Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Metab Brain Dis ; 25(2): 191-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20437086

ABSTRACT

Glutaryl-CoA dehydrogenase deficiency or glutaric acidemia type I (GA I) is an inherited neurometabolic disorder biochemically characterized by tissue accumulation of predominantly glutaric (GA) and 3-hydroxyglutaric (3OHGA) acids and clinically by severe neurological symptoms and structural brain abnormalities, manifested as progressive cerebral atrophy and acute striatum degeneration following encephalopathic crises, whose pathophysiology is still in debate. Considering that reactive astrogliosis is a common finding in brain of GA I patients, in the present study we investigated the effects of GA and 3OHGA on glial activity determined by S100B release by rat C6-glioma cells. We also evaluated the effects of these organic acids on some parameters of oxidative stress in these astroglial cells. We observed that GA and 3OHGA significantly increased S100B secretion and thiobarbituric acid-reactive substances (lipid peroxidation), whereas GA markedly decreased reduced glutathione levels in these glioma cells. This is the first report demonstrating that the major metabolites accumulating in GA I activate S100B secretion in astroglial cells, indicating activation of these cells. We also showed that GA and 3OHGA induced oxidative stress in C6 lineage cells, confirming previous findings observed in brain fresh tissue. It is therefore presumed that reactive glial cells and oxidative damage may underlie at least in part the neuropathology of GA I.


Subject(s)
Amino Acid Metabolism, Inborn Errors/metabolism , Astrocytes/metabolism , Brain Diseases, Metabolic/metabolism , Glutarates/metabolism , Glutaryl-CoA Dehydrogenase/deficiency , Nerve Growth Factors/metabolism , S100 Proteins/metabolism , Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Metabolism, Inborn Errors/pathology , Animals , Astrocytes/pathology , Atrophy , Brain Diseases, Metabolic/enzymology , Brain Diseases, Metabolic/pathology , Cell Line, Tumor , Corpus Striatum/enzymology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Gliosis/enzymology , Gliosis/metabolism , Gliosis/pathology , Glutarates/pharmacology , Glutathione/antagonists & inhibitors , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , S100 Calcium Binding Protein beta Subunit , Thiobarbituric Acid Reactive Substances/metabolism
2.
Neurosci Res ; 57(2): 277-88, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17126438

ABSTRACT

Quinolinic acid (QA) is found at increased concentrations in brain of patients affected by various common neurodegenerative disorders, including Huntington's and Alzheimer's diseases. Considering that the neuropathology of these disorders has been recently attributed at least in part to energy deficit, in the present study we investigated the in vitro effect of QA (0.1-100 microM) on various parameters of energy metabolism, such as glucose uptake, (14)CO(2) production and lactate production, as well as on the activities of the respiratory chain complexes I-V, the citric acid cycle (CAC) enzymes, creatine kinase (CK), lactate dehydrogenase (LDH) and Na(+),K(+)-ATPase and finally the rate of oxygen consumption in brain of 30-day-old rats. We initially observed that QA significantly increased glucose uptake (55%), whereas (14)CO(2) generation from glucose, acetate and citrate was inhibited (up to 60%). Furthermore, QA-induced increase of brain glucose uptake was prevented by the NMDA receptor antagonist MK-801. Complex II activity was also inhibited (up to 35%) by QA, whereas the other activities of the respiratory chain complexes, CAC enzymes, CK and Na(+),K(+)-ATPase were not affected by the acid. Furthermore, inhibition of complex II activity was fully prevented by pre-incubating cortical homogenates with catalase plus superoxide dismutase, indicating that this effect was probably mediated by reactive oxygen species. In addition, lactate production was also not altered by QA, in contrast to the conversion of pyruvate to lactate catalyzed by LDH, which was significantly decreased (17%) by this neurotoxin. We also observed that QA did not change state III, state IV and the respiratory control ratio in the presence of glutamate/malate or succinate, suggesting that its effect on cellular respiration was rather weak. The data provide evidence that QA provokes a mild impairment of brain energy metabolism in vitro and does not support the view that the brain energy deficiency associated to certain neurodegenerative disorders could be solely endorsed to QA accumulation.


