Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-39028331

ABSTRACT

Acrylamide (ACR) is a toxic, probably carcinogenic compound commonly found in fried foods and used in the production of many industrial consumer products. ACR-induced acute kidney injury is mediated through several signals. In this research, we investigated, for the first time, the therapeutic effects of phytochemicals apocynin (APO) and/or umbelliferone (UMB) against ACR-induced nephrotoxicity in rats and emphasized the underlying molecular mechanism. To achieve this goal, five groups of rats were randomly assigned: the control group received vehicle (0.5% CMC; 1 ml/rat), ACR (40 mg/kg, i.p.), ACR + APO (100 mg/kg, P.O.), ACR + UMB (50 mg/kg, P.O.), and combination group for 10 days. In ACR-intoxicated rats, there was a significant reduction in weight gain while the levels of blood urea, uric acid, creatinine, and Kim-1 were elevated, indicating renal injury. Histopathological injury was also observed in the kidneys of ACR-intoxicated rats, confirming the biochemical data. Moreover, MDA, TNF-α, and IL-1ß levels were raised; and GSH and SOD levels were decreased. In contrast, treatment with APO, UMB, and their combination significantly reduced the kidney function biomarkers, prevented tissue damage, and decreased inflammatory cytokines and MDA. Mechanistically, it suppressed the expression of NLRP-3, ASC, GSDMD, caspase-1, and IL-1ß, while it upregulated Nrf-2 and HO-1 in the kidneys of ACR-intoxicated rats. In conclusion, APO, UMB, and their combination prevented ACR-induced nephrotoxicity in rats by attenuating oxidative injury and inflammation, suppressing NLRP-3 inflammasome signaling, enhancing antioxidants, and upregulating Nrf-2 and HO-1 in the kidneys of ACR-induced rats.

2.
Molecules ; 26(22)2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34834085

ABSTRACT

Osteoarthritis (OA) is a complex disease characterized by structural, functional, and metabolic deteriorations of the whole joint and periarticular tissues. In the current study, we aimed to investigate the possible effects of tempol on knee OA induced by the chemical chondrotoxic monosodium iodoacetate (MIA) which closely mimics both the pain and structural changes associated with human OA. Rats were administrated oral tempol (100 mg/kg) one week post-MIA injection (3 mg/50 µL saline) at the right knee joints for 21 consecutive days. Tempol improved motor performance and debilitated the MIA-related radiological and histological alterations. Moreover, it subsided the knee joint swelling. Tempol decreased the cartilage degradation-related biomarkers as matrix metalloproteinase-13, bone alkaline phosphatase (bone ALP), and fibulin-3. The superoxide dismutase mimetic effect of tempol was accompanied by decreased NADPH oxidase 4 (NOX4), inflammatory mediators, nuclear factor-kappa B (NF-κB), over-released transforming growth factor-ß1 (TGF-ß1). Tempol decreased the expression of chemokine (C-C motif) ligand 2 (CCL2). On the molecular level, tempol reduced the phosphorylated protein levels of p38 mitogen-activated protein kinase (MAPK), and small mother against decapentaplegic 3 homologs (SMAD3). These findings suggest the promising role of tempol in ameliorating MIA-induced knee OA in rats via collateral suppression of the catabolic signaling cascades including TGF-ß1/SMAD3/NOX4, and NOX4/p38MAPK/NF-κB and therefore modulation of oxidative stress, catabolic inflammatory cascades, chondrocyte metabolic homeostasis.


