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










Publication year range
1.
Phytomedicine ; 57: 84-94, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30668326

ABSTRACT

BACKGROUND: In December 2016, WHO released a report stating that in 2015 there were 383,000 deaths caused by asthma and 235 million people suffering from asthma. As there are many adverse effects associated with the currently-used asthma drugs, new anti-asthmatic drugs need to be developed. PURPOSE: In order to find new drug candidates with safe and low side effects, the anti-asthmatic function and mechanism of C. japonica oil were evaluated, and its active ingredients were analyzed for use in an ovalbumin asthma murine model. STUDY DESIGN AND METHODS: The study consisted of six groups: control; ovalbumin group; and dexamethasone group as a positive control; and 10, 100, and 500 mg/kg C. japonica oil treatment groups. In order to measure the anti-asthmatic effect of C. japonica oil, WBC and differential cell count in BALF, IgE in serum, morphological changes in pulmonary system, and gene and protein levels such as IFN-γ, IL-12p40, IL-4, IL-5, IL-6, TNF-α, and IL-6 were all evaluated. RESULTS: C. japonica oil had an anti-asthmatic effect and significantly controlled eosinophil in BALF, Th2-related factors such as GATA-3 that is Th2 cell transcription factor, IL-4, IL-5, and IL-13, and TNF-α in the lung. It also dose-dependently modulated inflammatory cells, T-bet, IL-12p40, and IL-6. Oleci acid was the major gradient (52.89%) in C. japonica oil and also had anti-asthmatic effects such as the downregulation of inflammatory cells, WBC, and eosinophil in BALF, IgE in serum, and morphological changes in the lung. CONCLUSION: We concluded that C. japonica oil is a new anti-asthmatic drug candidate and that oleic acid is the major anti-asthmatic ingredient in C. japonica oil.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Camellia/chemistry , Plant Oils/pharmacology , Animals , Anti-Asthmatic Agents/chemistry , Asthma/metabolism , Bronchoalveolar Lavage Fluid , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Eosinophils/drug effects , Female , GATA3 Transcription Factor/metabolism , Interleukin-4/metabolism , Lung/drug effects , Lung/metabolism , Mice, Inbred BALB C , Oleic Acid/analysis , Plant Oils/administration & dosage , Plant Oils/chemistry
2.
Toxicol Ind Health ; 28(3): 203-15, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21730038

ABSTRACT

As chronic exposure to welding fumes causes pulmonary diseases, such as pneumoconiosis, public concern has increased regarding continued exposure to these hazardous gases in the workplace. In a previous study, the inflammatory response to welding fume exposure was analysed in rat lungs in the case of recurrent exposure and recovery periods. Thus using lung samples, well-annotated by histological observation and biochemical analysis, this study examines the gene expression profiles to identify phenotype-anchored genes corresponding to lung inflammation and the repair phenomenon after recurrent welding fume exposure. Seven genes (Mmp12, Cd5l, LOC50101, LOC69183, Spp1, and Slc26a4) were found to be significantly up-regulated according to the severity of the lung injury. In addition, the transcription and translation of Trem2, which was up-regulated in response to the repair process, were validated using a real-time polymerase chain reaction, Western blotting, and immunohistochemistry. The differentially expressed genes in the exposure and recovery groups were also classified using k-means and hierarchical clustering, plus their toxicological function and canonical pathways were further analysed using Ingenuity Pathways Analysis Software. As a result, this comprehensive and integrative analysis of the transcriptional changes that occur during repeated exposure provides important information on the inflammation and repair processes after welding-fume-induced lung injury.


Subject(s)
Air Pollutants, Occupational/toxicity , Inhalation Exposure/analysis , Lung Injury/chemically induced , Transcriptome , Welding , Analysis of Variance , Animals , Blotting, Western , Bronchoalveolar Lavage Fluid/chemistry , Cluster Analysis , Gene Expression Profiling , Immunohistochemistry , Lung/chemistry , Lung/drug effects , Lung Injury/immunology , Lung Injury/metabolism , Male , Rats , Rats, Sprague-Dawley , Signal Transduction , Up-Regulation
3.
Arch Pharm Res ; 33(5): 769-73, 2010 May.
Article in English | MEDLINE | ID: mdl-20512476

