Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters











Publication year range
1.
Psychiatry Res ; 281: 112577, 2019 11.
Article in English | MEDLINE | ID: mdl-31586841

ABSTRACT

Sleep plays a critical role in body health maintenance, whereas sleep deprivation (SD) negatively affects cognitive function. Cognitive defects mainly memory impairment resulting from sleep deprivation were related to an increase in the level of oxidative stress in the body, including the brain hippocampus region. Edaravone is a potent free radical scavenger having antioxidant effect. In the current study, edaravone's ability to prevent SD induced cognitive impairment was tested in rats. Animals were sleep deprived 8 h/day for 4 weeks. Concurrently, edaravone was administrated intraperitoneally for four weeks. Animals performance during cognitive testing was evaluated to display if edaravone has a role in the prevention of sleep deprivation induced memory impairment. Additionally, the role of antioxidant biomarkers glutathione peroxidase (GPx), catalase, glutathione (GSH), oxidized glutathione (GSSG), GSH/GSSG in this effect was investigated. The results showed that SD impaired both short- and long- term memories, and chronic edaravone administration prevented such effect. Additionally, edaravone prevented decreases in hippocampal GPx, catalase, GSH/GSSG ratio and normalized increases in GSSG levels, which were impaired by SD model. In conclusion, current result showed a protective effect of edaravone administration against SD induction that could be related to edaravone's ability to normalizing mechanisms related to oxidative balance.


Subject(s)
Antioxidants/pharmacology , Edaravone/pharmacology , Memory Disorders/prevention & control , Neuroprotective Agents/pharmacology , Sleep Deprivation/psychology , Animals , Catalase/drug effects , Glutathione/drug effects , Glutathione Disulfide/drug effects , Glutathione Peroxidase/drug effects , Hippocampus/metabolism , Male , Maze Learning/drug effects , Memory/drug effects , Memory, Long-Term/drug effects , Oxidative Stress/drug effects , Rats , Rats, Wistar
2.
Neurochem Res ; 44(5): 1167-1181, 2019 May.
Article in English | MEDLINE | ID: mdl-30806880

ABSTRACT

Menadione (2-methyl-1,4-naphthoquinone) is a synthetic derivative of vitamin K that allows rapid redox cycling in cells and thereby generates reactive oxygen species (ROS). To test for the consequences of a treatment of brain astrocytes with menadione, we incubated primary astrocyte cultures with this compound. Incubation with menadione in concentrations of up to 30 µM did not affect cell viability. In contrast, exposure of astrocytes to 100 µM menadione caused a time-dependent impairment of cellular metabolism and cell functions as demonstrated by impaired glycolytic lactate production and strong increases in the activity of extracellular lactate dehydrogenase and in the number of propidium iodide-positive cells within 4 h of incubation. In addition, already 5 min after exposure of astrocytes to menadione a concentration-dependent increase in the number of ROS-positive cells as well as a concentration-dependent and transient accumulation of cellular glutathione disulfide (GSSG) were observed. The rapid intracellular GSSG accumulation was followed by an export of GSSG that was prevented in the presence of MK571, an inhibitor of the multidrug resistance protein 1 (Mrp1). Menadione-induced glutathione (GSH) oxidation and ROS formation were found accelerated after glucose-deprivation, while the presence of dicoumarol, an inhibitor of the menadione-reducing enzyme NQO1, did not affect the menadione-dependent GSSG accumulation. Our study demonstrates that menadione rapidly depletes cultured astrocytes of GSH via ROS-induced oxidation to GSSG that is subsequently exported via Mrp1.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , Astrocytes/drug effects , Brain/drug effects , Glutathione Disulfide/drug effects , Vitamin K 3/pharmacology , Animals , Astrocytes/metabolism , Brain/metabolism , Cell Survival/drug effects , Cells, Cultured , Glutathione/drug effects , Glutathione/metabolism , Glutathione Disulfide/metabolism , Glycolysis/drug effects , Hydrogen Peroxide/metabolism , Oxidation-Reduction , Rats, Wistar
3.
Psychiatry Res ; 270: 430-437, 2018 12.
Article in English | MEDLINE | ID: mdl-30316170

ABSTRACT

Post-traumatic stress disorder (PTSD) may occur after exposure to stressful, fearful or troubling events. Until now, there is no curable medication for this disorder. Cerebrolysin is a neuropeptide, which has an important role in the treatment of vascular dementia. In this study, the probable protective effect of cerebrolysin on PTSD-induced memory impairment was investigated. To induce PTSD, the single prolonged stress (SPS) model was used. Rats were allocated into four groups: control (vehicle-treated), CBL (administrated cerebrolysin 2.5 ml/kg by intraperitoneal route for 4 weeks), SPS (as a model of PTSD and administered vehicle), and CBL-SPS (exposed to SPS and administered cerebrolysin for 4 weeks). Learning and memory were assessed using the radial arm water maze (RAWM). Results showed that SPS impaired both short- and long- term memories; and chronic cerebrolysin administration prevented such effect. Cerebrolysin also prevented decreases in hippocampal GSH levels and GSH/GSSG ratios, and increased GSSG and TBARs, levels induced by PTSD. In conclusion, a protective effect of cerebrolysin administration against SPS model of PTSD induced short- and long- term memory impairment was characterized. This protection could be accomplished, at least partly, by prevention of PTSD induced increase in oxidative stress in the hippocampus via the use of cerebrolysin.


