Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Brain Res ; 1748: 147080, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32866546

ABSTRACT

Glutamate (Glu) is the main mammalian brain neurotransmitter. Concerning the glutamatergic neurotransmission, excessive levels of glutamate in the synaptic cleft are extremally harmful. This phenomenon, named as excitotoxicity is involved in various acute and chronic brain diseases. Guanosine (GUO), an endogenous guanine nucleoside, possesses neuroprotective effects in several experimental models of glutamatergic excitotoxicity, an effect accompanied by an increase in astrocytic glutamate uptake. Therefore, the objective of this study was to investigate the involvement of an additional putative parameter, glutamate oxidation to CO2, involved in ex-vivo GUO neuroprotective effects in mouse hippocampal slices submitted to glutamatergic excitotoxicity. Mice were sacrificed by decapitation, the hippocampi were removed and sliced. The slices were incubated for various times and concentrations of Glu and GUO. First, the concentration of Glu that produced an increase in L-[14C(U)]-Glu oxidation to CO2 without cell injury was determined at different time points (between 0 and 90 min); 1000 µM Glu increased Glu oxidation between 30 and 60 min of incubation without cell injury. Under these conditions (Glu concentration and incubation time), 100 µM GUO increased Glu oxidation (35%). Additionally, 100 µM GUO increased L-[3,4-3H]-glutamate uptake (45%) in slices incubated with 1000 µM Glu (0-30 min). Furthermore, 1000  µM Glu increased reactive species levels, SOD activity, and decreased GPx activity, and GSH content after 30 and 60 min; 100 µM GUO prevented these effects. This is the first study demonstrating that GUO simultaneously promoted an increase in the uptake and utilization of Glu in excitotoxicity-like conditions preventing redox imbalance.


Subject(s)
Antioxidants/pharmacology , Glutamic Acid/pharmacology , Guanosine/pharmacology , Hippocampus/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Animals , Energy Metabolism/drug effects , Hippocampus/metabolism , Male , Mice , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
2.
Andrologia ; 2018 Feb 01.
Article in English | MEDLINE | ID: mdl-29388232

ABSTRACT

Although boar semen productivity is affected by seasonality, its effects are not equal among different regions which raise concerns regarding the profitability of boar stud farms. Therefore, the goals of this study were (i) to evaluate the seasonal effect on semen production in a commercial boar stud farm located in a subtropical climate region and (ii) to verify whether the activities of superoxide dismutase and glutathione peroxidase in spermatozoa and seminal plasma were associated with seminal traits of fresh and cooled semen. Nine boars were collected twice per season, and routine seminal parameter analyses were performed together with superoxide dismutase and glutathione peroxidase activities in seminal plasma and spermatozoa. Despite a reduction in sperm concentration in spring and summer, most seminal parameters were constant year-round. Temperature-humidity index was higher in the summer compared to spring, autumn and winter (p < .05). Superoxide dismutase activity in spermatozoa was increased in summer compared to autumn and winter (p < .05). The activities of both enzymes in seminal plasma and spermatozoa glutathione peroxidase remained unaltered throughout the seasons. In conclusion, seasonality showed little influence in overall boar seminal parameters despite microclimatic differences among seasons, and spermatozoa collected during summer increased superoxide dismutase activity.

3.
Neuroscience ; 310: 578-88, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26431622

ABSTRACT

Several physiological processes in the CNS are regulated by the endocannabinoid system (ECS). Cannabinoid receptors (CBr) and CBr agonists have been involved in the modulation of the N-methyl-D-aspartate receptor (NMDAr) activation. Glutaric (GA), 3-hydroxyglutaric (3-OHGA), methylmalonic (MMA) and propionic (PA) acids are endogenous metabolites produced and accumulated in the brain of children affected by severe organic acidemias (OAs) with neurodegeneration. Oxidative stress and excitotoxicity have been involved in the toxic pattern exerted by these organic acids. Studying the early pattern of toxicity exerted by these metabolites is crucial to explain the extent of damage that they can produce in the brain. Herein, we investigated the effects of the synthetic CBr agonist WIN 55,212-2 (WIN) on early markers of GA-, 3-OHGA-, MMA- and PA-induced toxicity in brain synaptosomes from adult (90-day-old) and adolescent (30-day-old) rats. As pre-treatment, WIN exerted protective effects on the GA- and MMA-induced mitochondrial dysfunction, and prevented the reactive oxygen species (ROS) formation and lipid peroxidation induced by all metabolites. Our findings support a protective and modulatory role of cannabinoids in the early toxic events elicited by toxic metabolites involved in OAs.