ABSTRACT

Lengthening of the Q-T interval and proarrhythmia are adverse effects associated with antimalarial agents. Also, lengthening of the Q-T interval is a definite outcome when patients are administered with an overdose of primaquine. Inhibition of potassium current I (Kr) and resultant QT prolongation is suggested as the reason behind drug-induced arrhythmias. The present study investigated the molecular mechanisms of voltage-dependent inhibition of human Ether-a-go-go Related Gene (hERG) delayed rectifier K(+) channels expressed in HEK-293 cells by primaquine. Primaquine inhibited hERG current in a concentration-dependent manner with the half-maximal inhibitory concentration (IC(50)) of 21.5 microM. The voltage-dependent inhibition of hERG current resulted in the activation curve to be shifted to a negative voltage after primaquine exposure in a dose-dependent manner. Blockade of hERG by primaquine was also found to be time-dependent, occurring rather rapidly. Blockade of wild-type hERG channel by primaquine was similar to those of both the S6 residue hERG mutants (F656A and Y652A) and the pore region mutants (T623A). In conclusion, these results indicate that primaquine preferentially inhibits the hERG potassium channel, but blockade of hERG channel by primaquine may not be related to the S6 residue or the pore region, but may be induced through other pathways such as binding other region or effect by drug binding receptor which indicates a need for further exploration.


Subject(s)
Antimalarials/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Primaquine/pharmacology , Cell Line , Dose-Response Relationship, Drug , Ether-A-Go-Go Potassium Channels/genetics , Humans , Mutagenesis, Site-Directed , Patch-Clamp Techniques/methods , Time Factors
4.
Int J Radiat Oncol Biol Phys ; 77(3): 858-66, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20510196

ABSTRACT

PURPOSE: Despite the widespread use of radiotherapy as a local and regional modality for the treatment of cancer, some non-small-cell lung cancers commonly develop resistance to radiation. We thus sought to clarify the molecular mechanisms underlying resistance to radiation. METHODS AND MATERIALS: We established the radioresistant cell line H460R from radiosensitive parental H460 cells. To identify the radioresistance-related genes, we performed microarray analysis and selected several candidate genes. RESULTS: Clonogenic and MTT assays showed that H460R was 10-fold more resistant to radiation than H460. Microarray analysis indicated that the expression levels of 1,463 genes were altered more than 1.5-fold in H460R compared with parental H460. To evaluate the putative functional role, we selected one interesting gene tumor protein p53-inducible protein 3 (TP53I3), because that this gene was significantly downregulated in radioresistant H460R cells and that it was predicted to link p53-dependent cell death signaling. Interestingly, messenger ribonucleic acid expression of TP53I3 differed in X-ray-irradiated H460 and H460R cells, and overexpression of TP53I3 significantly affected the cellular radiosensitivity of H460R cells. CONCLUSIONS: These results show that H460R may be useful in searching for candidate genes that are responsible for radioresistance and elucidating the molecular mechanism of radioresistance.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Gene Expression Profiling , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Radiation Tolerance/genetics , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Line, Tumor , Genetic Markers , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/radiotherapy , Proto-Oncogene Proteins/metabolism
5.
Arch Toxicol ; 84(8): 609-18, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20221588

ABSTRACT

We investigated the effects of astemizole, a second-generation antihistamine, on the heart and peripheral blood mononuclear cells (PBMCs) and identified the early markers of its cardiotoxicity using gene expression profiling. Astemizole causes torsades de pointes, which is a type of ventricular tachycardia. We administered astemizole (dosage: 20, 60 mg/kg) to male Sprague-Dawley rats, using an oral gavage. Cardiac tissue and PBMCs were collected from the rats 4 h after treatment. Gene expression profiles were obtained using an Affymetrix GeneChip. The most deregulated genes were associated with energy metabolism pathways and calcium ion homeostasis in the heart of astemizole-treated rats. The most altered genes in the PBMCs were those involved in developmental processes and cardiotoxicity. Genes related to the response to oxidative stress, reactive oxygen species, heat shock proteins, hypoxia, immunity, and inflammation were also deregulated in the heart and PBMCs. These data provide further insight into the genetic pathways affected by astemizole. In addition, the simultaneously deregulated genes identified herein may be further studied. It will be interesting to find out whether single genes or certain sets of these genes could finally serve as biomarkers for cardiotoxicity of astemizole or other similar antihistamine drugs.