Subject(s)
Cyclic N-Oxides/pharmacology , Iodoacetic Acid/adverse effects , MAP Kinase Signaling System/drug effects , NADPH Oxidase 4/metabolism , Osteoarthritis, Knee , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Iodoacetic Acid/pharmacology , Male , Osteoarthritis, Knee/chemically induced , Osteoarthritis, Knee/drug therapy , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , Rats , Rats, Wistar , Spin Labels
3.
Life Sci ; 266: 118882, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33310046

ABSTRACT

AIMS: Cilostazol (Cilo), a phosphodiesterase-III inhibitor, has signified its efficacy against different ischemia/reperfusion (IS/RE) models. Nevertheless, it has not fully illuminated its potential effect against intestinal IS/RE-induced lung injury. Consequently, the study was fashioned to evaluate the feasible mechanism of action of Cilo against intestinal IS/RE-induced lung injury. MAIN METHODS: Wistar rats were treated with Cilo (0.1 g/kg, p.o.) or with a vehicle for 14 days prior to IS/RE, induced by clamping of the superior mesenteric artery for 30 min with subsequent clamp removal for 2 h. KEY FINDINGS: The mechanistic study disclosed that Cilo protected the two studied organs, viz., lung, and intestine partially by intensifying the expression/content of PPAR-γ accompanied by reducing the expression/content of NF-қB-p65 and STAT3. In addition to normalizing MDA, iNOS, and NOx, the Cilo antioxidant power was confirmed by intensifying tissues content of the total antioxidant capacity. With regard to the anti-inflammatory effect, Cilo reduced the effects of TNF-α, IL-6, and ICAM-1, which were reflected in MPO activity. Furthermore, Cilo had an anti-apoptotic attribute demonstrated by enhancing Bcl-2 content and lessening caspase-3 level. SIGNIFICANCE: Cilo provided conceivable protective mechanisms to modulate events concomitant with mesenteric IS/RE partly by modulating oxidative stress, inflammation, and apoptosis feasibly via the participation of PPAR-γ, STAT3, and NF-κB p65 signaling pathways.


Subject(s)
Cilostazol/pharmacology , Lung Diseases/prevention & control , Mesenteric Ischemia/complications , NF-kappa B/metabolism , PPAR gamma/metabolism , Reperfusion Injury/complications , STAT3 Transcription Factor/metabolism , Animals , Bronchodilator Agents/pharmacology , Gene Expression Regulation , Lung Diseases/etiology , Lung Diseases/metabolism , Lung Diseases/pathology , Male , NF-kappa B/genetics , Oxidative Stress , PPAR gamma/genetics , Rats , Rats, Wistar , STAT3 Transcription Factor/genetics , Signal Transduction
4.
Int Immunopharmacol ; 80: 106131, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31981960

ABSTRACT

BACKGROUND: Hepatic ischemia/reperfusion (I/R) injury occurs in different clinical settings as hepatic transplantation, and different types of shock. I/R injury is the main cause of hepatic damage and failure due to the production of reactive oxygen species (ROS) and inflammatory cytokines. Dimethyl fumarate (DMF), an immunomodulatory drug, activates cellularantioxidantsignaling pathways exerting cytoprotective properties. Curcumin (CUR), a natural phenolic compound, possesses antioxidant and anti-inflammatory properties. METHOD: To study potential protective effects of DMF with CUR against hepatic I/R injury in rats, animals were randomly allocated into seven groups as follows: (1) Sham; (2) DMF (25 mg/Kg, p.o); (3) CUR (400 mg/Kg, p.o.); (4) I/R; (5) DMF + I/R; (6) CUR + I/R; and combination (COM) therapy + I/R. Drugs were given for 14 days before I/R. RESULTS: Compared with I/R group, COM group showed the best amelioration in hepatic injury induced by I/R insult. This was confirmed by a significant reduction in serum ALT and AST activity with improved histopathological results when compared to every single treatment. Hepatic protection afforded by DMF was mediated by activating Nrf2/HO-1 signaling and increasing GSH and TAC contents. CUR treatment improved the inflammatory markers (TNF-α, IL-1ß, Il-6 and iNOS) as well as neutrophilic infiltration assessed as MPO. Moreover, CUR potentiated Nrf2/HO-1 signaling induced by DMF with significant suppression in lipid peroxidation. CONCLUSION: We concluded that combining DMF and CUR has more efficient hepatoprotective effects against hepatic-induced IRI via potentiating antioxidant and anti-inflammatory properties mediated by Nrf2/HO-1 pathway.