Subject(s)
Acids, Acyclic/metabolism , Acids, Acyclic/toxicity , Amino Acid Metabolism, Inborn Errors/metabolism , Benzoxazines/pharmacology , Brain Diseases, Metabolic/metabolism , Brain/metabolism , Cannabinoid Receptor Agonists/pharmacology , Glutaryl-CoA Dehydrogenase/deficiency , Morpholines/pharmacology , Naphthalenes/pharmacology , Oxidative Stress/drug effects , Animals , Brain/drug effects , Glutarates/metabolism , Glutarates/toxicity , Glutaryl-CoA Dehydrogenase/metabolism , Lipid Peroxidation/drug effects , Male , Methylmalonic Acid/metabolism , Methylmalonic Acid/toxicity , Mitochondria/drug effects , Mitochondria/metabolism , Propionates/metabolism , Propionates/toxicity , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism
4.
Neuroscience ; 308: 64-74, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26343296

ABSTRACT

The brain of children affected by organic acidemias develop acute neurodegeneration linked to accumulation of endogenous toxic metabolites like glutaric (GA), 3-hydroxyglutaric (3-OHGA), methylmalonic (MMA) and propionic (PA) acids. Excitotoxic and oxidative events are involved in the toxic patterns elicited by these organic acids, although their single actions cannot explain the extent of brain damage observed in organic acidemias. The characterization of co-adjuvant factors involved in the magnification of early toxic processes evoked by these metabolites is essential to infer their actions in the human brain. Alterations in the kynurenine pathway (KP) - a metabolic route devoted to degrade tryptophan to form NAD(+) - produce increased levels of the excitotoxic metabolite quinolinic acid (QUIN), which has been involved in neurodegenerative disorders. Herein we investigated the effects of subtoxic concentrations of GA, 3-OHGA, MMA and PA, either alone or in combination with QUIN, on early toxic endpoints in rat brain synaptosomes. To establish specific mechanisms, we pre-incubated synaptosomes with different protective agents, including the endogenous N-methyl-d-aspartate (NMDA) receptor antagonist kynurenic acid (KA), the antioxidant S-allylcysteine (SAC) and the nitric oxide synthase (NOS) inhibitor nitro-l-arginine methyl ester (l-NAME). While the incubation of synaptosomes with toxic metabolites at subtoxic concentrations produced no effects, their co-incubation (QUIN+GA, +3-OHGA, +MMA or +PA) decreased the mitochondrial function and increased reactive oxygen species (ROS) formation and lipid peroxidation. For all cases, this effect was partially prevented by KA and l-NAME, and completely avoided by SAC. These findings suggest that early damaging events elicited by organic acids involved in metabolic acidemias can be magnified by toxic synergism with QUIN, and this process is mostly mediated by oxidative stress, and in a lesser extent by excitotoxicity and nitrosative stress. Therefore, QUIN can be hypothesized to contribute to the pathophysiology of brain degeneration in children with metabolic acidemias.


Subject(s)
Amino Acid Metabolism, Inborn Errors/metabolism , Brain Diseases, Metabolic/metabolism , Brain/metabolism , Glutarates/metabolism , Glutaryl-CoA Dehydrogenase/deficiency , Quinolinic Acid/metabolism , Synaptosomes/metabolism , Animals , Brain/drug effects , Disease Models, Animal , Glutarates/toxicity , Glutaryl-CoA Dehydrogenase/metabolism , Lipid Peroxidation/drug effects , Male , Methylmalonic Acid/metabolism , Methylmalonic Acid/toxicity , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Propionates/metabolism , Propionates/toxicity , Quinolinic Acid/toxicity , Rats, Wistar , Reactive Oxygen Species/metabolism , Synaptosomes/drug effects
5.
Neuroscience ; 304: 122-32, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26188285

ABSTRACT

Phytanic acid (Phyt) accumulates in various peroxisomal diseases including Refsum disease (RD) and Zellweger syndrome (ZS). Since the pathogenesis of the neurological symptoms and especially the cerebellar abnormalities in these disorders are poorly known, we investigated the effects of in vivo intracerebral administration of Phyt on a large spectrum of redox homeostasis parameters in the cerebellum of young rats. Malondialdehyde (MDA) levels, sulfhydryl oxidation, carbonyl content, nitrite and nitrate concentrations, 2',7'-dichlorofluorescein (DCFH) oxidation, total (tGS) and reduced glutathione (GSH) levels and the activities of important antioxidant enzymes were determined at different periods after Phyt administration. Immunohistochemical analysis was also carried out in the cerebellum. Phyt significantly increased MDA and nitric oxide (NO) production and decreased GSH levels, without altering tGS, DCFH oxidation, sulfhydryl oxidation, carbonyl content and the activities of glutathione peroxidase (GPx), superoxide dismutase (SOD), catalase (CAT), glutathione reductase (GR) and glucose-6-phosphate dehydrogenase (G6PD). Furthermore, immunohistochemical analysis revealed that Phyt caused astrogliosis and protein nitrosative damage in the cerebellum. It was also observed that the NO synthase inhibitor Nω-Nitro-L-arginine methyl ester (L-NAME) prevented the increase of MDA and NO production as well as the decrease of GSH and the immunohistochemical alterations caused by Phyt, strongly suggesting that reactive nitrogen species (RNS) were involved in these effects. The present data provide in vivo solid evidence that Phyt disrupts redox homeostasis and causes astrogliosis in rat cerebellum probably mediated by RNS production. It is therefore presumed that disequilibrium of redox status may contribute at least in part to the cerebellum alterations characteristic of patients affected by RD and other disorders with Phyt accumulation.


