Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters










Publication year range
1.
J Toxicol Environ Health B Crit Rev ; 26(8): 417-441, 2023 11 17.
Article in English | MEDLINE | ID: mdl-37606035

ABSTRACT

Buthionine sulfoximine (BSO) is a synthetic amino acid that blocks the biosynthesis of reduced glutathione (GSH), an endogenous antioxidant cellular component present in tumor cells. GSH levels have been associated with tumor cell resistance to chemotherapeutic drugs and platinum compounds. Consequently, by depleting GSH, BSO enhances the cytotoxicity of chemotherapeutic agents in drug-resistant tumors. Therefore, the aim of this study was to conduct a systematic review with meta-analysis of preclinical studies utilizing BSO in cancer treatments. The systematic search was carried out using the following databases: PubMed, Web of Science, Scopus, and EMBASE up until March 20, 2023, in order to collect preclinical studies that evaluated BSO, alone or in association, as a strategy for antineoplastic therapy. One hundred nine investigations were found to assess the cytotoxic potential of BSO alone or in combination with other compounds. Twenty-one of these met the criteria for performing the meta-analysis. The evidence gathered indicated that BSO alone exhibits cytotoxic activity. However, this compound is generally used in combination with other antineoplastic strategies, mainly chemotherapy ones, to improve cytotoxicity to carcinogenic cells and treatment efficacy. Finally, this review provides important considerations regarding BSO use in cancer treatment conditions, which might optimize future studies as a potential adjuvant antineoplastic therapeutic tool.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Buthionine Sulfoximine/pharmacology , Buthionine Sulfoximine/therapeutic use , Methionine Sulfoximine/therapeutic use , Methionine Sulfoximine/toxicity , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
2.
Antonie Van Leeuwenhoek ; 110(9): 1157-1168, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28631172

ABSTRACT

In the present work we studied the expression of genes from nitrogen central metabolism in the yeast Dekkera bruxellensis and under regulation by the Nitrogen Catabolite Repression mechanism (NCR). These analyses could shed some light on the biological mechanisms involved in the adaptation and survival of this yeast in the sugarcane fermentation process for ethanol production. Nitrogen sources (N-sources) in the form of ammonium, nitrate, glutamate or glutamine were investigated with or without the addition of methionine sulfoximine, which inhibits the activity of the enzyme glutamine synthetase and releases cells from NCR. The results showed that glutamine might act as an intracellular sensor for nitrogen availability in D. bruxellensis, by activating NCR. Gene expression analyses indicated the existence of two different GATA-dependent NCR pathways, identified as glutamine-dependent and glutamine-independent mechanisms. Moreover, nitrate is sensed as a non-preferential N-source and releases NCR to its higher level. After grouping genes according to their regulation pattern, we showed that genes for ammonium assimilation represent a regulon with almost constitutive expression, while permease encoding genes are mostly affected by the nitrogen sensor mechanism. On the other hand, nitrate assimilation genes constitute a regulon that is primarily subjected to induction by nitrate and, to a lesser extent, to a repressive mechanism by preferential N-sources. This observation explains our previous reports showing that nitrate is co-consumed with ammonium, a trait that enables D. bruxellensis cells to scavenge limiting N-sources in the industrial substrate and, therefore, to compete with Saccharomyces cerevisiae in this environment.


Subject(s)
Catabolite Repression/physiology , Dekkera/metabolism , Gene Expression Regulation, Fungal , Glutamine/metabolism , Nitrogen/metabolism , Ammonium Compounds/metabolism , Catabolite Repression/genetics , Dekkera/genetics , Dekkera/growth & development , Glutamate-Ammonia Ligase/metabolism , Glutamic Acid/metabolism , Glutamine/biosynthesis , Industrial Microbiology , Methionine Sulfoximine/metabolism , Methionine Sulfoximine/toxicity , Nitrates/metabolism , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Regulon
3.
Exp Neurol ; 288: 122-133, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27769717