Subject(s)
Astemizole/toxicity , Gene Expression/drug effects , Heart/drug effects , Histamine H1 Antagonists, Non-Sedating/toxicity , Leukocytes, Mononuclear/drug effects , Animals , Gene Expression Profiling , Gene Expression Regulation/drug effects , Leukocytes, Mononuclear/metabolism , Male , Myocardium/metabolism , Rats , Rats, Sprague-Dawley
6.
Int J Cardiol ; 145(2): 272-274, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-19913309

ABSTRACT

KCNH-2 and KCNE-2 may encode the channel-forming alpha- and regulatory beta-subunits, respectively, of I(Kr) channels, which are involved in inherited or acquired long QT syndrome. However, in contrast to other multimeric channels, the stoichiometry of KCNH-2 and KCNE-2, which should be reasonably maintained if they are to be accepted as the components of a multi-molecular complex, has not been established, yet. In this study, we found that the protein expression of KCNE-2 was adequate to support the formation of a complex with KCNH-2; however, the level of transcription was not. This finding, together with previous data from electrophysiological and molecular biological studies, supports that KCNH-2 and KCNE-2 are molecular components of I(Kr) channels.


Subject(s)
Ether-A-Go-Go Potassium Channels/chemistry , Potassium Channels, Voltage-Gated/chemistry , Animals , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/biosynthesis , Potassium Channels, Voltage-Gated/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/chemistry , Rats , Rats, Sprague-Dawley , Stereoisomerism
7.
Toxicol Res ; 26(2): 137-47, 2010 Jun.
Article in English | MEDLINE | ID: mdl-24278517

ABSTRACT

Pulmonary fibrosis is a common consequence of many lung diseases and a leading cause of morbidity and mortality. The molecular mechanisms underlying the development of pulmonary fibrosis remain poorly understood. One model used successfully to study pulmonary fibrosis over the past few decades is the bleomycin-induced pulmonary fibrosis model. We aimed to identify the genes associated with fibrogenesis using an Affymetrix GeneChip system in a bleomycin-induced rat model for pulmonary fibrosis. To confirm fibrosis development, several analyses were performed, including cellular evaluations using bronchoalveolar lavage fluid, measurement of lactate dehydrogenase activity, and histopathological examinations. Common aspects of pulmonary fibrosis such as prolonged inflammation, immune cell infiltration, emergence of fibroblasts, and deposition of extracellular matrix and connective tissue elements were observed. Global gene expression analysis revealed significantly altered expression of genes (≥ 1.5-fold, p < 0.05.) in a time-dependent manner during the development of pulmonary fibrosis. Our results are consistent with previous results of well-documented gene expression. Interestingly, the expression of triggering receptor expressed on myeloid cells 2 (Trem2) , secreted phosphoprotein 1 (Spp1) , and several proteases such as Tpsab1, Mcpt1, and Cma1 was considerably induced in the lung after bleomycin treatment, despite little evidence that they are involved in pulmonary fibrogenesis. These data will aid in our understanding of fibrogenic mechanisms and contribute to the identification of candidate biomarkers of fibrotic disease development.

8.
Toxicol Res ; 25(2): 85-92, 2009 Jun.
Article in English | MEDLINE | ID: mdl-32038824

ABSTRACT

4,4'-Methylenedianiline (MDA) is an aromatic amine that is widely used in the industrial synthetic process. Genotoxic MDA forms DNA adducts in the liver and is known to induce liver damage in human and rats. To elucidate the molecular mechanisms associated with MDA-induced hepatotoxicity, we have identified genes differentially expressed by microarray approach. BALB/c male mice were treated once daily with MDA (20 mg/kg) up to 7 days via intraperitoneal injection (i.p.) and hepatic damages were revealed by histopathological observation and elevation of serum marker enzymes such as AST, ALT, ALP, cholesterol, DBIL, and TBIL. Microarray analysis showed that 952 genes were differentially expressed in the liver of MDA-treated mice and their biological functions and canonical pathways were further analyzed using Ingenuity Pathways Analysis (IPA). Toxicological functional analysis showed that genes related to hepatotoxicity such hyperplasia/hyperproliferation (Timpl), necrosis/cell death (Cd14, Mt1f, Timpl, and Pmaipl), hemorrhaging (Mt1f), cholestasis (Akr1c3, Hpx, and Slc10a2), and inflammation (Cd14 and Hpx) were differentially expressed in MDA-treated group. This gene expression profiling should be useful for elucidating the genetic events associated with aromatic amine-induced hepatotoxicity and for discovering the potential biomarkers for hepatotoxicity.