Subject(s)
Astrocytes/metabolism , Cerebellum/metabolism , Oxidative Stress/physiology , Peroxisomal Disorders/physiopathology , Phytanic Acid/metabolism , Reactive Nitrogen Species/metabolism , Animals , Astrocytes/pathology , Cerebellum/growth & development , Cerebellum/pathology , Disease Models, Animal , Gliosis/pathology , Gliosis/physiopathology , Homeostasis/physiology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Neuroprotective Agents/pharmacology , Peroxisomal Disorders/pathology , Phytanic Acid/administration & dosage , Rats, Wistar , Time Factors
6.
Neuroscience ; 277: 281-93, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25043325

ABSTRACT

High accumulation of D-2-hydroxyglutaric acid (D-2-HG) is the biochemical hallmark of patients affected by the inherited neurometabolic disorder D-2-hydroxyglutaric aciduria (D-2-HGA). Clinically, patients present neurological symptoms and basal ganglia injury whose pathophysiology is poorly understood. We investigated the ex vivo effects of intrastriatal administration of D-2-HG on important parameters of redox status in the striatum of weaning rats. D-2-HG in vivo administration increased malondialdehyde (MDA) and carbonyl formation (lipid and protein oxidative damage, respectively), as well as the production of reactive nitrogen species (RNS). D-2-HG also compromised the antioxidant defenses by decreasing reduced glutathione (GSH) concentrations, as well as the activities of superoxide dismutase (SOD) and glutathione peroxidase (GPx). Increased amounts of oxidized glutathione (GSSG) with no significant alteration of total glutathione (tGS) were also found. Furthermore, D-2-HG-induced lipid oxidation and reduction of GSH concentrations and GPx activity were prevented by the N-methyl-d-aspartate (NMDA) receptor antagonist dizocilpine maleate (MK-801) and the nitric oxide synthase (NOS) inhibitor N(ω)-nitro-l-arginine methyl ester (l-NAME), suggesting the participation of NMDA receptors and nitric oxide derivatives in these effects. Creatine also impeded D-2-HG-elicited MDA increase, but did not change the D-2-HG-induced diminution of GSH and of the activities of SOD and GPx. We also found that DCFH oxidation and H2O2 production were not altered by D-2-HG, making unlikely an important role for reactive oxygen species (ROS) and reinforcing the participation of RNS in the oxidative damage and the reduction of antioxidant defenses provoked by this organic acid. Vacuolization, lymphocytic infiltrates and macrophages indicating brain damage were also observed in the striatum of rats injected with D-2-HG. The present data provide in vivo solid evidence that D-2-HG disrupts redox homeostasis and causes histological alterations in the rat striatum probably mediated by NMDA overstimulation and RNS production. It is therefore presumed that disturbance of redox status may contribute at least in part to the basal ganglia alterations characteristic of patients affected by D-2-HGA.


Subject(s)
Corpus Striatum/drug effects , Glutarates/toxicity , Animals , Corpus Striatum/metabolism , Corpus Striatum/pathology , Creatine/pharmacology , Dizocilpine Maleate/pharmacology , Glutarates/metabolism , Glutarates/pharmacology , Glutathione/metabolism , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Malondialdehyde/metabolism , N-Methylaspartate/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism
7.
Free Radic Res ; 48(6): 659-69, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24580146

ABSTRACT

Hyperammonemia is a common finding in children with methylmalonic acidemia and propionic acidemia, but its contribution to the development of the neurological symptoms in the affected patients is poorly known. Considering that methylmalonic acid (MMA) and propionic acid (PA) predominantly accumulate in these disorders, we investigated the effects of hyperammonemia induced by urease treatment in 30-day-old rats receiving an intracerebroventricular (ICV) injection of MMA or PA on important parameters of redox homeostasis in cerebral cortex and striatum. We evaluated glutathione (GSH) concentrations, sulfhydryl content, nitrate and nitrite concentrations, 2',7'-dichlorofluorescein (DCFH) oxidation, and the activity of antioxidant enzymes. MMA decreased GSH concentrations and sulfhydryl content and increased nitrate and nitrite concentrations in cerebral cortex and striatum from hyperammonemic rats, whereas MMA or ammonia per se did not alter these parameters. MMA plus hyperammonemia also decreased glutathione reductase activity in rat cerebral cortex, but did not affect catalase, superoxide dismutase and glutathione peroxidase activities, neither DCFH oxidation. Furthermore, ICV PA administration alone or combined with hyperammonemia did not alter any of the evaluated parameters. We also found that pre-treatment with antioxidants prevented GSH reduction and sulfhydryl oxidation, whereas N(ω)-nitro-L-arginine methyl ester (L-NAME) prevented the increased nitrate and nitrite concentrations provoked by MMA plus ammonia treatments. Histological alterations, including vacuolization, ischemic neurons, and pericellular edema, were observed in brain of hyperammonemic rats injected with MMA. The data indicate a synergistic effect of MMA and ammonia disturbing redox homeostasis and causing morphological brain abnormalities in rat brain.


