Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters

Publication year range
1.
J Cell Physiol ; 234(5): 7384-7394, 2019 05.
Article in English | MEDLINE | ID: mdl-30362550

ABSTRACT

Excess reactive oxygen species (ROS) generated in embryos during in vitro culture damage cellular macromolecules and embryo development. Glutathione (GSH) scavenges ROS and optimizes the culture system. However, how exogenous GSH influences intracellular GSH and improves the embryo developmental rate is poorly understood. In this study, GSH or GSX (a stable GSH isotope) was added to the culture media of bovine in vitro fertilization embryos for 7 days. The cleavage rate, blastocyst rate, and total cell number of blastocysts were calculated. Similarly to GSH, GSX increased the in vitro development rate and embryo quality. We measured intracellular ROS, GSX, and GSH for 0-32-hr postinsemination (hpi) in embryos (including zygotes at G1, S, and G2 phases and cleaved embryos) cultured in medium containing GSX. Intracellular ROS significantly decreased with increasing intracellular GSH in S-stage zygotes (18 hpi) and cleaved embryos (32 hpi). ƎĀ³-Glutamyltranspeptidase ( GGT) and glutathione synthetase ( GSS) messenger RNA expression increased in zygotes (18 hpi) and cleaved embryos treated with GSH, consistent with the tendency of overall GSH content. GGT activity increased significantly in 18 hpi zygotes. GGT and GCL enzyme inhibition with acivicin and buthionine sulfoximine, respectively, decreased cleavage rate, blastocyst rate, total cell number, and GSH and GSX content. All results indicated that exogenous GSH affects intracellular GSH levels through the ƎĀ³-glutamyl cycle and improves early embryo development, enhancing our understanding of the redox regulation effects and transport of GSH during embryo culture in vitro.


Subject(s)
Cleavage Stage, Ovum/drug effects , Glutathione Synthase/metabolism , Glutathione/pharmacology , Zygote/drug effects , gamma-Glutamyltransferase/metabolism , Animals , Cattle , Cleavage Stage, Ovum/metabolism , Embryo Culture Techniques , Enzyme Inhibitors/pharmacology , Female , Fertilization in Vitro , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Glutathione/metabolism , Glutathione Synthase/antagonists & inhibitors , Glutathione Synthase/genetics , Male , Oxidation-Reduction , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Time Factors , Zygote/metabolism , gamma-Glutamyltransferase/antagonists & inhibitors , gamma-Glutamyltransferase/genetics
2.
Toxicol Ind Health ; 32(1): 162-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-24081639

ABSTRACT

This article reports in silico analysis of methyl isocyanate (MIC) on different key immune proteins against Mycobacterium tuberculosis. The analysis shows that MIC is released in the Bhopal gas tragedy in 1984, which is highly toxic and extremely hazardous to human health. In this study, we have selected immune proteins to perform molecular docking with the help of Autodock 4.0. Results show that the CD40 ligand and alpha5beta1 integrin have higher inhibition compared to plasminogen activator urokinase, human glutathione synthetase, mitogen-activated protein kinase (P38 MAPK 14), surfactant protein-B, -D (SP-D), and pulmonary SP-D. MIC interacted with His-125, Try-146 residue of CD40 ligand and Ala-149, and Arg-152 residue of alpha5beta1 integrin and affects the proteins functioning by binding on their active sites. These inhibitory conformations were energetically and statistically favored and supported the evidence from wet laboratory experiments reported in the literature. We can conclude that MIC directly or indirectly affects these proteins, which shows that survivals of the disaster suffer from the diseases like tuberculosis infection and lung cancer.


Subject(s)
CD40 Ligand/antagonists & inhibitors , Immune System/drug effects , Integrin alpha5beta1/antagonists & inhibitors , Isocyanates/toxicity , Molecular Docking Simulation , Glutathione Synthase/antagonists & inhibitors , Humans , Lung Neoplasms , Pulmonary Surfactant-Associated Protein B/antagonists & inhibitors , Pulmonary Surfactant-Associated Protein D/antagonists & inhibitors , Tuberculosis , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
3.
Eur J Oral Sci ; 123(4): 282-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25968591

ABSTRACT

2-Hydroxyethyl methacrylate (HEMA) is a methacrylate monomer used in polymer-based dental-restorative materials. In this study, the viability of human lung epithelial cells, BEAS-2B, was investigated after exposure to this monomer. Exposure to HEMA reduced the viability of the BEAS-2B cells as a result of increased apoptosis, interruption of the cell cycle, and decreased cell proliferation. Depletion of cellular glutathione and increased levels of reactive oxygen species (ROS) were seen after exposure of BEAS-2B cells to HEMA. The glutathione synthase inhibitor, L-buthioninesulfoximine (BSO), was used to study whether the reduced viability was caused by glutathione depletion and increased levels of ROS. Similarly to incubation with HEMA, incubation with BSO resulted in glutathione depletion and increased ROS levels, without increasing cell death or inhibiting cell growth. The results indicate that HEMA-induced cell damage is not caused exclusively by these mechanisms. Mechanisms other than glutathione depletion and ROS formation seem to be of importance for the toxic effect of HEMA on lung epithelial cells.