Subject(s)
Curcumin/pharmacology , Dimethyl Fumarate/pharmacology , Liver Failure, Acute/drug therapy , Reperfusion Injury/complications , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , Curcumin/therapeutic use , Dimethyl Fumarate/therapeutic use , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination/methods , Heme Oxygenase (Decyclizing)/metabolism , Humans , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Liver Failure, Acute/immunology , Male , NF-E2-Related Factor 2/metabolism , Rats , Reperfusion Injury/drug therapy , Reperfusion Injury/immunology , Signal Transduction/drug effects , Signal Transduction/immunology
5.
Oncotarget ; 8(12): 20145-20164, 2017 Mar 21.
Article in English | MEDLINE | ID: mdl-28423620

ABSTRACT

Exosomes have emerged as important mediators of diverse biological functions including tumor suppression, tumor progression, invasion, immune escape and cell-to-cell communication, through the release of molecules such as mRNAs, miRNAs, and proteins. Here, we identified differentially expressed exosomal miRNAs between normal epithelial ovarian cell line and both resistant and sensitive ovarian cancer (OC) cell lines. We found miR-940 as abundant in exosomes from SKOV3-IP1, HeyA8, and HeyA8-MDR cells. The high expression of miR-940 is associated with better survival in patients with ovarian serous cystadenocarcinoma. Ectopic expression of miR-940 inhibited proliferation, colony formation, invasion, and migration and triggered G0/G1 cell cycle arrest and apoptosis in OC cells. Overexpression of miR-940 also inhibited tumor cell growth in vivo. We showed that proto-oncogene tyrosine-protein kinase (SRC) is directly targeted by miR-940 and that miR-940 inhibited SRC expression at mRNA and protein levels. Following this inhibition, the expression of proteins downstream of SRC, such as FAK, paxillin and Akt was also reduced. Collectively, our results suggest that OC cells secrete the tumor-suppressive miR-940 into the extracellular environment via exosomes, to maintain their invasiveness and tumorigenic phenotype.


Subject(s)
Biomarkers, Tumor/metabolism , Exosomes/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Ovarian Neoplasms/pathology , src-Family Kinases/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Female , Genes, Tumor Suppressor , Humans , Mice , Mice, Nude , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Proto-Oncogene Mas , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , src-Family Kinases/genetics
6.
Int J Mol Sci ; 18(3)2017 Mar 02.
Article in English | MEDLINE | ID: mdl-28257101

ABSTRACT

Intercellular communication via cell-released vesicles is a very important process for both normal and tumor cells. Cell communication may involve exosomes, small vesicles of endocytic origin that are released by all types of cells and are found in abundance in body fluids, including blood, saliva, urine, and breast milk. Exosomes have been shown to carry lipids, proteins, mRNAs, non-coding RNAs, and even DNA out of cells. They are more than simply molecular garbage bins, however, in that the molecules they carry can be taken up by other cells. Thus, exosomes transfer biological information to neighboring cells and through this cell-to-cell communication are involved not only in physiological functions such as cell-to-cell communication, but also in the pathogenesis of some diseases, including tumors and neurodegenerative conditions. Our increasing understanding of why cells release exosomes and their role in intercellular communication has revealed the very complex and sophisticated contribution of exosomes to health and disease. The aim of this review is to reveal the emerging roles of exosomes in normal and pathological conditions and describe the controversial biological role of exosomes, as it is now understood, in carcinogenesis. We also summarize what is known about exosome biogenesis, composition, functions, and pathways and discuss the potential clinical applications of exosomes, especially as biomarkers and novel therapeutic agents.