Subject(s)
Ammonia/toxicity , Cerebral Cortex/pathology , Corpus Striatum/pathology , Hyperammonemia/pathology , Methylmalonic Acid/toxicity , Animals , Antioxidants , Catalase/metabolism , Fluoresceins/metabolism , Glutathione/biosynthesis , Glutathione Peroxidase/metabolism , Glutathione Reductase/biosynthesis , Homeostasis , Hyperammonemia/chemically induced , Infusions, Intraventricular , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitrates/analysis , Nitrites/analysis , Oxidation-Reduction , Rats , Rats, Wistar , Sulfhydryl Compounds/analysis , Superoxide Dismutase/metabolism , Urease/pharmacology
8.
Free Radic Res ; 47(12): 1066-75, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24127998

ABSTRACT

3-Hydroxy-3-methylglutaryl-CoA lyase (HL) deficiency is a disorder biochemically characterized by the predominant accumulation of 3-hydroxy-3-methylglutarate (HMG), 3-methylglutarate (MGA), 3-methylglutaconate and 3-hydroxyisovalerate in tissues and biological fluids of the affected patients. Neurological symptoms and hepatopathy are commonly found in HL deficiency, especially during metabolic crises. Since the mechanisms of tissue damage in this disorder are not well understood, in the present study we evaluated the ex vivo effects of acute administration of HMG and MGA on important parameters of oxidative stress in cerebral cortex and liver from young rats. In vivo administration of HMG and MGA provoked an increase of carbonyl and carboxy-methyl-lysine formation in cerebral cortex, but not in liver, indicating that these metabolites induce protein oxidative damage in the brain. We also verified that HMG and MGA significantly decreased glutathione concentrations in both cerebral cortex and liver, implying a reduction of antioxidant defenses. Furthermore, HMG and MGA increased 2',7'-dichlorofluorescin oxidation, but did not alter nitrate and nitrite content in cerebral cortex and liver, indicating that HMG and MGA effects are mainly mediated by reactive oxygen species. HMG and MGA also increased the activities of superoxide dismutase and catalase in cerebral cortex and liver, whereas MGA decreased glutathione peroxidase activity in cerebral cortex. Our present data showing a disruption of redox homeostasis in cerebral cortex and liver caused by in vivo administration of HMG and MGA suggest that this pathomechanism may possibly contribute to the brain and liver abnormalities observed in HL-deficient patients.


Subject(s)
Acetyl-CoA C-Acetyltransferase/deficiency , Amino Acid Metabolism, Inborn Errors/metabolism , Antioxidants/metabolism , Cerebral Cortex/drug effects , Glutathione Peroxidase/metabolism , Liver/drug effects , Acetyl-CoA C-Acetyltransferase/metabolism , Animals , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Homeostasis , Liver/enzymology , Liver/metabolism , Oxidation-Reduction , Oxidative Stress/physiology , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
9.
Neuroscience ; 246: 28-39, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23639877

ABSTRACT

Physical exercise during pregnancy has been considered beneficial to mother and child. Recent studies showed that maternal swimming improves memory in the offspring, increases hippocampal neurogenesis and levels of neurotrophic factors. The objective of this work was to investigate the effect of maternal swimming during pregnancy on redox status and mitochondrial parameters in brain structures from the offspring. Adult female Wistar rats were submitted to five swimming sessions (30 min/day) prior to mating with adult male Wistar rats, and then trained during the pregnancy (five sessions of 30-min swimming/week). The litter was sacrificed when 7 days old, when cerebellum, parietal cortex, hippocampus, and striatum were dissected. We evaluated the production of reactive species and antioxidant status, measuring the activities of superoxide-dismutase (SOD), catalase (CAT) and glutathione-peroxidase (GPx), as well as non-enzymatic antioxidants. We also investigated a potential mitochondrial biogenesis regarding mitochondrion mass and membrane potential, through cytometric approaches. Our results showed that maternal swimming exercise promoted an increase in reactive species levels in cerebellum, parietal cortex, and hippocampus, demonstrated by an increase in dichlorofluorescein oxidation. Mitochondrial superoxide was reduced in cerebellum and parietal cortex, while nitrite levels were increased in cerebellum, parietal cortex, hippocampus, and striatum. Antioxidant status was improved in cerebellum, parietal cortex, and hippocampus. SOD activity was increased in parietal cortex, and was not altered in the remaining brain structures. CAT and GPx activities, as well as non-enzymatic antioxidant potential, were increased in cerebellum, parietal cortex, and hippocampus of rats whose mothers were exercised. Finally, we observed an increased mitochondrial mass and membrane potential, suggesting mitochondriogenesis, in cerebellum and parietal cortex of pups subjected to maternal swimming. In conclusion, maternal swimming exercise induced neurometabolic programing in the offspring that could be of benefit to the rats against future cerebral insults.