Subject(s)
Lung/drug effects , Methacrylates/toxicity , Oxidative Stress/physiology , Apoptosis/drug effects , Buthionine Sulfoximine/pharmacology , Cell Cycle/drug effects , Cell Death/drug effects , Cell Division/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Glutathione/drug effects , Glutathione Synthase/antagonists & inhibitors , Humans , Lung/cytology , Materials Testing , Reactive Oxygen Species/analysis
4.
Mol Cell Biochem ; 360(1-2): 159-68, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21918827

ABSTRACT

Increased arginase activity in the vasculature has been implicated in the regulation of nitric oxide (NO) homeostasis, leading to the development of vascular disease and the promotion of tumor cell growth. Recently, we showed that cysteine, in the presence of iron, promotes arginase activity by driving the Fenton reaction. In the present report, we showed that induction of oxidative stress in erythroleukemic cells with the thiol-specific oxidant, diamide, led to an increase in arginase activity by 42% (P = 0.02; vs. control). By using specific antibodies, it was demonstrated that this increase correlated with an increase in arginase-1 levels in the cells and with corresponding decreases in glutathione and protein thiol levels. Treatment of cells with aurothiomalate (ATM), a protein thiol-complexing agent, diminished the activity of arginase and arginase-1 levels by 19.5 and 35.2%, respectively (vs. control) and significantly decreased both glutathione and protein thiol levels, further implicating the thiol redox system in the cellular activation of arginase. Furthermore, diamide significantly altered the kinetics of arginase, resulting in the doubling of its V(max) (vs. control). Our presented data demonstrate, for the first time that the intracellular arginase activation is may be enhanced in part, via a cellular thiol-mediated mechanism.


Subject(s)
Arginase/metabolism , Cysteine/metabolism , Diamide/pharmacology , Enzyme Activation/drug effects , Oxidants/pharmacology , Animals , Arginase/isolation & purification , Buthionine Sulfoximine/pharmacology , Cattle , Cell Line, Tumor , Cell Survival/drug effects , Glutathione/metabolism , Glutathione Synthase/antagonists & inhibitors , Humans , Kinetics , Ornithine/biosynthesis , Oxidation-Reduction , Oxidative Stress
5.
New Phytol ; 192(2): 496-506, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21726232

ABSTRACT

Legumes form a symbiotic interaction with bacteria of the Rhizobiaceae family to produce nitrogen-fixing root nodules under nitrogen-limiting conditions. We examined the importance of glutathione (GSH) and homoglutathione (hGSH) during the nitrogen fixation process. Spatial patterns of the expression of the genes involved in the biosynthesis of both thiols were studied using promoter-GUS fusion analysis. Genetic approaches using the nodule nitrogen-fixing zone-specific nodule cysteine rich (NCR001) promoter were employed to determine the importance of (h)GSH in biological nitrogen fixation (BNF). The (h)GSH synthesis genes showed a tissue-specific expression pattern in the nodule. Down-regulation of the ƎĀ³-glutamylcysteine synthetase (ƎĀ³ECS) gene by RNA interference resulted in significantly lower BNF associated with a significant reduction in the expression of the leghemoglobin and thioredoxin S1 genes. Moreover, this lower (h)GSH content was correlated with a reduction in the nodule size. Conversely, ƎĀ³ECS overexpression resulted in an elevated GSH content which was correlated with increased BNF and significantly higher expression of the sucrose synthase-1 and leghemoglobin genes. Taken together, these data show that the plant (h)GSH content of the nodule nitrogen-fixing zone modulates the efficiency of the BNF process, demonstrating their important role in the regulation of this process.


Subject(s)
Glutathione/analogs & derivatives , Medicago truncatula/metabolism , Nitrogen Fixation/physiology , Down-Regulation , Gene Expression Profiling , Gene Expression Regulation, Plant , Glutathione/biosynthesis , Glutathione/metabolism , Glutathione Synthase/antagonists & inhibitors , Medicago truncatula/genetics , Medicago truncatula/microbiology , Nitrogen Fixation/genetics , Plant Growth Regulators/metabolism , Plant Roots/metabolism , Plants, Genetically Modified , Root Nodules, Plant/genetics , Root Nodules, Plant/metabolism , Root Nodules, Plant/microbiology , Sinorhizobium meliloti/metabolism , Symbiosis/genetics , Symbiosis/physiology
6.
Int J Biol Macromol ; 161: 1230-1239, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32544581

