Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Free Radic Res ; 47(9): 740-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23805793

ABSTRACT

Bleomycin (BLM), a glycopeptide antibiotic from Streptomyces verticillus, is an effective antineoplastic drug. However, its clinical use is restricted due to the wide range of associated toxicities, especially pulmonary toxicity. Oxidative stress has been implicated as an important factor in the development of BLM-induced pulmonary toxicity. Previous studies have indicated disruption of thiol-redox status in lungs (lung epithelial cells) upon BLM treatment. Therefore, this study focused on (1) investigating the oxidative effects of BLM on lung epithelial cells (A549) and (2) elucidating whether a well-known thiol antioxidant, N-acetylcysteine amide (NACA), provides any protection against BLM-induced toxicity. Oxidative stress parameters, such as glutathione (GSH), malondialdehyde (MDA), and antioxidant enzyme activities were altered upon BLM treatment. Loss of mitochondrial membrane potential (ΔΨm), as assessed by fluorescence microscopy, indicated that cytotoxicity is possibly mediated through mitochondrial dysfunction. Pretreatment with NACA reversed the oxidative effects of BLM. NACA decreased the reactive oxygen species (ROS) and MDA levels and restored the intracellular GSH levels. Our data showed that BLM induced A549 cell death by a mechanism involving oxidative stress and mitochondrial dysfunction. NACA had a protective role against BLM-induced toxicity by inhibiting lipid peroxidation, scavenging ROS, and preserving intracellular GSH and ΔΨm. NACA can potentially be developed into a promising adjunctive therapeutic option for patients undergoing chemotherapy with BLM.


Subject(s)
Acetylcysteine/analogs & derivatives , Antioxidants/pharmacology , Bleomycin/adverse effects , Epithelial Cells/drug effects , Oxidative Stress/drug effects , Acetylcysteine/pharmacology , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Bleomycin/therapeutic use , Cell Line , Cell Survival/drug effects , Epithelial Cells/metabolism , Glutathione/metabolism , Humans , Lipid Peroxidation/drug effects , Lung/cytology , Lung/drug effects , Lung/metabolism , Reactive Oxygen Species/metabolism
2.
Hum Exp Toxicol ; 31(9): 931-44, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22354084

ABSTRACT

Methamphetamine (METH), a highly addictive drug used worldwide, induces oxidative stress in various animal organs, especially the brain. This study evaluated oxidative damage caused by METH to tissues in CD-1 mice and identified a therapeutic drug that could protect against METH-induced toxicity. Male CD-1 mice were pretreated with a novel thiol antioxidant, N-acetylcysteine amide (NACA, 250 mg/kg body weight) or saline. Following this, METH (10 mg/kg body weight) or saline intraperitoneal injections were administered every 2 h over an 8-h period. Animals were killed 24 h after the last exposure. NACA-treated animals exposed to METH experienced significantly lower oxidative stress in their kidneys, livers, and brains than the untreated group, as indicated by their levels of glutathione, malondialdehyde, and protein carbonyl and their catalase and glutathione peroxidase activity. This suggests that METH induces oxidative stress in various organs and that a combination of NACA as a neuro- or tissue-protective agent, in conjunction with current treatment, might effectively treat METH abusers.


Subject(s)
Acetylcysteine/analogs & derivatives , Antioxidants/pharmacology , Central Nervous System Stimulants/toxicity , Methamphetamine/toxicity , Oxidative Stress/drug effects , Acetylcysteine/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Catalase/metabolism , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Kidney/drug effects , Kidney/metabolism , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/metabolism , Male , Malondialdehyde/metabolism , Mice , Protein Carbonylation
3.
Toxicology ; 212(1): 1-9, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-15905016

