Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Artif Cells Nanomed Biotechnol ; 44(1): 128-34, 2016.
Article in English | MEDLINE | ID: mdl-24959911

ABSTRACT

Quercetin (3,5,7,3',4'-pentahydroxyflavone) is a natural bio-flavonoid originating from fruits, vegetables, seeds, berries, and tea. The antioxidant activity of quercetin and its protective effects against cardiovascular disorders, anti-cancer, anti-inflammatory, and anti-viral activities have been extensively documented; however, the clinical request of quercetin in cancer treatment is significantly limited due to its very poor delivery features. In order to increase the hydrophilicity and drug delivery capability, we encapsulated quercetin into liposomes. Our data indicated that liposomal quercetin can significantly improve the solubility and bioavailability of quercetin and can be used as an effective antioxidant for ROS protection within the polar cytoplasm, and the nano-sized quercetin encapsulated by liposomes enhanced the cellular uptake (cancer cell human MCF_7). Quercetin has many pharmaceutical applications, many of which arise from its potent antioxidant properties. The present research examined the antioxidant activities of quercetin in polar solvents by a comparative study using reduction of ferric iron in aqueous medium, intracellular ROS/toxicity assays, and reducing DPPH assays. Cell viability and ROS assays demonstrated that quercetin was able to penetrate into the polar medium inside the cells and to protect them against the highly toxic and deadly belongings of cumene hydroperoxide. The purpose of this study was to determine whether a liposomal formulation of quercetin can suggestively improve its solubility and bioavailability and can be a possible request in the treatment of tumor. The authors encapsulated quercetin in a liposomal delivery system. They studied the in vitro effects of this compound on proliferation using human MCF-7 carcinoma cells. The activity of liposomal quercetin was equal to or better than that of free quercetin at equimolar concentrations. Our data indicated that liposomal quercetin can significantly improve the solubility and bioavailability of quercetin and can be a potential application in the treatment of tumor.


Subject(s)
Free Radical Scavengers/pharmacology , Free Radicals/antagonists & inhibitors , Liposomes/chemical synthesis , Quercetin/pharmacology , Benzene Derivatives/antagonists & inhibitors , Benzene Derivatives/pharmacology , Biological Availability , Biphenyl Compounds/antagonists & inhibitors , Biphenyl Compounds/chemistry , Cell Survival/drug effects , Free Radical Scavengers/chemistry , Free Radicals/chemistry , Humans , Iron/chemistry , MCF-7 Cells , Oxidation-Reduction , Picrates/antagonists & inhibitors , Picrates/chemistry , Quercetin/chemistry , Solubility
2.
Toxicol Mech Methods ; 24(8): 529-35, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24861666

ABSTRACT

Diphenyl ditelluride (PhTe)2 is a versatile molecule used in the organic synthesis and it is a potential prototype for the development of novel biologically active molecules. The mechanism(s) involved in (PhTe)2 toxicity is(are) elusive, but thiol oxidation of critical proteins are important targets. Consequently, the possible remedy of its toxicity by thiol-containing compounds is of experimental and clinical interest. The present study aimed to investigate putative mechanisms underlying the toxicity of (PhTe)2 in vivo. We assessed behavioral and oxidative stress parameters in mice, including the modulation of antioxidant enzymatic defense systems. In order to mitigate such toxicity, N-acetylcysteine (NAC) was administered before (3 d) and simultaneously with (PhTe)2 (7 d). Mice were separated into six groups receiving daily injections of (1) TFK (2.5 ml/kg, intraperitonealy (i.p.)) plus canola oil (10 ml/kg, subcutaneously (s.c.)), (2) NAC (100 mg/kg, i.p.) plus canola oil s.c., (3) TFK i.p. plus (PhTe)2 (10 µmol/kg, s.c.), (4) TFK i.p. plus (PhTe)2 (50 µmol/kg, s.c.), (5) NAC plus (PhTe)2 (10 µmol/kg, s.c.), and (6) NAC plus (PhTe)2 (50 µmol/kg, s.c.). (PhTe)2 treatment started on the fourth day of treatment with NAC. Results demonstrated that (PhTe)2 induced behavioral alterations and inhibited important selenoenzymes (thioredoxin reductase and glutathione peroxidase). Treatments produced no or minor effects on the activities of antioxidant enzymes catalase and glutathione reductase. Contrary to expected, NAC co-administration did not protect against the deleterious effects of (PhTe)2. Other low-molecular-thiol containing molecules should be investigated to determine whether or not they can be effective against ditellurides.


Subject(s)
Benzene Derivatives/toxicity , Environmental Pollutants/toxicity , Glutathione Peroxidase/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Neurotoxicity Syndromes/enzymology , Organometallic Compounds/toxicity , Oxidative Stress/drug effects , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Acetylcysteine/administration & dosage , Acetylcysteine/therapeutic use , Animals , Antioxidants/administration & dosage , Antioxidants/therapeutic use , Behavior, Animal/drug effects , Benzene Derivatives/administration & dosage , Benzene Derivatives/antagonists & inhibitors , Brain/drug effects , Brain/enzymology , Dose-Response Relationship, Drug , Environmental Pollutants/administration & dosage , Environmental Pollutants/antagonists & inhibitors , Glutathione Peroxidase/metabolism , Injections, Intraperitoneal , Injections, Subcutaneous , Male , Mice , Motor Activity/drug effects , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/enzymology , Neurotoxicity Syndromes/prevention & control , Organometallic Compounds/administration & dosage , Organometallic Compounds/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/metabolism , Toxicity Tests, Acute
3.
Angew Chem Int Ed Engl ; 52(9): 2599-601, 2013 Feb 25.
Article in English | MEDLINE | ID: mdl-23355270

ABSTRACT

Good substrate gone bad! BN/CC isosterism of ethylbenzene leads to N-ethyl-1,2-azaborine and B-ethyl-1,2-azaborine. In contrast to ethylbenzene, which is the substrate for ethylbenzene dehydrogenase (EbDH), N-ethyl-1,2-azaborine (see scheme; Fc=Ferricenium tetrafluoroborate) and B-ethyl-1,2-azaborine are strong inhibitors of EbDH. Thus, the changes provided by BN/CC isosterism can lead to new biochemical reactivity.