ABSTRACT

Polydatin (PD) is a bio-active ingredient with known anti-tumor effects. However, its specific protein targets yet have not been systematically screened, and the molecular anti-tumor mechanism is still unclear. Here, proteomic-chip was efficiently used to screen potential targets of PD. First, we investigated through animal experiment and proteomics studies, and found that polydatin play an important role in tumor cells. Then, the red-green fluorescent of polydatin was compared comprehensively to screen its targets on chip, followed by bioinformatics analysis. Glutathione synthetase (GSS) was selected as candidate research target. After a series of molecular biological experiments GSS was confirmed a target protein for PD in vitro. Moreover, we also found that PD can significantly inhibit the activity of GSS in vitro and live cells. Our findings reveal that PD could be a selective small-molecule GSS enzyme activity inhibitor and GSS could be a potential therapeutic target in cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Glucosides/pharmacology , Glutathione Synthase/antagonists & inhibitors , High-Throughput Screening Assays , Proteome , Proteomics , Stilbenes/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans
7.
J Cell Biochem ; 108(2): 424-32, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19623661

ABSTRACT

Pyrrolizidine alkaloid (PA) clivorine, isolated from traditional Chinese medicinal plant Ligularia hodgsonii Hook, has been shown to induce apoptosis in hepatocytes via mitochondrial-mediated apoptotic pathway in our previous research. The present study was designed to observe the protection of N-acetyl-cysteine (NAC) on clivorine-induced hepatocytes apoptosis. Our results showed that 5 mM NAC significantly reversed clivorine-induced cytotoxicity via MTT and Trypan Blue staining assay. DNA apoptotic fragmentation analysis and Western-blot results showed that NAC decreased clivorine-induced apoptotic DNA ladder and caspase-3 activation. Further results showed that NAC inhibited clivorine-induced Bcl-xL decrease, mitochondrial cytochrome c release and caspase-9 activation. Intracellular glutathione (GSH) is an important ubiquitous redox-active reducing sulfhydryl (--SH) tripeptide, and our results showed that clivorine (50 microM) decreased cellular GSH amounts and the ratio of GSH/GSSG in the time-dependent manner, while 5 mM NAC obviously reversed this depletion. Further results showed that GSH synthesis inhibitor BSO augmented clivorine-induced cytotoxicity, while exogenous GSH reversed its cytotoxicity on hepatocytes. Clivorine (50 microM) significantly induced cellular reactive oxygen species (ROS) generation. Further results showed that 50 microM Clivorine decreased glutathione peroxidase (GPx) activity and increased glutathione S transferase (GST) activity, which are both GSH-related antioxidant enzymes. Thioredoxin-1 (Trx) is also a ubiquitous redox-active reducing (--SH) protein, and clivorine (50 microM) decreased cellular expression of Trx in a time-dependent manner, while 5 mM NAC reversed this decrease. Taken together, our results demonstrate that the protection of NAC is major via maintaining cellular reduced environment and thus prevents clivorine-induced mitochondrial-mediated hepatocytes apoptosis.


Subject(s)
Acetylcysteine/pharmacology , Chemical and Drug Induced Liver Injury/prevention & control , Cytotoxins/toxicity , Hepatocytes/drug effects , Pyrrolizidine Alkaloids/toxicity , Acetylcysteine/metabolism , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line , Cell Survival/drug effects , Cytochromes c/metabolism , Cytotoxins/isolation & purification , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Glutathione Synthase/antagonists & inhibitors , Glutathione Transferase/metabolism , Humans , Pyrrolizidine Alkaloids/isolation & purification , Reactive Oxygen Species/metabolism , Thioredoxins/metabolism , bcl-X Protein/metabolism
8.
Toxicon ; 53(5): 584-6, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19673104

ABSTRACT

Patulin (PAT), a mycotoxin produced by certain species of Penicillium, Aspergillus and Byssochlamys, is mainly found in ripe apple and apple products. In our present study, a significant increase of the micronuclei frequency induced by PAT was found in human hepatoma HepG2 cells. To elucidate the role of glutathione (GSH) in the effect, the intracellular GSH level was modulated by pre-treatment with buthionine-(S, R)-sulfoximine (BSO), a specific GSH synthesis inhibitor, and by pre-treatment with N-acetylcysteine (NAC), a GSH precursor. It was found that depletion of GSH in HepG2 cells with BSO dramatically increased the PAT-induced micronuclei frequencies and that when the intracellular GSH content was elevated by NAC, the chromosome damage induced by PAT was significantly prevented in our test concentrations (0.19-0.75 microM). These results indicate that GSH play an important role in cellular defense against PAT-induced genotoxicity.


Subject(s)
DNA Damage , Glutathione/metabolism , Patulin/toxicity , Cell Line , Cell Proliferation/drug effects , Glutathione/physiology , Glutathione Synthase/antagonists & inhibitors , Humans , Micronuclei, Chromosome-Defective
9.
Free Radic Biol Med ; 44(1): 44-55, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18045546