ABSTRACT

Epidemiological data supports lead exposure as a risk factor for cataract development. Previous studies which demonstrated oxidative imbalances in the lens following in vivo Pb(2+) exposure support the idea that lead exposure can alter the lens biochemical homeostasis which may ultimately lead to loss of lens clarity with time. alpha-Crystallin, a major lens structural protein and molecular chaperone, undergoes various post-translational modifications upon aging which may contribute to decreased chaperone function and contribute to loss of lens clarity. This study evaluated the impact of 5 weeks of oral Pb(2+) exposure (peripheral Pb(2+) level approximately 30 microg/dL) on the alphaA-crystallin protein profile of the lens from Fisher 344 rats. Decreases in relative protein spot intensity of more acidic forms of alphaA- and betaA(4)-crystallin and of truncated forms of alphaA-crystallin were noted. This data indicates that changes in post-translational processing of crystallins do occur in vivo following short courses of clinically relevant Pb(2+)-exposure. In addition, organ culture of lenses from 4.5-month-old rats in 5 microM Pb(2+) resulted in opacities, demonstrating that lead is toxic to the lens and can induce a loss of lens clarity.


Subject(s)
Cataract/chemically induced , Cataract/genetics , Lens, Crystalline/drug effects , Organometallic Compounds/toxicity , Protein Array Analysis/methods , alpha-Crystallin A Chain/genetics , Animals , Cataract/metabolism , Lens, Crystalline/metabolism , Organ Culture Techniques , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , alpha-Crystallin A Chain/biosynthesis
4.
Arch Environ Contam Toxicol ; 44(3): 417-20, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12712304

ABSTRACT

Lead poisoning has been extensively studied over the years. Many adverse physiological and behavioral impacts on the human body have been reported due to the entry of this heavy metal. It especially affects the neural development of children. The current study investigates the effect of lead exposure in young (1.5 months) and adult (10 months) male Fisher 344 rats. Five weeks of lead administration resulted in a profound change in the lead levels in the red blood cells (RBCs) of the young lead-exposed group (37.0 +/- 4.47 microg/dl) compared to the control (<1 microg/dl) and adult (27.4 +/- 8.38 microg/dl) lead-exposed groups. Therefore, this study confirms the fact that gastrointestinal absorption of lead in young is greater than that of adults. Furthermore, glutathione and glutathione disulfide (GSSG) levels in RBCs, liver, and brain tissues were measured to determine thiol status; malondialdehyde (MDA) levels of lipid peroxidation and catalase activity were measured to assess changes in oxidative stress parameters. Liver GSSG and MDA levels were significantly higher in the young lead-exposed group than those in the adult lead-exposed group. In RBCs and brains, however, adult lead-exposed animals have shown more elevated MDA levels than young animals exposed to the same lead treatment.


Subject(s)
Aging/metabolism , Lead Poisoning/physiopathology , Lead/toxicity , Oxidative Stress/physiology , Animals , Brain/metabolism , Catalase/metabolism , Disease Models, Animal , Glutathione/metabolism , Glutathione Disulfide/metabolism , Intestinal Absorption , Lead/blood , Lead/pharmacokinetics , Lead Poisoning/metabolism , Liver/metabolism , Male , Oxidative Stress/drug effects , Rats , Rats, Inbred F344
5.
Biomed Chromatogr ; 15(7): 427-32, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11746237

ABSTRACT

Captopril, a well-known angiotensin converting enzyme (ACE) inhibitor, is widely used for treatment of arterial hypertension. Recent studies suggest that it may also act as a scavenger of free radicals because of its thiol group. Therefore, the present study describes a rapid, sensitive and relatively simple method for the detection of captopril in biological tissues with reverse-phase HPLC. Captopril was first derivatized with ThioGlo 3 [3H-Naphto[2,1-b]pyran,9-acetoxy-2-(4-(2,5-dihydro-2,5-dioxo-1H-pyrrol-1-yl)phenyl-3-oxo-)]. It was then detected by fluorescence-HPLC using an Astec C(18) column as the stationary phase and a water:acetonitrile:acetic acid:phosphoric acid mixture (50:50; 1 mL/L acids) as the mobile phase (excitation wavelength, 365 nm; emission wavelength, 445 nm). The calibration curve for captopril was linear over a range of 10-2500 nM and the coefficient of variation acquired for the within- and between-run precision for captopril was 0.5 and 3.8%, respectively. The detection limit of captopril with this method was found to be 200 fmol/20 microL injection volume. Its relative recovery from biological samples was determined to the range from 93.3 to 105.3%. Based on these results, we believe that our method is advantageous for captopril determination.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/analysis , Captopril/analysis , Chromatography, High Pressure Liquid/methods , Fluorescent Dyes , Pyrans , Pyrroles , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/blood , Animals , Calibration , Captopril/administration & dosage , Captopril/blood , Kidney/chemistry , Liver/chemistry , Lung/chemistry , Male , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity
6.
Arch Environ Contam Toxicol ; 41(4): 397-402, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11598776

ABSTRACT

Oxidative stress is proposed as a molecular mechanism in lead toxicity, which suggests that antioxidants might play a role in the treatment of lead poisoning. The present study was designed to investigate whether taurine has a beneficial effect both on Chinese hamster ovary (CHO) cells and on Fisher 344 (F344) rats following lead exposure. Therefore, oxidative stress parameters (glutathione, malondialdehyde levels, catalase, and glucose-6-phosphate dehydrogenase [G6PD] activities) of lead-exposed CHO cells and F344 rats were determined following taurine treatment. Taurine was found to be effective in (1) increasing glutathione levels that had been diminished by lead; (2) reducing malondialdehyde levels, an end-product of lipid peroxidation; (3) decreasing catalase and erythrocyte G6PD activity, which had been increased by lead exposure; and (4) improving cell survival of CHO cells. However, taurine had no effect on blood and tissue lead levels when 1.1 g/kg/day taurine was administered to F344 rats for 7 days, following 5 weeks of lead exposure (2,000 ppm lead acetate). As a result, taurine seems to be capable of fortifying cells against lead-induced oxidative attack without decreasing lead levels. Therefore, administration of taurine, accompanied by a chelating agent, might increase its effectiveness in the treatment of lead poisoning.


Subject(s)
Antioxidants/pharmacology , Lead Poisoning/drug therapy , Oxidative Stress , Taurine/pharmacology , Animals , CHO Cells , Chelating Agents/pharmacology , Cricetinae , Disease Models, Animal , Male , Rats , Rats, Inbred F344
7.
J Chromatogr B Biomed Sci Appl ; 753(2): 287-92, 2001 Apr 05.
Article in English | MEDLINE | ID: mdl-11334342

ABSTRACT

The importance of thiols has stimulated the development of a number of methods for determining glutathione and other biologically significant thiols. Methods that are currently available, however have some limitations, such as being time consuming and complex. In the present study, a new high-performance liquid chromatography (HPLC) method for determining biological thiols was developed by using 9-Acetoxy-2-(4-(2,5-dihydro-2,5-dioxo-1H-pyrrol-1-yl)phenyl)-3-oxo-3H-naphtho[2,1-b]pyran (ThioGlo3) as a derivatizing agent. ThioGlo reacts selectively and rapidly with the thiols to yield fluorescent adducts which can be detected fluorimetrically (lambda(ex) = 365 nm, lambda(em) = 445 nm). The within-run coefficient of variation for glutathione (GSH) by this method ranges from 1.08 to 2.94% whereas the between-run coefficient of variation for GSH is 4.31-8.61%. For GSH, the detection limit is around 50 fmol and the GSH derivatives remain stable for 1 month, if kept at 4 degrees C. Results for GSSG and cysteine are also included. The ThioGlo method is compared to our previous method in which N-(1-pyrenyl)maleimide (NPM) is used to derivatize thiol-containing compounds. The present method offers various advantages over the currently accepted techniques, including speed and sensitivity.


Subject(s)
Chromatography, High Pressure Liquid/methods , Indicators and Reagents/chemistry , Sulfhydryl Compounds/analysis , Animals , Calibration , Fluorescent Dyes , Pyrans , Pyrroles , Rats , Rats, Inbred F344 , Reproducibility of Results , Sensitivity and Specificity
8.
Curr Top Med Chem ; 1(6): 529-39, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11895129