Subject(s)
Exosomes/physiology , Neoplasms/pathology , Neurodegenerative Diseases/pathology , Biological Transport , Cell Communication , Drug Delivery Systems , Humans , Neoplasms/metabolism , Neoplasms/therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/therapy
7.
Eur J Gastroenterol Hepatol ; 28(5): 553-7, 2016 May.
Article in English | MEDLINE | ID: mdl-26872109

ABSTRACT

BACKGROUND: Ezetimibe has been reported to inhibit viral entry and to reduce BMI and has been proposed as a novel therapeutic agent for chronic hepatitis C (CHC), potentiating the effects of pegylated interferon and ribavirin (peg-IFN/RBV). OBJECTIVE: The aim of the study was to assess the effects of ezetimibe coadministration with peg-IFN/RBV combination on the early virological response (EVR) rates in nonobese and obese patients with CHC genotype 4 (CHC-4). PATIENTS AND METHODS: A total of 144 CHC-4 patients were divided into two groups; group 1 included nonobese patients (n=76) and group 2 included obese patients (n=68). Each group was further subclassified into equal control and treated groups. The control groups received peg-IFN/RBV combination for 24 weeks, and the treated groups received peg-IFN/RBV plus ezetimibe for 12 weeks and then only peg-IFN/RBV for the remaining 12 weeks. RESULTS: The study revealed that EVR significantly improved in the obese patients (85.3 vs. 64.7% in the treated and control groups, respectively, at P<0.05) without any significant improvement in the nonobese patients. Biochemical responses (defined as normalization of alanine aminotransferase at week 12) were markedly improved in the treated groups in both the nonobese and obese groups compared with their respective controls. CONCLUSION: The addition of ezetimibe to peg-IFN/RBV combination significantly improves EVR rates in obese patients compared with nonobese patients, and remarkably improves the biochemical responses in both obese and nonobese patients with CHC-4. This may shed light on a new strategy for the treatment of CHC, particularly in obese Egyptian patients.


Subject(s)
Antiviral Agents/therapeutic use , Ezetimibe/therapeutic use , Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Interferon-alpha/therapeutic use , Obesity/complications , Polyethylene Glycols/therapeutic use , Ribavirin/therapeutic use , Adult , Antiviral Agents/adverse effects , Drug Therapy, Combination , Egypt , Ezetimibe/adverse effects , Female , Hepacivirus/pathogenicity , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/diagnosis , Humans , Interferon-alpha/adverse effects , Male , Middle Aged , Obesity/diagnosis , Polyethylene Glycols/adverse effects , Prospective Studies , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use , Ribavirin/adverse effects , Single-Blind Method , Time Factors , Treatment Outcome , Viral Load , Virus Internalization/drug effects
8.
Environ Toxicol ; 31(8): 913-22, 2016 Aug.
Article in English | MEDLINE | ID: mdl-25533183

ABSTRACT

The present study was aimed to investigate the mechanistic aspect of Aroclor 1260-induced hepatotoxicity and its protection by lipoic acid. The adult male Albino rats were divided into six groups. Group I served as control. Group II received lipoic acid (35 mg/kg/day). Aroclor 1260 was given to rats by oral gavage at doses 20, 40, or 60 mg/kg/day (Groups III, IV, and V, respectively). Group VI was pretreated with lipoic acid (35 mg/kg/day) 24 h before Aroclor 1260 (40 mg/kg/day). Treatment in all groups was continued for further 15 consecutive days. Serum alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, and lactate dehydrogenase activities and total bilirubin, total cholesterol, and triglycerides were significantly increased while total protein, total albumin, and high-density lipoprotein were significantly decreased. Hydrogen peroxide production and lipid peroxidation were significantly increased while superoxide dismutase and catalase activities and reduced glutathione (GSH) content was significantly decreased in liver. Caspase-3 & -9 activities were significantly increased in liver. Lipoic acid pretreatment significantly reverted all these abnormalities toward their normal levels. In conclusion, Aroclor 1260 induced liver dysfunction, at least in part, by induction of oxidative stress. Apoptotic effect of hepatic cells is involved in Aroclor 1260-induced liver injury. Lipoic acid could protect rats against Aroclor 1260-induced hepatotoxicity. © 2014 Wiley Periodicals, Inc. Environ Toxicol 31: 913-922, 2016.