ABSTRACT

Loss of intracellular neuronal glutathione (GSH) is an important feature of neurodegenerative disorders including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. The consequences of GSH depletion include increased oxidative damage to proteins, lipids, and DNA and subsequent cytotoxic effects. GSH is also an important modulator of cellular copper (Cu) homeostasis and altered Cu metabolism is central to the pathology of several neurodegenerative diseases. The cytotoxic effects of Cu in cells depleted of GSH are not well understood. We have previously reported that depletion of neuronal GSH levels results in cell death from trace levels of extracellular Cu due to elevated Cu(I)-mediated free radical production. In this study we further examined the molecular pathway of trace Cu toxicity in neurons and fibroblasts depleted of GSH. Treatment of primary cortical neurons or 3T3 fibroblasts with the glutathione synthetase inhibitor buthionine sulfoximine resulted in substantial loss of intracellular GSH and increased cytotoxicity. We found that both neurons and fibroblasts revealed increased expression and activation of p53 after depletion of GSH. The increased p53 activity was induced by extracellular trace Cu. Furthermore, we showed that in GSH-depleted cells, Cu induced an increase in oxidative stress resulting in DNA damage and activation of p53-dependent cell death. These findings may have important implications for neurodegenerative disorders that involve GSH depletion and aberrant Cu metabolism.


Subject(s)
Copper/metabolism , Glutathione , Tumor Suppressor Protein p53/metabolism , 3T3 Cells , Animals , Buthionine Sulfoximine/pharmacology , Cell Death/physiology , Cerebral Cortex/cytology , DNA Damage , Enzyme Inhibitors/pharmacology , Free Radicals/metabolism , Gene Expression Regulation , Glutathione/deficiency , Glutathione Synthase/antagonists & inhibitors , Mice , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurons/cytology , Neurons/metabolism , Tumor Suppressor Protein p53/genetics
10.
Crit Care Med ; 36(7): 2106-16, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18552690

ABSTRACT

OBJECTIVE: Antioxidant therapy has shown some promise in critical care medicine in which glutathione depletion and heart failure are often seen in critically ill patients. This study was designed to examine the impact of glutathione depletion and the free radical scavenger, metallothionein (MT), on cardiac function. DESIGN: Friend virus B and MT transgenic mice were given the glutathione synthase inhibitor buthionine sulfoximine (buthionine sulfoximine [BSO], 30 mmol/L) in drinking water for 2 wks. MEASUREMENTS: Echocardiographic and cardiomyocyte functions were evaluated, including myocardial geometry, fraction shortening, peak shortening, time-to-90% relengthening (TR90), maximal velocity of shortening/relengthening (+/-dL/dt), intracellular Ca2+ rise, sarcoplasmic reticulum Ca2+ release, and intracellular Ca2+ decay rate. Sacro (endo)plasmic reticulum Ca2+-ATPase function was evaluated by 45Ca uptake. Highly reactive oxygen species, caspase-3, and aconitase activity were detected by fluorescent probe and colorimetric assays. MAIN RESULT: BSO elicited lipid peroxidation, protein carbonyl formation, mitochondrial damage, and apoptosis. BSO also reduced wall thickness, enhanced end systolic diameter, depressed fraction shortening, peak shortening, +/-dL/dt, sarcoplasmic reticulum Ca2+ release, 45Ca uptake, and intracellular Ca2+ decay, leading to prolonged TR90. BSO-induced mitochondrial loss and myofilament aberration. MT transgene itself had little effect on myocardial mechanics and ultrastructure. However, it alleviated BSO-induced myocardial functional, morphologic, and carbonyl changes. Western blot analysis showed reduced expression of sacro (endo)plasmic reticulum Ca2+-ATPase2a, Bcl-2 and phosphorylated GSK-3beta, enhanced calreticulin, Bax, p53, myosin heavy chain-beta isozyme switch, and IkappaB phosphorylation in FVB-BSO mice, all of which with the exception of p53 were nullified by MT. CONCLUSION: Our findings suggest a pathologic role of glutathione depletion in cardiac dysfunction and the therapeutic potential of antioxidants.


Subject(s)
Buthionine Sulfoximine/pharmacology , Cardiomyopathies/drug therapy , Enzyme Inhibitors/pharmacology , Glutathione/deficiency , Metallothionein/therapeutic use , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/etiology , Glutathione Synthase/antagonists & inhibitors , Lipid Peroxidation/drug effects , Male , Mice , Mice, Transgenic , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Ultrasonography
11.
Toxicol Lett ; 291: 184-193, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29679711

ABSTRACT

Rhabdomyolysis is one of the serious side effects of ciprofloxacin (CPFX), a widely used antibacterial drug; and occasionally, acute kidney injury (AKI) occurs. Often, rhabdomyolysis has occurred in patients taking CPFX co-administered with statins. The purpose of this study is to establish a mouse model of drug-induced rhabdomyolysis by co-administration of CPFX and atorvastatin (ATV) and to clarify the mechanisms of its pathogenesis. C57BL/6J mice treated with L-buthionine-(S,R)-sulfoximine (BSO), a glutathione synthesis inhibitor, were orally administered with CPFX and ATV for 4Ć¢Ā€ĀÆdays. Plasma levels of creatinine phosphokinase (CPK) and aspartate aminotransferase (AST) were significantly increased in the CPFX and ATV-co-administered group. Histopathological examination of skeletal muscle observed degeneration in gastrocnemius muscle and an increased number of the satellite cells. Expressions of skeletal muscle-specific microRNA and mRNA in plasma and skeletal muscle, respectively, were significantly increased. The area under the curve (AUC) of plasma CPFX was significantly increased in the CPFX and ATV-co-administered group. Furthermore, cytoplasmic vacuolization and a positively myoglobin-stained region in kidney tissue and high content of myoglobin in urine were observed. These results indicated that AKI was induced by myoglobin that leaked from skeletal muscle. The established mouse model in the present study would be useful for predicting potential rhabdomyolysis risks in preclinical drug development.