ABSTRACT

Toxic metals (lead, cadmium, mercury and arsenic) are widely found in our environment. Humans are exposed to these metals from numerous sources, including contaminated air, water, soil and food. Recent studies indicate that transition metals act as catalysts in the oxidative reactions of biological macromolecules therefore the toxicities associated with these metals might be due to oxidative tissue damage. Redox-active metals, such as iron, copper and chromium, undergo redox cycling whereas redox-inactive metals, such as lead, cadmium, mercury and others deplete cells' major antioxidants, particularly thiol-containing antioxidants and enzymes. Either redox-active or redox-inactive metals may cause an increase in production of reactive oxygen species (ROS) such as hydroxyl radical (HO.), superoxide radical (O2.-) or hydrogen peroxide (H2O2). Enhanced generation of ROS can overwhelm cells' intrinsic antioxidant defenses, and result in a condition known as "oxidative stress". Cells under oxidative stress display various dysfunctions due to lesions caused by ROS to lipids, proteins and DNA. Consequently, it is suggested that metal-induced oxidative stress in cells can be partially responsible for the toxic effects of heavy metals. Several studies are underway to determine the effect of antioxidant supplementation following heavy metal exposure. Data suggest that antioxidants may play an important role in abating some hazards of heavy metals. In order to prove the importance of using antioxidants in heavy metal poisoning, pertinent biochemical mechanisms for metal-induced oxidative stress should be reviewed.


Subject(s)
Metals, Heavy/toxicity , Oxidative Stress/drug effects , Animals , Antioxidants/metabolism , Environmental Pollutants/pharmacology , Environmental Pollutants/toxicity , Humans , Metals, Heavy/pharmacology , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism
9.
Biomed Chromatogr ; 14(8): 535-40, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11113937

ABSTRACT

D-Penicillamine (2-amino-3-mercapto-3-methylbutanoic acid), a well-known heavy metal chelator, is the drug of choice in the treatment of Wilson's disease and is also effective for the treatment of several disorders including rheumatoid arthritis, primary biliary cirrhosis, scleroderma, fibrotic lung diseases and progressive systemic sclerosis. The method proposed incorporates a technique, previously developed in our laboratory, that utilizes the derivatizing agent N-(1-pyrenyl)maleimide (NPM) and reversed-phase high-performance liquid chromatography (HPLC). The coefficients of variation for within-run precision and between-run precision for 500 nM standard D-penicillamine (D-pen) were 2.27% and 2.23%, respectively. Female Sprague-Dawley rats were given 1 g/kg D-pen i.p. and the amounts of D-pen in liver, kidney, brain and plasma were subsequently analyzed. This assay is rapid, sensitive and reproducible for determining D-pen in biological samples.


Subject(s)
Chromatography, High Pressure Liquid/methods , Maleimides/chemistry , Penicillamine/analysis , Animals , Brain Chemistry , Calibration , Female , Kidney/chemistry , Liver/chemistry , Male , Penicillamine/blood , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity
10.
Free Radic Biol Med ; 29(10): 927-45, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11084283

ABSTRACT

Recent studies have shown that lead causes oxidative stress by inducing the generation of reactive oxygen species, reducing the antioxidant defense system of cells via depleting glutathione, inhibiting sulfhydryl-dependent enzymes, interfering with some essential metals needed for antioxidant enzyme activities, and/or increasing susceptibility of cells to oxidative attack by altering the membrane integrity and fatty acid composition. Consequently, it is plausible that impaired oxidant/antioxidant balance can be partially responsible for the toxic effects of lead. Where enhanced oxidative stress contributes to lead-induced toxicity, restoration of a cell's antioxidant capacity appears to provide a partial remedy. Several studies are underway to determine the effect of antioxidant supplementation following lead exposure. Data suggest that antioxidants may play an important role in abating some hazards of lead. To explain the importance of using antioxidants in treating lead poisoning the following topics are addressed: (i) Oxidative damage caused by lead poisoning; (ii) conventional treatment of lead poisoning and its side effects; and (iii) possible protective effects of antioxidants in lead toxicity.