Subject(s)
Antioxidants/pharmacokinetics , Aroclors/toxicity , Chemical and Drug Induced Liver Injury/prevention & control , Environmental Pollutants/toxicity , Thioctic Acid/pharmacology , Alanine Transaminase/blood , Alkaline Phosphatase/blood , Animals , Antioxidants/therapeutic use , Aspartate Aminotransferases/blood , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/enzymology , Drug Evaluation, Preclinical , Glutathione/metabolism , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/enzymology , Liver/pathology , Male , Oxidative Stress/drug effects , Rats , Rats, Wistar , Superoxide Dismutase/blood , Thioctic Acid/therapeutic use
9.
PLoS One ; 10(7): e0132497, 2015.
Article in English | MEDLINE | ID: mdl-26196679

ABSTRACT

Mangiferin (MF), a xanthonoid from Mangifera indica, has been proved to have antisecretory and antioxidant gastroprotective effects against different gastric ulcer models; however, its molecular mechanism has not been previously elucidated. Therefore, the aim of this study was to test its modulatory effect on several signaling pathways using the ischemia/reperfusion model for the first time. Animals were treated with MF, omeprazole (OMP), and the vehicle. The mechanistic studies revealed that MF mediated its gastroprotective effect partly via inducing the expression of Nrf2, HO-1 and PPAR-γ along with downregulating that of NF-κB. Surprisingly, the effect of MF, especially the high dose, exceeded that mediated by OMP except for Nrf2. The molecular results were reflected on the biomarkers measured, where the antioxidant effect of MF was manifested by increasing total antioxidant capacity and glutathione, besides normalizing malondialdehyde level. Additionally, MF decreased the I/R-induced nitric oxide elevation, an effect that was better than that of OMP. In the serum, MF, dose dependently, enhanced endothelial nitric oxide synthase, while reduced the inducible isoform. Regarding the anti-inflammatory effect of MF, it reduced serum level of IL-1ß and sE-selectin, effects that were mirrored on the tissue level of myeloperoxidase, the neutrophil infiltration marker. In addition, MF possessed an antiapoptotic character evidenced by elevating Bcl-2 level and reducing that of caspase-3 in a dose related order. As a conclusion, the intimated gastroprotective mechanisms of MF are mediated, partially, by modulation of oxidative stress, inflammation and apoptosis possibly via the Nrf2/HO-1, PPAR-γ/NF-κB signaling pathways.


Subject(s)
Anti-Ulcer Agents/therapeutic use , Antioxidants/therapeutic use , Omeprazole/therapeutic use , Reperfusion Injury/drug therapy , Stomach Ulcer/drug therapy , Xanthones/therapeutic use , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Antioxidants/chemistry , Gene Expression Regulation/drug effects , Heme Oxygenase-1/genetics , Male , Mangifera/chemistry , NF-E2-Related Factor 2/genetics , NF-kappa B/genetics , PPAR gamma/genetics , Rats , Rats, Wistar , Reperfusion Injury/blood , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction/drug effects , Stomach Ulcer/blood , Stomach Ulcer/genetics , Stomach Ulcer/pathology , Xanthones/chemistry
10.
Fitoterapia ; 90: 151-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23892001

ABSTRACT

Increased consumption of green tea (GT) without enough scientific data has raised safety concerns. Epigallocatechin 3-gallate (EGCG) is the most prominent polyphenol of GT that has antioxidant activity. However, higher doses of EGCG have been shown to cause liver injury. This study was initiated to determine the effect of GT extracts in a mouse model. We also investigated the effects of EGCG in normal and health-compromised mice. Different doses of GT fractions and EGCG were administered for 5 days to mice. Also, a single dose of lipopolysaccharide (LPS) was combined with EGCG in order to investigate its effect in the presence of fever. Plasma ALT and ALP levels were determined along with liver histopathology. Combining a single high IG dose of EGCG with a single IP dose of LPS initiated liver injury. Furthermore, repeated administration of high IG doses of EGCG showed mild liver injury, but it was augmented under febrile conditions induced by LPS. This study confirms the safety of reasonable consumption of GT over a short term. However, it highlights a caution that high doses of EGCG can lead to mild liver injury, and this may be markedly enhanced under febrile conditions.