Subject(s)
Anti-Bacterial Agents/toxicity , Atorvastatin/toxicity , Ciprofloxacin/toxicity , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Rhabdomyolysis/chemically induced , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Animals , Anti-Bacterial Agents/blood , Aspartate Aminotransferases/metabolism , Atorvastatin/blood , Buthionine Sulfoximine/pharmacology , Ciprofloxacin/blood , Creatine Kinase/metabolism , Disease Models, Animal , Drug Interactions , Female , Glutathione Synthase/antagonists & inhibitors , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Mice , Mice, Inbred C57BL , Muscle, Skeletal/pathology , Rhabdomyolysis/pathology
12.
Life Sci ; 80(9): 873-8, 2007 Feb 06.
Article in English | MEDLINE | ID: mdl-17137603

ABSTRACT

A close relationship between oxidative stress, endothelial dysfunction, and hypoadiponectinemia has been observed. The present study was performed to investigate how glutathione depletion via buthionine sulfoximine (BSO) administration affects endothelial function and adiponectin levels in rats. Acetylcholine (Ach)-induced vasodilation was significantly enhanced in BSO-treated rats, compared with control rats. This was completely abolished by L-NAME, and Ach-induced vasodilation was not observed in the aorta without endothelium. These results suggest that Ach-induced hyper-relaxation of the aorta in BSO-treated rats is completely dependent on the presence of endothelium and mediated by changes in eNOS activity. Catalase significantly inhibited this relaxation to Ach and no effect of catalase on sodium nitroprusside-induced relaxation of the aorta without endothelium was observed in BSO-treated rats. Thus, hyper-relaxation of the aorta in BSO-treated rats is likely caused by H2O2 in addition to NO produced by the endothelium via an eNOS-dependent mechanism. Hypoadiponectinemia and decreased levels of adiponectin mRNA in adipose tissue were observed in BSO-treated rats. Protein expression of eNOS and SODs (SOD-1 and SOD-2) in the aorta was increased and plasma NOx levels were decreased in BSO-treated rats. Our results suggest that oxidative stress induced by BSO causes eNOS uncoupling and hyper-relaxation by producing H2O2, and that BSO-induced oxidative stress causes hypoadiponectinemia, probably by increasing H2O2 production in adipose tissue.


Subject(s)
Adiponectin/blood , Buthionine Sulfoximine/pharmacology , Enzyme Inhibitors/pharmacology , Glutathione/metabolism , Oxidative Stress/drug effects , Vasodilation/drug effects , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Biopterins/analogs & derivatives , Biopterins/metabolism , Blood Pressure/drug effects , Glutathione Synthase/antagonists & inhibitors , Heart Rate/drug effects , Hydrogen Peroxide/metabolism , Immunoblotting , Liver/drug effects , Liver/metabolism , Male , Nitric Oxide/blood , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
13.
FEBS Lett ; 591(23): 3881-3894, 2017 12.
Article in English | MEDLINE | ID: mdl-29127710

ABSTRACT

Buthionine sulfoximine (BSO) induces decreased glutathione (GSH) and trypanothione [T(SH)2 ] pools in trypanosomatids, presumably because only gamma-glutamylcysteine synthetase (ƎĀ³ECS) is blocked. However, some BSO effects cannot be explained by exclusive ƎĀ³ECS inhibition; therefore, its effect on the T(SH)2 metabolism pathway in TrypanosomaĀ cruzi was re-examined. Parasites exposed to BSO did not synthesize T(SH)2 even when supplemented with cysteine or GSH, suggesting trypanothione synthetase (TryS) inhibition by BSO. Indeed, recombinant ƎĀ³ECS and TryS, but not GSH synthetase, were inhibited by BSO and kinetics and docking analyses on a TcTryS 3D model suggested BSO binding at the GSH site. Furthermore, parasites overexpressing ƎĀ³ECS and TryS showed ~Ā 50% decreased activities after BSO treatment. These results indicated that BSO is also an inhibitor of TryS.