Subject(s)
Antioxidants/therapeutic use , Lead Poisoning/drug therapy , Aminolevulinic Acid/metabolism , Animals , Antioxidants/metabolism , Cell Membrane/drug effects , Chelating Agents/adverse effects , Chelating Agents/therapeutic use , Hemoglobins/drug effects , Humans , Lead/toxicity , Lead Poisoning/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
11.
Arch Environ Contam Toxicol ; 39(2): 251-6, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10871428

ABSTRACT

Evidence implicating oxidative stress in toxicity during lead intoxication in vivo has opened new avenues for investigation of the mechanisms of lead-induced immunosuppression. The current study explores the possibility that lead-induced oxidative stress contributes to the immunosuppression observed during lead poisoning. Fisher 344 rats were exposed to 2,000 ppm lead acetate in their drinking water for 5 weeks. One week following removal of lead from the drinking water, significant reductions in serum levels of IgA, IgM, and IgG were detected. Significant increases in oxidative damage, based on malondialdehyde (MDA) content, were observed in peripheral blood mononuclear cells (PMCs) collected during the same experiments. In addition, MDA content increased in livers from lead-exposed rats. Following 5 weeks of lead exposure, administration of either 5.5 mmol/kg N-acetylcysteine (NAC) or 1 mmol/kg meso-2,3-dimercaptosuccinic acid (DMSA) in the drinking water for 1 week significantly reversed the inhibitory effects of lead on serum immunoglobulin (Ig) levels. Also, all parameters indicative of oxidative stress returned to control levels. These results suggest that oxidative stress contributes to suppressed serum Ig levels during lead intoxication in vivo, and that intervention with either a thiol antioxidant (NAC) or a metal chelator (DMSA) will alleviate this lead-induced suppression by correcting the prooxidant/antioxidant imbalance caused by lead exposure.


Subject(s)
Immunoglobulins/drug effects , Immunosuppression Therapy , Immunosuppressive Agents/toxicity , Lead Poisoning/immunology , Lead/toxicity , Oxidative Stress , Animals , Antibody Formation/drug effects , Body Weight/drug effects , Catalase/metabolism , Chromatography, High Pressure Liquid , Immunodiffusion , Immunoglobulins/blood , Lead/metabolism , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/metabolism , Male , Malondialdehyde/metabolism , Monocytes/drug effects , Monocytes/metabolism , Oxidative Stress/physiology , Rats , Rats, Inbred F344
12.
Arch Environ Contam Toxicol ; 37(4): 434-9, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10508890

ABSTRACT

The toxicities and oxidative stress-inducing actions of (-)-nicotine and smokeless tobacco extract (STE), containing equivalent amounts of nicotine, were studied. Toxicities were determined by colony formation assays using Chinese hamster ovary (CHO) cells. Results indicated that nicotine is less toxic than smokeless tobacco extract that contained the same amount of nicotine. The generation of reactive oxygen species, following treatment with smokeless tobacco extract and nicotine, was assessed by measurement of changes in glutathione (GSH) and malondialdehyde (MDA) levels. CHO cells (5 x 10(5) cells/5 ml media) were incubated with 4, 0.8, and 0.08 mg of nicotine and STE containing the same amounts of nicotine. All preparations of smokeless tobacco extract significantly decreased GSH levels and increased MDA generation. However, 0.08 mg of nicotine treatment did not result in a significant change in GSH level, and only 4 mg of nicotine were sufficient to increase MDA generation. Addition of free radical scavenging enzymes, superoxide dismutase (SOD) and catalase (CAT), and an intracellular GSH precursor, N-acetyl-L-cysteine (NAC), replenished the GSH levels in nicotine-treated cells. GSH levels in cells exposed to smokeless tobacco extract containing 4 and 0.8 mg nicotine remained significantly lower than the control with the addition of SOD and CAT. However, co-addition of NAC with smokeless tobacco extract preparations returned the GSH levels to the control level. Lactate dehydrogenase (LDH) activities were measured in the media to establish the membrane damage following exposure to smokeless tobacco extract and nicotine. Treatment of cells with 4 mg nicotine caused a significant increase in LDH activity, which was returned to control level in the presence of the antioxidant enzymes and NAC. Smokeless tobacco extract did not change the LDH activity. http://link.springer-ny. com/link/service/journals/00244/bibs/37n4p434.html

Subject(s)
Nicotine/toxicity , Oxidative Stress/drug effects , Plants, Toxic , Tobacco, Smokeless/toxicity , 3,4-Methylenedioxyamphetamine/metabolism , Acetylcysteine/metabolism , Animals , CHO Cells , Catalase/metabolism , Cell Survival/drug effects , Cricetinae , Glutathione/metabolism , L-Lactate Dehydrogenase/metabolism , Plant Extracts/toxicity , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
13.
Free Radic Biol Med ; 27(1-2): 75-81, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10443922

ABSTRACT

The assumption of oxidative stress as a mechanism in lead toxicity suggests that antioxidants might play a role in the treatment of lead poisoning. The present study was designed to investigate the efficacy of lipoic acid (LA) in rebalancing the increased prooxidant/antioxidant ratio in lead-exposed Chinese hamster ovary (CHO) cells and Fischer 344 rats. Furthermore, LA's ability to decrease lead levels in the blood and tissues of lead-treated rats was examined. LA administration resulted in a significant improvement in the thiol capacity of cells via increasing glutathione levels and reducing malondialdehyde levels in the lead-exposed cells and animals, indicating a strong antioxidant shift on lead-induced oxidative stress. Furthermore, administration of LA after lead treatment significantly decreased catalase and red blood cell glucose-6-phosphate dehydrogenase activity. In vitro administration of LA to cultures of CHO cells significantly increased cell survival, that was inhibited by lead treatment in a concentration-dependent manner. Administration of LA was not effective in decreasing blood or tissue lead levels compared to a well-known chelator, succimer, that was able to reduce them to control levels. Hence, LA seems to be a good candidate for therapeutic intervention of lead poisoning, in combination with a chelator, rather than as a sole agent.


Subject(s)
Antioxidants , Lead Poisoning , Thioctic Acid/physiology , Animals , CHO Cells , Catalase/metabolism , Cricetinae , Glucosephosphate Dehydrogenase/metabolism , Glutathione/metabolism , Lead/metabolism , Malondialdehyde/metabolism , Rats , Rats, Inbred F344 , Thioctic Acid/administration & dosage
14.
Toxicol Lett ; 106(1): 41-7, 1999 May 20.
Article in English | MEDLINE | ID: mdl-10378449

ABSTRACT

5-Aminolevulinic acid (ALA) is a heme precursor that accumulates in acute intermittent porphyria and lead poisoning. It has been shown that ALA induces free radical generation and may cause damage to proteins and DNA. In the present study, the effects of ALA on DNA damage and its prevention by N-acetyl-L-cysteine (NAC) and the antioxidant enzymes catalase (CAT) and superoxide dismutase (SOD) are investigated. Oxidative damage to DNA was quantitated by measuring the increase in 8-hydroxy-2'-deoxyguanosine (oh8dG) formation. The time-course study demonstrated that ALA causes a linear increase in oh8dG levels in Chinese hamster ovary (CHO) cells. However, direct lead exposure did not cause any measurable increase in oh8dG levels. In the presence of either NAC (1 mM) or antioxidant enzymes (10 u/ml SOD and 10 u/ml CAT), oh8dG levels returned to the corresponding control levels. This suggests a protective role for NAC and the antioxidant enzymes. To determine the effect of ALA on cell proliferation, cell numbers were counted at the end of 24 h of incubation in the presence and absence of ALA at different concentrations. Results showed that levels of ALA up to 5 mM do not inhibit cell proliferation.


Subject(s)
Acetylcysteine/pharmacology , Aminolevulinic Acid/toxicity , Guanosine/analogs & derivatives , Animals , CHO Cells , Catalase/pharmacology , Cricetinae , DNA Damage , Guanosine/metabolism , Oxidation-Reduction , Superoxide Dismutase/pharmacology
15.
Hum Exp Toxicol ; 18(1): 27-32, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10025365

ABSTRACT

Recent studies suggest that lead induces oxidative stress in various tissues. Captopril ([2S]-1-[3-mercapto-2-methylpropionyl]-L-proline), an angiotensin-converting enzyme inhibitor, is a well-known antihypertensive agent and is also believed to function as an antioxidant. In the present study the antioxidant effect of captopril on lead-induced oxidative stress was studied in Fischer 344 rats. Their liver, brain and kidneys were assayed for glutathione (GSH), glutathione disulfide (GSSG), malondialdehyde concentrations, and catalase activities. The results from animals treated with captopril were compared to results of control and lead-exposed non-treated groups. The captopril-treated samples showed higher GSH:GSSG ratios in the liver, brain and kidneys, as well as slightly decreased malondialdehyde concentrations. The catalase activity was not significantly affected.