Subject(s)
Antioxidants/metabolism , Brain/metabolism , Mitochondria/metabolism , Physical Conditioning, Animal/physiology , Prenatal Exposure Delayed Effects/metabolism , Swimming/physiology , Animals , Animals, Newborn , Female , Male , Membrane Potential, Mitochondrial/physiology , Organelle Biogenesis , Pregnancy , Rats , Rats, Wistar
10.
J Inherit Metab Dis ; 30(5): 800-10, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17885820

ABSTRACT

Animal models of inborn errors of metabolism are useful for investigating the pathogenesis associated with the corresponding human disease. Since the mechanisms involved in the pathophysiology of succinate semialdehyde dehydrogenase (SSADH) deficiency (Aldh5a1; OMIM 271980) are still not established, in the present study we evaluated the tissue antioxidant defences and lipid peroxidation in various cerebral structures (cortex, cerebellum, thalamus and hippocampus) and in the liver of SSADH-deficient mice. The parameters analysed were total radical-trapping antioxidant potential (TRAP) and glutathione (GSH) levels, the activities of the antioxidant enzymes superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx), as well as thiobarbituric acid-reactive substances (TBARS). We first observed that the tissue nonenzymatic antioxidant defences were significantly reduced in the SSADH-deficient animals, particularly in the liver (decreased TRAP and GSH) and in the cerebral cortex (decreased GSH), as compared to the wild-type mice. Furthermore, SOD activity was significantly increased in the liver and cerebellum, whereas the activity of CAT was significantly higher in the thalamus. In contrast, GPx activity was significantly diminished in the hippocampus. Finally, we observed that lipid peroxidation (TBARS levels) was markedly increased in the liver and cerebral cortex, reflecting a high lipid oxidative damage in these tissues. Our data showing an imbalance between tissue antioxidant defences and oxidative attack strongly indicate that oxidative stress is involved in the pathophysiology of SSADH deficiency in mice, and likely the corresponding human disorder.


Subject(s)
Antioxidants/metabolism , Brain Diseases, Metabolic, Inborn/metabolism , Brain/metabolism , Lipid Peroxidation , Liver/metabolism , Oxidative Stress , Succinate-Semialdehyde Dehydrogenase/deficiency , Animals , Brain/enzymology , Brain Diseases, Metabolic, Inborn/enzymology , Brain Diseases, Metabolic, Inborn/genetics , Catalase/metabolism , Cerebellum/enzymology , Cerebellum/metabolism , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Disease Models, Animal , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Hippocampus/enzymology , Hippocampus/metabolism , Liver/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Succinate-Semialdehyde Dehydrogenase/genetics , Superoxide Dismutase/metabolism , Thalamus/enzymology , Thalamus/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
11.
Neuroscience ; 135(1): 111-20, 2005.
Article in English | MEDLINE | ID: mdl-16111821

ABSTRACT

3-Hydroxyglutaric acid (3HGA) accumulates in the inherited neurometabolic disorder known as glutaryl-CoA dehydrogenase deficiency. The disease is clinically characterized by severe neurological symptoms, frontotemporal atrophy and striatum degeneration. Because of the pathophysiology of the brain damage in glutaryl-CoA dehydrogenase deficiency is not completed clear, we investigated the in vitro effect of 3HGA (0.01-5.0mM) on critical enzyme activities of energy metabolism, including the respiratory chain complexes I-V, creatine kinase isoforms and Na(+),K(+)-ATPase in cerebral cortex and striatum from 30-day-old rats. Complex II activity was also studied in rat C6-glioma cells exposed to 3HGA. The effect of 3HGA was further investigated on the rate of oxygen consumption in mitochondria from rat cerebrum. We observed that 1.0mM 3HGA significantly inhibited complex II in cerebral cortex and C6 cells but not the other activities of the respiratory chain complexes. Creatine kinase isoforms and Na(+),K(+)-ATPase were also not affected by the acid. Furthermore, no inhibition of complex II activity occurred when mitochondrial preparations from cerebral cortex or striatum homogenates were used. In addition, 3HGA significantly lowered the respiratory control ratio in the presence of glutamate/malate and succinate under stressful conditions or when mitochondria were permeabilized with digitonin. Since 3HGA stimulated oxygen consumption in state IV and compromised ATP formation, it can be presumed that this organic acid might act as an endogenous uncoupler of mitochondria respiration. Finally, we observed that 3HGA changed C6 cell morphology from a round flat to a spindle-differentiated shape, but did not alter cell viability neither induced apoptosis. The data provide evidence that 3HGA provokes a moderate impairment of brain energy metabolism and do not support the view that 3HGA-induced energy failure would solely explain the characteristic brain degeneration observed in glutaryl-CoA dehydrogenase deficiency patients.


Subject(s)
Brain Chemistry/drug effects , Energy Metabolism/drug effects , Glutarates/pharmacology , Animals , Ca(2+) Mg(2+)-ATPase/metabolism , Cell Line, Tumor/metabolism , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Cell Survival/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Creatine Kinase/metabolism , Cytosol/enzymology , Electron Transport/drug effects , Glioma/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , Neostriatum/drug effects , Neostriatum/metabolism , Nerve Tissue Proteins/biosynthesis , Neurons/drug effects , Neurons/ultrastructure , Oxygen Consumption/drug effects , Rats , Sodium-Potassium-Exchanging ATPase/metabolism
12.
J Inherit Metab Dis ; 28(4): 501-15, 2005.
Article in English | MEDLINE | ID: mdl-15902553

ABSTRACT

Mitochondrial beta-ketothiolase and 2-methyl-3-hydroxybutyryl-CoA dehydrogenase (MHBD) deficiencies are inherited neurometabolic disorders affecting isoleucine catabolism. Biochemically, beta-ketothiolase deficiency is characterized by intermittent ketoacidosis and urinary excretion of 2-methyl-acetoacetate (MAA), 2-methyl-3-hydroxybutyrate (MHB) and tiglylglycine (TG), whereas in MHBD deficiency only MHB and tiglylglycine accumulate. Lactic acid accumulation and excretion are also observed in these patients, being more pronounced in MHBD-deficient individuals, particularly during acute episodes of decompensation. Patients affected by MHBD deficiency usually manifest severe mental retardation and convulsions, whereas beta-ketothiolase-deficient patients present encephalopathic crises characterized by metabolic acidosis, vomiting and coma. Considering that the pathophysiological mechanisms responsible for the neurological alterations of these disorders are unknown and that lactic acidosis suggests an impairment of energy production, the objective of the present work was to investigate the in vitro effect of MAA and MHB, at concentrations varying from 0.01 to 1.0 mmol/L, on several parameters of energy metabolism in cerebral cortex from young rats. We observed that MAA markedly inhibited CO2 production from glucose, acetate and citrate at concentrations as low as 0.01 mmol/L. In addition, the activities of the respiratory chain complex II and succinate dehydrogenase were mildly inhibited by MAA. MHB, at 0.01 mmol/L and higher concentrations, strongly inhibited CO2 production from all tested substrates, as well as the respiratory chain complex IV activity. The other activities of the respiratory chain were not affected by these metabolites. The data indicate a marked blockage in the Krebs cycle and a mild inhibition of the respiratory chain caused by MAA and MHB. Furthermore, MHB inhibited total and mitochondrial creatine kinase activities, which was prevented by the use of the nitric-oxide synthase inhibitor L-NAME and glutathione (GSH). These data indicate that the effect of MHB on creatine kinase was probably mediated by oxidation or other modification of essential thiol groups of the enzyme by nitric oxide and other by-products derived from this organic acid. In contrast, MAA did not affect creatine kinase activity. Taken together, these observations indicate that aerobic energy metabolism is inhibited by MAA and to a greater extent by MHB, a fact that may be related to lactic acidaemia occurring in patients affected by MHBD and beta-ketothiolase deficiencies. If the in vitro effects detected in the present study also occur in vivo, it is tempting to speculate that they may contribute, at least in part, to the neurological dysfunction found in these disorders.


Subject(s)
Acetoacetates/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/embryology , Energy Metabolism , Hydroxybutyrates/pharmacology , 3-Hydroxyacyl CoA Dehydrogenases , Acetates/metabolism , Acetyl-CoA C-Acyltransferase/metabolism , Acidosis/metabolism , Alcohol Oxidoreductases/metabolism , Animals , Brain/metabolism , Carbon Dioxide/chemistry , Carbon Dioxide/metabolism , Cerebral Cortex/metabolism , Citrates/metabolism , Creatine Kinase/metabolism , Dose-Response Relationship, Drug , Electron Transport , Glucose/metabolism , Glutathione/metabolism , Glycine/analogs & derivatives , Glycine/metabolism , In Vitro Techniques , Intellectual Disability , Lactic Acid/metabolism , NG-Nitroarginine Methyl Ester/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Oxygen/metabolism , Rats , Rats, Wistar , Time Factors
13.
J Inherit Metab Dis ; 28(1): 57-67, 2005.
Article in English | MEDLINE | ID: mdl-15702406

ABSTRACT

The pathophysiology of the striatum degeneration characteristic of patients affected by the inherited neurometabolic disorder glutaryl-CoA dehydrogenase deficiency (GDD), also known as glutaric aciduria type I, is still in debate. We have previously reported that 3-hydroxyglutaric acid (3-OH-GA) considered the main neurotoxin in this disorder, induces oxidative stress in rat cerebral cotex. In the present work, we extended these studies by investigating the in vitro effect of 3-OH-GA, at concentrations ranging from 0.01 to 1.0 mmol/L on the brain antioxidant defences by measuring total radical-trapping antioxidant potential (TRAP), total antioxidant reactivity (TAR) and glutathione (GSH) levels, and on the production of hydrogen peroxide (H(2)O(2)), nitric oxide (NO) and malondialdehyde in striatum homogenates from young rats. We observed that TRAP, TAR and GSH levels were markedly reduced (by up to 50%) when striatum homogenates were treated with 3-OH-GA. In contrast, H(2)O(2) (up to 44%), NO (up to 95%) and malondialdehyde levels (up to 28%) were significantly increased by 3-OH-GA. These data indicate that total nonenzymatic antioxidant defences (TRAP) and the tissue capacity to handle an increase of reactive species (TAR) were reduced by 3-OH-GA in the striatum. Furthermore, the results also reflect an increase of lipid peroxidation, probably secondary to 3-OH-GA-induced free radical production. Thus, it may be presumed that oxidative stress is involved in the neuropathology in GDD.


Subject(s)
Corpus Striatum/metabolism , Glutarates/metabolism , Oxidative Stress , Oxidoreductases Acting on CH-CH Group Donors/deficiency , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Dose-Response Relationship, Drug , Glutaryl-CoA Dehydrogenase , Glutathione/metabolism , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Lipid Metabolism , Lipid Peroxidation , Male , Malondialdehyde/pharmacology , Nitric Oxide/metabolism , Rats , Rats, Wistar , Thiobarbituric Acid Reactive Substances/metabolism , Time Factors
14.
Eur J Clin Invest ; 33(10): 840-7, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14511354

ABSTRACT

BACKGROUND: Tissue accumulation of high amounts of D-2-hydroxyglutaric acid (DGA) is the biochemical hallmark of the inherited neurometabolic disorder D-2-hydroxyglutaric aciduria (DHGA). Patients affected by this disease usually present hypotonia, muscular weakness, hypertrophy and cardiomyopathy, besides severe neurological findings. However, the underlying mechanisms of muscle injury in this disorder are virtually unknown. MATERIALS AND METHODS: In the present study we have evaluated the in vitro role of DGA, at concentrations ranging from 0.25 to 5.0 mM, on total, cytosolic and mitochondrial creatine kinase activities from skeletal and cardiac muscle of 30-day-old Wistar rats. We also tested the effects of various antioxidants on the effects elicited by DGA. RESULTS: We first verified that total creatine kinase (CK) activity from homogenates was significantly inhibited by DGA (22-24% inhibition) in skeletal and cardiac muscle, and that this activity was approximately threefold higher in skeletal muscle than in cardiac muscle. We also observed that CK activities from mitochondrial (Mi-CK) and cytosolic (Cy-CK) preparations from skeletal muscle and cardiac muscle were also inhibited (12-35% inhibition) by DGA at concentrations as low as 0.25 mm, with the effect being more pronounced in cardiac muscle preparations. Finally, we verified that the DGA-inhibitory effect was fully prevented by preincubation of the homogenates with reduced glutathione and cysteine, suggesting that this effect is possibly mediated by modification of essential thiol groups of the enzyme. Furthermore, alpha-tocopherol, melatonin and the inhibitor of nitric oxide synthase L-NAME were unable to prevent this effect, indicating that the most common reactive oxygen and nitrogen species were not involved in the inhibition of CK provoked by DGA. CONCLUSION: Considering the importance of creatine kinase activity for cellular energy homeostasis, our results suggest that inhibition of this enzyme by increased levels of DGA might be an important mechanism involved in the myopathy and cardiomyopathy of patients affected by DHGA.


Subject(s)
Creatine Kinase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Glutarates/pharmacology , Heart/drug effects , Muscle, Skeletal/drug effects , Animals , Antioxidants/pharmacology , Creatine Kinase/metabolism , Creatine Kinase, Mitochondrial Form , Cytosol/enzymology , Dose-Response Relationship, Drug , Glutarates/antagonists & inhibitors , In Vitro Techniques , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Muscle, Skeletal/enzymology , Myocardium/enzymology , Rats , Rats, Wistar
15.
Semin Thromb Hemost ; 25(1): 49-55, 1999.
Article in English | MEDLINE | ID: mdl-10327221

ABSTRACT

In contrast to other European countries, in Germany more than 90% of oral anticoagulated patients are controlled by general practitioners. The International Normalized Ratio (INR) system in laboratory control is not in widespread use, often leading to misinterpretations of prothrombin time (PT) measurements. To improve the management of anticoagulated patients, a model was developed, consisting of different questionnaires and on the base of the INR system. Since 1993, 60 patients in our Department's outpatient anticoagulant clinic and since 1996 16 patients in the office of a general practitioner were followed for 146.32 patient years. There were no thromboembolic events and no major bleedings during follow-up. A total of 126 minor bleedings occurred in 30 patients. There were no significant differences in INR values and stable phases between the two centers; however, significantly shorter stable phases in patients with bleeding episodes were noted. Thus, this model seems to be useful also in general practitioners' hands.