Subject(s)
Amino Acids/pharmacology , Hippocampus/drug effects , Memory Disorders/prevention & control , Memory/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Stress Disorders, Post-Traumatic/psychology , Stress, Psychological/psychology , Animals , Disease Models, Animal , Fear , Glutathione/drug effects , Glutathione/metabolism , Glutathione Disulfide/drug effects , Glutathione Disulfide/metabolism , Hippocampus/metabolism , Male , Maze Learning/drug effects , Random Allocation , Rats , Rats, Sprague-Dawley , Restraint, Physical , Swimming , Thiobarbituric Acid Reactive Substances/metabolism
4.
Sci Rep ; 7(1): 13828, 2017 10 23.
Article in English | MEDLINE | ID: mdl-29062059

ABSTRACT

Cold atmospheric pressure plasmas are gaining increased interest in the medical sector and clinical trials to treat skin diseases are underway. Plasmas are capable of producing several reactive oxygen and nitrogen species (RONS). However, there are open questions how plasma-generated RONS interact on a molecular level in a biological environment, e.g. cells or cell components. The redox pair glutathione (GSH) and glutathione disulphide (GSSG) forms the most important redox buffer in organisms responsible for detoxification of intracellular reactive species. We apply Raman spectroscopy, mass spectrometry, and molecular dynamics simulations to identify the time-dependent chemical modifications on GSH and GSSG that are caused by dielectric barrier discharge under ambient conditions. We find GSSG, S-oxidised glutathione species, and S-nitrosoglutathione as oxidation products with the latter two being the final products, while glutathione sulphenic acid, glutathione sulphinic acid, and GSSG are rather reaction intermediates. Experiments using stabilized pH conditions revealed the same main oxidation products as were found in unbuffered solution, indicating that the dominant oxidative or nitrosative reactions are not influenced by acidic pH. For more complex systems these results indicate that too long treatment times can cause difficult-to-handle modifications to the cellular redox buffer which can impair proper cellular function.


Subject(s)
Glutathione Disulfide/chemistry , Glutathione/chemistry , Plasma Gases/pharmacology , Glutathione/analysis , Glutathione/drug effects , Glutathione Disulfide/analysis , Glutathione Disulfide/drug effects , Humans , Mass Spectrometry , Oxidation-Reduction , Oxidative Stress , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism
5.
Thyroid ; 27(2): 271-278, 2017 02.
Article in English | MEDLINE | ID: mdl-27824294

ABSTRACT

BACKGROUND: A recent clinical trial has shown a beneficial effect of the antioxidant agent selenium in Graves' orbitopathy (GO). In order to shed light on the cellular mechanisms on which selenium may act, this study investigated its effects in cultured orbital fibroblasts. METHODS: Primary cultures of orbital fibroblasts from six GO patients and six control subjects were established. Cells were treated with H2O2 to induce oxidative stress, after pre-incubation with selenium-(methyl)selenocysteine (SeMCys). The following assays were performed: glutathione disulfide (GSSG), as a measure of oxidative stress, glutathione peroxidase (GPX) activity, cell proliferation, hyaluronic acid (HA), and pro-inflammatory cytokines. RESULTS: H2O2 induced an increase in cell GSSG and fibroblast proliferation, which were reduced by SeMCys. Incubation of H2O2-treated cells with SeMCys was followed by an increase in glutathione peroxidase activity, one of the antioxidant enzymes into which selenium is incorporated. At the concentrations used (5 µM), H2O2 did not significantly affect HA release, but it was reduced by SeMCys. H2O2 determined an increase in endogenous cytokines involved in the response to oxidative stress and GO pathogenesis, namely tumor necrosis factor alpha, interleukin 1 beta, and interferon gamma. The increases in tumor necrosis factor alpha and interferon gamma were blocked by SeMCys. While the effects of SeMCys on oxidative stress and cytokines were similar in GO and control fibroblasts, they were exclusive to GO fibroblasts in terms of inhibiting proliferation and HA secretion. CONCLUSIONS: Selenium, in the form of SeMCys, abolishes some of the effects of oxidative stress in orbital fibroblasts, namely increased proliferation and secretion of pro-inflammatory cytokines. SeMCys reduces HA release in GO fibroblasts in a manner that seems at least in part independent from H2O2-induced oxidative stress. Some effects of SeMCys are specific for GO fibroblasts. These findings reveal some cellular mechanisms by which selenium may act in patients with GO.