Subject(s)
Antioxidants/pharmacology , Brain Chemistry/drug effects , Captopril/pharmacology , Kidney/drug effects , Lead/toxicity , Liver/drug effects , Oxidative Stress/drug effects , 3,4-Methylenedioxyamphetamine/analysis , Animals , Catalase/drug effects , Catalase/metabolism , Glutathione/analysis , Glutathione/drug effects , Glutathione Disulfide/analysis , Glutathione Disulfide/drug effects , Lead/blood , Male , Molecular Structure , Rats , Rats, Inbred F344
17.
Free Radic Biol Med ; 26(1-2): 239-43, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9890658

ABSTRACT

Lead (Pb) is known to negatively affect glutathione (GSH) metabolism in the lens. The present study examined the effects of Captopril, Taurine, and alpha-Lipoic acid on the Pb-induced GSH depletion and lipid peroxide increase in the lenticular system. Captopril administration returned the GSH, cysteine (CYS), and malondialdehyde (MDA) levels to near normal. Following Taurine administration the GSH, CYS and MDA levels were intermediate between the control group and the Pb group levels. Alpha-Lipoic acid administration, however, only increased the CYS levels. No significant changes in oxidized glutathione (GSSG) levels were observed in any treatment group.


Subject(s)
Antioxidants/pharmacology , Lead/toxicity , Lens, Crystalline/drug effects , Lens, Crystalline/metabolism , Animals , Antioxidants/chemistry , Captopril/pharmacology , Cysteine/metabolism , Glutathione/metabolism , Glutathione Disulfide/metabolism , Lipid Peroxides/metabolism , Male , Malondialdehyde/metabolism , Oxidative Stress/drug effects , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism , Sulfur/chemistry , Taurine/pharmacology , Thioctic Acid/pharmacology
18.
Toxicology ; 130(2-3): 155-65, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9865482

ABSTRACT

Nicotine affects a variety of cellular processes ranging from induction of gene expression to secretion of hormones and modulation of enzymatic activities. The objective of this study was to characterize the toxicity of nicotine enantiomers as well as their ability to induce oxidative stress in an in vitro model using Chinese hamster ovary (CHO) cells. Colony formation assay has demonstrated that (-)-nicotine is the more toxic of the enantiomers. At 6 mM concentrations, (-)-nicotine was found to be approximately 28- and 19-fold more potent than (+)-, and (+/-)-nicotine (racemic), respectively. Results also indicated that the toxicity of (+/-)-nicotine is higher than that of (+)-nicotine. (-)-Nicotine at a 10 mM concentration substantially decreased glutathione (GSH) levels (46% decrease). In addition, a 3-fold increase in malondialdehyde (MDA) level was evident in cells after exposure to 10 mM (-)-nicotine. Increased lactate dehydrogenase (LDH) activities in the media demonstrated that cellular membrane integrity was disturbed in nicotine treated cells. In the presence of superoxide dismutase (SOD) and catalase (CAT), the LDH activities returned to control value in 24 h with all concentrations of (-)-, (+)-, and (+/-)-nicotine. The decreases in LDH activities in the presence of the radical scavenging enzymes SOD and CAT suggest that membrane damage may be due to free radical generation.


Subject(s)
CHO Cells/drug effects , Nicotine/toxicity , Oxidative Stress/drug effects , Animals , CHO Cells/metabolism , Catalase/pharmacology , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Cricetinae , Glutathione/metabolism , L-Lactate Dehydrogenase/metabolism , Malondialdehyde/metabolism , Nicotine/analogs & derivatives , Stereoisomerism , Superoxide Dismutase/pharmacology
19.
Toxicology ; 130(2-3): 167-74, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9865483