Subject(s)
Anticoagulants , Thrombosis/drug therapy , Administration, Oral , Anticoagulants/administration & dosage , Anticoagulants/adverse effects , Anticoagulants/therapeutic use , Germany , Humans , International Normalized Ratio , Quality Control
17.
Thromb Res ; 62(1-2): 75-81, 1991 Apr.
Article in English | MEDLINE | ID: mdl-1906642

ABSTRACT

The pharmacokinetics of urokinase (two-chain urokinase-type plasminogen activator, tcu-PA) and single-chain urokinase-type plasminogen activator (scu-PA) were studied in 20 patients with acute myocardial infarction (AMI). Ten consecutive patients received 2.5 million units tcu-PA by bolus injection within 5 min during the first 6 h after AMI (group I). Ten further consecutive patients received 250,000 U tcu-PA within 5 min, followed by 4.5 million U scu-PA by intravenous infusion over 40 min (group II). An enzyme immunoassay was developed for urokinase antigen determinations, and a fibrin plate assay for determinations of fibrinolytic activity was applied. Using a 3-compartment model, in group I 98% of urokinase antigen were cleared with a half-life of 60.8 min. After scu-PA, urokinase antigen was cleared with half-lives (area under the curve in parentheses) of 6.9 min (74.8%), 26.5 min (23.6%), and 329.7 min (2.2%). The half-disappearance times of fibrinolytic activity were 18 and 8 min in group I and II, respectively. A more pronounced decrease of plasminogen was observed after tcu-PA.


Subject(s)
Myocardial Infarction/metabolism , Plasminogen Activators/pharmacokinetics , Urokinase-Type Plasminogen Activator/pharmacokinetics , Half-Life , Humans , Middle Aged , Molecular Weight , Myocardial Infarction/drug therapy , Plasminogen Activators/therapeutic use , Urokinase-Type Plasminogen Activator/therapeutic use
18.
Vasa Suppl ; 33: 318-9, 1991.
Article in German | MEDLINE | ID: mdl-1788734

ABSTRACT

The method of nuclear magnetic resonance (NMR) spectroscopy compared to the parameters absolute-, pain-free walking distance and ankle/arm coefficient in Doppler pressure was used for observation of patients with peripheral arterial occlusive disease stage IIb according to Fontaine. While the classic parameters (walking distances, ankle/arm coefficient) described a homogenous group, NMR-spectroscopy parameters showed marked inter- and intraindividual variations during exercise. Further studies on high magnetic power fields, exercise patterns and muscle recreation analysis have to be carried out to develop a reliable system of non invasive muscle energy monitoring in vascular diseases.


Subject(s)
Arterial Occlusive Diseases/diagnosis , Energy Metabolism/physiology , Ischemia/diagnosis , Magnetic Resonance Spectroscopy , Muscles/blood supply , Phosphates/metabolism , Aged , Aged, 80 and over , Arterial Occlusive Diseases/classification , Arterial Occlusive Diseases/physiopathology , Female , Follow-Up Studies , Humans , Ischemia/physiopathology , Male , Middle Aged
20.
Acta Med Austriaca ; 18 Suppl 1: 52-5, 1991.
Article in German | MEDLINE | ID: mdl-1719732

ABSTRACT

Hemorheological therapy through hemodilution has been gaining importance for several years and been applied to an ever increasing degree in stationary as well as in ambulant treatment. While renal insufficiency without previously established nephropathy is known to be a side effect of dextrans, cases of HES-induced nephropathy have so far not been reported. Two cases are presented in which in the course of stationary hemodilution therapy with HES an acute deterioration of an already exiting nephropathy was noted. Possible pathophysiological causes for such a deterioration are most likely to be found in an increased permeability of the glomerular basal lamina. Hydroxyethyl starch molecules are filtered above the physiological renal threshold which increases the viscosity of the primary urine. This can be counteracted by increasing diuresis. This conclusion can be drawn from our own observations which proved that renal insufficiency can be avoided through sufficient fluid intake (approx. 3 liters/day). In patients with creatinine values above 1.5/dl and arterial hypertension the indication for hemodilution therapy must be analysed carefully. If hemodilution therapy proves to be necessary, sufficient fluid intake must be guaranteed. Retention parameters must be controlled every other day in the course of the therapy. As an alternative, the administration of gelatin preparations should be considered as it does not cause cumulation.


Subject(s)
Acute Kidney Injury/chemically induced , Diabetic Angiopathies/therapy , Diabetic Nephropathies/complications , Diabetic Retinopathy/therapy , Hemodilution/adverse effects , Hydroxyethyl Starch Derivatives/adverse effects , Acute Kidney Injury/blood , Aged , Cerebral Infarction/blood , Cerebral Infarction/therapy , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/complications , Diabetic Angiopathies/blood , Diabetic Nephropathies/blood , Diabetic Retinopathy/blood , Female , Hemodilution/methods , Humans , Hydroxyethyl Starch Derivatives/administration & dosage , Risk Factors , Urea/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...