Subject(s)
Buthionine Sulfoximine/pharmacology , Glutathione/analogs & derivatives , Spermidine/analogs & derivatives , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/metabolism , Amide Synthases/antagonists & inhibitors , Amide Synthases/chemistry , Amide Synthases/genetics , Animals , Enzyme Inhibitors/pharmacology , Glutamate-Cysteine Ligase/antagonists & inhibitors , Glutamate-Cysteine Ligase/genetics , Glutathione/biosynthesis , Glutathione/metabolism , Glutathione Synthase/antagonists & inhibitors , Glutathione Synthase/genetics , Humans , Kinetics , Metabolic Networks and Pathways/drug effects , Molecular Docking Simulation , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spermidine/biosynthesis , Trypanosoma cruzi/genetics
14.
Mol Nutr Food Res ; 50(6): 530-42, 2006 May.
Article in English | MEDLINE | ID: mdl-16671059

ABSTRACT

Ochratoxin A (OTA), a nephrotoxic mycotoxin probably implicated in human Balkan endemic nephropathy and associated urothelial tumors, induces renal carcinomas in rodents and nephrotoxicity in pigs. OTA induces DNA-adduct formation, but the structure of the adducts and their role in nephrotoxicity and carcinogenicity have only partly been elucidated. In vivo, 2-mercaptoethane sulfonate (MESNA) protects rats against OTA-induced nephrotoxicity but not against carcinogenicity, indicating two different mechanisms leading to nephrotoxicity or carcinogenicity. To better understand how DNA-adduct could be generated, opossum kidney cells (OK) have been treated by OTA alone or in presence of several compounds such as MESNA or N-acetylcysteine (another agent that, like MESNA, reduces oxidative stress by increasing of free thiols in kidney), buthionine sulfoximine (BSO) (an inhibitor of glutathione-synthase), and alpha amino-3-chloro-4,5-dihydro-5-isoxazole acetic acid (ACIVICIN) (an inhibitor of gamma glutamyl transpeptidase). Cytotoxicity of OTA on OK cells was evaluated by applying the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. None of the listed agents diminished OTA cytotoxicity significantly; ACIVICIN even increases OTA cytotoxicity. In contrast, analysis of the HPLC profiles of OTA metabolites produced during these incubations indicated that the pattern, the quantity of metabolites, and the nature of the derivatives were modulated by these agents. Ochratoxin B (OTB), open-ring ochratoxin A (OP-OA), 4 hydroxylated OTA, 10 hydroxylated OTA, OTA without phenylalanine, OTB without phenylalanine, and a dechlorinated OTA metabolite could be identified by nano-ESI-IT-MS.


Subject(s)
Chlorine/analysis , DNA Adducts/biosynthesis , Glutathione/drug effects , Kidney/metabolism , Ochratoxins/chemistry , Ochratoxins/metabolism , Acetylcysteine/pharmacology , Animals , Buthionine Sulfoximine/pharmacology , Cell Survival/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid , Enzyme Inhibitors/pharmacology , Glutathione/metabolism , Glutathione Synthase/antagonists & inhibitors , Isoxazoles/pharmacology , Kidney/chemistry , Kidney/drug effects , Mesna/pharmacology , Ochratoxins/toxicity , Opossums , Oxidative Stress/drug effects , Spectrometry, Mass, Electrospray Ionization , gamma-Glutamyltransferase/antagonists & inhibitors
15.
Oncogene ; 22(9): 1349-57, 2003 Mar 06.
Article in English | MEDLINE | ID: mdl-12618760

ABSTRACT

It is well known that intracellular antioxidant glutathione (GSH) plays major roles in the maintenance of redox status and defense of oxidative stress. Ras, a small GTP-binding protein, may send growth-stimulating message to the nucleus through downstream Rac oncoprotein and superoxide (O(2*-)). These findings led us to investigate the effects of GSH and melatonin, a free-radical scavenger, on Ras-Rac-O(2*-)-related growth signal transduction. Our results demonstrate that overexpression of the inducible Ha-ras oncogene by isopropyl-beta-D-thiogalactoside (IPTG) increases the levels of reactive oxygen species (ROS, including O(2*-) and hydrogen peroxide (H(2)O(2))) and GSH in an Ha-ras-transformed NIH/3T3 fibroblast cell line. On the contrary, melatonin significantly suppresses ras-triggered cell growth by inhibiting the increase of ROS and GSH. Moreover, severe apoptosis of this transformed cell line occurred when the cell redox balance between ROS and GSH was dramatically changed in the presence of IPTG and L-buthionine-[S,R]-sulfoximine (BSO, a specific inhibitor of GSH synthetase). That BSO-induced cell apoptosis needs Ras to increase the ROS level was demonstrated by the free-radical scavenger melatonin. It effectively blocked cell apoptosis, but cell growth was also slowed without affecting Ras expression. Based on our studies, two approaches can be applied to treating ras-related cancers. One is utilizing melatonin to suppress cancer cell proliferation, and the other is utilizing BSO to induce cancer-cell apoptosis. Cotreatment of ras-related cancer cells with melatonin and BSO stops cell growth as well as apoptosis. Whether these cancer cells will undergo further regression or become recurrent merits investigation.