Subject(s)
Camellia sinensis/chemistry , Catechin/analogs & derivatives , Chemical and Drug Induced Liver Injury , Liver/drug effects , Plant Extracts/adverse effects , Polyphenols/adverse effects , Animals , Camellia sinensis/adverse effects , Catechin/adverse effects , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/pathology , Fever/chemically induced , Lipopolysaccharides , Liver/enzymology , Liver/pathology , Male , Mice , Mice, Inbred Strains , Tea
11.
J Biochem Mol Toxicol ; 25(6): 386-92, 2011.
Article in English | MEDLINE | ID: mdl-21823216

ABSTRACT

Acrylonitrile is a potent hepatotoxic, mutagen, and carcinogen. A role for free radical-mediated lipid peroxidation in the toxicity of acrylonitrile has been suggested. The present study was designed to assess the hepatoprotective effect of quercetin against acrylonitrile-induced hepatotoxicity in rats. Liver damage was induced by oral administration of acrylonitrile (50 mg/kg/day/5 weeks). Acrylonitrile produced a significant elevation of malondialdehyde (138.9%) with a marked decrease in reduced glutathione (72.4%), and enzymatic antioxidants; superoxide dismutase (81%), and glutathione peroxidase (53.2%) in the liver. Serum aspartate aminotransferase, alanine aminotransferases, direct bilirubin, and total bilirubin showed a significant increase in acrylonitrile alone treated rats (115.5%, 110.8%, 1006.8%, and 1000.8%, respectively). Pretreatment with quercetin (70 mg/kg/day/6 weeks) and its coadministration with acrylonitrile prevented acrylonitrile-induced alterations in hepatic lipid peroxides and enzymatic antioxidants as well as serum aminotransferases and bilirubin. Histopathological findings supported the biochemical results. We suggest that querectin possess hepatoprotective effect against acrylonitrile-induced hepatotoxicity through its antioxidant activity.


Subject(s)
Acrylonitrile/toxicity , Antioxidants/therapeutic use , Chemical and Drug Induced Liver Injury/prevention & control , Liver/drug effects , Quercetin/therapeutic use , Animals , Antioxidants/administration & dosage , Biomarkers/blood , Biomarkers/metabolism , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Lipid Peroxides/metabolism , Liver/enzymology , Liver/metabolism , Liver/pathology , Liver Function Tests , Male , Quercetin/administration & dosage , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
12.
J Biochem Mol Toxicol ; 22(4): 268-73, 2008.
Article in English | MEDLINE | ID: mdl-18752313

ABSTRACT

Acrylonitrile (ACN) is a volatile, toxic liquid used as a monomer in the manufacture of synthetic rubber, styrene plastics, acrylic fiber, and adhesives. ACN is a potent neurotoxin. A role for free radical mediated lipid peroxidation in the toxicity of ACN has been suggested. We examined the ability of hesperidin, an antioxidant flavonoid, to attenuate ACN-induced alterations in lipid peroxidation in rat brains. The daily oral administration of ACN to male albino rats in a dose of 50 mg/kg bwt for a period of 28 days produced a significant elevation in brain lipid peroxides measured as malondialdehyde (MDA) amounting to 107%, accompanied by a marked decrease in brain-reduced glutathione (GSH) content reaching 63%. In addition, ACN administration resulted in significant reductions in the enzymatic antioxidant parameters of brain; superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), and glutathione-S-transferase (GST) recording 43%, 64%, 52%, and 43%, respectively. On the other hand, pretreatment with hesperidin and its coadministration with ACN once daily in a dose of 200 mg/kg bwt i.p. for 28 days ameliorated ACN-induced alterations in brain lipid peroxidation. These results suggest that hesperidin may have a beneficial role against ACN-induced oxidative stress in the brain; an effect that is mainly attributed to the antioxidant property of hesperidin.