9.
J Pharmacol Sci ; 99(1): 117-20, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16141634

ABSTRACT

Accumulating evidence indicates that growth hormone (GH) might be effective at preventing the development of Alzheimer's disease. However, exogenous GH treatment has exhibited side effects for clinical application; thus supplementation with amino acids to promote the release of GH could be a possible alternative treatment. In this study, mice that were fed with a diet of GH-releasing supplements had significantly attenuated memory impairments and hippocampal changes in the acetylcholinesterase activity and acetylcholine level induced by amyloid beta protein (Abeta) (1 - 42). Our results suggest that the use of GH-releasing supplement exerts beneficial effects on the memory impairment induced by Abeta (1 - 42).


Subject(s)
Aging/physiology , Amino Acids/pharmacology , Amyloid beta-Peptides/antagonists & inhibitors , Growth Hormone/blood , Memory Disorders/prevention & control , Memory/drug effects , Peptide Fragments/antagonists & inhibitors , Acetylcholine/metabolism , Acetylcholinesterase/metabolism , Amino Acids/administration & dosage , Animals , Brain/drug effects , Brain/enzymology , Brain Chemistry , Growth Hormone/pharmacology , Male , Memory Disorders/chemically induced , Mice , Mice, Inbred C57BL , Time Factors
10.
J Med Primatol ; 34(2): 96-100, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15860116

ABSTRACT

The effects of ketamine anesthesia on both hematological and serum biochemical variables were investigated in 19 male and 15 female cynomolgus monkeys. Blood samples were obtained from the cephalic vein within 30 minutes of an intramuscular injection of ketamine hydrochloride (10 mg/kg). Ketamine anesthesia caused a reduction in leukocyte counts and a significant reduction in lymphocytes percentages. Ketamine anesthesia also increased the serum activities of aspartate aminotransferase (AST), alanine aminotransferase (ALT) and creatine phosphokinase (CPK), but reduced the serum concentrations of glucose, inorganic phosphate, sodium and potassium. The alterations of hematological and serum biochemical values will be discussed. These alterations should be considered when designing studies for and interpreting data from cynomolgus monkeys.


Subject(s)
Anesthesia/veterinary , Anesthetics, Dissociative/adverse effects , Ketamine/adverse effects , Macaca fascicularis/blood , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Blood Glucose/analysis , Creatine Kinase/blood , Female , Leukocyte Count , Lymphocyte Count , Male , Phosphates/blood , Potassium/blood , Sodium/blood
11.
FASEB J ; 19(7): 783-5, 2005 May.
Article in English | MEDLINE | ID: mdl-15758042

ABSTRACT

The PMCA (protein misfolding cyclic amplification) technique has been shown to drive the amplification of misfolded prion protein by PrP(Sc) seeds during several cycles of incubation-sonication. Here, we report that cyclic amplification of normal hamster brain homogenates treated with a number of transition metals (manganese [Mn], copper [Cu], and iron [Fe]) leads to conversion of PrP(C) into protease-resistant PrP(res). The efficiency of PrP(res) formation and the glycoforms induced by Mn were different from those obtained by Cu and Fe. Previous results have shown higher Mn and lower Cu levels in the affinity-purified PrP(Sc) from the brain of prion diseases compared with normal hamster brain homogenates. We focused on Mn because we observed higher levels of Mn in whole brain, mitochondria, and scrapie-associated fibril-enriched fractions from the brains of animals with prion disease. In the presence of minute quantities of Mn-induced PrP(res) template with a large amount of PrP(C), PrP(res) amplification is observed. A metal chelater, EDTA reverses the effect of Mn on PrP(res) amplification, suggesting that Mn may play a role in the formation of PrP(res). It has been proposed that metal-catalyzed oxidation of PrP leads to the oxidation of amino acids and extensive aggregation of oxidized PrP. Carboxyl acids such as deoxycholic acid (DA) are oxidized molecules produced by 3' oxidation pathway. In in vitro studies, the potent effect of Mn on PrP(res) amplification is augmented by DA in a dose-dependent manner. On the basis of the evidence of the elevated Mn levels in scrapie-associated fibril (SAF)-enriched preparations from the brains of animals with prion disease, Mn-loaded PrP and oxidized molecules such as carboxyl acids may contribute to the formation of the scrapie isoform of PrP in prion diseases.