Subject(s)
Antioxidants/pharmacology , Fibroblasts/drug effects , Graves Ophthalmopathy/drug therapy , Oxidative Stress/drug effects , Selenium/pharmacology , Selenocysteine/analogs & derivatives , Aged , Case-Control Studies , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/drug effects , Cytokines/metabolism , Female , Glutathione Disulfide/drug effects , Glutathione Disulfide/metabolism , Glutathione Peroxidase/drug effects , Glutathione Peroxidase/metabolism , Graves Ophthalmopathy/metabolism , Humans , Hyaluronic Acid/metabolism , Hydrogen Peroxide/pharmacology , Male , Middle Aged , Orbit/cytology , Oxidants/pharmacology , Selenocysteine/pharmacology
6.
Inflamm Bowel Dis ; 17(8): 1674-84, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21744422

ABSTRACT

BACKGROUND: Intestinal subepithelial myofibroblasts (ISEMFs) produce inflammatory cytokines in response to certain stimuli. In the intestine of patients with Crohn's disease (CD), cytokine synthesis is modified and an increased number of myofibroblasts has been observed. The intracellular redox state influences cytokine production and oxidative stress is present in the intestinal mucosa of CD patients. METHODS: This study was performed in ISEMFs isolated from the colon of patients with active CD and in a myofibroblast cell line derived from human colonic mucosa: 18Co cells. Cellular glutathione (GSH) levels were modulated by treatment with buthionine sulfoximine, an inhibitor of GSH synthesis, or N-acetylcysteine, a GSH precursor. GSH and oxidized glutathione (GSSG) levels were measured by high-performance liquid chromatography (HPLC) methods. Interleukin (IL)-6 production was detected by enzyme-linked immunosorbent assay (ELISA). RESULTS: ISEMFs of CD patients exhibited an increased oxidative state due to a decrease in the GSH/GSSG ratio, which is related to an increase in basal IL-6 production or is stimulated by tumor necrosis factor alpha (TNFα) or bacterial products. This relationship was also confirmed in 18Co cells. Phosphorylation and activation of ERK1/2 and p38 MAPK, which are signaling factors involved in the IL-6 synthesis, were also increased when there is oxidative stress in ISEMFs. CONCLUSIONS: This study shows for the first time in ISEMFs of CD patients an increased production of IL-6 synthesis related to the decrease in the GSH/GSSH ratio, suggesting redox regulation with the involvement of specific kinase activation. The present data shed light on the pathogenesis of inflammatory chronic processes and relapses that occur in this pathology.


Subject(s)
Crohn Disease/metabolism , Glutathione Disulfide/metabolism , Glutathione/metabolism , Interleukin-6/metabolism , Myofibroblasts/metabolism , Oxidative Stress/physiology , Acetylcysteine/pharmacology , Adult , Buthionine Sulfoximine/pharmacology , Cell Line , Colon/metabolism , Colon/physiopathology , Crohn Disease/physiopathology , Female , Glutathione/drug effects , Glutathione Disulfide/drug effects , Humans , Ileum/metabolism , Ileum/physiopathology , Lipopolysaccharides/pharmacology , Male , Middle Aged , Mitogen-Activated Protein Kinase 3/metabolism , Myofibroblasts/drug effects , Myofibroblasts/physiology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Oxidative Stress/drug effects , Phosphorylation/drug effects , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
7.
J Physiol Biochem ; 67(1): 35-42, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20924736

ABSTRACT

Telomeres are specialized DNA-protein complexes found at the tips of linear chromosomes. In this study, we investigated the effects of oxidative stress on telomeric length distribution of proliferating vascular smooth muscle cells following balloon injury in single or combined treatment of rabbits with either buthionine sulfoximine or taurine. Exposure to oxidative stress increased the balloon injury whereas taurine treatment significantly diminished L-buthionine-sulfoximine-related intimal hyperplasia. Our results also showed that both variables had a significant influence on mean telomeric length distribution.


Subject(s)
Angioplasty, Balloon/adverse effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Oxidative Stress , Telomere/pathology , Vascular System Injuries/etiology , Animals , Arteries/anatomy & histology , Arteries/drug effects , Arteries/injuries , Atherosclerosis/pathology , Buthionine Sulfoximine/pharmacology , Cell Proliferation/drug effects , Glutathione/blood , Glutathione/drug effects , Glutathione Disulfide/blood , Glutathione Disulfide/drug effects , Glutathione Peroxidase/blood , Glutathione Peroxidase/drug effects , Hyperplasia/pathology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/injuries , Myocytes, Smooth Muscle/pathology , Rabbits , Taurine/pharmacology , Telomere/drug effects , Tunica Intima/drug effects , Tunica Intima/pathology , Vascular System Injuries/prevention & control
8.
Toxicol In Vitro ; 22(8): 1918-26, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18845235

ABSTRACT

The present study was conducted to investigate the role of arjunolic acid (AA) against sodium fluoride (NaF)-induced cytotoxicity and necrotic cell death in murine hepatocytes. Dose-dependent studies suggest that incubation of hepatocytes with NaF (100mM) for 1h significantly decreased the cell viability as well as intracellular antioxidant power. Besides, NaF administration increased the activities of the membrane leakage enzymes and accumulation of intracellular reactive oxygen species; decreased the activities of the antioxidant enzymes, the glutathione (GSH) and total thiol contents; and elevated the level of oxidised glutathione (GSSG), lipid peroxidation end products as well as protein carbonyl content. In addition to the oxidative impairments, fluoride exposure caused hepatic cell death mainly via the necrotic pathway as supported by the flowcytometric and DNA fragmentation analyses. Incubation with AA (100 microg/ml) both prior to and in combination with NaF almost normalized the altered activities of antioxidant indexes. AA treatment enhanced the cellular antioxidant capability and protected hepatocytes against NaF-induced cytotoxicity and necrotic death. The cytoprotective activity of AA was found to be comparable to that of a known antioxidant, vitamin C. Combining, data suggest that AA plays a protective role against NaF-induced cellular damage and prevents hepatocytes from necrotic death.