Subject(s)
Acrylonitrile/antagonists & inhibitors , Acrylonitrile/toxicity , Antioxidants/pharmacology , Brain/drug effects , Brain/metabolism , Hesperidin/pharmacology , Oxidative Stress/drug effects , Animals , Antioxidants/metabolism , Brain/pathology , Catalase/metabolism , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Glutathione Transferase/metabolism , Lipid Peroxidation/drug effects , Male , Malondialdehyde/metabolism , Rats , Superoxide Dismutase/metabolism
13.
Pak J Pharm Sci ; 21(3): 255-61, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18614421

ABSTRACT

This study has been initiated to determine whether captopril, an angiotensin-converting enzyme (ACE) inhibitor containing sulfhydryl (-SH) group can protect against cisplatin-induced nephrotoxicity in rats. A single dose of cisplatin (7.5 mg/kg bwt) injected i.p. caused a significant increase in blood urea nitrogen (BUN) and creatinine levels amounting to 402% and 573%, respectively with a marked elevation in lipid peroxides measured as malondialdehyde (MDA) content (54%), accompanied by a significant decrease in reduced glutathione (GSH) content (27%) of kidney tissue as compared to control group. In addition, there were marked increases in kidney tissue content of nitric oxide (NO) (43%) and plasma endothelin-1(ET-1) (37%). On the other hand, administration of captopril (60 mg/kg bwt, i.p.) 1 h before cisplatin protected the kidney as indicated by restoration of BUN, creatinine, MDA, GSH, NO and ET-1. These results indicate that captopril, an ACEI, has a protective effect against cisplatin-induced damage to kidney. This reflects the beneficial role of captopril in treatment of renovascular hypertention and congestive heart failure; an effect that may be related to its free radicals scavenging and antioxidant effects which are sulfhydryl dependent.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Captopril/pharmacology , Free Radical Scavengers/pharmacology , Kidney Diseases/prevention & control , Kidney/drug effects , Animals , Antineoplastic Agents , Blood Urea Nitrogen , Cisplatin , Creatinine/blood , Disease Models, Animal , Endothelin-1/blood , Glutathione/metabolism , Kidney/enzymology , Kidney/metabolism , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Lipid Peroxidation/drug effects , Male , Nitric Oxide/metabolism , Rats , Rats, Wistar
14.
Biotechnol Appl Biochem ; 46(Pt 1): 35-40, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16848766

ABSTRACT

The aim of this experimental study was to investigate the effect of a standardized preparation of Ginkgo biloba extract (EGb 761) on the hyperlipidaemic nephrotoxicity and oxidative stress induced by a single intravenous injection (5 mg/kg) of adriamycin. EGb 761 was received daily thereafter by a gavage at the dose of 100 mg/kg for 35 consecutive days. EGb 761 administration significantly attenuated adriamycin-induced renal dysfunction, as assessed by measuring serum lipid profile, serum total protein, serum urea and Ccr (creatinine clearance). Furthermore, urinary excretions of protein and NAG (N-acetyl-beta-D-glucosaminidase; a marker of renal tubular injury) were significantly inhibited following EGb 761 administration. EGb 761 supplementation significantly prevented the generation of TBARS (thiobarbituric acid-reacting substances) with a marked improvement in terms of GSH content and activity of antioxidant enzymes in the kidney homogenate. Moreover, EGb 761 treatment significantly reduced both renal-tissue and urine total NO (nitric oxide) levels. The results suggest that the protective potential of EGb 761 in the prevention of adriamycin-induced hyperlipidaemic nephrotoxicity in rats was associated with the decrease in the oxidative stress and the total NO levels of renal tissues. Likewise, the present study demonstrates the ability of EGb 761 to reduce the hyperlipidaemia and proteinuria associated with this nephropathy, which might be beneficial to enhance the therapeutic index of adriamycin.