Subject(s)
Brain/metabolism , Deoxycholic Acid/pharmacology , Peptide Hydrolases/metabolism , Prions/metabolism , Transition Elements/pharmacology , Animals , Brain/drug effects , Brain Chemistry , Chelating Agents/pharmacology , Copper/pharmacology , Cricetinae , Edetic Acid/pharmacology , Endopeptidase K/pharmacology , Iron/pharmacology , Male , Manganese/analysis , Manganese/pharmacology , Mesocricetus , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/pharmacology , Prions/chemistry , Protein Folding , Scrapie/metabolism
12.
Biol Pharm Bull ; 27(5): 723-6, 2004 May.
Article in English | MEDLINE | ID: mdl-15133254

ABSTRACT

Oxidative stress caused by an elevation in reactive oxygen species (ROS) plays an important role in Alzheimer's disease and other neurodegenerative diseases. In this study, we examined the neuroprotective effect of danthron (1,8-dihydroxyanthraquinone) against neurotoxicities induced by beta-amyloid (25-35), excitotoxins, apoptosis, and oxidative stress in primary cortical cultures. Danthron dose-dependently reduced neuronal injury induced by 30 microM beta-amyloid (25-35). Danthron significantly inhibited oxidative injury induced by 100 microM Fe(3+) and decreased membrane lipid peroxidation induced by 100 microM Fe(3+) as measured by thiobarbituric-acid-reactive substance (TBARS). Danthron (0.5-50 microM) ameliorated the effects of buthionine sulfoximine (BSO, 1 mM), which depletes endogenous glutathione by 10-73%. Danthron also dose-dependently inhibited neuronal injury mediated by nitric oxide (NO) radicals, but failed to inhibit injury due to superoxide radicals (O(2-)). These results suggest danthron treatment may, in part, reduce neurotoxicity related to beta-amyloid protein by both dominant inhibitory effects on membrane lipid peroxidation and glutathione deprivation.


Subject(s)
Amyloid beta-Peptides/toxicity , Anthraquinones/pharmacology , Cerebral Cortex/drug effects , Oxidative Stress/drug effects , Peptide Fragments/toxicity , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Dose-Response Relationship, Drug , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Mice , Mice, Inbred ICR , Oxidative Stress/physiology , Peptide Fragments/antagonists & inhibitors
13.
Exp Anim ; 53(1): 27-30, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14993737

ABSTRACT

The chronic hepatotoxic effects of carbon tetrachloride (CCl(4)) in heat-shock protein (HSP) 70 knock out (HSP70-/-) mice were examined. After repeated intraperitoneal injections of CCl(4) for six weeks, the level of ALT and weight ratio of the liver to body were lower in HSP70-/- mice than in the control (WT) mice. The levels of HSP25 and HSP47 were lowered in HSP70-/- mice as compared with WT mice. The grades of hepatic necrosis and neutrophil infiltration were not significantly different between HSP70-/- and WT mice. The collagen content was not affected significantly by CCl(4) treatment.


Subject(s)
Alanine Transaminase/blood , Carbon Tetrachloride/toxicity , HSP70 Heat-Shock Proteins/genetics , Liver/drug effects , Animals , Blotting, Western , Body Weights and Measures , Electrophoresis, Polyacrylamide Gel , HSP47 Heat-Shock Proteins , Heat-Shock Proteins/metabolism , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Molecular Chaperones , Neoplasm Proteins/metabolism , Neutrophil Infiltration , Neutrophils/pathology
14.
Neurosci Lett ; 355(3): 231-5, 2004 Jan 30.
Article in English | MEDLINE | ID: mdl-14732473

ABSTRACT

We examined the effects of the antioxidant propolis on seizures induced by kainic acid (KA). Sprague-Dawley rats received propolis (75 and 150 mg/kg, p.o.) five times at 12 h intervals. KA (10 mg/kg, i.p.) was injected 1 h after the last propolis treatment. Pretreatment with propolis significantly attenuated KA-induced seizures and KA-induced increases in hippocampal AP-1 DNA binding activity in a dose-dependent manner. KA induced increases in the levels of malondialdehyde and protein carbonyl, and a decrease in the ratio of GSH/GSSG. These oxidative stresses and neuronal degenerations were significantly attenuated by pretreatment with propolis. The neuroprotective effects of propolis appeared to be counteracted by adenosine receptor antagonists [A1 antagonist, 8-cyclopentyl-1,3-dimethylxanthine (25 or 50 microg/kg); A2A antagonist, 1,3,7-trimethyl-8-(3-chlorostyryl)xanthine (0.5 or 1 mg/kg); and A2B antagonist, alloxazine (1.5 or 3.0 mg/kg)]. However, this counteraction was most pronounced in the presence of the A1 antagonist. Our results suggest that the protective effect of propolis against KA-induced neurotoxic oxidative damage is, at least in part, via adenosine A1 receptor modulation.


Subject(s)
Antioxidants/pharmacology , Kainic Acid/toxicity , Propolis/pharmacology , Receptor, Adenosine A1/physiology , Adenosine A1 Receptor Antagonists , Animals , Flavins/pharmacology , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiology , Kainic Acid/antagonists & inhibitors , Male , Rats , Rats, Sprague-Dawley
15.
Exp Anim ; 52(5): 371-5, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14625400

ABSTRACT

Agaricus blazei (H1 strain) was tested for its anticancer activity using a sarcoma 180 (S180) inoculation model and the changing patterns of splenocyte subsets were examined. Its hot-water extract was administered orally to ICR and KSN nude mice that were inoculated with S180. The growth of S180 was significantly inhibited in A.blazei treated groups. Pan T cells significantly increased in all treated groups compared to controls, even in KSN nude mice. Splenocyte subset changes were slightly different between ICR and KSN nude mice. This S180 inoculation model proved to be effective in screening the antitumor effect of basidiomycetes and allowed comparisons of immunological cellular changes between the mouse strains.


Subject(s)
Agaricus , Phytotherapy , Plant Extracts/administration & dosage , Sarcoma 180/drug therapy , Administration, Oral , Animals , Disease Models, Animal , Lymphocyte Subsets , Male , Mice , Mice, Inbred ICR , Mice, Nude , Neoplasm Transplantation , Sarcoma 180/immunology , Sarcoma 180/pathology , Spleen/immunology , Water
16.
Biol Pharm Bull ; 26(4): 544-6, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12673040

ABSTRACT

The possible role of quercetin, a naturally occurring plant flavonoid, in protecting against oxygen-glucose deprivation (OGD)-, excitotoxins-, and free radical-induced neuronal injury in mouse cortical cell cultures was investigated. Pre- and co-treatment with quercetin (100 microM) inhibited 50 min OGD-, 20 microM N-methyl-D-aspartate (NMDA)-, and 50 microM kainate-induced neurotoxicity by 36, 22, and 61%, respectively. Quercetin significantly ameliorated free radical-induced neuronal injury caused by buthionine sulfoximine, sodium nitroprusside, ZnCl(2), and FeCl(2). These results suggest that quercetin may contribute a neuroprotective action against ischemic neural injury, partially via antioxidant actions.


Subject(s)
Cerebral Cortex/drug effects , Excitatory Amino Acid Agonists/toxicity , Glucose/deficiency , Oxygen/metabolism , Quercetin/pharmacology , Animals , Antioxidants/pharmacology , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cells, Cultured , Cerebral Cortex/pathology , Cerebral Cortex/physiology , Dose-Response Relationship, Drug , Kainic Acid/toxicity , Mice , Mice, Inbred ICR , N-Methylaspartate/toxicity , Neuroglia/drug effects , Neuroglia/pathology , Neuroglia/physiology , Neurons/drug effects , Neurons/pathology , Neurons/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...