Subject(s)
Necrosis/prevention & control , Oxidative Stress/drug effects , Sodium Fluoride/toxicity , Triterpenes/pharmacology , Animals , Antioxidants/metabolism , Cell Survival/drug effects , Cytoprotection/drug effects , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Flow Cytometry , Glutathione/drug effects , Glutathione/metabolism , Glutathione Disulfide/drug effects , Glutathione Disulfide/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , In Vitro Techniques , Lipid Peroxidation/drug effects , Male , Mice , Necrosis/chemically induced , Reactive Oxygen Species/metabolism , Time Factors , Triterpenes/administration & dosage
9.
Toxicol In Vitro ; 22(8): 1832-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18801422

ABSTRACT

The objective was to investigate the regulation of glutathione (GSH) turnover in porcine aortic endothelial cells (PAECs) treated with sodium arsenite (NaAsO(2)), arsenic trioxide (As(2)O(3)) or sodium arsenate (Na(2)HAsO(4)) up to 72 hr at 0, 1, 5, and 10 microM, respectively. Intracellular GSH and glutathione disulfide (GSSG) contents, as well as the activities and mRNA levels of glutamate-cysteine lyase (GCL; gamma-glutamylcysteine synthetase) and gamma-glutamyl transpeptidase (GGT), were examined. The trivalent arsenic compounds increased GSH and GSSG contents in PAECs. An increase in GCL activity was observed at 24hr whereas an increase in GCL mRNA level was observed at 72 hr. The increase in GGT activity was only observed at 72 hr. In addition, a tendency of increase in GGT mRNA level was observed. Na(2)HAsO(4) treatment did not affect GSH content and the turnover-related enzymes. A differential GSH modulation in PAECs by trivalent arsenic compounds was found. The regulatory mechanism responsible for the As(2)O(3)-induced GSH increase is related to the GSH-turnover enzymes, GCL and GGT, while that for the NaAsO(2)-induced GSH increase may not be related to expression of GSH-turnover enzymes.


Subject(s)
Arsenates/toxicity , Arsenites/toxicity , Glutathione/drug effects , Oxides/toxicity , Sodium Compounds/toxicity , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Arsenic Trioxide , Arsenicals , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Glutamate-Cysteine Ligase/drug effects , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Glutathione Disulfide/drug effects , Glutathione Disulfide/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Swine , Time Factors , gamma-Glutamyltransferase/drug effects , gamma-Glutamyltransferase/metabolism
10.
Toxicology ; 247(1): 6-10, 2008 May 02.
Article in English | MEDLINE | ID: mdl-18336984

ABSTRACT

The present study shows the use of confocal autofluorescence spectroscopy coupled with the time-resolved fluorescence decay analysis to measure changes in FAD/NAD[P]H and free/bound NAD[P]H in HepG(2) cells at 0.5, 1.5, 3 and 4.5h after exposure to cadmium chloride (Cd). These changes were compared to changes in GSSG/GSH and production of reactive oxygen radicals (ROS) production. The results demonstrated that both FAD/NAD[P]H and GSSG/GSH increased significantly upon exposure to Cd. The change in GSSG/GSH occurred as early as 1.5h after treatment while the change in FAD/NAD[P]H did not occur until 3h after exposure. Production of ROS was also increased at 1.5h. The ratio of free/bound NAD[P]H was studied. It was demonstrated that free/bound NAD[P]H increased significantly as early as 0.5h and remained elevated until 4.5h after treatment with Cd. The present study provides novel data to show that changes in NAD[P]H metabolism precedes the increase in ROS production and cellular oxidative stress (increase GSSG/GSH, FAD/NAD[P]H). It is suggested that Cd causes a release of NAD[P]H, an important cofactor for electron transfer, from its normal protein binding sites. This may result in a disruption of the activity of the enzyme and proteins, and may lead to the subsequent toxic events.


Subject(s)
Cadmium Chloride/toxicity , Flavin-Adenine Dinucleotide/metabolism , NADP/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Binding Sites/drug effects , Cell Line, Tumor , Glutathione/drug effects , Glutathione/metabolism , Glutathione Disulfide/drug effects , Glutathione Disulfide/metabolism , Humans , NADP/metabolism , Protein Binding/drug effects , Spectrometry, Fluorescence , Time Factors
11.
Basic Clin Pharmacol Toxicol ; 100(3): 151-6, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17309517

ABSTRACT

The objective of the study was to investigate the effect of epigallocatechin-3-gallate (EGCG) on ethanol (EtOH)-induced cytotoxicity in human Chang liver cells. Cells were incubated with either 30 mM EtOH alone or together in presence of (25 microM) EGCG for 24 hr. Assays were performed in treated cells to evaluate the ability of EGCG to prevent the toxic effects of EtOH. EtOH exposure suppressed the growth of Chang liver cells and induced lactate dehydrogenase leakage, oxygen radical formation, peroxidation of lipids, mitochondrial dysfunction and apoptosis. Reduced glutathione (GSH) concentration was significantly decreased (P < 0.05) while oxidized glutathione (GSSG) concentration was significantly elevated in EtOH-treated cells as compared to normal cells. Incubation of EGCG along with EtOH significantly prevented EtOH-dependent cell loss and lactate dehydrogenase leakage. This was associated with a reduction in oxidative damage as reflected by a reduction in the generation of reactive oxygen species, and in lipid peroxidation and maintenance of intracellular GSH/GSSG ratio. EGCG decreased the accumulation of sub-G(1) phase cells and reduced apoptosis. The findings suggest that EGCG exerts a protective action during EtOH-induced liver cell damage.