Subject(s)
Benzene Derivatives/chemistry , Boron Compounds/chemistry , Boron Compounds/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Benzene Derivatives/antagonists & inhibitors , Benzene Derivatives/metabolism , Hydroxylation/drug effects , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/chemistry , Oxidoreductases/metabolism
4.
Psychopharmacology (Berl) ; 222(4): 709-19, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22451092

ABSTRACT

RATIONALE: The transition to menopause is associated with an increased risk of depressed mood. OBJECTIVES: This study was conducted to investigate whether diphenyl diselenide [(PhSe)2] treatment could reduce the effects of postmenopausal depression-like behavior in ovariectomized female mice submitted to subchronic stress exposure. METHODS: Mice were divided into four groups: sham, (PhSe)2, ovariectomy (OVX), and OVX + (PhSe)2. Animals were ovariectomized/sham-operated and subjected to stress session once a day for 7 days from the fifth to the 11th day after OVX. The behavioral tests (open field, tail suspension (TST), and forced swimming (FST)) were performed on the 14th day after OVX. Mice were treated orally once a day with vehicle (canola oil, 10 ml/kg) or (PhSe)2 (10 mg/kg; 10 ml/kg) 30 min before being exposed to subchronic stress, or from the 11th to the 14th day. Paroxetine (8 mg/kg i.p.) and pargyline (30 mg/kg i.p.) were used as positive controls. The involvement of serotonergic receptor subtypes in the antidepressant-like effect of (PhSe)2 was assessed in the FST using WAY 100635 (0.1 mg/kg s.c.), ritanserin (1 mg/kg i.p.), and ondansetron (1 mg/kg i.p.) as serotonergic antagonists. Monoamine oxidase (MAO) A and B activities were also determined. RESULTS: The prolongation of immobility time in TST and FST in OVX mice submitted to subchronic stress was prevented by (PhSe)2 treatment. Ritanserin and ondansetron blocked the antidepressive-like effect of (PhSe)2, suggesting the involvement of 5-HT(2A/2C) and 5-HT3 receptor subtypes. Both paroxetine and pargyline were effective in reducing the immobility time of stressed OVX mice in the FST. No alterations in locomotor activity were observed. Although (PhSe)2 had inhibited in vitro MAO-A and MAO-B activities, none of the groups presented alterations neither in ex vivo MAO-A nor in MAO-B activity. CONCLUSIONS: (PhSe)2 treatment could influence mood and behavior, indicating a promising role of this organoselenium compound in the management of postmenopausal depressive symptoms.


Subject(s)
Benzene Derivatives/therapeutic use , Depression/enzymology , Menopause/physiology , Organoselenium Compounds/therapeutic use , Ovariectomy/psychology , Serotonergic Neurons/drug effects , Serotonergic Neurons/enzymology , Stress, Psychological/drug therapy , Animals , Antidepressive Agents/therapeutic use , Benzene Derivatives/antagonists & inhibitors , Benzene Derivatives/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Depression/complications , Depression/drug therapy , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Female , Hippocampus/drug effects , Hippocampus/enzymology , Immobility Response, Tonic/drug effects , Mice , Mice, Inbred Strains , Mitochondria/enzymology , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/therapeutic use , Ondansetron/pharmacology , Organoselenium Compounds/antagonists & inhibitors , Organoselenium Compounds/pharmacology , Pargyline/pharmacology , Pargyline/therapeutic use , Paroxetine/pharmacology , Paroxetine/therapeutic use , Piperazines/pharmacology , Pyridines/pharmacology , Ritanserin , Serotonin Antagonists/pharmacology , Stress, Psychological/complications , Stress, Psychological/enzymology
5.
Eur J Pharmacol ; 638(1-3): 90-8, 2010 Jul 25.
Article in English | MEDLINE | ID: mdl-20406629

ABSTRACT

Previously, it was shown that intravenous (i.v.) treatment with the essential oil of Aniba canelilla (EOAC) elicited a hypotensive response that is due to active vascular relaxation rather than to the withdrawal of sympathetic tone. The present study investigated mechanisms underlying the cardiovascular responses to 1-nitro-2-phenylethane, the main constituent of the EOAC. In pentobarbital-anesthetized normotensive rats, 1-nitro-2-phenylethane (1-10mg/kg, i.v.) elicited dose-dependent hypotensive and bradycardiac effects which were characterized in two periods (phases 1 and 2). The first rapid component (phase 1) evoked by 1-nitro-2-phenylethane (10mg/kg) was fully abolished by bilateral vagotomy, perineural treatment of both cervical vagus nerves with capsaicin (250 microg/ml) and was absent after left ventricle injection. However, pretreatment with capsazepine (1mg/kg, i.v.) or ondansetron (30 microg/kg, i.v.) did not alter phase 1 of the cardiovascular responses to 1-nitro-2-phenylethane (10mg/kg, i.v.). In conscious rats, 1-nitro-2-phenylethane (1-10mg/kg, i.v.) evoked rapid hypotensive and bradycardiac (phase 1) effects that were fully abolished by methylatropine (1mg/kg, i.v.). It is concluded that 1-nitro-2-phenylethane induces a vago-vagal bradycardiac and depressor reflex (phase 1) that apparently results from the stimulation of vagal pulmonary rather than cardiac C-fiber afferents. The transduction mechanism of the 1-nitro-2-phenylethane excitation of C-fiber endings is not fully understood and does not appear to involve activation of either Vanilloid TPRV(1) or 5-HT(3) receptors. The phase 2 hypotensive response to 1-nitro-2-phenylethane seems to result, at least in part, from a direct vasodilatory effect since 1-nitro-2-phenylethane (1-300 microg/ml) induced a concentration-dependent reduction of phenylephrine-induced contraction in rat endothelium-containing aorta preparations.