ABSTRACT

Lead (Pb) is known to disrupt the pro-oxidant/anti-oxidant balance of tissues which leads to biochemical and physiological dysfunction. The present study investigated the effects of exposure on the redox status of the lenses of Fisher 344 rats and examined whether antioxidant or chelator administration reversed these changes. Animals were given 5 weeks of 2000 ppm Pb exposure followed by 1 week of either antioxidant, chelator or distilled water administration. Glutathione (GSH) and cysteine (CYS) levels decreased in the Pb-exposed group. N-acetylcysteine or 2,3-dimercaptopsuccinic acid (Succimer) supplementation following Pb intoxication resulted in increases in the GSH and CYS levels. Protein bound glutathione (PSSG) and cysteine (PSSC) increased following Pb exposure. In the Succimer-treated animals, the PSSG decreased significantly. The glutathione disulfide (GSSG) levels remained unchanged. Malondialdehyde (MDA) levels, a major lipid peroxidation byproduct, increased following Pb exposure and decreased following Succimer treatment. Our results suggest that antioxidant supplementation, as well as chelation, following Pb exposure may enhance the reductive status of lenses.


Subject(s)
Acetylcysteine/pharmacology , Chelating Agents/pharmacology , Free Radical Scavengers/pharmacology , Lens, Crystalline/drug effects , Organometallic Compounds/toxicity , Oxidative Stress/drug effects , Succimer/pharmacology , Animals , Chromatography, High Pressure Liquid , Cysteine/metabolism , Glutathione/metabolism , Glutathione Disulfide/metabolism , Lens, Crystalline/metabolism , Male , Malondialdehyde/metabolism , Organometallic Compounds/blood , Rats , Rats, Inbred F344
20.
Toxicology ; 128(3): 181-9, 1998 Jul 17.
Article in English | MEDLINE | ID: mdl-9750041

ABSTRACT

This study examined whether lead-induced alterations in selected parameters that are indicative of oxidative stress accompany the toxic effects of lead in red blood cells (RBCs) in vivo. It also explored the possibility that treatment with N-acetylcysteine (NAC) or succimer (meso-2,3-dimercaptosuccinic acid) was capable of reversing parameters indicative of lead-induced oxidative stress. Fisher 344 rats were given 2000 ppm lead acetate in their drinking water for 5 weeks. The lead was then removed and the animals were given NAC (800 mg/kg/day) or succimer (90 mg/kg/day) in their drinking water for 1 week, after which the RBCs were harvested. Animals not given lead and those given lead, but not NAC or succimer, served as negative and positive controls, respectively. At the end of the experiment, blood-lead levels were 35 +/- 4 microg/dl in lead-treated animals, which were reduced to 2.5 +/- 1 microg/dl by treatment with succimer and to 25 +/- 3 microg/dl by treatment with NAC. Lead-exposed animals demonstrated signs of anemia as evidenced by anisocytosis, poikilocytosis, and alterations in hemoglobin, hematocrit, and mean corpuscular volume. Lipid peroxidation, as evidenced by increased malondialdehyde (MDA) content, as well as decreases in reduced glutathione (GSH) and increases in catalase and glucose 6-phosphate dehydrogenase (G6PD) activity were noted in RBCs from lead-treated rats, suggesting that the lead induced oxidative stress. In addition, a significant reduction in blood delta-aminolevulinic acid dehydratase (ALAD) activity suggested that accumulation and autooxidation of delta-aminolevulinic acid might contribute to lead-induced oxidative stress. Treatment with either NAC or succimer reversed lead-induced alterations in MDA and GSH content, but only succimer appeared to partially restore ALAD activity. These results provide in vivo evidence supporting the hypothesis that lead induces oxidative stress in RBCs, which is reversible by treatment with a thiol antioxidant (NAC), as well as a chelating agent (succimer).


Subject(s)
Acetylcysteine/pharmacology , Antioxidants/pharmacology , Erythrocytes/metabolism , Lead/toxicity , Succimer/pharmacology , Animals , Blood Cell Count/drug effects , Erythrocytes/drug effects , Erythrocytes/enzymology , Glutathione/blood , Lead/blood , Male , Malondialdehyde/blood , Oxidative Stress/drug effects , Rats , Rats, Inbred F344
SELECTION OF CITATIONS
SEARCH DETAIL
...