Subject(s)
Antioxidants/pharmacology , Catechin/analogs & derivatives , Central Nervous System Depressants/toxicity , Ethanol/toxicity , Liver/cytology , Apoptosis/drug effects , Catechin/pharmacology , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Glutathione/drug effects , Glutathione/metabolism , Glutathione Disulfide/drug effects , Glutathione Disulfide/metabolism , Humans , L-Lactate Dehydrogenase/drug effects , L-Lactate Dehydrogenase/metabolism , Lipid Peroxidation/drug effects , Liver/drug effects , Mitochondria/drug effects , Reactive Oxygen Species/metabolism
12.
Free Radic Res ; 41(1): 15-24, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17164175

ABSTRACT

Heart mitochondria from female senescence-accelerated (SAMP8) and senescence-resistant (SAMR1) mice of 5 or 10 months of age, were studied. Mitochondrial oxidative stress was determined by measuring the levels of lipid peroxidation, glutathione and glutathione disulfide and glutathione peroxidase and reductase activities. Mitochondrial function was assessed by measuring the activity of the respiratory chain complexes and ATP content. The results show that the age-dependent mitochondrial oxidative damage in the heart of SAMP8 mice was accompanied by a reduction in the electron transport chain complex activities and in ATP levels. Chronic melatonin administration between 1 and 10 months of age normalized the redox and the bioenergetic status of the mitochondria and increased ATP levels. The results support the presence of significant mitochondrial oxidative stress in SAM mice at 10 months of age, and they suggest a beneficial effect of chronic pharmacological intervention with melatonin, which reduces the deteriorative and functional oxidative changes in cardiac mitochondria with age.


Subject(s)
Aging , Antioxidants/pharmacology , Melatonin/pharmacology , Mitochondria/drug effects , Adenosine Triphosphate/metabolism , Animals , Electron Transport Chain Complex Proteins/drug effects , Electron Transport Chain Complex Proteins/metabolism , Female , Glutathione/drug effects , Glutathione/metabolism , Glutathione Disulfide/drug effects , Glutathione Disulfide/metabolism , Glutathione Peroxidase/drug effects , Glutathione Peroxidase/metabolism , Glutathione Reductase/drug effects , Glutathione Reductase/metabolism , Heart/drug effects , Lipid Peroxidation/drug effects , Mice , Mitochondria/pathology , Myocardium/metabolism , Oxidative Stress/drug effects
13.
Free Radic Res ; 41(1): 89-96, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17164182

ABSTRACT

We have previously reported that isomeric Zn(II) N-methylpyridylporphyrins (ZnTM-2(3,4)-PyP4 + ) can act as photosensitizers with efficacy comparable to that of hematoporphyrin derivative (HpD) in preventing cell proliferation and causing cell death in vitro. To better understand the biochemical basis of this activity, the effects of photo-activated ZnTM-3-PyP4 + on GSH/GSSG ratio, lipid peroxidation, membrane permeability, oxidative DNA damage, and the activities of SOD, catalase, glutathione reductase, and glutathione peroxidase were evaluated. Light exposure of ZnTM-3-PyP4 + -treated colon adenocarcinoma cells caused a wide spectrum of oxidative damage including depletion of GSH, inactivation of glutathione reductase and glutathione peroxidase, oxidative DNA damage and peroxidation of membrane lipids. Cell staining with Hoechst-33342 showed morphological changes consistent with both necrotic and apoptotic death sequences, depending upon the presence of oxygen.


Subject(s)
Metalloporphyrins/toxicity , Oxidative Stress/drug effects , Photosensitizing Agents/toxicity , Catalase/drug effects , Catalase/metabolism , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , DNA Damage/drug effects , DNA Damage/physiology , Glutathione/drug effects , Glutathione/metabolism , Glutathione Disulfide/drug effects , Glutathione Disulfide/metabolism , Glutathione Peroxidase/drug effects , Glutathione Peroxidase/metabolism , Glutathione Reductase/drug effects , Glutathione Reductase/metabolism , Humans , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Oxidative Stress/physiology , Photosensitivity Disorders , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism
14.
Exp Biol Med (Maywood) ; 231(8): 1430-8, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16946412

