. Results show that arsenic preexposure increased the susceptibility of rats to hepatic oxidative stress induced by the lower dose of acetaminophen, but reduced the oxidative stress induced by the higher dose.


Subject(s)
Acetaminophen/toxicity , Analgesics/toxicity , Arsenites/toxicity , Liver/drug effects , Oxidative Stress/drug effects , Sodium Compounds/toxicity , Water Pollutants, Chemical/toxicity , Acetaminophen/metabolism , Administration, Oral , Analgesics/metabolism , Animals , Antioxidants/metabolism , Biomarkers/blood , Cytochrome P-450 CYP2E1/drug effects , Cytochrome P-450 CYP2E1/metabolism , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/drug effects , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Lipid Peroxidation/drug effects , Liver/enzymology , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Oxidoreductases/metabolism , Rats , Rats, Wistar
17.
Ecotoxicol Environ Saf ; 73(1): 94-100, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19782400

ABSTRACT

Arsenic is an environmental contaminant, while acetaminophen is an extensively used nonsteroidal analgesic-antipyretic drug. We evaluated whether subacute co-exposure to arsenic and acetaminophen would produce more toxicity than that caused by exposure to either of the xenobiotics in rats. Toxicity was evaluated through changes in body weight, feed consumption, liver weight and microsomal drug-metabolizing enzymes, lipid peroxidation and antioxidants in liver. Arsenic had no effect on body weight and feed consumption. Acetaminophen-mediated decrease in body weight was attenuated in the co-exposed rats. Acetaminophen alone or its co-administration with arsenic decreased feed consumption. Arsenic reduced acetaminophen-mediated increase in the activities of drug-metabolizing enzymes. The co-exposure caused lesser lipid peroxidation than the individual exposure. Arsenic or acetaminophen given alone depleted GSH and decreased the activities of superoxide dismutase, catalase, glutathione peroxidase, glutathione reductase and glutathione-S-transferase and these effects remained mostly unaffected after co-exposure. The results suggest that co-exposure to arsenic and acetaminophen may be less hazardous than their independent exposure in rats.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Arsenic/toxicity , Water Pollutants, Chemical/toxicity , Animals , Body Weight/drug effects , Cytochrome P-450 Enzyme System/metabolism , Eating/drug effects , Glutathione/analysis , Lipid Peroxidation , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Oxidative Stress , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
18.
Eur J Pharmacol ; 626(2-3): 205-12, 2010 Jan 25.
Article in English | MEDLINE | ID: mdl-19782680

ABSTRACT

Ischemia/reperfusion injury ends up in the cascade of excitotoxic stimulation of superoxide and nitric oxide formation leading to the generation of highly reactive products, including peroxinitrite and hydroxyl radical, which are capable of damaging lipids, proteins and DNA. Several polyphenolic compounds scavenge the radicals and protect from injury. 5,7,3',4',5'-pentahydroxy dihdroflavanol-3-O-(2''-O-galloyl)-beta-d-glucopyranoside (AP1), a polyphenolic compound, isolated from Anogeissus pendula Edgew was tested for its neuroprotective effect in transient focal cerebral ischemia in rats. Transient focal cerebral ischemia was produced by middle cerebral artery occlusion for 2h for studying infarct volume, brain edema, apoptosis and oxidative stress. AP1 was tested for in vitro protection from glutamate and hydrogen peroxide-induced damage to Neuro-2a cells by MTT assay. It was also tested for its in vitro antioxidant, lipid peroxidation inhibition, NO scavenging and cyclooxygenase inhibitory activities. AP1 treatment (30 mg/kg i.p.) before reperfusion injury (0 h) significantly reduced the infarct volume, cerebral edema, number of apoptotic cells in penumbra and neurobehavioural abnormality score and lipid peroxidation, protein carbonyl levels and total thiols in brain. Increased catalase activity and NOx levels in ischemic animals were significantly reduced by AP1 treatment. AP1 (3 microg/ml) protected Neuro-2a cells to H2O2 and glutamate-induced damage. In in vitro studies, AP1 was found to possess reducing and NO scavenging activities. It also reduced lipid peroxidation and inhibited cyclooxygenase activity (cyclooxygenase-1 and cyclooxygenase-2). AP1 can be used as a neuroprotective agent in stroke as it reduced apoptosis and found to be a good antioxidant and anti-inflammatory compound.


Subject(s)
Brain Ischemia/prevention & control , Flavonoids/pharmacology , Glucosides/pharmacology , Neuroprotective Agents/pharmacology , Phenols/pharmacology , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Apoptosis/drug effects , Behavior, Animal/drug effects , Brain Edema/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cerebral Infarction/drug therapy , Cerebral Infarction/pathology , Flavonoids/therapeutic use , Glucosides/therapeutic use , Glutamic Acid/pharmacology , Hydrogen Peroxide/pharmacology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Phenols/therapeutic use , Polyphenols , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Rats, Wistar , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...