Subject(s)
Doxorubicin/toxicity , Ginkgo biloba/chemistry , Hyperlipidemias/chemically induced , Kidney/drug effects , Nitric Oxide/biosynthesis , Plant Extracts/pharmacology , Animals , Kidney/physiopathology , Kidney Function Tests , Male , Rats , Rats, Sprague-Dawley , Thiobarbituric Acid Reactive Substances/metabolism
15.
Food Chem Toxicol ; 45(1): 88-92, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17011106

ABSTRACT

Thymoquione (TQ), the main constituent of the volatile oil of Nigella sativa seeds, has been shown to protect mice against benzo(a)pyrene [B(a)P]-induced forestomach carcinogenesis. The present investigation was undertaken to study the possible chemopreventive activity of TQ, supplemented in the drinking water, against B(a)P-induced chromosomal aberrations (CAs) in mouse bone marrow cells. Male Swiss albino mice received TQ (0.01% in drinking water) daily for 28 days. The daily dose of TQ was estimated to be 10mg/kg based on the calculated average daily water consumption by mice. From day 9, the carcinogen, B(a)P, was given by gastric intubation at dose level of 50mg/kg on alternative days for a total of 8 doses. On day 29, all mice were transferred to a normal drinking tap water. Control groups received corn oil vehicle, TQ alone or B(a)P alone. All mice were sacrificed at 12 weeks after the end of the treatment. Chromosome preparations were made of bone marrow. Cytogenetic end points screened were the frequencies of CAs and damaged cells induced. Daily intake of TQ after and before or during exposure to B(a)P significantly reduced the frequencies of CAs and damaged cells compared to the highly clastogenic activity of B(a)P alone.


Subject(s)
Antimutagenic Agents/pharmacology , Benzo(a)pyrene/toxicity , Benzoquinones/pharmacology , Chromosome Aberrations/drug effects , Mutagens/toxicity , Nigella/chemistry , Administration, Oral , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Chemoprevention , Chromosome Aberrations/chemically induced , Male , Mice , Seeds/chemistry , Water Supply
16.
J Egypt Natl Canc Inst ; 17(4): 291-300, 2005 Dec.
Article in English | MEDLINE | ID: mdl-17102822

ABSTRACT

BACKGROUND: Doxorubicin is one of the most active cytotoxic agents in current use. It has proven efficacy in various malignancies either alone or combined with other cytocidal agents. The clinical usefulness of the anthracycline drug has been precluded by cardiac toxicity. Many therapeutic interventions have been attempted to improve the therapeutic benefits of the drug. Few, however, have been efficacious in this setting. PURPOSE: We have addressed in the current study the possible protective effects of naringenin, a flavonoid known to have anti-oxidant properties, on doxorubicin-induced cardiac toxicity in male Swiss albino rats. METHODS: Forty male Swiss albino rats were used in this study. Naringenin (25 mg/kg body weight) was administered daily by gavage for 7 consecutive days before a cumulative single dose of doxorubicin (15 mg/kg body weight, ip). RESULTS: Doxorubicin induced marked biochemical alterations characteristic of cardiac toxicity including, elevated activities of serum total lactate dehydrogenase (LDH) and creatine phosphokinase (CPK), enhanced lipid peroxidation measured as malondialdehyde (MDA). The anthracycline drug has also reduced the cardiac enzymatic activities of superoxide dismutase (SOD), glutathione-S-transferase (GST) and catalase (CAT). Besides, it reduced significantly the reduced glutathione (GSH) level, but it increased the total NO content in heart tissue. Prior administration of naringenin ahead of doxorubicin challenge ameliorated all these biochemical markers. CONCLUSIONS: Taken together, one could conclude that naringenin has a protective role in the abatement of doxorubicin-induced cardiac toxicity that resides, at least in part, on its anti-radical effects and regulatory role on NO production.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Antioxidants/therapeutic use , Doxorubicin/adverse effects , Flavanones/therapeutic use , Heart Diseases/chemically induced , Heart Diseases/prevention & control , Animals , Catalase/drug effects , Catalase/metabolism , Creatine Kinase/blood , Creatine Kinase/drug effects , Glutathione Transferase/drug effects , Glutathione Transferase/metabolism , L-Lactate Dehydrogenase/blood , L-Lactate Dehydrogenase/drug effects , Lipid Peroxidation/drug effects , Male , Nitric Oxide/metabolism , Rats , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...