ABSTRACT

Rhabdomyolysis-induced oxidative stress is associated with morphological and functional damage to the kidney and other organs, but applications of this model in the lung are still lacking. The aim of the present study was to determine the relationship between oxidative stress and the morphological changes occurring in the lungs of rats subjected to rhabdomyolysis. Rhabdomyolysis was induced by intramuscular glycerol injection (50% v/v, 10 ml/kg), and the control group was injected with saline vehicle. Arterial blood samples were drawn at 0, 2, 4, and 6 hrs for measurement of arterial gases, creatine kinase activity, and plasma free F2-isoprostane levels. Six hours later, the lungs were removed to determine the wet-to-dry weight ratio, reduced glutathione (GSH) and GSH disulfide (GSSG) levels, and activity of antioxidant enzymes (catalase [CAT], superoxide dismutase [SOD], and GSH peroxidase [GSH-Px]). Protein carbonylation and lipid peroxidation were assessed in the lungs by measurement of carbonyl and malondialdehyde (MDA) production, respectively. Bronchoalveolar lavage, cell counts, and lung ultrastructural studies were also performed. Six hours after glycerol injection, arterial PO2 and PCO2 were 23% and 38% lower, respectively, and plasma free F2-isoprostane levels were 72% higher, compared with control values. In lungs, protein carbonyl and MDA production were 58% and 12% higher, respectively; the GSH:GSSG ratio and GSH-Px activity were 43% and 60% lower, respectively; and activities of CAT and SOD showed no significant differences compared with controls. Rhabdomyolysis-induced ultrastructural impairment of the lung showed Type II cell damage, extracytoplasmic lamellar bodies and lack of tubular myelin reorganization, endothelial cellular edema, and no disruption of the alveolar-capillary barrier. These results provide evidence that rhabdomyolysis could induce tissue injury associated with increased oxidative stress, suggesting the contribution of oxidative stress to the pathogenic mechanism of acute lung injury.


Subject(s)
Lung Diseases/etiology , Lung Diseases/metabolism , Lung/ultrastructure , Oxidative Stress/physiology , Rhabdomyolysis/complications , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Carbon Dioxide/blood , Catalase/analysis , Catalase/metabolism , F2-Isoprostanes/blood , Glutathione/analysis , Glutathione/metabolism , Glutathione Disulfide/analysis , Glutathione Disulfide/drug effects , Glutathione Peroxidase/analysis , Glutathione Peroxidase/metabolism , Glycerol/toxicity , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Lung/chemistry , Lung/metabolism , Lung Diseases/pathology , Male , Microscopy, Electron, Transmission , Oxygen/blood , Protein Carbonylation/drug effects , Protein Carbonylation/physiology , Rats , Rats, Wistar , Respiratory Function Tests , Rhabdomyolysis/chemically induced , Superoxide Dismutase/analysis , Superoxide Dismutase/metabolism
15.
Free Radic Biol Med ; 40(2): 316-22, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16413413

ABSTRACT

The aim of this study was to determine seizure-induced oxidative stress by measuring hippocampal glutathione (GSH) and glutathione disulfide (GSSG) levels in tissue and mitochondria. Kainate-induced status epilepticus (SE) in rats resulted in a time-dependent decrease of GSH/GSSG ratios in both hippocampal tissue and mitochondria. However, changes in GSH/GSSG ratios were more dramatic in the mitochondrial fractions compared to hippocampal tissue. This was accompanied by a mild increase in glutathione peroxidase activity and a decrease in glutathione reductase activity in hippocampal tissue and mitochondria, respectively. Since coenzyme A (CoASH) and its disulfide with GSH (CoASSG) are primarily compartmentalized within mitochondria, their measurement in tissue was undertaken to overcome problems associated with GSH/GSSG measurement following subcellular fractionation. Hippocampal tissue CoASH/CoASSG ratios were decreased following kainate-induced SE, the time course and magnitude of change paralleling mitochondrial GSH/GSSG levels. Cysteine, a rate-limiting precursor of glutathione was decreased following kainate administration in both hippocampal tissue and mitochondrial fractions. Together these changes in altered redox status provide further evidence for seizure-induced mitochondrial oxidative stress.


Subject(s)
Glutathione Disulfide/metabolism , Glutathione/metabolism , Mitochondria/metabolism , Oxidative Stress , Seizures/metabolism , Status Epilepticus/metabolism , Animals , Coenzyme A/drug effects , Coenzyme A/metabolism , Cysteine/drug effects , Cysteine/metabolism , Disease Models, Animal , Glutathione/drug effects , Glutathione Disulfide/drug effects , Glutathione Peroxidase/drug effects , Glutathione Peroxidase/metabolism , Glutathione Reductase/drug effects , Glutathione Reductase/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Kainic Acid , Male , Mitochondria/drug effects , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Seizures/chemically induced , Status Epilepticus/chemically induced , Time Factors
16.
Med Sci Sports Exerc ; 37(2): 234-9, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15692318