Subject(s)
Benzene Derivatives/pharmacology , Bradycardia/chemically induced , Cryptocarya , Hypotension/chemically induced , Oils, Volatile/pharmacology , Reflex/drug effects , Vagus Nerve/drug effects , Animals , Aorta/drug effects , Atropine Derivatives/pharmacology , Benzene Derivatives/antagonists & inhibitors , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Dose-Response Relationship, Drug , Herb-Drug Interactions , In Vitro Techniques , Male , Oils, Volatile/isolation & purification , Ondansetron/pharmacology , Phenylephrine/antagonists & inhibitors , Phenylephrine/pharmacology , Rats , Rats, Wistar , Vagus Nerve/surgery , Vasoconstriction/drug effects
6.
Food Chem Toxicol ; 48(3): 854-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20036300

ABSTRACT

The protective effects of different concentrations of aqueous extract of Rhus coriaria L. fruit (75 and 100 microg/ml) and also gallic acid (100 microM) as one of its main components were examined against oxidative stress toxicity induced by cumene hydroperoxide (CHP) in isolated rat hepatocytes. Both extract concentrations and gallic acid (100 microM) significantly (P<0.05) protected the hepatocyte against all oxidative stress markers including cell lysis, ROS generation, lipid peroxidation, glutathione depletion, mitochondrial membrane potential decrease, lysosomal membrane oxidative damage and cellular proteolysis. Aqueous extracts of Rhus coriaria L. (75 and 100 microg/ml) were more effective than gallic acid (100 microM) in protecting hepatocytes against CHP induced lipid peroxidation (P<0.05). On the other hand gallic acid (100 microM) acted more effective than aqueous extracts of Rhus coriaria L. (75 and 100 microg/ml) at preventing hepatocyte membrane lysis (P<0.05). In addition H(2)O(2) scavenging effect of both extract concentrations (75 and 100 microg/ml) were determined in hepatocytes and compared with gallic acid (100 microM). Gallic acid (100 microM) was more effective than aqueous extracts of Rhus coriaria L. (75 and 100 microg/ml) at H(2)O(2) scavenging activity (P<0.05).


Subject(s)
Cell Survival/drug effects , Chemical and Drug Induced Liver Injury/prevention & control , Oxidative Stress/drug effects , Protective Agents , Rhus/chemistry , Animals , Benzene Derivatives/antagonists & inhibitors , Benzene Derivatives/toxicity , Fruit/chemistry , Gallic Acid/chemistry , Glutathione/metabolism , Hepatocytes/drug effects , Lipid Peroxidation/drug effects , Lysosomes/drug effects , Male , Membrane Potentials/drug effects , Oxidants/toxicity , Plant Extracts/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
7.
Chem Biol Interact ; 179(2-3): 94-102, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19135433

ABSTRACT

Macrophages, the major phagocytes of body, are largely dependent on membrane for their apposite functioning. Cum-OOH, a catalyst used in chemical and pharmaceutical industry, is a peroxidative agent, which may induce oxidative stress in macrophages hampering the integrity of their membrane. Alpha-tocopherol is known to protect the membrane from oxidative modulation and preserve its integrity. In the present study, we investigated the effect of Cum-OOH on physiology of macrophages and evaluated the protective effect of alpha-tocopherol against Cum-OOH-induced functional impairment. An in vitro exposure to 10-200 microM Cum-OOH altered redox balance of murine peritoneal macrophages and led to a severe physiological impairment. It markedly augmented the release of proinflammatory cytokines (tumor necrosis factor-alpha, interleukin-1beta and prostaglandin E(2)), lipopolysaccharide primed nitric oxide release and inducible nitric oxide synthase expression, and lysosomal hydrolases secretion. It mitigated respiratory burst and phagocytosis and intracellular killing of yeast (Saccharomyces cerevisiae). Mannose receptor, a major macrophage phagocytic receptor (also implicated in S. cerevisiae phagocytosis), exhibited a hampered recycling with its number being reduced to about 54% of the untreated, control cells following Cum-OOH exposure. A 24-h pretreatment of macrophages with 25 microM alpha-tocopherol preserved most of the assessed functions close to their corresponding control values. These data suggest that exposure to Cum-OOH may impair the physiology of immune cells such as macrophages and that supplementation with alpha-tocopherol can safeguard these cells against Cum-OOH toxicity.