Subject(s)
3T3 Cells/drug effects , Apoptosis/drug effects , Free Radical Scavengers/pharmacology , Glutathione/pharmacology , Melatonin/pharmacology , Proto-Oncogene Proteins p21(ras)/physiology , 3T3 Cells/cytology , Animals , Buthionine Sulfoximine/pharmacology , Cell Division/drug effects , Cell Line, Transformed/cytology , Cell Line, Transformed/drug effects , Genes, ras , Glutathione Synthase/antagonists & inhibitors , Isopropyl Thiogalactoside/pharmacology , Mice , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Signal Transduction/drug effects , Superoxides/metabolism
16.
Mol Plant Microbe Interact ; 18(3): 254-9, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15782639

ABSTRACT

Legumes form a symbiotic interaction with bacteria of the Rhizobiaceae family to produce nitrogen-fixing root nodules under nitrogen-limiting conditions. This process involves the recognition of the bacterial Nod factors by the plant which mediates the entry of the bacteria into the root and nodule organogenesis. We have examined the importance of the low molecular weight thiols, glutathione (GSH) and homoglutathione (hGSH), during the nodulation process in the model legume Medicago truncatula. Using both buthionine sulfoximine, a specific inhibitor of GSH and hGSH synthesis, and transgenic roots expressing GSH synthetase and hGSH synthetase in an antisense orientation, we showed that deficiency in GSH and hGSH synthesis inhibited the formation of the root nodules. This inhibition was not correlated to a modification in the number of infection events or to a change in the expression of the Rhizobium sp.-induced peroxidase rip1, indicating that the low level of GSH or hGSH did not alter the first steps of the infection process. In contrast, a strong diminution in the number of nascent nodules and in the expression of the early nodulin genes, Mtenod12 and Mtenod40, were observed in GSH and hGSH-depleted plants. In conclusion, GSH and hGSH appear to be essential for proper development of the root nodules during the symbiotic interaction.


Subject(s)
Glutathione/analogs & derivatives , Glutathione/metabolism , Medicago truncatula/metabolism , Medicago truncatula/microbiology , Buthionine Sulfoximine/pharmacology , DNA, Antisense/genetics , Glutathione Synthase/antagonists & inhibitors , Glutathione Synthase/genetics , Medicago truncatula/growth & development , Nitrogen Fixation , Peptide Synthases/antagonists & inhibitors , Peptide Synthases/genetics , Plant Roots/growth & development , Plant Roots/metabolism , Plant Roots/microbiology , Plants, Genetically Modified , Symbiosis
17.
Comb Chem High Throughput Screen ; 18(5): 492-504, 2015.
Article in English | MEDLINE | ID: mdl-26220832

ABSTRACT

Malaria is the world's most fatal disease - causing up to 2.7 million deaths annually all over the world. The ability of organisms to develop resistance against existing antimalarial drugs exacerbates the problem. There is a clear cut need for more effective, affordable and accessible drugs that act by novel modes of action. Glutathione synthetase (GS) from Plasmodium falciparum represents an important potential drug target due to its defensive role; hence ceasing the respective metabolic step will destroy the parasite. A three dimensional model of Plasmodium GS was constructed by de novo modelling method and potential GS inhibitors were identified from a library of glutathione (GSH) analogues retrieved from Ligand-info database and filtered using Lipinski and ADME rules. Two common feature pharmacophore models were generated from the individual inhibitor clusters to provide insight into the key pharmacophore features that are crucial for the GS inhibition. Molecular docking of selective compounds into the predicted GS binding site revealed that the compound CMBMB was the best GS inhibitor when compared to the standard reference Chloroquine (CQ). This was taken as indicating that CMBMB was the best effective and safest drug against P. falciparum.


Subject(s)
Antimalarials/pharmacology , Enzyme Inhibitors/pharmacology , Glutathione Synthase/antagonists & inhibitors , Glutathione/pharmacology , Plasmodium falciparum/drug effects , Amino Acid Sequence , Antimalarials/chemistry , Binding Sites/drug effects , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Glutathione/chemistry , Glutathione Synthase/chemistry , Glutathione Synthase/metabolism , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Sequence Data , Molecular Structure , Parasitic Sensitivity Tests , Plasmodium falciparum/enzymology , Sequence Alignment , Structure-Activity Relationship
18.
Eur J Cancer ; 36(3): 428-34, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10708946