Subject(s)
Cerebral Cortex/drug effects , Energy Metabolism/drug effects , Quinolinic Acid/pharmacology , Analysis of Variance , Animals , Brain Chemistry/drug effects , Cell Membrane/drug effects , Cerebral Cortex/cytology , Creatine Kinase/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Glucose/metabolism , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , Rats , Rats, Wistar , Sodium-Potassium-Exchanging ATPase/metabolism , Synaptic Membranes/drug effects
3.
J Neurosci Res ; 74(1): 103-10, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-13130512

ABSTRACT

L-2-hydroxyglutaric acid (LGA) is the biochemical hallmark of L-2-hydroxyglutaric aciduria (L-OHGA), an inherited neurometabolic disorder characterized by progressive neurodegeneration with cerebellar and pyramidal signs, mental deterioration, epilepsy, and subcortical leukoencephalopathy. Because the underlying mechanisms of the neuropathology of this disorder are virtually unknown, in this study we tested the in vitro effect of LGA on various parameters of oxidative stress, namely, chemiluminescence, thiobarbituric acid-reactive substances (TBA-RS), protein carbonyl formation (PCF), total radical-trapping antioxidant potential (TRAP), total antioxidant reactivity (TAR), and the activities of the antioxidant enzymes catalase, glutathione peroxidase, and superoxide dismutase in cerebellum and cerebral cortex of 30-day-old rats. LGA significantly increased chemiluminescence, TBA-RS, and PCF measurements and markedly decreased TAR values in cerebellum, in contrast to TRAP and the activity of the antioxidant enzymes, which were not altered by the acid. Similar but less pronounced effects were provoked by LGA in cerebral cortex. Moreover, the LGA-induced increase of TBA-RS was significantly attenuated by melatonin (N-acetyl-5-methoxytryptamine) and by the combinations of ascorbic acid plus Trolox (soluble alpha-tocopherol) and of superoxide dismutase plus catalase but not by the inhibitor of nitric oxide synthase Nomega-nitro-L-arginine methyl ester (L-NAME), creatine, or superoxide dismutase or catalase alone in either cerebral structure. The data indicate that LGA provokes oxidation of lipids and proteins and reduces the brain capacity to modulate efficiently the damage associated with an enhanced production of free radicals, possibly by inducing generation of superoxide and hydroxyl radicals, which are trapped by the scavengers used. Thus, in case these findings can be extrapolated to human L-OHGA, it may be presumed that oxidative stress is involved in the pathophysiology of the brain damage observed in this disorder.


Subject(s)
Brain/drug effects , Glutarates/pharmacology , Lipid Peroxidation/drug effects , Oxidative Stress/drug effects , Animals , Antioxidants/metabolism , Brain/metabolism , Dose-Response Relationship, Drug , Glutarates/chemistry , Lipid Peroxidation/physiology , Oxidative Stress/physiology , Rats , Rats, Wistar
4.
Eur J Neurosci ; 17(10): 2017-22, 2003 May.
Article in English | MEDLINE | ID: mdl-12786967

ABSTRACT

Large amounts of d-2-hydroxyglutaric acid (DGA) accumulate in d-2-hydroxyglutaric aciduria (D-2-OHGA), an inherited neurometabolic disorder characterized by severe neurological dysfunction and cerebral atrophy. Despite the significant brain abnormalities, the neurotoxic mechanisms of brain injury in this disease are virtually unknown. In this work, the in vitro effect of DGA on various parameters of oxidative stress was investigated; namely chemiluminescence, thiobarbituric acid-reactive substances (TBA-RS), total radical-trapping antioxidant potential (TRAP), total antioxidant reactivity (TAR) and the activities of the antioxidant enzymes catalase, glutathione peroxidase and superoxide dismutase in cerebral cortex from 30-day-old-rats. DGA significantly increased chemiluminescence and TBA-RS and decreased TAR values in the cortical supernatants. In contrast, TRAP and the antioxidant enzyme activities were not altered by the metabolite. Furthermore, the DGA-induced increase of TBA-RS was fully prevented by the free radical scavengers ascorbic acid plus Trolox (water-soluble alpha-tocopherol) and attenuated by the inhibitor of nitric oxide synthase Nomega-nitro-L-arginine methyl ester (L-NAME), suggesting the role of superoxide, hydroxyl and nitric oxide radicals in this action. The data indicate a stimulation of lipid peroxidation through the production of free radicals and a reduction of the brain capacity to efficiently modulate the damage associated with the enhanced generation of free radicals by DGA. In the case that these findings also occur in human D-2-OHGA, it is feasible that oxidative stress may be involved in the pathophysiology of the brain injury observed in patients with this disease.


Subject(s)
Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Glutarates/pharmacology , Oxidative Stress/physiology , Age Factors , Animals , Catalase/metabolism , Free Radicals/metabolism , Glutathione Peroxidase/metabolism , Lipid Peroxidation/drug effects , Male , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...