Subject(s)
Benzene Derivatives/pharmacology , Macrophages/drug effects , Oxidative Stress/drug effects , alpha-Tocopherol/pharmacology , Animals , Benzene Derivatives/antagonists & inhibitors , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Glucuronidase/metabolism , Glutathione/analysis , Interleukin-1beta/metabolism , Lectins, C-Type/drug effects , Macrophages/physiology , Male , Mannose Receptor , Mannose-Binding Lectins/drug effects , Muramidase/metabolism , Oxygen/metabolism , Phagocytosis/drug effects , Rats , Rats, Wistar , Receptors, Cell Surface/drug effects , Saccharomyces cerevisiae/drug effects , Tumor Necrosis Factor-alpha/metabolism
8.
J Am Chem Soc ; 128(51): 16432-3, 2006 Dec 27.
Article in English | MEDLINE | ID: mdl-17177355

ABSTRACT

A new class of thermally activated chain-breaking antioxidants is presented. Dimers of persistent carbon-centered radicals are able to inhibit the autoxidation of cumene and styrene with better rate constants than the commercial antioxidant Irganox HP-136 and 3,5-di-tert-butyl-4-hydroxyanisole. A dramatic increase in antioxidant activity is observed with increasing temperature as more dimers dissociate to their corresponding persistent radicals.


Subject(s)
Antioxidants/chemistry , Carbon/chemistry , Lactones/chemistry , Benzene Derivatives/antagonists & inhibitors , Benzene Derivatives/chemistry , Dimerization , Free Radicals/chemistry , Molecular Structure , Oxidation-Reduction , Styrene/antagonists & inhibitors , Styrene/chemistry , Time Factors
9.
Drug Metab Dispos ; 34(1): 61-8, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16221755

ABSTRACT

2,6-Dichlorophenyl methylsulfone (2,6-diClPh-MeSO2) is a potent olfactory toxicant reported to induce endoplasmic reticulum (ER) stress, caspase activation, and extensive cell death in mice. The aim of the present study was to examine cytochrome P450 (P450)-dependent bioactivation, nonprotein sulfhydryl (NP-SH) levels, and early ultrastructural changes in mouse olfactory mucosa following an i.p. injection of 2,6-diClPh-MeSO2 (32 mg/kg). A high covalent binding of 2,6-diClPh-14C-MeSO2 in olfactory mucosa S9 fraction was observed, and the CYP2A5/CYP2G1 substrates coumarin and dichlobenil significantly decreased the binding, whereas the CYP2E1 substrate chlorzoxazone had no effects. An increased bioactivation was detected in liver microsomes of mice pretreated with pyrazole, known to induce CYP2A4, 2A5, 2E1, and 2J, and addition of chlorzoxazone reduced this binding. 2,6-DiClPh-14C-MeSO2 showed a marked covalent binding to microsomes of recombinant yeast cells expressing mouse CYP2A5 or human CYP2A6 compared with wild type. One and 4 h after a single injection of 2,6-diClPh-MeSO2, the NP-SH levels in the olfactory mucosa were significantly reduced compared with control, whereas there was no change in the liver. Ultrastructural studies revealed that ER, mitochondria, and secretory granules in nonneuronal cells were early targets 1 h after injection. We propose that lesions induced by 2,6-diClPh-MeSO2 in the mouse olfactory mucosa were initiated by a P450-mediated bioactivation in the Bowman's glands and depletion of NP-SH levels, leading to disruption of ion homeostasis, organelle swelling, and cell death. The high expression of CYP2A5 in the olfactory mucosa is suggested to play a key role for the tissue-specific toxicity induced by 2,6-diClPh-MeSO2.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Benzene Derivatives/toxicity , Mixed Function Oxygenases/metabolism , Olfactory Mucosa/drug effects , Sulfones/toxicity , Animals , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Benzene Derivatives/antagonists & inhibitors , Benzene Derivatives/metabolism , Chlorzoxazone/pharmacology , Coumarins/pharmacology , Cytochrome P-450 CYP2A6 , Cytochrome P450 Family 2 , Dose-Response Relationship, Drug , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/ultrastructure , Female , Injections, Intraperitoneal , Liver/chemistry , Liver/drug effects , Liver/metabolism , Metyrapone/pharmacology , Mice , Microscopy, Electron, Transmission , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Mixed Function Oxygenases/antagonists & inhibitors , Nitriles/pharmacology , Olfactory Mucosa/metabolism , Olfactory Mucosa/ultrastructure , Pyrazoles/pharmacology , Sulfhydryl Compounds/metabolism , Sulfones/administration & dosage , Sulfones/antagonists & inhibitors , Sulfones/metabolism
10.
Life Sci ; 77(19): 2398-411, 2005 Sep 23.
Article in English | MEDLINE | ID: mdl-15932762

ABSTRACT

Organoselenium compounds have a potential thiol peroxidase-like activity. Diphenyl diselenide (DPDS) is an electrophilic reagent used in the synthesis of a variety of pharmacologically active organic selenium compounds. Using TRAP assay of chemiluminescense we have shown that diphenyl diselenide clearly possesses a pro-oxidant property. For an investigation on the mechanisms of this property, we used mutant strains of Saccharomyces cerevisiae defective in antioxidant defenses, i.e. in superoxide dismutase, in biosynthesis of glutathione, and the transcription factor yAP-1-lacking yap 1 mutant that cannot activate genes of the oxidative stress response. Exposure of growing cultures to the drug increased cell sensitivity to oxidizing agents. The pro-oxidant effect was independent of the metabolic condition or of the oxidative mutagen tested. N-acetylcysteine, a precursor of glutathione biosynthesis, could neutralize the pro-oxidant effects of diphenyl diselenide by stimulating an increase of endogenous glutathione biosynthesis or by directly binding to the drug. Vitamin E (Trolox), a known antioxidant, was also able to protect S. cerevisiae against the pro-oxidant effect of diphenyl diselenide. In vitro assays showed that diphenyl diselenide interacts non-enzymatically with the thiol group of glutathione.