ABSTRACT

Loss of glutamine synthetase (GS) in hippocampal astrocytes has been implicated in the causation of human mesial temporal lobe epilepsy (MTLE). However, the mechanism by which the deficiency in GS leads to epilepsy is incompletely understood. Here we ask how hippocampal GS inhibition affects seizure phenotype and neuronal activation during epilepsy development (epileptogenesis). Epileptogenesis was induced by infusing the irreversible GS blocker methionine sulfoximine (MSO) unilaterally into the hippocampal formation of rats. We then used continuous video-intracranial electroencephalogram (EEG) monitoring and c-Fos immunohistochemistry to determine the type of seizures and spatial distribution of neuronal activation early (1-5days postinfusion) and late (16-43days postinfusion) in epileptogenesis. Early in epileptogenesis, seizures were preferentially mild (stage 1-2), activating neurons in the entorhinal-hippocampal area, the basolateral amygdala, the piriform cortex, the midline thalamus, and the anterior olfactory area. Late in epileptogenesis, the seizures were generally more severe (stages 4-5) with neuronal activation extending to the neocortex, the bed nucleus of the stria terminalis, the mediodorsal thalamu\s, and the central nucleus of the amygdala. Our findings demonstrate that inhibition of GS focally in the hippocampal formation triggers a process of epileptogenesis characterized by gradual worsening of seizure severity and involvement of progressively larger neuronal populations over a period of several weeks. Knowledge about the underlying mechanism of epileptogenesis is important because such knowledge may result in more specific and efficacious treatments of MTLE by moving away from large and poorly specific surgical resections to highly targeted surgical or pharmacological interventions of the epileptogenic process.


Subject(s)
Enzyme Inhibitors/toxicity , Epilepsy/chemically induced , Hippocampus/cytology , Hippocampus/drug effects , Methionine Sulfoximine/toxicity , Neurons/pathology , Animals , Disease Models, Animal , Electroencephalography , Glutamate-Ammonia Ligase/metabolism , Hippocampus/physiology , Male , Muscarinic Agonists/toxicity , Neurons/drug effects , Pilocarpine/toxicity , Rats , Rats, Sprague-Dawley , Video Recording
4.
Neurobiol Dis ; 47(3): 331-7, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22659305

ABSTRACT

Increased extracellular brain glutamate has been implicated in the pathophysiology of human refractory temporal lobe epilepsy (TLE), but the cause of the excessive glutamate is unknown. Prior studies by us and others have shown that the glutamate degrading enzyme glutamine synthetase (GS) is deficient in astrocytes in the epileptogenic hippocampal formation in a subset of patients with TLE. We have postulated that the loss of GS in TLE leads to increased glutamate in astrocytes with elevated concentrations of extracellular glutamate and recurrent seizures as the ultimate end-points. Here we test the hypothesis that the deficiency in GS leads to increased glutamate in astrocytes. Rats were chronically infused with methionine sulfoximine (MSO, n=4) into the hippocampal formation to induce GS deficiency and recurrent seizures. A separate group of rats was infused with 0.9% NaCl (saline) as a control (n=6). At least 10days after the start of infusion, once recurrent seizures were established in the MSO-treated rats, the concentration of glutamate was assessed in CA1 of the hippocampal formation by immunogold electron microscopy. The concentration of glutamate was 47% higher in astrocytes in the MSO-treated vs. saline-treated rats (p=0.02), and the ratio of glutamate in astrocytes relative to axon terminals was increased by 74% in the MSO-treated rats (p=0.003). These data support our hypothesis that a deficiency in GS leads to increased glutamate in astrocytes. We additionally propose that the GS-deficient astrocytes in the hippocampal formation in TLE lead to elevated extracellular brain glutamate either through decreased clearance of extracellular glutamate or excessive release of glutamate into the extracellular space from these cells, or a combination of the two.


Subject(s)
Astrocytes/metabolism , Epilepsy, Temporal Lobe/pathology , Glutamic Acid/metabolism , Animals , Astrocytes/ultrastructure , Brain Waves/drug effects , Brain Waves/physiology , Disease Models, Animal , Electric Stimulation/adverse effects , Electroencephalography , Epilepsy, Temporal Lobe/etiology , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/ultrastructure , Male , Methionine Sulfoximine/toxicity , Microscopy, Immunoelectron , Rats , Rats, Sprague-Dawley
5.
Indian J Exp Biol ; 48(8): 858-60, 2010 Aug.
Article in English | MEDLINE | ID: mdl-21341546

ABSTRACT

Methionine-sulfoximine (MSO), a convulsant is known to increase the activity of histamine N-methyl transferase. The effect of a selective H3 receptor agonist R- (alpha) methylhistamine (RAMH) and antagonist (thioperamide, THP) and some antiepileptic drugs (gabapentin and sodium valproate) have been evaluated on MSO-induced convulsions in mice. The effect of THP was also evaluated in combination with these antiepileptic drugs. Sodium valproate (300 mg/kg, po) and gabapentin (400 mg/kg, po) offered protection against MSO-induced convulsions as evidenced by a significant prolongation of latency to abnormal dorsoflexion and complete protection against mortality within 6 h of administration. THP (15 mg/kg, ip) alone and in combination with sub-effective doses of gabapentin (75 mg/kg, po) and sodium valproate (75 mg/kg, po) revealed no significant differences from the control group or either drug alone. Hence, the convulsant action of MSO does not appear to be mediated via histaminergic mechanisms.


Subject(s)
Anticonvulsants/pharmacology , Brain/drug effects , Methionine Sulfoximine/toxicity , Piperidines/pharmacology , Seizures/drug therapy , Amines/pharmacology , Animals , Cyclohexanecarboxylic Acids/pharmacology , Drug Combinations , Gabapentin , Male , Mice , Seizures/chemically induced , Valproic Acid/pharmacology , gamma-Aminobutyric Acid/pharmacology
6.
Toxicon ; 50(2): 189-95, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17462687

ABSTRACT

Since several years, in the area of Kabrousse in Casamance (Senegal), a neurotoxic syndrome has caused more than 50 human deaths. Field studies showed that epidemic could be due to consumption of leave decoction of Cnestis ferruginea, a tropical plant belonging to the Connaraceae family. An ethnobotanical study has been conducted in order to investigate the traditional uses of C. ferruginea, and describe the circumstances and the symptoms of this plant poisoning. As a first experimental approach, the leave decoction was tested for its ability to induce cytotoxic effects using the XTT method. A phytochemical approach revealed the presence of methionine sulfoximine (MSX), a neurotoxic amino acid, in the plant extract by gas chromatography-mass spectrometry (GC-MS). The description of this poisoning, the cytotoxic activity of the decoction and the occurence of MSX in leaves of C. ferruginea constituted the first etiological data on this poisoning.


Subject(s)
Connaraceae/poisoning , Neurotoxicity Syndromes/physiopathology , Animals , CHO Cells , Cell Survival/drug effects , Chromatography, Thin Layer , Connaraceae/chemistry , Cricetinae , Cricetulus , Ethnobotany , Gas Chromatography-Mass Spectrometry , Humans , Methionine Sulfoximine/chemistry , Methionine Sulfoximine/isolation & purification , Methionine Sulfoximine/toxicity , Plant Leaves/chemistry , Plant Leaves/poisoning , Senegal , Tetrazolium Salts
7.
Respir Physiol Neurobiol ; 147(1): 19-30, 2005 May 12.
Article in English | MEDLINE | ID: mdl-15848120

ABSTRACT

Recent in vitro data suggest that astrocytes may modulate respiration. To examine this question in vivo, we treated 5-day-old rat pups with methionine sulfoximine (MS), a compound that alters carbohydrate and glutamate metabolism in astrocytes, but not neurons. MS-treated pups displayed a reduced breathing frequency (f) in baseline conditions relative to saline-treated pups. Hypercapnia (5% CO(2)) increased f in both groups, but f still remained significantly lower in the MS-treated group. No differences between treatment groups in the responses to hypoxia (8% O(2)) were observed. Also, MS-treated rats showed an enhanced accumulation of glycogen in neurons of the facial nucleus, the nucleus ambiguus, and the hypoglossal nucleus, structures that regulate respiratory activity and airway patency. An altered transfer of nutrient molecules from astrocytes to neurons may underlie these effects of MS, although direct effects of MS upon neurons or upon peripheral structures that regulate respiration cannot be completely ruled out as an explanation.


Subject(s)
Astrocytes/drug effects , Brain/drug effects , Hypercapnia/physiopathology , Methionine Sulfoximine/toxicity , Respiration/drug effects , Animals , Animals, Newborn , Brain/metabolism , Brain/pathology , Glycogen/metabolism , In Situ Hybridization , Neurons/drug effects , Neurons/pathology , RNA, Messenger/analysis , Rats , Receptors, Neurokinin-1/drug effects , Receptors, Neurokinin-1/metabolism , Respiratory Function Tests
8.
Brain Res Bull ; 57(1): 11-5, 2002 Jan 01.
Article in English | MEDLINE | ID: mdl-11827732

ABSTRACT

The convulsant methionine sulfoximine (MSO) is a byproduct of the agenized flour commonly used for feeding domestic animals decades ago. MSO is a powerful glycogenic and epileptogenic agent, and it is an irreversible inhibitor of glutamine synthetase. This latter effect was hypothesized to be responsible for the increase in the incidence of some neuropathologies in humans, such as Alzheimer's disease or Parkinson's disease. In order to test this hypothesis, we chronically administered MSO to two inbred strains of mice, C57BL/6J and BALB/cJ, and analyzed possible alterations in learning and memory features of these mice. Mice were given 20 mg/kg of MSO three times a week for 10 weeks. Spatial learning capabilities assessed with a radial maze were not affected by the long-term MSO treatment, although activity was significantly decreased in BALB/cJ mice. Thus, our data suggest that long-term administration of non-convulsive and non-glycogenic doses of MSO do not alter the spatial memory of mice. Our results do not support the hypothesis that chronic treatment with MSO influences hippocampus-dependent learning abilities in mice.


Subject(s)
Brain Chemistry/drug effects , Glutamate-Ammonia Ligase/antagonists & inhibitors , Glutamic Acid/metabolism , Learning Disabilities/chemically induced , Memory Disorders/chemically induced , Methionine Sulfoximine/toxicity , Neurodegenerative Diseases/chemically induced , Animals , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Brain Chemistry/physiology , Glutamate-Ammonia Ligase/metabolism , Learning Disabilities/metabolism , Learning Disabilities/physiopathology , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/metabolism , Memory Disorders/physiopathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Neurons/drug effects , Neurons/metabolism
9.
Toxicology ; 153(1-3): 179-87, 2000 Nov 16.
Article in English | MEDLINE | ID: mdl-11090956

ABSTRACT

Methionine sulfoximine is a xenobiotic amino acid derived from methionine. One of its major properties is to display a glycogenic activity in the brain. After studying this property, we investigate here a possible action of this xenobiotic on the expression of genes related to carbohydrate anabolism in the brain. Glycogen was studied by the means of electron microscopy. Astrocytes were cultured and the influence of methionine sulfoximine on carbohydrate anabolism in these cells was investigated. In vivo, methionine sulfoximine induced a large increase in glycogen accumulation. It also enhanced the glycogen accumulation in cultured astrocytes principally, when the medium was enriched in glucose. The gluconeogenic enzyme fructose-1,6-bisphosphatase may account for glycogen accumulation. Plasmids were built using antisens cDNA to permanently block the expression of fructose-1,6-bisphosphatase. An eukaryotic vector was used and the expression of fructose-1,6-bisphosphatase gene was under the control of the promoter of the glial fibrillary acidic protein. In this case, the glycogen content in cultured astrocytes largely decreased. This work shows that methionine sulfoximine enhances energy carbohydrate synthesis in the brain. Since this xenobiotic also enhances the expression of some genes related to one of the key step of glucose synthesis, it is possible that genes may be one target of methionine sulfoximine. Next investigations will study the actual effect of methionine sulfoximine in the cells.


Subject(s)
Brain Chemistry/drug effects , Brain Chemistry/genetics , Carbohydrate Metabolism , Gene Expression/drug effects , Methionine Sulfoximine/toxicity , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cells, Cultured , Cloning, Molecular , Fructose-Bisphosphatase/biosynthesis , Glycogen/metabolism , Male , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Transfection
10.
Neurotoxicology ; 21(4): 573-9, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11022864

ABSTRACT

Astrocytes occupy approximately 25% of the CNS volume. Their "foot" processes are closely associated with synapses, nodes of Ranvier, axonal tracts, and capillaries. Astrocytic functions include neurotrophic factor secretion, control of extracellular pH, inactivation of glutamate, as well as uptake and metabolism of neurotransmitters. Astrocyte-neuron interactions provide strategic sites for actions of numerous chemical compounds. In this manuscript, we discuss examples of toxins that directly affect astrocyte function (methylmercury, ammonia, methionine sulfoximine, and alcohol), leading to an altered homeostatic control of the extracellular milieu and neuronal dysfunction. In addition, the potential role of astrocytic proteins, the metallothioneins, in attenuating the neurotoxicity of methylmercury is discussed.


Subject(s)
Astrocytes/drug effects , Central Nervous System Depressants/toxicity , Ethanol/toxicity , Methionine Sulfoximine/toxicity , Methylmercury Compounds/toxicity , Neurotoxicity Syndromes/pathology , Quaternary Ammonium Compounds/toxicity , Animals , Astrocytes/pathology , Humans
11.
Can J Physiol Pharmacol ; 77(11): 871-7, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10593660

ABSTRACT

Methionine sulfoximine (MSO) is a rare amino acid. It occurs in nature or as a by-product of some forms of food processing. A notable example of the latter was a former method for bleaching wheat flour, using nitrogen trichloride, the "agene process," in use for most of the first 50 years of this century. "Agenized" flour was found to be responsible for various neurological disorders in animals, and MSO was identified as the toxic factor. The agene process was subsequently discontinued in the United States and the United Kingdom circa 1950. MSO inhibits the synthesis of both glutathione and glutamine, and it is possible that its actions on the nervous system arise from alterations in the amount or distribution of these molecules. Structurally, MSO resembles glutamate, an observation that has also raised the possibility that it might have more direct glutamate-like actions on neurons. In the present investigation, we report excitatory and toxic actions of MSO in an in vitro preparation of adult rat cortex. Field potential recordings in this preparation show that MSO application evokes a sustained depolarization, which can be blocked by the N-methyl-D-aspartate (NMDA) antagonist L-(+)-2-amino-5-phosphonovalerate (AP5). However, competition assays using MSO on [3H]CGP-39653 (DL-(E)-2-amino-4-propyl-1-phosphono-3-pentenoate) binding in rat cortical homogenates show only 20% displacement of total binding, suggesting that MSO is acting indirectly, perhaps by releasing glutamate. To investigate this possibility, we measured glutamate release during MSO application. Time course and dose-response experiments with MSO showed significant [3H]glutamate release, which was partially attenuated by AP5. To assess cellular toxicity, we measured lactate dehydrogenase (LDH) release from cortical sections exposed to MSO. MSO treatment led to a rapid increase in LDH activity, which could be blocked by AP5. These data suggest that MSO acts by increasing glutamate release, which then activates NMDA receptors, leading to excitotoxic cell death. These data suggest the possibility that MSO in processed flour had excitotoxic actions that may have been contributing factors to some human neuronal disorders.


Subject(s)
Cerebral Cortex/drug effects , Excitatory Amino Acids/pharmacology , Methionine Sulfoximine/pharmacology , 2-Amino-5-phosphonovalerate/analogs & derivatives , 2-Amino-5-phosphonovalerate/metabolism , 2-Amino-5-phosphonovalerate/pharmacology , Animals , Binding, Competitive , Cerebral Cortex/metabolism , Cerebral Cortex/physiology , Evoked Potentials/drug effects , Excitatory Amino Acid Antagonists/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acids/metabolism , Excitatory Amino Acids/toxicity , Glutamic Acid/metabolism , In Vitro Techniques , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/metabolism , Male , Methionine Sulfoximine/metabolism , Methionine Sulfoximine/toxicity , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism
12.
Ecotoxicol Environ Saf ; 34(1): 70-5, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8793322

ABSTRACT

L-Methionine sulfoximine (MSO) at concentration 1.25 mM in vivo causes the inhibition of glutamine synthetase (GS) in both roots and leaves of young seedlings of kidney bean following the accumulation of high levels of ammonia and decrease in amounts of free amino acids that is more pronounced in leaves. The inhibition of GS by MSO in leaves in the case of externally supplied 5 mM (15NH4)2SO4 assimilation leads to ammonia accumulation and the decrease in the amounts of glutamine and glutamic acid and the intensity of the incorporation of 15N into them. In roots the inhibition of GS is not followed by the decrease of 15N content into glutamate. It is concluded that the pathway of ammonia primary assimilation in leaves is via GS and glutamate synthase (GOGAT), while in roots glutamate dehydrogenase also plays an important role in this process.


Subject(s)
Fabaceae/metabolism , Glutathione Synthase/antagonists & inhibitors , Methionine Sulfoximine/toxicity , Nitrogen/metabolism , Plants, Medicinal , Quaternary Ammonium Compounds/metabolism , Amino Acid Oxidoreductases/metabolism , Amino Acids/metabolism , Ammonium Sulfate/metabolism , Fabaceae/drug effects , Ferredoxins/metabolism , Glutamate Dehydrogenase/metabolism , Isotope Labeling , Kinetics , Methionine Sulfoximine/metabolism , NAD/metabolism , Plant Leaves/enzymology , Plant Roots/enzymology
13.
Biochem Biophys Res Commun ; 221(2): 422-9, 1996 Apr 16.
Article in English | MEDLINE | ID: mdl-8619871

ABSTRACT

This study revealed a marked stereospecificity in the prevention of buthionine sulfoximine-induced cataract, and in the protection of lens antioxidants, in newborn rats by alpha-lipoate, R- and racemic alpha-lipoate decreased cataract formation from 100% (buthionine sulfoximine only) to 55% (buthionine sulfoximine + R-alpha-lipoic acid) and 40% (buthionine sulfoximine + rac-alpha-lipoic acid) (p<0.05 compared to buthionine sulfoximine only). S-alpha-lipoic acid had no effect on cataract formation induced by buthionine sulfoximine. The lens antioxidants glutathione, ascorbate, and vitamin E were depleted to 45, 62, and 23% of control levels, respectively, by buthionine sulfoximine treatment, but were maintained at 84-97% of control levels when R-alpha-lipoic acid or rac-alpha-lipoic acid were administered with buthionine sulfoximine; S-alpha-lipoic acid administration had no protective effect on lens antioxidants. When enantiomers of alpha-lipoic acid were administered to animals, R-alpha-lipoic acid was taken up by lens and reached concentrations 2- to 7-fold greater than those of S-alpha-lipoic acid, with rac-alpha-lipoic acid reaching levels midway between the R-isomer and racemic form. Reduced lipoic acid, dihydrolipoic acid, reached the highest levels in lens of the rac-alpha-lipoic acid-treated animals and the lowest levels in S-alpha-lipoic acid-treated animals. These results indicate that the protective effects of alpha-lipoic acid against buthionine sulfoximine-induced cataract are probably due to its protective effects on lens antioxidants, and that the stereospecificity exhibited is due to selective uptake and reduction of R-alpha-lipoic acid by lens cells.


Subject(s)
Cataract/prevention & control , Methionine Sulfoximine/analogs & derivatives , Thioctic Acid/pharmacology , Animals , Animals, Newborn , Ascorbic Acid/metabolism , Buthionine Sulfoximine , Cataract/chemically induced , Female , Glutathione/analogs & derivatives , Glutathione/deficiency , Glutathione/metabolism , Glutathione Disulfide , Lens, Crystalline/metabolism , Methionine Sulfoximine/toxicity , Pregnancy , Rats , Rats, Sprague-Dawley , Stereoisomerism , Thioctic Acid/chemistry , Vitamin E/metabolism
14.
Mol Cell Biochem ; 156(1): 17-24, 1996 Mar 09.
Article in English | MEDLINE | ID: mdl-8709971

ABSTRACT

The role of glutathione (GSH) in myocardial antioxidant defense was investigated in Swiss-Webster mice either performing swim exercise to exhaustion or rested in both the GSH adequate (GSH-A) and GSH deficient (GSH-D) states. GSH deficiency was accomplished by injecting mice with L-buthionine [S,R]sulfoximine (BSO; 2 nmol/kg body wt, i.p.) and providing BSO (20 mM) in drinking water for 12 days. GSH and glutathione disulfide (GSSG) contents in the GSH-D hearts were decreased to 10 and 8%, respectively, of those in the GSH-A mice. This decrease was associated with a significant decline of the total glutathione level in the liver, skeletal muscle and plasma. Myocardial GSH peroxidase and GSH sulfur-transferase activities decreased significantly following GSH deficiency, whereas superoxide dismutase activity was significantly elevated. GSH deficiency did not affect exercise endurance performance. However, exhaustive exercise decreased GSH content in the myocardium of the GSH-A and GSH-D mice by 22 and 44% (p < 0.05), respectively. The GSH:GSSG ratio was not altered significantly following exercise because of a concomitant decrease in GSSG (p < 0.05). gamma-Glutamyltranspeptidase activity was significantly increased after exercise, especially in the GSH-D hearts (72%; p < 0.05). GSH content after exercise correlated negatively with exercise time in both GSH-A and GSH-D mice (p < 0.05). These data indicate that GSH is actively used in the myocardium during prolonged exercise at moderate intensity and that GSH deficiency is tolerated by the heart, possibly compensated for by an increased GSH uptake from the plasma.


Subject(s)
Glutathione/deficiency , Heart/physiopathology , Myocardium/metabolism , Physical Exertion/physiology , Animals , Buthionine Sulfoximine , Fatigue/physiopathology , Glutathione Peroxidase/metabolism , Glutathione Transferase/metabolism , Lipid Peroxidation , Male , Methionine Sulfoximine/analogs & derivatives , Methionine Sulfoximine/toxicity , Mice , Muscle Proteins/metabolism , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species , gamma-Glutamyltransferase/metabolism
15.
Mol Cell Biochem ; 156(1): 79-85, 1996 Mar 09.
Article in English | MEDLINE | ID: mdl-8709979

ABSTRACT

The aim of this study was to test the hypothesis that a decreased myocardial concentration of reduced glutathione (GSH) during ischemia renders the myocardium more susceptible to injury by reactive oxygen species generated during early reperfusion. To this end, rats were pretreated with L-buthionine-S,R-sulfoximine (2 mmol/kg), which depleted myocardial GSH by 55%. Isolated buffer-perfused hearts were subjected to 30 min of either hypothermic or normothermic no-flow ischemia followed by reperfusion. Prior depletion of myocardial GSH did not lead to oxidative stress during reperfusion, as myocardial concentration of glutathione disulfide (GSSG) was not increased after 5 and 30 min of reperfusion. In addition, prior depletion of GSH did not exacerbate myocardial enzyme release, nor did it impair the recoveries of tissue ATP, coronary flow rate and left ventricular developed pressure during reperfusion after either hypothermic or normothermic ischemia. Even administration of the prooxidant cumene hydroperoxide (20 microM) to postischemic GSH-depleted hearts during the first 10 min of reperfusion did not aggravate postischemic injury, although this prooxidant load induced oxidative stress, as indicated by an increased myocardial concentration of GSSG. These results do not support the hypothesis that a reduced myocardial concentration of GSH during ischemia increases the susceptibility to injury mediated by reactive oxygen species generated during reperfusion. Apparently, myocardial tissue possesses a large excess of GSH compared to the quantity of reactive oxygen species generated upon reperfusion.


Subject(s)
Glutathione/physiology , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Adenosine Triphosphate/metabolism , Animals , Benzene Derivatives/toxicity , Buthionine Sulfoximine , Glutathione/deficiency , Male , Methionine Sulfoximine/analogs & derivatives , Methionine Sulfoximine/toxicity , Oxidation-Reduction , Oxidative Stress , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
16.
J Clin Oncol ; 14(1): 249-56, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8558205

ABSTRACT

PURPOSE AND METHODS: Resistance to alkylating agents and platinum compounds is associated with elevated levels of glutathione (GSH). Depletion of GSH by buthionine sulfoximine (BSO) restores the sensitivity of resistant tumors to melphalan in vitro and in vivo. In a phase I trial, each patient received two cycles as follows: BSO alone intravenously (i.v.) every 12 hours for six doses, and 1 week later the same BSO as cycle one with melphalan (L-PAM) 15 mg/m2 i.v. 1 hour after the fifth dose. BSO doses were escalated from 1.5 to 17 g/m2 in 41 patients. RESULTS: The only toxicity attributable to BSO was grade I or II nausea/vomiting in 50% of patients. Dose-related neutropenia required an L-PAM dose reduction to 10 mg/m2 at BSO 7.5 g/m2. We measured GSH in peripheral mononuclear cells (PMN), and in tumor biopsies when available, at intervals following BSO dosing. In PMNs, GSH content decreased over 36 to 72 hours to reach a nadir on day 3; at the highest dose, recovery was delayed beyond day 7. The mean PMN GSH nadirs were approximately 10% of control at BSO doses > or = 7.5 g/m2; at 13 and 17 g/m2, all but two patients had nadir values in this range. GSH was depleted in sequential tumor biopsies to a variable extent, but with a similar time course. At BSO doses > or = 13 g/m2, tumor GSH was < or = 20% of starting values on day 3 in five of seven patients; recovery had not occurred by day 5. We measured plasma concentrations of R- and S-BSO by high-performance liquid chromatography (HPLC) in 22 patients throughout the dosing period. Total-body clearance (CLt) and volume of distribution at steady-state (Vss) for both isomers were dose-independent. The CLt of S-BSO was significantly less than that of R-BSO at all doses, but no significant differences in Vss were observed between the racemates. Harmonic mean half-lives were 1.39 hours and 1.89 hours for R-BSO and S-BSO, respectively. CONCLUSION: A biochemically appropriate dose of BSO for use on this schedule is 13 g/m2, which will be used in phase II trials to be conducted in ovarian cancer and melanoma.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Methionine Sulfoximine/analogs & derivatives , Neoplasms/drug therapy , Adult , Aged , Antimetabolites, Antineoplastic/pharmacokinetics , Antimetabolites, Antineoplastic/toxicity , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/toxicity , Bone Marrow/drug effects , Buthionine Sulfoximine , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Female , Glutathione/blood , Glutathione/drug effects , Humans , Linear Models , Male , Melphalan/administration & dosage , Methionine Sulfoximine/administration & dosage , Methionine Sulfoximine/pharmacokinetics , Methionine Sulfoximine/toxicity , Middle Aged , Nausea/chemically induced , Neoplasm Recurrence, Local/diagnostic imaging , Neoplasm Recurrence, Local/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Neutropenia/chemically induced , Neutrophils/drug effects , Radiography , Vomiting/chemically induced
17.
Neuroreport ; 7(1): 93-6, 1995 Dec 29.
Article in English | MEDLINE | ID: mdl-8742425

ABSTRACT

Free radical-mediated damage to cultured cortical neurons was induced by a 24 h exposure to Fe2+ (30 microM) or an inhibitor of gamma-glutamylcysteine synthetase, L-buthionine-[S,R]-sulfoximine (BSO, 1 mM). As expected, neuronal death was blocked by inclusion of the free radical scavenger trolox during the Fe2+ or BSO exposure. However, unexpectedly, pretreatment of the cultures with BDNF or IGF-I markedly potentiated neuronal death. This growth factor-potentiated death was still blocked by trolox, but was insensitive to glutamate antagonists. Concurrent addition of cycloheximide with the growth factors prevented injury potentiation. Present findings suggest that growth factors may increase free radical-induced neuronal death by mechanisms dependent upon protein synthesis.


Subject(s)
Cerebral Cortex/drug effects , Insulin-Like Growth Factor I/pharmacology , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins/pharmacology , Neurons/drug effects , Animals , Brain-Derived Neurotrophic Factor , Buthionine Sulfoximine , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/pathology , Drug Synergism , Enzyme Inhibitors/toxicity , Ferric Compounds/toxicity , Glutamate-Cysteine Ligase/antagonists & inhibitors , Methionine Sulfoximine/analogs & derivatives , Methionine Sulfoximine/toxicity , Mice , Neurons/pathology
18.
J Pathol ; 177(3): 303-7, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8551393

ABSTRACT

This study utilized two samples of amosite asbestos which differ in their length, but not in their diameter and which have been shown previously to have very different abilities to cause pathology in rats exposed by instillation or inhalation. The activity of these amosite samples in causing chromosomal aberrations in Chinese hamster ovary cells in culture was examined, along with the effect of the glutathione (GSH) synthesis-inhibiting agent buthionine sulphoximine. The incidence of chromosomal aberrations in cells treated with the short fibre sample was similar to control levels; the long amosite sample caused significantly more chromosomal aberrations than the short fibre sample. When cells were treated with buthionine sulphoximine to decrease the levels of intracellular glutathione, the incidence of chromosomal aberrations was increased in the control cells, but also on treatment with both short and long amosite, the long sample again being considerably more active than the short. The pathogenicity of the long amosite may result from the ability of the fibres to cause chromosome damage, while the enhancement of this damage caused by decreasing intracellular glutathione suggests that the asbestos fibres may impose an oxidant stress on the cells which contributes to these aberrations.


Subject(s)
Asbestos, Amosite/toxicity , Chromosome Aberrations , Animals , Buthionine Sulfoximine , CHO Cells , Cricetinae , Cricetulus , Drug Synergism , Glutathione/physiology , Methionine Sulfoximine/analogs & derivatives , Methionine Sulfoximine/toxicity , Mineral Fibers/toxicity , Mitotic Index
20.
Toxicol Appl Pharmacol ; 133(2): 249-61, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7645021

ABSTRACT

Glutathione depletion achieved by continuous exposure of mitogen-activated human T lymphocytes to L-buthionine-(S,R)-sulfoximine, a specific inhibitor of gamma-glutamylcysteine synthetase, leads to a marked inhibition of the proliferative response. Concanavalin A-activated T cells treated with buthionine sulfoximine failed to exhibit the increase in glutathione content normally observed in activated T cells and were depleted of cellular glutathione over 4 days of culture. On Day 3 of culture, DNA synthesis was inhibited by greater than 75%. In addition, total RNA synthesis was dramatically reduced in the glutathione-depleted cells being inhibited by 26, 61, and 82% on Days 2, 3, and 4, respectively. Despite this global reduction in RNA synthesis, no specific effects on mRNA expression of a number of critical T cell genes required for activation and/or proliferation were detected. In contrast to a recent report of GSH depletion leading to down-regulation of ras mRNA expression in a number of transformed cell lines, glutathione depletion did not influence N-ras mRNA expression in T lymphocytes. No influence of glutathione depletion on the induction of histone mRNA expression was observed. However, consistent with previous studies on regulation of histone mRNA expression, histone transcript levels were reduced when DNA synthesis was markedly inhibited. A cellular stress response, characterized by an increase in mRNA levels of the two stress response genes, HSP70 and gadd 153, was evident in glutathione-depleted unstimulated cells. Additionally, in these cells at 48 hr, we observed a 3.5-fold increase in the steady-state level of mRNA encoding the catalytic subunit of gamma-glutamylcysteine synthetase, the enzyme inhibited by buthionine sulfoximine.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Gene Expression Regulation/genetics , Glutathione/metabolism , Methionine Sulfoximine/analogs & derivatives , T-Lymphocytes/metabolism , Blotting, Northern , Buthionine Sulfoximine , Cell Survival/drug effects , Cells, Cultured , DNA/biosynthesis , DNA Probes , Down-Regulation , Glutamate-Cysteine Ligase/antagonists & inhibitors , Glutamate-Cysteine Ligase/genetics , HSP70 Heat-Shock Proteins/genetics , Humans , Lymphocyte Activation/genetics , Methionine Sulfoximine/toxicity , Oxidative Stress/genetics , RNA, Messenger/biosynthesis , T-Lymphocytes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...