ABSTRACT

PURPOSE: This study was designed to ascertain the effects of a combination antioxidant therapy on plasma protein carbonyls (PC), malondialdehyde (MDA), and whole blood total (TGSH), oxidized (GSSG), and reduced (GSH) glutathione in non-resistance trained females after eccentric resistance exercise. METHODS: Eighteen women (aged 19-31 yr) were randomized in a double-blind manner to either an antioxidant supplement (N = 9; 400 IU vitamin E, 1 g vitamin C, and 90 mug selenium per day) or a lactose placebo (N = 9) for 14 d before and for 2 d after eccentric elbow flexor exercise. Blood samples taken before and immediately, 2, 6, 24, and 48 h postexercise were analyzed for PC, MDA, TGSH, and GSSG. RESULTS: No treatment by time interaction was noted for any variable, with all blood markers experiencing a change after the exercise in both conditions. Time main effects were observed for PC, MDA, and GSSG, with values elevated above preexercise after the eccentric exercise, whereas GSH concentration decreased after the eccentric exercise. Antioxidant supplementation resulted in a condition main effect for PC and MDA, with lower values compared with placebo. The antioxidant treatment attenuated the rise in both PC (75%) and MDA (100%). CONCLUSION: These data suggest that eccentric resistance exercise can increase blood biomarkers of oxidative stress in non-resistance trained females, and this vitamin E, C, and selenium supplementation can attenuate the rise in PC and MDA.


Subject(s)
Antioxidants/administration & dosage , Antioxidants/pharmacology , Exercise , Oxidative Stress/drug effects , Adult , Ascorbic Acid/administration & dosage , Ascorbic Acid/pharmacology , Biomarkers/blood , Double-Blind Method , Drug Therapy, Combination , Female , Glutathione/blood , Glutathione/drug effects , Glutathione Disulfide/blood , Glutathione Disulfide/drug effects , Humans , Malondialdehyde/blood , Protein Carbonylation/drug effects , Selenium/administration & dosage , Selenium/pharmacology , Treatment Outcome , Vitamin E/administration & dosage , Vitamin E/pharmacology , Women's Health
17.
Am J Physiol Cell Physiol ; 288(4): C840-9, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15561762

ABSTRACT

Persistent inhibition of cytochrome-c oxidase, a terminal enzyme of the mitochondrial electron transport chain, by excessive nitric oxide (NO) derived from inflammation, polluted air, and tobacco smoke contributes to enhanced oxidant production and programmed cell death or apoptosis of lung cells. We sought to determine whether the long-term exposure of pulmonary artery endothelial cells (PAEC) to pathophysiological concentrations of NO causes persistent inhibition of complex IV through redox modification of its key cysteine residues located in a putative NO-sensitive motif. Prolonged exposure of porcine PAEC to 1 mM 2,2'-(hydroxynitrosohydrazino)-bis-ethanamine (NOC-18; slow-releasing NO donor, equivalent to 1-5 microM NO) resulted in a gradual, persistent inhibition of complex IV concomitant with a reduction in ratios of mitochondrial GSH and GSSG. Overexpression of thioredoxin in mitochondria of PAEC attenuated NO-induced loss of complex IV activities, suggesting redox regulation of complex IV activity. Sequence analysis of complex IV subunits revealed a novel putative NO-sensitive motif in subunit II (S2). There are only two cysteine residues in porcine complex IV S2, located in the putative motif. Immunoprecipitation and Western blot analysis and "biotin switch" assay demonstrated that exposure of PAEC to 1 mM NOC-18 increased S-nitrosylation of complex IV S2 by 200%. Site-directed mutagenesis of these two cysteines of complex IV S2 attenuated NO-increased nitrosylation of complex IV S2. These results demonstrate for the first time that NO nitrosylates active site cysteines of complex IV, which is associated with persistent inhibition of complex IV. NO inhibition of complex IV via nitrosylation of NO-sensitive cysteine residues can be a novel upstream event in NO-complex IV signaling for NO toxicity in lung endothelial cells.


Subject(s)
Cysteine/chemistry , Electron Transport Complex IV/metabolism , Endothelial Cells/drug effects , Mitochondria/enzymology , Nitric Oxide/pharmacology , Animals , Binding Sites , Blotting, Western , Cells, Cultured , Cysteine/drug effects , Endothelial Cells/metabolism , Glutathione/drug effects , Glutathione/metabolism , Glutathione Disulfide/drug effects , Glutathione Disulfide/metabolism , Humans , Immunoprecipitation , Lung/drug effects , Lung/metabolism , Mutagenesis, Site-Directed , Nitric Oxide Donors/pharmacology , Nitroso Compounds/pharmacology , Oxidation-Reduction , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Sulfhydryl Compounds/metabolism , Swine , Thioredoxins/metabolism
18.
Free Radic Res ; 38(10): 1101-6, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15512798

ABSTRACT

The blood reduced glutathione (GSH)/GSH disulfide (GSSG) ratio is an index of the oxidant/antioxidant balance of the whole body. Nevertheless, data indicating GSH and GSSG physiological levels are still widely divergent, especially those on GSSG, probably due to its low concentration. Standardization in methodological protocols and sample manipulation could help to minimize these discrepancies. Therefore, we have investigated how plasma reduced GSH, which is rapidly oxidized after blood withdrawal, could alter the blood GSSG measurement if the sample is not suitably processed. We have observed that an increase in plasma GSH concentration, due to red blood cell hemolysis, is responsible for a significant overestimation of blood GSSG level. Our results show that, before performing blood GSSG determination, thiols have to be rapidly blocked, to avoid possible pitfalls in GSSG measurement, in particular when hemolysis is present.