ABSTRACT

Multidrug resistance (MDR) is frequently associated with the overexpression of P-glycoprotein (Pgp) and/or multidrug resistance associated protein (MRP1), both members of the ABC superfamily of transporters. Pgp and MRP1 function as ATP-dependent efflux pumps that extrude cytotoxic drugs from tumour cells. Glutathione (GSH) has been considered to play an important role in the MRP1-mediated MDR. In our study, we examined the effects of buthionine sulphoximine (BSO), an inhibitor of GSH biosynthesis, on the nuclear accumulation of daunorubicin (DNR), in etoposide (VP16) and doxorubicin (ADR) resistant MCF7 cell lines, overexpressing respectively MRP1 (MCF7/VP) and Pgp (MCF7/ADR). The study of DNR transport was carried out using scanning confocal microspectrofluorometry. This technique allows the determination of the nuclear accumulation of anthracyclines in single living tumour cells. Treatment of MCF7/VP cells with BSO increased the sensitivity of these cells to DNR whilst the cytotoxicity of the drug in MCF7/ADR cells remained unchanged. In MCF7 resistant cells treated with BSO, their GSH level decreased as observed by confocal microscopy. DNR nuclear accumulation in MCF7/VP cells was increased by BSO whereas in MCF7/ADR cells BSO was unable to significantly increase the DNR nuclear accumulation. These data suggest a requirement for GSH in MRP1-mediated resistance whilst the nuclear efflux of GSH conjugates is probably not the primary mechanism of Pgp-mediated MDR. Finally, BSO might be a useful agent in clinical assays for facilitating detection of MRP1 expression.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Breast Neoplasms/metabolism , Buthionine Sulfoximine/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Glutathione Synthase/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/metabolism , Antibiotics, Antineoplastic/analysis , Antibiotics, Antineoplastic/metabolism , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Daunorubicin/analysis , Daunorubicin/metabolism , Dose-Response Relationship, Drug , Doxorubicin , Enzyme Inhibitors/pharmacology , Etoposide , Female , Flow Cytometry , Glutathione/metabolism , Humans , Microscopy, Confocal , Multidrug Resistance-Associated Proteins , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Fluorescence , Tumor Cells, Cultured/metabolism
19.
J Biochem ; 101(1): 207-15, 1987 Jan.
Article in English | MEDLINE | ID: mdl-3553173

ABSTRACT

Glutathione synthetase from Escherichia coli B showed amino acid sequence homology with mammalian and bacterial dihydrofolate reductases over 40 residues, although these two enzymes are different in their reaction mechanisms and ligand requirements. The effects of ligands of dihydrofolate reductase on the reaction of E. coli B glutathione synthetase were examined to find resemblances in catalytic function to dihydrofolate reductase. The E. coli B enzyme was potently inhibited by 7,8-dihydrofolate, methotrexate, and trimethoprim. Methotrexate was studied in detail and proved to bind to an ATP binding site of the E. coli B enzyme with K1 value of 0.1 mM. The homologous portion of the amino acid sequence in dihydrofolate reductases, which corresponds to the portion coded by exon 3 of mammalian dihydrofolate reductase genes, provided a binding site of the adenosine diphosphate moiety of NADPH in the crystal structure of dihydrofolate reductase. These analyses would indicate that the homologous portion of the amino acid sequence of the E. coli B enzyme provides the ATP binding site. This report gives experimental evidence that amino acid sequences related by sequence homology conserve functional similarity even in enzymes which differ in their catalytic mechanisms.


Subject(s)
Escherichia coli/enzymology , Glutathione Synthase/analysis , Peptide Synthases/analysis , Tetrahydrofolate Dehydrogenase/analysis , Amino Acid Sequence , Binding Sites , Catalysis , Chromatography, DEAE-Cellulose , Chromatography, Ion Exchange , Electrophoresis, Polyacrylamide Gel , Folic Acid Antagonists , Glutathione Synthase/antagonists & inhibitors , Glutathione Synthase/isolation & purification , Kinetics , Methotrexate/pharmacology , Sodium Dodecyl Sulfate , Tetrahydrofolate Dehydrogenase/isolation & purification
20.
Curr Eye Res ; 3(7): 923-8, 1984 Jul.
Article in English | MEDLINE | ID: mdl-6547896

ABSTRACT

The activity of glutathione synthetase from bovine lens was examined as a functions of the concentration of L-gamma-glutamyl-L-alpha-aminobutyrate, the dipeptide substrate required in the formation of ophthalmic acid. Several significant anomalies of the glutathione synthetase-catalyzed formation of ophthalmic acid were found. Curvilinearity of double reciprocal plots occurred with this substrate; this curvilinearity shows substrate activation of the reaction which is likely a result of negative cooperativity. Both ATP4- and, to a lesser extent Mg2+ inhibited the reaction, whereas MgATP2- is the substrate; maximum activity occurred with 2 mM Mg2+ in excess of the concentration of added ATP. This investigation shows that it is necessary to establish a defined set of conditions for reporting enzyme activity and that the usual practice of using very large concentrations of Mg2+ relative to ATP, and 5- to 20-fold excess of the dipeptide will give less than optimum activity. The unit of enzyme activity is suggested to be that activity in ml using 2 mM ATP, 4 mM Mg2+, 30 mM glycine and 15 mM L-gamma-glutamyl-alpha-aminobutyrate, which results in the formation of 1 nmole/minute of ADP or P(i). In this study, 5'-AMP was for the first time, shown to be an inhibitor of the reaction with a K(i) of 0.9 mM.


Subject(s)
Dipeptides/metabolism , Glutathione Synthase/metabolism , Lens, Crystalline/enzymology , Magnesium/metabolism , Peptide Synthases/metabolism , Adenosine Triphosphate/metabolism , Animals , Cattle , Glutathione Synthase/antagonists & inhibitors , Kinetics , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL