Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 308
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Neurochem Int ; 129: 104493, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31220473

RESUMO

Glutamatergic excitotoxicity is crucial in the pathogenesis of epileptic seizures. Dexmedetomidine, a potent and highly selective α2 adrenoceptor agonist, inhibits glutamate release from nerve terminals in rat cerebrocortical nerve terminals. However, the ability of dexmedetomidine to affect glutamate-induced brain injury is still unknown. Therefore, the present study evaluated the protective effect of dexmedetomidine against brain damage by using a kainic acid (KA) rat model, a frequently used model for temporal lobe epilepsy. Rats were treated with dexmedetomidine (1 or 5 µg/kg, intraperitoneally) 30 min before the KA (15 mg/kg) intraperitoneal injection. KA-induced seizure score and elevations of glutamate release in rat hippocampi were inhibited by pretreatment with dexmedetomidine. Histopathological and TUNEL staining analyzes showed that dexmedetomidine attenuated KA-induced neuronal death in the hippocampus. Dexmedetomidine ameliorated KA-induced apoptosis, and this neuroprotective effect was accompanied by inhibited the KA-induced caspase-3 expression as well as MAPKs phosphorylation, and reversed Bcl-2 down-expression, coupled with increased Nrf2, BDNF and TrkB expression in KA-treated rats. The results suggest that dexmedetomidine protected rat brains from KA-induced excitotoxic damage by reducing glutamate levels, suppressing caspase-3 activation and MAPKs phosphorylation, and enhancing Bcl-2, Nrf2, BDNF and TrkB expression in the hippocampus. Therefore, dexmedetomidine may be beneficial for preventing or treating brain disorders associated with excitotoxic neuronal damage. In conclusion, these data suggest that dexmedetomidine has the therapeutic potential for treating epilepsy.


Assuntos
Anticonvulsivantes/uso terapêutico , Fator Neurotrófico Derivado do Encéfalo/agonistas , Dexmedetomidina/uso terapêutico , Ácido Glutâmico/metabolismo , Hipocampo/efeitos dos fármacos , Ácido Caínico/antagonistas & inibidores , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Convulsões/prevenção & controle , Animais , Anticonvulsivantes/farmacologia , Apoptose/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Fator Neurotrófico Derivado do Encéfalo/genética , Dexmedetomidina/farmacologia , Avaliação Pré-Clínica de Medicamentos , Hipocampo/fisiopatologia , Ácido Caínico/toxicidade , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Fator 2 Relacionado a NF-E2/biossíntese , Fator 2 Relacionado a NF-E2/genética , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Neurônios/fisiologia , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor trkB/biossíntese , Receptor trkB/genética , Convulsões/induzido quimicamente
2.
Planta Med ; 81(18): 1697-704, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26366753

RESUMO

Epilepsy is a complex neurological disorder characterized by the repeated occurrence of electrical activity known as seizures. This activity induces increased intracellular calcium, which ultimately leads to neuronal damage. Houttuyniae Herba, the aerial part of Houttuynia cordata, has various pharmacological effects and is widely used as a traditional herb. In the present study, we evaluated the protective effects of Houttuyniae Herba water extract on kainic acid-induced neurotoxicity. Kainic acid directly acts on calcium release, resulting in seizure behavior, neuronal damage, and cognitive impairment. In a rat primary hippocampal culture system, Houttuyniae Herba water extract significantly protected neuronal cells from kainic acid toxicity. In a seizure model where mice received intracerebellar kainic acid injections, Houttuyniae Herba water extract treatment resulted in a lower seizure stage score, ameliorated cognitive impairment, protected neuronal cells against kainic acid-induced toxicity, and suppressed neuronal degeneration in the hippocampus. In addition, Houttuyniae Herba water extract regulated increases in the intracellular calcium level, its related downstream pathways (reactive oxygen species production and mitochondrial dysfunction), and calcium/calmodulin complex kinase type II immunoreactivity in the mouse hippocampus, which resulted from calcium influx stimulation induced by kainic acid. These results demonstrate the neuroprotective effects of Houttuyniae Herba water extract through inhibition of calcium generation in a kainic acid-induced epileptic model.


Assuntos
Cálcio/metabolismo , Hipocampo/efeitos dos fármacos , Houttuynia/química , Fármacos Neuroprotetores/farmacologia , Convulsões/prevenção & controle , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Hipocampo/metabolismo , Ácido Caínico/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos ICR , Neurônios/efeitos dos fármacos , Neurotoxinas/antagonistas & inibidores , Ratos , Convulsões/induzido quimicamente
3.
Neurochem Res ; 40(1): 118-23, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25417010

RESUMO

Trans-caryophyllene (TC), a component of essential oil found in many flowering plants, has shown its neuroprotective effects in various neurological disorders. However, the effects of TC on epilepsy haven't been reported before. In this study, we investigated the effect of TC on kainic acid-induced seizure activity caused by oxidative stress and pro-inflammation. We found that TC pretreatment significantly decreased seizure activity score compared to kainic acid treated group. Importantly, TC pretreatment leads to lowering the mortality in kainic acid treated mice. In addition, TC was found to significantly inhibit KA-induced generation of malondialdehyde. TC pretreatment also preserved the activity of GPx, SOD, and CAT. Notably, our data shows that an important property of TC is its capacity to exert cerebral anti-inflammatory effects by mitigating the expression of proinflammatory cytokines, such as TNF-α and IL-1ß. These data suggest that TC has a potential protective effect on chemical induced seizure and brain damage.


Assuntos
Agonistas de Aminoácidos Excitatórios/toxicidade , Ácido Caínico/antagonistas & inibidores , Ácido Caínico/toxicidade , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Convulsões/prevenção & controle , Sesquiterpenos/farmacologia , Animais , Antioxidantes/metabolismo , Química Encefálica/efeitos dos fármacos , Citocinas/biossíntese , Peroxidação de Lipídeos/efeitos dos fármacos , Camundongos , Sesquiterpenos Policíclicos , Convulsões/induzido quimicamente
4.
Epilepsy Res ; 108(1): 11-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24246145

RESUMO

We investigated how nitric oxide (NO) synthase inhibitor modulates muscarinic receptor expression in epileptic rats. We found that subchronic treatment (4 days) with Nω-nitro-l-arginine reduced the down-regulation of muscarinic receptors induced by pilocarpine and kainic acid in rat fronto-parietal cortex, notwithstanding the dramatic potentiation of seizures induced by both convulsants. Furthermore, functional experiments in fronto-parietal cortex slices, showed that Nω-nitro-l-arginine reduces the down-regulating effect of pilocarpine on carbachol-induced phosphoinositol hydrolysis. Finally, Nω-nitro-l-arginine greatly potentiated the induction of basic fibroblast growth factor (FGF2) by pilocarpine. These data suggest a potential role of NO in a regulatory feedback loop to control muscarinic receptor signal during seizures. The dramatic potentiation of convulsions by NO synthase inhibitors in some animal models of seizures could derive from preventing this feedback loop.


Assuntos
Ácido Caínico/toxicidade , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Pilocarpina/toxicidade , Receptores Muscarínicos/metabolismo , Convulsões/enzimologia , Animais , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Inibidores Enzimáticos/farmacologia , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/enzimologia , Ácido Caínico/antagonistas & inibidores , Masculino , Óxido Nítrico Sintase/metabolismo , Técnicas de Cultura de Órgãos , Lobo Parietal/efeitos dos fármacos , Lobo Parietal/enzimologia , Pilocarpina/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente
5.
Toxicol Appl Pharmacol ; 271(2): 127-36, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23707761

RESUMO

Our previous findings suggest that mitochondrial dysfunction is the mechanism underlying cognitive deficits induced by domoic acid (DA). Ursolic acid (UA), a natural triterpenoid compound, possesses many important biological functions. Evidence shows that UA can activate PI3K/Akt signaling and suppress Forkhead box protein O1 (FoxO1) activity. FoxO1 is an important regulator of mitochondrial function. Here we investigate whether FoxO1 is involved in the oxidative stress-induced mitochondrial dysfunction in DA-treated mice and whether UA inhibits DA-induced mitochondrial dysfunction and cognitive deficits through regulating the PI3K/Akt and FoxO1 signaling pathways. Our results showed that FoxO1 knockdown reversed the mitochondrial abnormalities and cognitive deficits induced by DA in mice through decreasing HO-1 expression. Mechanistically, FoxO1 activation was associated with oxidative stress-induced JNK activation and decrease of Akt phosphorylation. Moreover, UA attenuated the mitochondrial dysfunction and cognitive deficits through promoting Akt phosphorylation and FoxO1 nuclear exclusion in the hippocampus of DA-treated mice. LY294002, an inhibitor of PI3K/Akt signaling, significantly decreased Akt phosphorylation in the hippocampus of DA/UA mice, which weakened UA actions. These results suggest that UA could be recommended as a possible candidate for the prevention and therapy of cognitive deficits in excitotoxic brain disorders.


Assuntos
Transtornos Cognitivos/induzido quimicamente , Transtornos Cognitivos/prevenção & controle , Inibidores de Ciclo-Oxigenase/uso terapêutico , Ácido Caínico/análogos & derivados , Fármacos Neuromusculares Despolarizantes/efeitos adversos , Fármacos Neuromusculares Despolarizantes/toxicidade , Triterpenos/uso terapêutico , Animais , Animais Geneticamente Modificados , Transtornos Cognitivos/psicologia , Dependovirus , Imunofluorescência , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/metabolismo , Vetores Genéticos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Ácido Caínico/antagonistas & inibidores , Ácido Caínico/toxicidade , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , Doenças Mitocondriais/induzido quimicamente , Doenças Mitocondriais/prevenção & controle , Proteína Oncogênica v-akt/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Desempenho Psicomotor/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
6.
Brain Res ; 1486: 27-38, 2012 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-22985671

RESUMO

Glia cell line-derived neurotrophic factor (GDNF) is a potent survival factor for several neuron types. In this study, we have evaluated the utility of adenovirus-based vectors (Ad) and hippocampal neural stem cells (NSCs) as genetic tools for the delivery of a therapeutic protein, GDNF, in hippocampus tissues damaged by kainic acid (KA)-induced excitotoxicity. The experimental animals were treated with KA 3 days prior to exposure to Ad-GDNF, NSCs, and NSCs infected with Ad-GDNF (Ad-GDNF-NSCs). Seven days after the treatments with Ad-GDNF, NSCs and Ad-GDNF-NSCs, the effects of the treatments were evaluated. GAD-67 labeled cells originating from the transplanted NSCs were observed at increased levels in the Ad-GDNF-NSCs-treated rats as compared to the NSCs-only rats. In situ apoptosis assays showed that the levels of TUNEL-positive cells were slightly, but not significantly, reduced in the Ad-GDNF and Ad-GDNF-NSCs groups, as compared to the saline and NSCs only groups. GDNF expression by NSCs and Ad-GDNF was upregulated as the consequence of adenoviral gene delivery in the NSCs and Ad-GDNF-treated rats, and the transplanted NSCs were shown to have migrated to the hippocampal regions in Ad-GDNF-NSCs rats to a greater degree than in the NSCs-only rats. Furthermore, in the region in which the NSCs were detected, GDNF and GAD-67 expression were increased. These results indicate that the migration and differentiation of NSCs may be associated with the expression of GDNF. However, cell death consequent to KA administration was not prevented by upregulated GDNF and NSCs transplantation. Collectively, our results indicate that GDNF may exert effects on the migration and differentiation of NSCs, but there are no protective properties with regard to excitotoxically damaged hippocampal tissue.