Subject(s)
Glutathione Disulfide/blood , Glutathione/blood , Hemoglobins/metabolism , Hemolysis , Animals , Glutathione/pharmacology , Glutathione Disulfide/drug effects , Humans , Rats
19.
Biol Cell ; 94(4-5): 251-65, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12489694

ABSTRACT

Arachidonic acid (AA)-induced cytotoxicity was evaluated in leukocytes: the human leukemia cell lines HL-60, Jurkat and Raji and in rat lymphocytes. Such cytotoxicity was dose- and time-dependent. At concentrations below 5 microM, AA was not toxic; at 10-400 microM, AA induced apoptosis and at concentrations beyond 400 microM, necrosis. The minimum exposure time to trigger cell death was of around 1 h, but the effect was increased by longer exposure times until 6-24 h. Apoptosis was morphologically characterized by a decrease in cell and nuclear volume, chromatin condensation and DNA fragmentation and the presence of lipid bodies, without changes in organelle integrity. Biochemically, AA-induced apoptosis was associated with internucleosomal fragmentation and caspase activation, evaluated by PARP cleavage and the use of a caspase inhibitor. Necrosis was characterized by increased cell volume, presence of loose chromatin, appearance of vacuoles, loss of membrane integrity and of the definition of organelles. The apoptotic effect of AA was studied as to oxidative-reductive imbalance and the participation of eicosanoids. Apoptotic AA treatment was accompanied by an increase in the quantity of thiobarbituric acid reactive substances (TBARS), low-level chemiluminescence and in the glutathione disulfide/reduced glutathione ratio, indicating oxidative stress. The addition of tocopherol, ascorbate, prostaglandin E2 and lipoxygenase inhibitors delayed cell death, whereas the inhibition of cyclooxygenase promoted AA-induced cell death. Cell treatment with AA was accompanied by increased cellular production of LTB4. AA, therefore, is cytotoxic at physiological and supraphysiological concentrations, causing apoptosis and necrosis. Cell treatment with apoptotic concentrations of AA involves oxidative stress and changes in eicosanoid biosynthesis.


Subject(s)
Apoptosis/drug effects , Arachidonic Acid/toxicity , Cytotoxins/toxicity , Eicosanoids/biosynthesis , Leukocytes/drug effects , Oxidative Stress/drug effects , Animals , Antioxidants/pharmacology , Apoptosis/physiology , Arachidonic Acid/metabolism , Caspases/drug effects , Caspases/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cyclooxygenase Inhibitors/pharmacology , Cytotoxins/metabolism , DNA Fragmentation/drug effects , DNA Fragmentation/physiology , Dose-Response Relationship, Drug , Drug Administration Schedule , Glutathione Disulfide/drug effects , Glutathione Disulfide/metabolism , HL-60 Cells , Humans , Jurkat Cells , Leukocytes/metabolism , Leukotriene B4/metabolism , Necrosis , Oxidative Stress/physiology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases , Proteins/drug effects , Proteins/metabolism , Rats , Thiobarbituric Acid Reactive Substances/metabolism
20.
J Pediatr Gastroenterol Nutr ; 35(3): 344-9, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12352525

ABSTRACT

BACKGROUND: Although bilirubin, which crosses the blood-brain barrier, can cause irreversible brain damage, it also possesses antioxidant properties that may be protective against oxidative stress. Intestinal ischemia-reperfusion (IR) injury results in cell destruction, mediated via the generation of reactive oxygen species. Although increased serum bilirubin is correlated with increased antioxidant potential in the face of hyperoxia, evidence of bilirubin-associated protective effect against IR injury remains nonspecific. We therefore sought to investigate whether hyperbilirubinemia would be protective against IR injury to the intestine. METHODS: Young adult rats were randomly assigned to one of three groups: 1) IR/control (n = 12); 2) IR/hyperbilirubinemia (n = 10), in which IR was generated while the rats were treated with a continuous infusion of bilirubin; and 3) hyperbilirubinemia controls (n = 10). Blood and intestinal tissue samples were obtained to determine serial thiobarbituric acid reducing substances (index of lipid peroxidation) and for xanthine oxidase/xanthine dehydrogenase and glutathione/glutathione disulfide ratios. Intestinal histopathology was graded from 1 (normal) to 4 (severe necrotic lesions). RESULTS: Histopathologic scoring and circulating and tissue thiobarbituric acid reducing substances were highest in the IR/control animals compared with either the IR/hyperbilirubinemics or the controls. All of these are consistent with the most severe injury in this group. Xanthine oxidase/xanthine dehydrogenase ratios were not significantly different among the groups. CONCLUSION: Hyperbilirubinemia ameliorates the extent of intestinal IR injury in our model and appears to act as an antioxidant. This study supports the concept that bilirubin possesses some beneficial properties in vivo, although no direct clinical conclusions can be drawn from these data.


Subject(s)
Bilirubin/therapeutic use , Intestines/pathology , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Animals , Bilirubin/blood , Disease Models, Animal , Enterocolitis, Necrotizing/pathology , Glutathione Disulfide/drug effects , Rats , Thiobarbituric Acid Reactive Substances/analysis , Time Factors , Xanthine Dehydrogenase/drug effects , Xanthine Oxidase/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL