Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
ACS Appl Mater Interfaces ; 12(18): 20234-20242, 2020 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-32285658

RESUMO

Metal-organic frameworks (MOFs) can be applied in biology and medicine as drug delivery systems by carrying drugs on their surfaces or releasing bioactive ligands. To investigate the therapeutic potential of hydrogels that contain MOFs, three MOFs containing glutarate and 1,2-bis(4-pyridyl)ethylene ligands were synthesized by the previously reported hydrothermal or solvothermal reactions: Cu-MOF 1, Co-MOF 2, and Zn-MOF 3. Bioactive MOF-embedded hydrogels (hydrogel@Cu-MOF 1, hydrogel@Co-MOF 2, and hydrogel@Zn-MOF 3) were prepared by UV light-mediated thiol-ene photopolymerization using diacrylated polyethylene glycol (PEG), 4-arm-thiolated PEG, and MOFs. The activities of the MOF-embedded hydrogels were tested against the Gram-negative bacterium Escherichia coli and the Gram-positive bacterium Staphylococcus aureus. These MOF-embedded hydrogels were observed to be very stable, based on the release test of MII ions, and both hydrogel@Cu-MOF 1 and hydrogel@Co-MOF 2 showed excellent antibacterial activity. Although, in human dermal fibroblasts, hydrogel@Cu-MOF 1 showed no cytotoxic effects, it exhibited 99.9% antibacterial effects at the minimum bactericidal concentration. Physical properties such as the surface area and dimension of MOFs with different central metals appeared to be more important than the chemical properties of the ligands in determining the effects on bacteria. These MOF-embedded hydrogels may be useful in antibacterial applications such as cosmetics, treatment of skin diseases, and drug delivery owing to their low cytotoxicity and high bactericidal activity.


Assuntos
Antibacterianos/farmacologia , Hidrogéis/farmacologia , Estruturas Metalorgânicas/farmacologia , Antibacterianos/química , Antibacterianos/toxicidade , Escherichia coli/efeitos dos fármacos , Glutaratos/química , Glutaratos/farmacologia , Glutaratos/toxicidade , Humanos , Hidrogéis/química , Hidrogéis/toxicidade , Ligantes , Estruturas Metalorgânicas/química , Estruturas Metalorgânicas/toxicidade , Testes de Sensibilidade Microbiana , Piridinas/química , Piridinas/farmacologia , Piridinas/toxicidade , Staphylococcus aureus/efeitos dos fármacos
2.
Biochimie ; 171-172: 187-196, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32169667

RESUMO

High urinary excretion and tissue accumulation of 3-methylglutaric acid (MGA) are observed in patients affected by 3-hydroxy-3-methylglutaric (HMGA) and 3-methylglutaconic (MGTA) acidurias. The pathomechanisms underlying the hepatic dysfunction commonly observed in these disorders are not fully elucidated so that we investigated here the effects of intraperitoneal administration of MGA on redox homeostasis, mitochondrial bioenergetics, biogenesis and dynamics in rat liver. The effects of a pre-treatment with the protective compound bezafibrate (BEZ) were also determined. Our data showed that MGA induced lipid peroxidation and altered enzymatic and non-enzymatic antioxidant defenses in liver, indicating redox homeostasis disruption. BEZ prevented most of these alterations induced by MGA. MGA also decreased the activities of the respiratory chain complexes II and IV and increased of II-III, whereas BEZ prevented the alteration in complex II activity. Furthermore, MGA decreased levels of nuclear PGC-1α and Sirt1, and increased levels of AMPKα1 and cytosolic PPARγ, which were blocked by BEZ. MGA augmented the levels of mitofusin-1 and dynamin-related protein 1, suggesting that both fusion and fission mitochondrial processes are enhanced by MGA. BEZ was able to prevent only the changes in mitofusin-1 levels. Collectively, these findings indicate that oxidative stress and mitochondrial dysfunction are mechanisms involved in the hepatic dysfunction found in HMGA and MGTA. It is also presumed that mitochondrial biogenesis stimulation may constitute an attractive approach to reduce MGA toxicity in liver of individuals affected by HMGA and MGTA.


Assuntos
Bezafibrato/uso terapêutico , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Glutaratos/toxicidade , Meglutol/análogos & derivados , Meglutol/toxicidade , Animais , Antioxidantes/uso terapêutico , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Feminino , Peroxidação de Lipídeos , Fígado/metabolismo , Masculino , Meglutol/metabolismo , Mitocôndrias/metabolismo , Biogênese de Organelas , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Wistar
3.
Cancer Lett ; 472: 151-164, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31846689

RESUMO

IDH1 (Isocitrate dehydrogenase 1) mutation occurring at codon 132 (R132) in prostate cancer (PCa) is considered as a classifier for a subgroup of PCas with accumulation of oncometabolite R-2HG (R-2-hydroxyglutarate). Here we found that adding R-2HG or the mutant IDH1 R132H could promote PCa cell invasion in androgen receptor (AR)-negative PC3 cells or suppressing the AR in AR-positive C4-2 cells. Mechanism dissection revealed that R-2HG could increase circRNA-51217 expression to sponge miRNA-646, which might then lead to increase TGFß1 expression and thus induce TGFß1/p-Smad2/3 signaling to increase PCa cell invasion. AR can suppress this R-2HG/circRNA-51217/miRNA-646/TGFß1/p-Smad2/3 signaling-increased PCa cell invasion via repressing TGFß1 transcription and inhibiting circRNA-51217 expression through regulating ADAR2 expression. Preclinical studies with an in vivo xenograft mouse model also revealed that PCa cells with the IDH1 R132H mutation have more invasive metastasis. This study demonstrates that IDH1 R132H mutation with increased oncometabolite R-2HG in PCa cells may play important roles to increase PCa cell invasion.


Assuntos
Isocitrato Desidrogenase/genética , Neoplasias da Próstata/genética , RNA Circular/sangue , Fator de Crescimento Transformador beta1/genética , Animais , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glutaratos/toxicidade , Xenoenxertos , Humanos , Masculino , Camundongos , MicroRNAs/efeitos dos fármacos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Próstata/efeitos dos fármacos , Próstata/patologia , Neoplasias da Próstata/induzido quimicamente , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , Receptores Androgênicos/genética , Proteína Smad2/genética , Proteína Smad3/genética
4.
Neuroscience ; 401: 84-95, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30668975

RESUMO

The endocannabinoid system (ECS) regulates several physiological processes in the Central Nervous System, including the modulation of neuronal excitability via activation of cannabinoid receptors (CBr). Both glutaric acid (GA) and quinolinic acid (QUIN) are endogenous metabolites that, under pathological conditions, recruit common toxic mechanisms. A synergistic effect between them has already been demonstrated, supporting potential implications for glutaric acidemia type I (GA I). Here we investigated the possible involvement of a cannabinoid component in the toxic model exerted by QUIN + GA in rat cortical slices and primary neuronal cell cultures. The effects of the CB1 receptor agonist anandamide (AEA), and the fatty acid amide hydrolase inhibitor URB597, were tested on cell viability in cortical brain slices and primary neuronal cultures exposed to QUIN, GA, or QUIN + GA. As a pre-treatment to the QUIN + GA condition, AEA prevented the loss of cell viability in both preparations. URB597 only protected in a moderate manner the cultured neuronal cells against the QUIN + GA-induced damage. The use of the CB1 receptor reverse agonist AM251 in both biological preparations prevented partially the protective effects exerted by AEA, thus suggesting a partial role of CB1 receptors in this toxic model. AEA also prevented the cell damage and apoptotic death induced by the synergic model in cell cultures. Altogether, these findings demonstrate a modulatory role of the ECS on the synergic toxic actions exerted by QUIN + GA, thus providing key information for the understanding of the pathophysiological events occurring in GA I.


Assuntos
Ácidos Araquidônicos/farmacologia , Córtex Cerebral/efeitos dos fármacos , Endocanabinoides/farmacologia , Glutaratos/toxicidade , Neurônios/efeitos dos fármacos , Alcamidas Poli-Insaturadas/farmacologia , Ácido Quinolínico/toxicidade , Animais , Benzamidas/farmacologia , Agonistas de Receptores de Canabinoides/farmacologia , Carbamatos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Sinergismo Farmacológico , Endocanabinoides/metabolismo , Feminino , Masculino , Neurônios/metabolismo , Piperidinas/farmacologia , Gravidez , Pirazóis/farmacologia , Ratos , Ratos Endogâmicos WF , Receptores de Canabinoides/metabolismo
5.
Mol Neurobiol ; 56(7): 4945-4959, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30421167

RESUMO

Dysfunction of basal ganglia neurons is a characteristic of glutaric acidemia type I (GA-I), an autosomal recessive inherited neurometabolic disease characterized by deficiency of glutaryl-CoA dehydrogenase (GCDH) and accumulation of glutaric acid (GA). The affected patients present clinical manifestations such as motor dysfunction and memory impairment followed by extensive striatal neurodegeneration. Knowing that there is relevant striatal dysfunction in GA-I, the purpose of the present study was to verify the performance of young rats chronically injected with GA in working and procedural memory test, and whether N-acetylcysteine (NAC) would protect against impairment induced by GA. Rat pups were injected with GA (5 µmol g body weight-1, subcutaneously; twice per day; from the 5th to the 28th day of life) and were supplemented with NAC (150 mg/kg/day; intragastric gavage; for the same period). We found that GA injection caused delay procedural learning; increase of cytokine concentration, oxidative markers, and caspase levels; decrease of antioxidant defenses; and alteration of acetylcholinesterase (AChE) activity. Interestingly, we found an increase in glial cell immunoreactivity and decrease in the immunoreactivity of nuclear factor-erythroid 2-related factor 2 (Nrf2), nicotinic acetylcholine receptor subunit alpha 7 (α7nAChR), and neuronal nuclei (NeuN) in the striatum. Indeed, NAC administration improved the cognitive performance, ROS production, neuroinflammation, and caspase activation induced by GA. NAC did not prevent neuronal death, however protected against alterations induced by GA on Iba-1 and GFAP immunoreactivities and AChE activity. Then, this study suggests possible therapeutic strategies that could help in GA-I treatment and the importance of the striatum in the learning tasks.


Assuntos
Acetilcisteína/uso terapêutico , Neurônios Colinérgicos/efeitos dos fármacos , Glutaratos/toxicidade , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/prevenção & controle , Neuroglia/efeitos dos fármacos , Acetilcisteína/farmacologia , Animais , Neurônios Colinérgicos/metabolismo , Masculino , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/metabolismo , Neuroglia/metabolismo , Ratos , Ratos Wistar
7.
J Inherit Metab Dis ; 41(1): 91-99, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29098534

RESUMO

Glutaric aciduria type I is a rare, autosomal recessive, inherited defect of glutaryl-CoA dehydrogenase. Deficiency of this protein in L-lysine degradation leads to the characteristic accumulation of nontoxic glutarylcarnitine and neurotoxic glutaric acid (GA), glutaryl-CoA, and 3-hydroxyglutaric acid. Untreated patients develop bilateral lesions of basal ganglia resulting in a complex movement disorder with predominant dystonia in infancy and early childhood. The current pathomechanistic concept strongly focuses on imbalanced neuronal energy metabolism due to accumulating metabolites, whereas little is known about the pathomechanistic role of astrocytes, which are thought to be in constant metabolic crosstalk with neurons. We found that glutaric acid (GA) causes astrocytic cell death under starvation cell culture conditions, i.e. low glucose, without glutamine and fetal calf serum. Glutamine completely abolished GA-induced toxicity, suggesting involvement of glutaminolysis. Increasing dependence on glutaminolysis by chemical induction of hypoxia signaling-potentiated GA-induced toxicity. We further show that GA disturbs glutamine degradation by specifically inhibiting glutamate dehydrogenase. Summarizing our study shows that pathologically relevant concentrations of GA block an important step in the metabolic crosstalk between neurons and astrocytes, ultimately leading to astrocytic cell death.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Astrócitos/efeitos dos fármacos , Encefalopatias Metabólicas/metabolismo , Inibidores Enzimáticos/toxicidade , Glutamato Desidrogenase/antagonistas & inibidores , Glutamina/metabolismo , Glutaratos/toxicidade , Glutaril-CoA Desidrogenase/deficiência , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Encefalopatias Metabólicas/patologia , Morte Celular/efeitos dos fármacos , Hipóxia Celular , Linhagem Celular , Glutamato Desidrogenase/metabolismo , Glutamina/farmacologia , Glutaril-CoA Desidrogenase/metabolismo , Ratos
8.
Mol Neurobiol ; 55(6): 5362-5376, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28936789

RESUMO

It has been shown that synergistic toxic effects of quinolinic acid (QUIN) and glutaric acid (GA), both in isolated nerve endings and in vivo conditions, suggest the contribution of these metabolites to neurodegeneration. However, this synergism still requires a detailed characterization of the mechanisms involved in cell damage during its occurrence. In this study, the effects of subtoxic concentrations of QUIN and/or GA were tested in neuronal cultures, co-cultures (neuronal cells + astrocytes), and mixed cultures (neuronal cells + astrocytes + microglia) from rat cortex and striatum. The exposure of different cortical and striatal cell cultures to QUIN + GA resulted in cell death and stimulated different markers of oxidative stress, including reactive oxygen species (ROS) formation; changes in the activity of antioxidant enzymes such as superoxide dismutase, catalase, and glutathione peroxidase; and depletion of endogenous antioxidants such as -SH groups and glutathione. The co-incubation of neuronal cultures with QUIN + GA plus the N-methyl-D-aspartate antagonist MK-801 prevented cell death but not ROS formation, whereas the antioxidant melatonin reduced both parameters. Our results demonstrated that QUIN and GA can create synergistic scenarios, inducing toxic effects on some parameters of cell viability via the stimulation of oxidative damage. Therefore, it is likely that oxidative stress may play a major causative role in the synergistic actions exerted by QUIN + GA in a variety of cell culture conditions involving the interaction of different neural types.


Assuntos
Glutaratos/toxicidade , Modelos Biológicos , Neurônios/metabolismo , Estresse Oxidativo , Ácido Quinolínico/toxicidade , Animais , Antioxidantes/metabolismo , Catalase/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Córtex Cerebral/patologia , Técnicas de Cocultura , Maleato de Dizocilpina/farmacologia , Feminino , Gliose/metabolismo , Gliose/patologia , Glutaratos/administração & dosagem , Glutationa/metabolismo , Melatonina/farmacologia , Neostriado/patologia , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Ácido Quinolínico/administração & dosagem , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo
9.
Toxicol In Vitro ; 42: 47-53, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28396261

RESUMO

d-2-hydroxyglutaric (D-2-HGA) and l-2-hydroxyglutaric (L-2-HGA) acidurias are rare neurometabolic disorders biochemically characterized by increased levels of d-2-hydroxyglutaric acid (D-2-HG) and l-2-hydroxyglutaric acid (L-2-HG) respectively, in biological fluids and tissues. These diseases are caused by mutations in the specific enzymes involved in the metabolic pathways of these organic acids. In the present work, we first investigated whether D-2-HG and L-2-HGA could provoke DNA oxidative damage in blood leukocytes and whether l-carnitine (LC) could prevent the in vitro DNA damage induced by these organic acids. It was verified that 50µM of D-2-HG and 30µM of L-2-HG significantly induced DNA damage that was prevented by 30 and 150µM of LC. We also evaluated oxidative stress parameters in urine of L-2-HGA patients and observed a significant increase of oxidized guanine species and di-tyrosine, biomarkers of oxidative DNA and protein damage, respectively. In contrast, no significant changes of urinary isoprostanes and reactive nitrogen species levels were observed in these patients. Taken together, our data indicate the involvement of oxidative damage, especially on DNA, in patients affected by these diseases and the protective effect of LC.


Assuntos
Encefalopatias Metabólicas Congênitas/genética , Carnitina/farmacologia , Dano ao DNA/efeitos dos fármacos , Glutaratos/toxicidade , Substâncias Protetoras/farmacologia , Adolescente , Adulto , Encefalopatias Metabólicas Congênitas/urina , Criança , Pré-Escolar , Ensaio Cometa , Dinoprosta/análogos & derivados , Dinoprosta/urina , Guanina/análogos & derivados , Guanina/urina , Humanos , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Nitrogênio/urina , Tirosina/análogos & derivados , Tirosina/urina , Adulto Jovem
10.
Neuroscience ; 310: 578-88, 2015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-26431622

RESUMO

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.


Assuntos
Ácidos Acíclicos/metabolismo , Ácidos Acíclicos/toxicidade , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Benzoxazinas/farmacologia , Encefalopatias Metabólicas/metabolismo , Encéfalo/metabolismo , Agonistas de Receptores de Canabinoides/farmacologia , Glutaril-CoA Desidrogenase/deficiência , Morfolinas/farmacologia , Naftalenos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Glutaratos/metabolismo , Glutaratos/toxicidade , Glutaril-CoA Desidrogenase/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Ácido Metilmalônico/metabolismo , Ácido Metilmalônico/toxicidade , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Propionatos/metabolismo , Propionatos/toxicidade , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo
11.
Neuroscience ; 308: 64-74, 2015 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-26343296

RESUMO

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.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Encefalopatias Metabólicas/metabolismo , Encéfalo/metabolismo , Glutaratos/metabolismo , Glutaril-CoA Desidrogenase/deficiência , Ácido Quinolínico/metabolismo , Sinaptossomos/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Modelos Animais de Doenças , Glutaratos/toxicidade , Glutaril-CoA Desidrogenase/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Ácido Metilmalônico/metabolismo , Ácido Metilmalônico/toxicidade , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Propionatos/metabolismo , Propionatos/toxicidade , Ácido Quinolínico/toxicidade , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Sinaptossomos/efeitos dos fármacos
12.
Neuroscience ; 277: 281-93, 2014 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-25043325

RESUMO

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.


Assuntos
Corpo Estriado/efeitos dos fármacos , Glutaratos/toxicidade , Animais , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Creatina/farmacologia , Maleato de Dizocilpina/farmacologia , Glutaratos/metabolismo , Glutaratos/farmacologia , Glutationa/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Peroxidação de Lipídeos/fisiologia , Malondialdeído/metabolismo , N-Metilaspartato/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Ratos , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo
13.
Neurotox Res ; 25(4): 381-91, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24297153

RESUMO

Glutaric acid (GA) is a neurotoxic metabolite that accumulates in the CNS of patients with glutaric acidemia-I (GA-I), a neurometabolic disease caused by deficient activity of glutaryl-CoA dehydrogenase. Most GA-I patients display characteristic CNS lesions, mainly in the gray and white matter of basal ganglia and cerebral cortex. Neurons and astrocytes are believed to be vulnerable to millimolar concentrations of GA. However, little is known about the effects of GA on oligodendrocytes (OL) and the myelination process in the postnatal brain. Here, we show that a single intracerebroventricular administration of GA to rat neonatal pups induced a selective and long-lasting myelination failure in the striatum but no deleterious effect in the myelination of the corpus callosum. At 45 days post-GA injection, the myelinated area of striatal axonal bundles was decreased by 35 %, and the expression of myelin basic protein and myelin-associated glycoprotein (MAG) reduced by 25 and 60 %, respectively. This was accompanied by long lasting cytopathology features in MAG and CC-1-expressing OLs, which was confirmed by transmission electron microscopy. Remarkably, GA did not induce acute loss of pre-OLs in the striatum as assessed by NG2 or PDGFRα immunohistochemistry, suggesting an indirect and progressive mechanism for OL damage. In accordance, GA-induced white matter injury was restricted to the striatum and associated to GA-induced astrocytosis and neuronal loss. In conclusion, the current evidence indicates a pathogenic mechanism by which GA can permanently affect myelin status.


Assuntos
Corpo Caloso/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Glutaratos/toxicidade , Bainha de Mielina/efeitos dos fármacos , Substância Branca/efeitos dos fármacos , Erros Inatos do Metabolismo dos Aminoácidos , Animais , Animais Recém-Nascidos , Encefalopatias Metabólicas , Morte Celular/efeitos dos fármacos , Corpo Caloso/crescimento & desenvolvimento , Corpo Caloso/metabolismo , Corpo Caloso/ultraestrutura , Corpo Estriado/crescimento & desenvolvimento , Corpo Estriado/metabolismo , Corpo Estriado/ultraestrutura , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Gliose/induzido quimicamente , Gliose/metabolismo , Gliose/patologia , Glutaril-CoA Desidrogenase/deficiência , Proteína Básica da Mielina/metabolismo , Bainha de Mielina/fisiologia , Glicoproteína Associada a Mielina/metabolismo , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Neurônios/fisiologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Oligodendroglia/ultraestrutura , Ratos , Ratos Sprague-Dawley , Substância Branca/metabolismo , Substância Branca/ultraestrutura
14.
Oxid Med Cell Longev ; 2013: 607610, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23606926

RESUMO

trans-Glutaconic acid (tGA) is an unsaturated C5-dicarboxylic acid which may be found accumulated in glutaric aciduria type I, whose pathophysiology is still uncertain. In the present work it was investigated the in vitro effect of increasing tGA concentrations on neurochemical and oxidative stress parameters in rat cerebral cortex. We observed that Na(+), K(+)-ATPase activity was reduced by tGA, but not creatine kinase, respiratory chain complex IV, and ATP synthase activities. On the other hand, tGA significantly increased lipid peroxidation (thiobarbituric acid-reactive species levels and spontaneous chemiluminescence), as well as protein oxidative damage (oxidation of sulfhydryl groups). tGA also significantly decreased nonenzymatic antioxidant defenses (TRAP and reduced glutathione levels). Our data suggest that tGA may be neurotoxic in rat brain.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Glutaratos/toxicidade , Complexos de ATP Sintetase/metabolismo , Animais , Córtex Cerebral/metabolismo , Creatina Quinase/metabolismo , Isomerismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Mitocôndrias/metabolismo , Proteínas/química , Proteínas/metabolismo , Ratos , Ratos Wistar , ATPase Trocadora de Sódio-Potássio/metabolismo , Compostos de Sulfidrila/metabolismo
15.
J Pharm Sci ; 101(9): 3292-304, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22674061

RESUMO

Paclitaxel (PTX) is a potent chemotherapy for many cancers but it suffers from very poor solubility. Consequently, the TAXOL formulation uses copious amounts of the surfactant Cremophor EL to solubilize the drug for injection, resulting in severe hypersensitivity and neutropenia. In contrast to Cremophor EL, presented is a way to solubilize PTX by conjugation of a dicarboxylic fatty acid for specific binding to the ubiquitous protein, serum albumin. The conjugation chemistry was simplified to a single step using the activated anhydride form of 3-pentadecylglutaric (PDG) acid, which is reactive to a variety of nucleophiles. The PDG derivative is less cytotoxic than the parent compound and was found to slowly hydrolyze to PTX (≈ 5% over 72 h) in serum, tumor cytosol, and tumor tissue homogenate. When injected intravenously to tumor-bearing mice, [(3) H]-PTX in the TAXOL formulation was cleared rapidly with a half-life of 7 h. In the case of the PDG derivative of PTX, the drug is quickly distributed and approximately 20% of the injected dose remained in the vasculature experiencing a 23 h half-life. These improvements from modifying PTX with the PDG fatty acid present the opportunity for PDG to become a generic modification for the improvement of many therapeutics.


Assuntos
Antineoplásicos Fitogênicos/farmacocinética , Neoplasias Colorretais/metabolismo , Portadores de Fármacos , Ácidos Graxos/química , Glutaratos/química , Paclitaxel/farmacocinética , Albumina Sérica/metabolismo , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/sangue , Antineoplásicos Fitogênicos/química , Biotransformação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Química Farmacêutica , Neoplasias Colorretais/patologia , Estabilidade de Medicamentos , Ácidos Graxos/toxicidade , Feminino , Glutaratos/toxicidade , Meia-Vida , Humanos , Hidrólise , Injeções Intravenosas , Camundongos , Camundongos Endogâmicos BALB C , Paclitaxel/administração & dosagem , Paclitaxel/análogos & derivados , Paclitaxel/sangue , Paclitaxel/química , Ligação Proteica , Albumina Sérica/química , Albumina Sérica/toxicidade , Albumina Sérica Humana , Tecnologia Farmacêutica/métodos , Distribuição Tecidual
16.
J Neurosci Res ; 90(9): 1723-31, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22535575

RESUMO

Glutaric acidemia type I (GA-I) is an inherited metabolic disease characterized by accumulation of glutaric acid (GA) and seizures. The intrastriatal GA administration in rats has been used as an animal model to mimic seizures presented by glutaric acidemic patients. m-Trifluoromethyl diphenyl diselenide, (m-CF(3) -C(6) H(4) Se)(2) , is an organoselenium compound that protects against seizures induced by pentylenetetrazole in mice. Thus, the aim of this study was to investigate whether (m-CF(3) -C(6) H(4) Se)(2) is effective against GA-induced seizures and oxidative stress in rat pups 21 days of age. Our findings demonstrate that (m-CF(3) -C(6) H(4) Se)(2) preadministration (50 mg/kg; p.o.) protected against the reduction in latency and the increased duration of GA (1.3 µmol/right striatum)-induced seizures in rat pups. In addition, (m-CF(3) -C(6) H(4) Se)(2) protected against the increase in reactive species generation and the reduction in antioxidant defenses glutathione peroxidase and glutathione S-transferase activities induced by GA. By contrast, no change in glutathione reductase or catalase activities was found. In addition, (m-CF(3) -C(6) H(4) Se)(2) was effective in protecting against inhibition of Na(+) ,K(+) -ATPase activity caused by GA in striatum of rat pups. This study showed for the first time that GA administration caused an increase in [(3) H]GABA uptake from striatum slices of rat pups and that (m-CF(3) -C(6) H(4) Se)(2) preadministration protected against this increase. A positive correlation between duration of seizures and [(3) H]GABA uptake levels was demonstrated. The results indicate that (m-CF(3) -C(6) H(4) Se)(2) protected against GA-induced seizures. Moreover, these findings suggest that the protection against oxidative stress, the inhibition of Na(+) ,K(+) -ATPase activity, and the increase in [(3) H]GABA uptake are possible mechanisms for the potential anticonvulsant action of (m-CF(3) -C(6) H(4) Se)(2).


Assuntos
Anticonvulsivantes/farmacologia , Glutaratos/toxicidade , Compostos de Organossilício/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Convulsões/tratamento farmacológico , Convulsões/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/complicações , Erros Inatos do Metabolismo dos Aminoácidos/tratamento farmacológico , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Animais , Encefalopatias Metabólicas/complicações , Encefalopatias Metabólicas/tratamento farmacológico , Encefalopatias Metabólicas/metabolismo , Modelos Animais de Doenças , Glutaril-CoA Desidrogenase/deficiência , Glutaril-CoA Desidrogenase/metabolismo , Masculino , Ratos , Ratos Wistar , Convulsões/induzido quimicamente , Ácido gama-Aminobutírico
17.
Int J Dev Neurosci ; 27(1): 65-72, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18930800

RESUMO

Glutaric acidemia type I (GA-I) is an inherited metabolic disease characterized by accumulation of glutaric acid (GA) and striatal degeneration. Although growing evidence suggests that excitotoxicity and oxidative stress play central role in the neuropathogenesis of this disease, mechanism underlying striatal damage in this disorder is not well established. Thus, we decided to investigate the in vitro effects of GA 10nM (a low concentration that can be present initial development this disorder) on l-[(3)H]glutamate uptake and reactive oxygen species (ROS) generation in synaptosomes from striatum of rats. GA reduced l-[(3)H]glutamate uptake in synaptosomes from 1 up to 30min after its addition. Furthermore, we also provided some evidence that GA competes with the glutamate transporter inhibitor l-trans-pyrrolidine-2,4-dicarboxylate (PDC), suggesting a possible interaction of GA with glutamate transporters on synaptosomes. Moreover, GA produced a significant decrease in the V(MAX) of l-[(3)H]glutamate uptake, but did not affect the K(D) value. Although the GA did not show oxidant activity per se, it increased the ROS generation in striatal synaptosomes. To evaluate the involvement of reactive species generation in the GA-induced l-[(3)H]glutamate uptake inhibition, trolox (0.3, 0.6 and 6muM) was added on the incubation medium. Statistical analysis showed that trolox did not decrease inhibition of GA-induced l-[(3)H]glutamate uptake, but decreased GA-induced reactive species formation in striatal synaptosomes (1, 3, 5, 10, 15 and 30min), suggesting that ROS generation appears to occur secondarily to glutamatergic overstimulation in this model of organic acidemia. Since GA induced DCFH oxidation increase, we evaluate the involvement of glutamate receptor antagonists in oxidative stress, showing that CNQX, but not MK-801, decreased the DCFH oxidation increase in striatal synaptosomes. Furthermore, the results presented in this report suggest that excitotoxicity elicited by low concentration of GA, could be in part by maintaining this excitatory neurotransmitter in the synaptic cleft by non-competitive inhibition of glutamate uptake. Thus the present data may explain, at least partly, initial striatal damage at birth, as evidenced by acute bilateral destruction of caudate and putamen observed in children with GA-I.


Assuntos
Corpo Estriado/metabolismo , Corpo Estriado/fisiopatologia , Ácido Glutâmico/metabolismo , Glutaratos/metabolismo , Estresse Oxidativo/fisiologia , Terminações Pré-Sinápticas/metabolismo , Sistema X-AG de Transporte de Aminoácidos/efeitos dos fármacos , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Animais , Antioxidantes/farmacologia , Ligação Competitiva/efeitos dos fármacos , Ligação Competitiva/fisiologia , Cromanos/farmacologia , Corpo Estriado/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Glutaratos/toxicidade , Masculino , Deficiência Múltipla de Acil Coenzima A Desidrogenase/metabolismo , Deficiência Múltipla de Acil Coenzima A Desidrogenase/fisiopatologia , Estresse Oxidativo/efeitos dos fármacos , Terminações Pré-Sinápticas/efeitos dos fármacos , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Sinaptossomos
18.
Behav Brain Res ; 187(2): 411-6, 2008 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-18023886

RESUMO

Glutaric acidemia type I (GA I) is an autosomal recessive metabolic disorder caused by glutaryl-CoA dehydrogenase deficiency leading to predominant accumulation of glutaric acid (GA), and to a lesser extent of 3-hydroxyglutaric acid (3HG) in body fluids and tissues. The clinical manifestations of GA I are predominantly neurological. Although the pathophysiological mechanisms responsible for the brain damage of this disease are virtually unknown, they are thought to be due to the neurotoxic actions of GA and 3HG. Therefore, in the present work we investigated whether chronic exposure of GA (5 micromol g of body weight(-1), twice per day), the major metabolite accumulating in GA I, during early development (from the 5th to the 28th day of life) could alter the cognitive performance of adult rats in the Morris water maze, open field and elevated plus maze tasks. Control rats were treated with saline in the same volumes. GA administration provoked an impairment of spatial performance in the water maze since adult rats pretreated with GA were not able to remember the previous location of the platform spending significantly less time in the training quadrant. In contrast, GA chronic administration did not affect rat performance in the open field and elevated plus maze tasks, indicating that motor activity and anxiety was not changed by GA. The results provide evidence that early chronic GA treatment induces long-lasting spatial behavioral deficit.


Assuntos
Encéfalo/efeitos dos fármacos , Transtornos Cognitivos/induzido quimicamente , Glutaratos/toxicidade , Aprendizagem em Labirinto/efeitos dos fármacos , Deficiência Múltipla de Acil Coenzima A Desidrogenase/fisiopatologia , Fatores Etários , Análise de Variância , Animais , Animais Recém-Nascidos , Esquema de Medicação , Masculino , Neurotoxinas/toxicidade , Ratos , Ratos Wistar , Estatísticas não Paramétricas
19.
Rev. Med. Univ. Navarra ; 51(3): 9-12, jul.-sept. 2007. ilus
Artigo em Es | IBECS | ID: ibc-057580

RESUMO

La acidosis glutárica tipo I (AGTI) es un trastorno poco frecuente del metabolismo de los ácidos orgánicos consecuencia de un défi cit congénito de la enzima mitocondrial glutaril-CoA deshidrogenasa. Existe una alteración del metabolismo de los aminoácidos triptófano, lisina e hidroxilisina y se produce un aumento de los niveles de ácido glutárico que clínicamente se manifi esta con crisis distónicas agudas en niños de corta edad. La acumulación de ácido glutárico produce neurotoxicidad en los ganglios de la base y corteza fronto-temporal lo que puede causar distonía progresiva, hipotonía, disartria y crisis epilépticas. El diagnóstico precoz de esta enfermedad es crucial ya que la dieta y el tratamiento pueden alterar su historia natural. Presentamos los hallazgos neuroradiológicos de una adolescente de 16 años con AGTI que presentaba un síndrome distónico crónico. La RM demostró afectación del putamen de forma bilateral y de la sustancia blanca periventricular, además de atrofi a temporal en ambos hemisferios y ensanchamiento de ambas cisuras de Silvio


Glutaric aciduria type I is a rare disorder of organic acid metabolism caused by defi ciency of glutaryl-CoA dehydrogenase, a mitochondrial enzyme. Improper degeneration of amino acids: tryptophan, lysine, and hydroxylysine, results in increased levels of glutaric acid, which typically becomes clinically manifest as an acute dystonic crisis in young children. Accumulation of glutaric acid causes neurotoxicity in the basal ganglia and fronto-temporal cortex which can lead to progressive dystonia, hypotonia, permanently impaired speech and seizures. Because dietary and drug therapy may alter the natural history of the disease, early diagnosis of such patients is critical. We report the magnetic resonance (MR) imaging fi ndings in a 16 year-old girl with this disorder who presented with a chronic dystonic syndrome and previously diagnosed of brain paralysis. MR imaging demonstrated bilateral involvement of the putamina and periventricular white matter, and bilateral temporal atrophy and widened Silvian fi ssures


Assuntos
Feminino , Adolescente , Humanos , Glutaratos/toxicidade , Distonia/etiologia , Triptofano/metabolismo , Lisina/metabolismo , Hidroxilisina/metabolismo
20.
Int J Dev Neurosci ; 25(6): 391-8, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17643899

RESUMO

Glutaric acidemia type I is an inherited metabolic disorder caused by a severe deficiency of the mitochondrial glutaryl-CoA dehydrogenase activity leading to accumulation of predominantly glutaric and 3-hydroxyglutaric acids in the brain tissue of the affected patients. Considering that a toxic role was recently postulated for quinolinic acid in the neuropathology of glutaric acidemia type I, in the present work we investigated whether the combination of quinolinic acid with glutaric or 3-hydroxyglutaric acids or the mixture of glutaric plus 3-hydroxyglutaric acids could alter brain energy metabolism. The parameters evaluated in cerebral cortex from young rats were glucose utilization, lactate formation and (14)CO(2) production from labeled glucose and acetate, as well as the activities of pyruvate dehydrogenase and creatine kinase. We first observed that glutaric (5 mM), 3-hydroxyglutaric (1 mM) and quinolinic acids (0.1 microM) per se did not alter these parameters. Similarly, no change of these parameters occurred when combining glutaric with quinolinic acids or 3-hydroxyglutaric with quinolinic acids. In contrast, co-incubation of glutaric plus 3-hydroxyglutaric acids increased glucose utilization, decreased (14)CO(2) generation from glucose, inhibited pyruvate dehydrogenase activity as well as total and mitochondrial creatine kinase activities. The glutaric plus 3-hydroxyglutaric acids-induced inhibitory effects on creatine kinase were prevented by the antioxidants glutathione and catalase plus superoxide dismutase, indicating the participation of reactive oxygen species. Our data indicate a synergic action of glutaric and 3-hydroxyglutaric acids disturbing energy metabolism in cerebral cortex of young rats.


Assuntos
Química Encefálica/fisiologia , Encefalopatias Metabólicas/metabolismo , Encéfalo/metabolismo , Metabolismo Energético/fisiologia , Glutaratos/metabolismo , Animais , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Química Encefálica/efeitos dos fármacos , Encefalopatias Metabólicas/fisiopatologia , Creatina Quinase/metabolismo , Sinergismo Farmacológico , Metabolismo Energético/efeitos dos fármacos , Glucose/metabolismo , Glutaratos/toxicidade , Ácido Láctico/metabolismo , Deficiência Múltipla de Acil Coenzima A Desidrogenase/metabolismo , Deficiência Múltipla de Acil Coenzima A Desidrogenase/fisiopatologia , Técnicas de Cultura de Órgãos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Complexo Piruvato Desidrogenase/metabolismo , Ácido Quinolínico/metabolismo , Ácido Quinolínico/toxicidade , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...