Subject(s)
Benzene Derivatives/toxicity , Organoselenium Compounds/toxicity , Oxidants/toxicity , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Antimetabolites, Antineoplastic/pharmacology , Benzene Derivatives/antagonists & inhibitors , Bleomycin/pharmacology , DNA Damage/drug effects , Fermentation , Free Radical Scavengers/pharmacology , Genes, Fungal/genetics , Glutathione/metabolism , Luminescent Measurements , Luminol/chemistry , Mutation/physiology , Organoselenium Compounds/antagonists & inhibitors , Oxygen/metabolism , Saccharomyces cerevisiae/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Vitamin E/pharmacology
11.
Neurotoxicology ; 26(3): 321-30, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15935204

ABSTRACT

Previously, increased formation of reactive oxygen species (ROS) has been demonstrated in cultured rat cerebellar granule cells (CGCs) exposed to t-butylcyclohexane, n-decane, and n-butylbenzene (Dreiem et al. Relationship between lipophilicity of C6-10 hydrocarbon solvents and their ROS-inducing potency in rat cerebellar granule cells. Neurotoxicology 2002;23:701-9). In the present paper, we have studied the effects of these hydrocarbons on the viability of CGCs. Cell death was assessed by measurement of lactate dehydrogenase (LDH) release and trypan blue staining. t-butylcyclohexane and n-butylbenzene induced cell death in rat CGCs in a time-dependent and concentration-dependent manner. In contrast, n-decane did not cause release of LDH from rat CGCs even at 1mM. Morphological studies revealed apoptotic morphology characterized by cell shrinkage and chromatin condensation after exposure to low concentrations of t-butylcyclohexane and n-butylbenzene. However, there was no internucleosomal DNA fragmentation and no protection by the pan-caspase inhibitor Boc-D-FMK or the protein synthesis inhibitor cycloheximide. This indicates that cell death after t-butylcyclohexane and n-butylbenzene exposure is an intermediate between classical apoptosis and necrosis. Treatment with the antioxidant alpha-tocopherol ameliorated hydrocarbon-induced cell death, indicating involvement of reactive oxygen species in the mechanism of hydrocarbon toxicity. The significance of ROS formation in relation to cell death is discussed.


Subject(s)
Cell Death/drug effects , Cerebellum/cytology , Hydrocarbons/toxicity , Reactive Oxygen Species/metabolism , Solvents/toxicity , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Benzene Derivatives/antagonists & inhibitors , Benzene Derivatives/toxicity , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Cells, Cultured , Cerebellum/drug effects , Cerebellum/metabolism , Cyclohexanes/antagonists & inhibitors , Cyclohexanes/toxicity , DNA/biosynthesis , DNA Fragmentation , Gas Chromatography-Mass Spectrometry , Hydrocarbons/antagonists & inhibitors , Hydrocarbons/chemistry , L-Lactate Dehydrogenase/metabolism , Neurons/drug effects , Neurons/ultrastructure , Nucleosomes/drug effects , Nucleosomes/ultrastructure , Rats , Structure-Activity Relationship
12.
Toxicology ; 191(2-3): 169-78, 2003 Sep 30.
Article in English | MEDLINE | ID: mdl-12965120

ABSTRACT

Organochalcogens are important intermediates and useful reagents in organic synthesis, which can increase human exposure risk to these chemicals in the workplace. As well, there are a number of reported cases of acute toxicity following organochalcogen ingestion of vitamins and dietary supplements. Since, the erythrocytic delta-ALA-D activity could be an important indicator of toxicity this report investigated the organochalcogens effects on blood delta-ALA-D in vitro. To investigate a possible involvement of cysteinyl groups in the inhibitory actions of diphenyl diselenide, diphenyl ditelluride and Ebselen (4-100 micro M), the effects of thiol reducing agents (0-3 mM) or zinc chloride (0-2 mM) were examined. Diphenyl ditelluride, diphenyl diselenide and Ebselen inhibited in a concentration-dependent manner delta-ALA-D activity from human erythrocytes. Ebselen was lesser delta-ALA-D inhibitor than (PhSe)(2) and (PhTe)(2), whereas the diorganoyldichalcogenides displayed similar inhibitory potency towards delta-ALA-D. Dithiothreitol, a hydrophobic SH-reducing agent, was able to reactivate and to protect inhibited delta-ALA-D. The pre-incubation of blood with the inhibitors changed considerably the reversing potency of thiols. From these findings we suggest that organochalcogens inactivate in vitro human erythrocyte delta-ALA-D by an interaction with the sulfhydryl group essential of the enzyme activity.


Subject(s)
Antioxidants/toxicity , Azoles/toxicity , Benzene Derivatives/toxicity , Disulfides/toxicity , Erythrocytes/drug effects , Organometallic Compounds/toxicity , Organoselenium Compounds/toxicity , Porphobilinogen Synthase/metabolism , Azoles/antagonists & inhibitors , Azoles/blood , Benzene Derivatives/antagonists & inhibitors , Benzene Derivatives/blood , Cysteine/pharmacology , Disulfides/antagonists & inhibitors , Disulfides/blood , Dithiothreitol/pharmacology , Drug Interactions , Erythrocytes/enzymology , Glutathione Transferase/pharmacology , Humans , Isoindoles , Organometallic Compounds/antagonists & inhibitors , Organometallic Compounds/blood , Organoselenium Compounds/antagonists & inhibitors , Organoselenium Compounds/blood , Porphobilinogen Synthase/antagonists & inhibitors , Zinc/pharmacology
13.
Int Arch Occup Environ Health ; 76(5): 382-6, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12739171

ABSTRACT

OBJECTIVES: The aim of the experiments was to evaluate the efficacy of skin barrier creams (SBCs) and protective gloves and its potential for reduction of percutaneous absorption of industrial solvents. METHODS: We assessed percutaneous absorption of ethylene glycol (EG), isopropyl alcohol (IA) and 1,2,4-trimethylbenzene (TMB), using static diffusion cells. These solvents were applied neat (EG, TMB) as well as in 10% and 50% aqueous solution (EG, IA) or in 10% and 50% ethanol-diluted solution (TMB). Furthermore, we tested the percutaneous absorption of IA mixed in one cleaning agent (CA), used in newspaper printing shops to clean the rollers of printing machines. Additionally, the penetration behaviour of 10% and 50% solutions of EG, IA and TMB was tested. The experiments were carried out on untreated and on SBC-treated excised human skin from one donor, and on protective gloves. Saline was used as receptor fluid for EG and IA, and neat ethanol for TMB. RESULTS: The penetration of 50% EG, IA and TMB solutions through SBC-treated skin was higher than in untreated skin (factor 3.9 for EG, 0.32 for IA and 0.06 for TMB). The penetration of IA in the IA-CA mixture was five-times higher through untreated skin as for the single compound in 10% aqueous solution. In skin, treated with SBC, we found a 17-fold penetration enhancement of IA in the IA-CA mixture. No appreciable penetration of EG and IA was observed through nitrile rubber gloves. CONCLUSIONS: Our in vitro experiments could not demonstrate an efficacy of SBC to protect skin penetration for the tested solvents. The percutaneous absorption of all solvents in 50% solution was increased through skin treated with SBCs. Furthermore, SBCs enhance the penetration rates of solvents from complex mixtures compared with the single solvents. The tested gloves showed sufficient protection for the hydrophilic solvents, but not for TMB.


Subject(s)
2-Propanol/pharmacokinetics , Benzene Derivatives/pharmacokinetics , Dermatitis, Occupational/prevention & control , Emollients/standards , Gloves, Protective/standards , Occupational Exposure/prevention & control , Skin Absorption/physiology , Skin Care/methods , Solvents/pharmacokinetics , 2-Propanol/antagonists & inhibitors , Benzene Derivatives/antagonists & inhibitors , Emollients/pharmacokinetics , Ethylene Glycol/antagonists & inhibitors , Ethylene Glycol/pharmacokinetics , Humans , In Vitro Techniques , Permeability , Printing , Workforce
14.
Toxicol Lett ; 105(1): 25-30, 1999 Mar 08.
Article in English | MEDLINE | ID: mdl-10092053

ABSTRACT

Recent metabolic studies have demonstrated the importance of reactive intermediates like quinones or semiquinone radicals in the covalent binding of halobenzenes to liver protein. The current studies were designed to examine if quinone intermediates are involved in the toxicity of hepatotoxic halobenzenes, bromobenzene (BB) and 1,2,4-trichlorobenzene (1,2,4-TCB). Two-electron reduction of the quinone intermediates by DT-diaphorase is considered to be a detoxication pathway since the resulting hydroquinone may be readily conjugated and excreted. Mice were pretreated with butylated hydroxyanisole (BHA; 0.5% in the diet, for 3 days), an inducer of DT-diaphorase, or dicoumarol (0.3 mmol/kg, p.o.), an inhibitor of this enzyme. The mice were then given BB (2.5 or 3.5 mmol/kg, i.p.) or 1,2,4-TCB (0.75 or 1.5 mmol/kg, i.p.). Dietary BHA markedly suppressed the hepatotoxicity caused by both BB and 1,2,4-TCB while dicoumarol significantly enhanced it, as judged by serum alanine aminotransferase activity. When mice were treated with BB at different times after the end of dietary BHA exposure, the degree of the protection against the hepatotoxicity appears to correlate to the extent of the induction of DT-diaphorase activity by BHA pretreatment. BHA pretreatment failed to protect against carbon tetrachloride-induced hepatotoxicity. These results seem to provide evidence for the involvement of the quinone metabolites in BB- and 1,2,4-TCB-induced hepatotoxicity and for the protective role of DT-diaphorase against the toxicity.


Subject(s)
Benzene Derivatives/toxicity , Butylated Hydroxyanisole/pharmacology , Chemical and Drug Induced Liver Injury/prevention & control , Dicumarol/pharmacology , Enzyme Inhibitors/pharmacology , NAD(P)H Dehydrogenase (Quinone)/metabolism , Alanine Transaminase/metabolism , Animals , Benzene Derivatives/antagonists & inhibitors , Bromobenzenes/antagonists & inhibitors , Bromobenzenes/toxicity , Carbon Tetrachloride Poisoning/enzymology , Carbon Tetrachloride Poisoning/prevention & control , Chemical and Drug Induced Liver Injury/enzymology , Chlorobenzenes/antagonists & inhibitors , Chlorobenzenes/toxicity , Diet , Enzyme Induction , Inactivation, Metabolic , Liver/drug effects , Liver/enzymology , Male , Mice , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , NAD(P)H Dehydrogenase (Quinone)/biosynthesis
15.
Drugs Exp Clin Res ; 23(5-6): 157-65, 1997.
Article in English | MEDLINE | ID: mdl-9515225

ABSTRACT

The protective effect of hydroxynimesulide, the main metabolite of the nonsteroidal antiinflammatory drug nimesulide, on red blood cells (RBCs, 0.2%; 3.5 x 10(7) cell/ml) hemolysis induced by cumene hydroperoxide (CuOOH; 50 microM) was evaluated by turbidimetric and morphological analyses. Hydroxynimesulide inhibits the CuOOH-induced hemolysis in a dose dependent fashion: the protective effect, calculated after 150 min incubation (100% hemolysis in the controls), starts at 1 micron (% hemolysis 85.2 +/- 3.4%) and increases at the higher concentrations (63.5 +/- 3.9% at 5 microM; 43.5 +/- 6.3% at 10 microM; and, 14.5 +/- 4.3% at 20 microM). In addition, in the samples protected with 10 microM and 20 microM, there is a significant delay (30 and 60 min) in the onset of the hemolytic response. Inhibition of hemolysis is the result of protection of RBC membrane integrity, both on lipid (cis-Parinaric acid fluorescence quenching was delayed by 53 +/- 10 sec vs. the controls at 1 micron, by 115 +/- 15 sec at 5 microM, with a lag phase of 240 +/C- 18 sec at 10 microM) and protein constituents, as determined by SDS-PAGE electrophoresis. In hemolysis experiments, the efficacy of hydroxynimesulide is comparable to that of alpha-tocopherol and a cooperative interaction between hydroxynimesulide and alpha-tocopherol (both at 10 microM) has been observed. These results indicate that hydroxynimesulide protects RBC membranes by directly quenching reactive oxygen species generated by hemoglobin/peroxide interaction. Evidence for a direct radical scavenging intervention of the metabolite comes from HPLC studies, which demonstrate a time-dependent consumption of hydroxynimesulide, with the concomitant formation of two main reaction (addition/oxidation) products.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzene Derivatives/toxicity , Erythrocytes/drug effects , Hemoglobins/toxicity , Hemolysis/drug effects , Sulfonamides/metabolism , Sulfonamides/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Benzene Derivatives/antagonists & inhibitors , Benzene Derivatives/metabolism , Chromatography, High Pressure Liquid , Drug Interactions , Erythrocyte Membrane/drug effects , Erythrocyte Membrane/metabolism , Hemoglobins/metabolism , Male , Membrane Proteins/blood , Oxidation-Reduction , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
16.
Chem Res Toxicol ; 9(6): 924-31, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8870978

ABSTRACT

Organic hydroperoxides are believed to be primarily detoxified in cells by the GSH peroxidase/GSSG reductase system and activated to cytotoxic radical species by non-heme iron. However, organic hydroperoxides seem to be bioactivated by cytochrome P450 (P450) in isolated hepatocytes as various P450 (particularly P450 2E1) inhibitors inhibited cumene hydroperoxide (CumOOH) metabolism and attenuated subsequent cytotoxic effects including antimycin A-resistant respiration, lipid peroxidation, iron mobilization, ATP depletion, and cell membrane disruption. CumOOH metabolism was also faster in P450 1A-induced hepatocytes and was inhibited by the P450 1A inhibitor alpha-naphthoflavone. The ferric chelator deferoxamine also prevented cytotoxicity even after CumOOH had been metabolized but had no effect on CumOOH metabolism. This emphasizes the toxicological significance of the iron released following hydroperoxide metabolic activation by cytochrome P450. The radical trap, 4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl (TEMPO), had no effect on CumOOH metabolism but prevented CumOOH-induced antimycin A-resistant respiration, lipid peroxidation, iron mobilization, and loss of membrane integrity. These results suggest that CumOOH is metabolically activated by some P450 enzymes (e.g., P450 2E1) in hepatocytes to form reactive radical metabolites or oxidants that cause lipid peroxidation and cytotoxicity.


Subject(s)
Benzene Derivatives/metabolism , Cytochrome P-450 CYP2E1/metabolism , Cytochrome P-450 Enzyme System/metabolism , Liver/enzymology , Oxidants/metabolism , Animals , Benzene Derivatives/antagonists & inhibitors , Benzene Derivatives/toxicity , Biotransformation , Cell Membrane/drug effects , Chlorzoxazone/pharmacology , Deferoxamine/pharmacology , Glutathione Peroxidase/antagonists & inhibitors , Imidazoles/pharmacology , Lipid Peroxidation/drug effects , Liver/cytology , Liver/drug effects , Male , Oxidants/toxicity , Rats , Rats, Sprague-Dawley
17.
J Mol Cell Cardiol ; 26(7): 877-87, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7966356

ABSTRACT

The primary defense mechanism of myocytes against peroxide-derived free radicals is the glutathione redox cycle. The purpose of the present study was to investigate whether desferrioxamine protects myocytes against peroxide-induced cell damage, and if so, whether the turnover rate of the glutathione redox cycle is involved in this protection. Neonatal rat heart cell cultures were subjected to a standardized oxidative stress by a 90 min incubation with 80 mumol/l cumene hydroperoxide. The consequences of this stress protocol were described in terms of cellular concentrations of GSH, GSSG, ATP, ATP-catabolites, and Ca2+, formation of malondialdehyde to quantify lipid peroxidation, and enzyme release to quantify the relative number of irreversibly injured cells. Following pretreatment of cell cultures with 10 mmol/l desferrioxamine mesylate for 1 h, 80 mumol/l cumene hydroperoxide caused less malondialdehyde formation (at 90 min: 0.34 v 2.35 nmol), less ATP depletion (at 60 min: 16.7 v 3.6 nmol), less Ca2+ overload (at 30 min: 40 v 1500 nM) and less enzyme release (at 90 min: 4.6 v 60.5% of the cells) compared to cell cultures subjected to cumene hydroperoxide without pretreatment. However, in desferrioxamine pretreated cell cultures cumene hydroperoxide caused cellular GSH depletion (at 60 min: 19.5 v 20.8 nmol) and GSSG efflux (at 60 min: 6.3 v 6.0 nmol) which was not different from cell cultures subjected to cumene hydroperoxide without pretreatment. Added to the finding that in a cell-free system cumene hydroperoxide is a substrate for glutathione peroxidase, we conclude that desferrioxamine, by chelating free iron ions (1), prevented the formation of cumene alkoxyl and peroxyl radicals associated with protection of the myocytes, and (2) did not diminish rapid glutathione redox cycling leading to GSH depletion and GSSG efflux.


Subject(s)
Benzene Derivatives/antagonists & inhibitors , Deferoxamine/pharmacology , Glutathione/metabolism , Heart/drug effects , Adenosine Triphosphate/metabolism , Animals , Calcium/pharmacology , Cell Death/drug effects , Free Radicals , Lipid Peroxidation/drug effects , Myocardial Contraction/drug effects , Myocardium/cytology , Myocardium/metabolism , Oxidation-Reduction , Oxidative Stress , Rats , Rats, Wistar , Reproducibility of Results
18.
Clin Chim Acta ; 223(1-2): 1-7, 1993 Dec 31.
Article in English | MEDLINE | ID: mdl-8143355

ABSTRACT

The increase of cytosolic free calcium in human umbilical vein endothelial cells caused by peroxides was used as a model to determine and compare the putative cytoprotective properties of substances known to interfere with the generation or metabolism of reactive oxygen species. Hydrophilic hydrogen peroxide and lipophilic cumene hydroperoxide were used as sources of reactive oxygen. [Ca2+]i in endothelial cells was measured by the FURA method and the resting level was found to be 129.3 +/- 14.1 nM. Both peroxides were found to increase cytosolic calcium with dependence on the concentration and on the presence of extracellular calcium. Among the substances tested, only catalase and N-acetyl-cysteine were able to exhibit a significant cytoprotective effect.


Subject(s)
Calcium/metabolism , Cytosol/drug effects , Endothelium, Vascular/drug effects , Peroxides/antagonists & inhibitors , Antioxidants/pharmacology , Benzene Derivatives/antagonists & inhibitors , Cells, Cultured , Cytosol/metabolism , Endothelium, Vascular/metabolism , Humans , Hydrogen Peroxide/antagonists & inhibitors , Peroxides/pharmacology , Umbilical Veins/cytology
19.
Pharmacol Toxicol ; 70(6 Pt 1): 407-11, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1438018

ABSTRACT

The ability of stobadine, a novel pyridoindole antioxidant, to inhibit lipid peroxidation induced by cumene hydroperoxide was investigated in rat liver microsomes. In the micromolar range stobadine effectively inhibited lipid peroxidation as measured by the formation of thiobarbituric acid reactive products. The peroxidation-related degradation of microsomal cytochrome P-450 was prevented by stobadine in the same pattern. Another line of evidence in support of the antioxidant action of stobadine was given by its inhibition of cumene hydroperoxide-induced oxygen consumption in microsomal incubations. Inhibition of lipid peroxidation was not a function of decreased bioactivation of cumene hydroperoxide, as stobadine did not affect the rate of cytochrome P-450 dependent cleavage of cumene hydroperoxide. Neither had stobadine any effect on cytochrome P-450 peroxidase function characterized by the rate of cumene hydroperoxide-dependent oxidation of TMPD, and no direct spectral interaction with microsomal cytochrome P-450 was observed in the micromolar region. We suggest that it is the ability of stobadine to scavenge alkoxyl and peroxyl radicals that is predominantly responsible for the observed antioxidant effect.


Subject(s)
Antioxidants/pharmacology , Benzene Derivatives/antagonists & inhibitors , Carbolines/pharmacology , Indoles/pharmacology , Lipid Peroxidation/drug effects , Microsomes, Liver/drug effects , Animals , Cytochrome P-450 Enzyme System/drug effects , Kinetics , Male , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Oxygen Consumption/drug effects , Rats , Rats, Wistar , Thiobarbiturates/metabolism
20.
Biochem Biophys Res Commun ; 185(2): 698-704, 1992 Jun 15.
Article in English | MEDLINE | ID: mdl-1610362

ABSTRACT

The mechanism by which the free radical scavenger butylated hydroxytoluene (BHT) prevents cumene hydroperoxide-induced Ca2+ release from rat liver mitochondria was studied. In Ca(2+)-loaded mitochondria cumene hydroperoxide induced a rapid oxidation and subsequent hydrolysis of the pyridine nucleotides. In the presence of BHT, pyridine nucleotide oxidation by cumene hydroperoxide occurred but was reversible as hydrolysis was prevented by BHT. However, the addition of BHT directly to rat liver submitochondrial particles did not inhibit NAD+ hydrolysis or the formation of ADP-ribose from NAD+. Thus, whilst BHT prevented NAD+ hydrolysis in isolated mitochondria, this appeared not to be due to a direct effect of BHT on the NADase. It is concluded that the mechanism of action of BHT on cumene hydroperoxide-induced Ca2+ release from mitochondria involves the inhibition of pyridine nucleotide hydrolysis by an indirect mechanism rather than the radical scavenging properties of BHT.


Subject(s)
Benzene Derivatives/antagonists & inhibitors , Butylated Hydroxytoluene/pharmacology , Calcium/metabolism , Mitochondria, Liver/metabolism , NADP/metabolism , NAD/metabolism , Adenosine Diphosphate Ribose/metabolism , Animals , Hydrolysis , In Vitro Techniques , Oxidation-Reduction , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...