Assuntos
Movimento Celular/fisiologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/biossíntese , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Ácido Caínico/toxicidade , Células-Tronco Neurais/metabolismo , Fármacos Neuroprotetores/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Ácido Caínico/antagonistas & inibidores , Masculino , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/patologia , Ratos , Ratos Sprague-Dawley , Transplante de Células-Tronco/métodos , Regulação para Cima/fisiologia
7.
Brain Res Bull ; 88(5): 467-70, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22542883

RESUMO

Docosahexaeonic acid (DHA) is the final compound in the omega-3 polyunsaturated fatty acids (PUFA) synthetic pathway and the most abundant PUFA found in the brain. DHA plays an essential role in the development of the brain, and the intakes in pregnancy and early life affect growth and cognitive performance later in childhood. Recently, it has been proposed that dietary intake of DHA could be a non-pharmacological interventional strategy for the treatment of seizures in humans. However, to date, the experimental approaches to study the antiepileptic effect of DHA have been exclusively restricted to rodent models during short-to-medium periods of treatment. The purpose of the present study was to test the chronic anticonvulsivant effects of DHA supplementation in zebrafish from the pre-spawning stage to aging, taking advantage of our recently described kainate-induced seizure model using this animal. To that end, two groups of adult female zebrafish were fed with standard or 200mg/kg DHA-enriched diets during 1 month previous to the spawning, and offspring subdivided in two categories, and subsequently fed with standard or DHA diets, generating 4 groups of animals that were aged until 20 months. Afterward, KA was intraperitoneally administered and epileptic score determined. All the DHA-enriched groups presented antiepileptic effects compared to the control group, showing that DHA presents an anticonvulsant potential. Among the studied groups, zebrafish fed with DHA from the pre-spawning stage to aging presented the best antiepileptic profile. These results show a neuroprotective benefit in zebrafish fed with DHA-enriched diet before birth and during the whole life.


Assuntos
Envelhecimento/efeitos dos fármacos , Envelhecimento/fisiologia , Ácidos Docosa-Hexaenoicos/administração & dosagem , Ácido Caínico/toxicidade , Convulsões/prevenção & controle , Animais , Ácidos Docosa-Hexaenoicos/fisiologia , Embrião não Mamífero , Feminino , Ácido Caínico/antagonistas & inibidores , Masculino , Convulsões/induzido quimicamente , Convulsões/embriologia , Peixe-Zebra
8.
Neurotox Res ; 21(4): 379-92, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22144346

RESUMO

Abundant evidence suggests that indirect inhibitory modulation of glutamatergic transmission, via metabotropic glutamatergic receptors (mGluR), may induce neuroprotection. The present study was designed to determine whether the selective antagonist of mGluR1 (3-ethyl-2-methyl-quinolin-6-yl)-(4-methoxy-cyclohexyl)-methanone methanesulfonate (EMQMCM), showed neuroprotection against the kainate (KA)-induced excitotoxicity in vitro and in vivo. In in vitro studies on mouse primary cortical and hippocampal neuronal cultures, incubation with KA (150 µM) induced strong degeneration [measured as lactate dehydrogenase (LDH) efflux] and apoptosis (measured as caspase-3 activity). EMQMCM (0.1-100 µM) added 30 min to 6 h after KA, significantly attenuated the KA-induced LDH release and prevented the increase in caspase-3 activity in the cultures. Those effects were dose- and time-dependent. In in vivo studies KA (2.5 nmol/1 µl) was unilaterally injected into the rat dorsal CA1 hippocampal region. Degeneration was calculated by counting surviving neurons in the CA pyramidal layer using stereological methods. It was found that EMQMCM (5-10 nmol/1 µl) injected into the dorsal hippocampus 30 min, 1 h, or 3 h (the higher dose only) after KA significantly prevented the KA-induced neuronal degeneration. In vivo microdialysis studies in rat hippocampus showed that EMQMCM (100 µM) significantly increased γ-aminobutyric acid (GABA) and decreased glutamate release. When perfused simultaneously with KA, EMQMCM substantially increased GABA release and prevented the KA-induced glutamate release. The obtained results indicate that the mGluR1 antagonist, EMQMCM, may exert neuroprotection against excitotoxicity after delayed treatment (30 min to 6 h). The role of enhanced GABAergic transmission in the neuroprotection is postulated.


Assuntos
Apoptose/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Ácido Caínico/antagonistas & inibidores , Fármacos Neuroprotetores/farmacologia , Quinolinas/farmacologia , Animais , Caspase 3/metabolismo , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Ácido Glutâmico/metabolismo , Hipocampo/patologia , Ácido Caínico/administração & dosagem , Ácido Caínico/toxicidade , L-Lactato Desidrogenase/metabolismo , Masculino , Camundongos , Microinjeções , Degeneração Neural/induzido quimicamente , Degeneração Neural/patologia , Cultura Primária de Células , Quinolinas/administração & dosagem , Ratos , Ratos Wistar , Ácido gama-Aminobutírico/metabolismo
9.
J Biomed Sci ; 18: 75, 2011 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-22014163

RESUMO

BACKGROUND: Pu-Erh tea is one of the most-consumed beverages due to its taste and the anti-anxiety-producing effect of the gamma-aminobutyric acid (GABA) if contains. However the protective effects of Pu-Erh tea and its constituent, GABA to kainic acid (KA)-induced seizure have not been fully investigated. METHODS: We analyzed the effect of Pu-Erh tea leaf (PETL) and GABA on KA-induced neuronal injury in vivo and in vitro. RESULTS: PETL and GABA reduced the maximal seizure classes, predominant behavioral seizure patterns, and lipid peroxidation in male FVB mice with status epilepticus. PETL extracts and GABA were effective in protecting KA-treated PC12 cells in a dose-dependent manner and they decreased Ca(2+) release, ROS production and lipid peroxidation from KA-stressed PC12 cells. Western blot results revealed that mitogen-activated protein kinases (MAPKs), RhoA and cyclo-oxygenase-2 (COX-2) expression were increased in PC12 cells under KA stress, and PETL and GABA significantly reduced COX-2 and p38 MAPK expression, but not that of RhoA. Furthermore, PETL and GABA reduced PGE(2) production from KA-induced PC12 cells. CONCLUSIONS: Taken together, PETL and GABA have neuroprotective effects against excitotoxins that may have clinical applications in epilepsy.


Assuntos
Camellia sinensis/química , Medicamentos de Ervas Chinesas/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Estresse Oxidativo/efeitos dos fármacos , Estado Epiléptico/tratamento farmacológico , Ácido gama-Aminobutírico/administração & dosagem , Animais , Cálcio/metabolismo , Caspase 3/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Ácido Caínico/antagonistas & inibidores , Ácido Caínico/farmacologia , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Células PC12 , Folhas de Planta/química , Ratos , Espécies Reativas de Oxigênio/metabolismo , Estado Epiléptico/induzido quimicamente
10.
Eur J Pharmacol ; 666(1-3): 61-71, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21596030

RESUMO

Systemic or intracerebral administration of kainic acid in rodents induces neuronal death followed by a cascade of neuroplastic changes in the hippocampus. Kainic acid-induced neuroplasticity is evidenced by alterations in hippocampal neurogenesis, dispersion of the granule cell layer and re-organisation of mossy fibres. Similar abnormalities are observed in patients with temporal lobe epilepsy and, therefore, kainic acid-induced hippocampal neuroplasticity might mimic pathological mechanisms leading to the formation of 'epileptic brain' in patients with temporal lobe epilepsy. Previous studies have demonstrated that selective serotonin re-uptake inhibitor antidepressants might reduce the severity of seizures in epileptic patients and reduce neuronal death in laboratory animal models of kainic acid-induced neurotoxicity. In the present study, we investigated whether kainic acid-induced neuroplasticity in mice is modulated by the repeated administration of citalopram, a selective serotonin reuptake inhibitor. We found that at the histopathological level, repeated citalopram treatment counteracted the kainic acid-induced neuronal loss and dispersion of young granule neurons expressing the polysialylated neural cell adhesion molecule within the granule cell layer of the hippocampus. Citalopram also counteracted the downregulation of reelin on both mRNA and protein levels induced by kainic acid administration. Our findings indicate that repeated administration of citalopram is able to prevent kainic acid-induced abnormal brain plasticity and thereby prevent the formation of an epileptic phenotype.


Assuntos
Moléculas de Adesão Celular Neuronais/deficiência , Citalopram/administração & dosagem , Citalopram/farmacologia , Proteínas da Matriz Extracelular/deficiência , Hipocampo/efeitos dos fármacos , Ácido Caínico/antagonistas & inibidores , Ácido Caínico/farmacologia , Proteínas do Tecido Nervoso/deficiência , Molécula L1 de Adesão de Célula Nervosa/imunologia , Serina Endopeptidases/deficiência , Ácidos Siálicos/imunologia , Animais , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/imunologia , Moléculas de Adesão Celular Neuronais/metabolismo , Contagem de Células , Citalopram/uso terapêutico , Regulação para Baixo/efeitos dos fármacos , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/imunologia , Proteínas da Matriz Extracelular/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Hipocampo/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fibras Musgosas Hipocampais/efeitos dos fármacos , Fibras Musgosas Hipocampais/metabolismo , Fibras Musgosas Hipocampais/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/imunologia , Proteínas do Tecido Nervoso/metabolismo , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Plasticidade Neuronal/efeitos dos fármacos , Neurotoxinas/antagonistas & inibidores , Neurotoxinas/farmacologia , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína Reelina , Convulsões/induzido quimicamente , Convulsões/metabolismo , Convulsões/patologia , Convulsões/fisiopatologia , Serina Endopeptidases/genética , Serina Endopeptidases/imunologia , Serina Endopeptidases/metabolismo , Ácidos Siálicos/metabolismo
11.
Neuroscience ; 175: 273-80, 2011 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-21093549

RESUMO

The pancreatitis-associated protein (PAP) family (also known as the regenerating gene (Reg) family) is a group of 16 kDa secretory proteins structurally classified as the calcium dependent-type lectin superfamily. Some PAP family members are expressed in neurons following peripheral nerve injury and traumatic brain injury. To determine whether PAP family members are expressed in non-traumatic brain injury, expressions were analyzed following kainic acid (KA)-induced seizure. PAP-I (also known as Reg2 in rat and RegIII-beta in mouse) and pancreatitis associated protein-III (PAP-III; RegIII-gamma in mouse) messenger ribonucleic acid (mRNA) was transiently expressed in some restricted areas, such as the hippocampus and parahippocampal area; expression was observed immediately at a maximal level 1 day after seizure. Expression disappeared within 3 days after seizure. In situ hybridization (ISH) and immunohistochemistry revealed neuronal PAP-I and PAP-III expression in the hippocampal dentate gyrus, perirhinal and entorhinal cortices, and the posterior cortical nucleus of the amygdala. The number of PAP-III mRNA-positive neurons was significantly greater than PAP-I mRNA-positive neurons. The majority of positive neurons co-localized with c-Jun, but not with glutamic acid decarboxylase (GAD). These results may suggest that PAP-I and PAP-III induction in non-GABAergic neurons would protect neurons against damage following seizure.


Assuntos
Aminopeptidases/biossíntese , Antígenos de Neoplasias/biossíntese , Biomarcadores Tumorais/biossíntese , Epilepsia do Lobo Temporal/metabolismo , Lectinas Tipo C/biossíntese , Convulsões/metabolismo , Lobo Temporal/metabolismo , Aminopeptidases/genética , Aminopeptidases/fisiologia , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/fisiologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/fisiologia , Modelos Animais de Doenças , Epilepsia do Lobo Temporal/prevenção & controle , Ácido Caínico/antagonistas & inibidores , Ácido Caínico/toxicidade , Lectinas Tipo C/genética , Lectinas Tipo C/fisiologia , Masculino , Neurotoxinas/antagonistas & inibidores , Neurotoxinas/toxicidade , Proteínas Associadas a Pancreatite , Ratos , Ratos Wistar , Convulsões/induzido quimicamente , Convulsões/prevenção & controle , Lobo Temporal/fisiopatologia , Ácido gama-Aminobutírico/fisiologia
12.
Neurochem Res ; 35(12): 2175-83, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21127971

RESUMO

Injection of rats with kainic acid (KA), a non-N-methyl-D-aspartate (NMDA) type glutamate receptor agonist, induces recurrent (delayed) convulsive seizures and subsequently hippocampal neurodegeneration, which is reminiscent of human epilepsy. The protective effect of anti-epileptic drugs on seizure-induced neuronal injury is well known; however, molecular basis of this protective effect has not yet been elucidated. In this study, we investigated the effect and signaling mediators of voltage-gated Na(+) channel blockers (Lamotrigine, Rufinamide, Oxcarbazepine, Valproic Acid, and Zonisamide) on KA-induced apoptosis in rat primary hippocampal neurons. Exposure of hippocampal neurons to 10 µM KA for 24 h caused significant increases in morphological and biochemical features of apoptosis, as determined by Wright staining and ApopTag assay, respectively. Analyses showed increases in expression and activity of cysteine proteases, production of reactive oxygen species (ROS), intracellular free [Ca(2+)], and Bax:Bcl-2 ratio during apoptosis. Cells exposed to KA for 15 min were then treated with Lamotrigine, Rufinamide, Oxcarbazepine, Valproic Acid, or Zonisamide. Post-treatment with one of these anti-epileptic drugs (500 nM) attenuated production of ROS and prevented apoptosis in hippocampal neurons. Lamotrigine, Rufinamide, and Oxcarbazepine appeared to be less protective when compared with Valproic Acid or Zonisamide. This difference may be due to blockade of T-type Ca(2+) channels also by Valproic Acid and Zonisamide. Our findings thus suggest that the anti-epileptic drugs that block both Na(+) channels and Ca(2+) channels are significantly more effective than agents that block only Na(+) channels for attenuating seizure-induced hippocampal neurodegeneration.


Assuntos
Apoptose/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Ativação do Canal Iônico , Ácido Caínico/antagonistas & inibidores , Neurônios/efeitos dos fármacos , Bloqueadores dos Canais de Sódio/farmacologia , Animais , Western Blotting , Calpaína/metabolismo , Células Cultivadas , Colorimetria , Ativação Enzimática , Hipocampo/citologia , Hipocampo/metabolismo , Ácido Caínico/farmacologia , Neurônios/metabolismo , Ratos , Espécies Reativas de Oxigênio , Proteína X Associada a bcl-2/metabolismo
13.
Brain Res ; 1360: 8-16, 2010 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-20833150

RESUMO

This study has assessed the neuroprotective efficacy of five AP-1 inhibitory peptides in an in vitro excitotoxicity model. The five AP-1 inhibitory peptides and controls of the JNK inhibitor peptide (JNKI-1D-TAT) and TAT cell-penetrating-peptide were administered to primary cortical neuronal cultures prior to kainic acid exposure. All five AP-1 inhibitory peptides and JNKI-1D-TAT provided significant neuroprotection from kainic acid induced neuronal cell death. Kainic acid exposure induced caspase and calpain activation in neuronal cultures, with caspase-induced cleavage of α-fodrin reduced by administration of the AP-1 inhibitory peptides. Sequence analysis of the AP-1 inhibitory peptides did not reveal the presence of any secondary structures; however two peptides shared 66% amino-acid sequence homology. As a result, truncated sequences were designed and synthesised to identify the active region of the peptides. All truncated peptides were significantly neuroprotective following kainic acid and glutamate exposure. We have shown for the first time the neuroprotective efficacy of full-length and truncated AP-1 inhibitory peptides in kainic acid and glutamate neuronal excitotoxicity models. The identification of therapeutic targets, such as the AP-1 complex, is an important step for the development of pharmaceuticals to reduce neuronal loss in disorders with a prevalence of excitotoxic cell death such as epilepsy, cerebral ischaemia, and traumatic brain injury.


Assuntos
Córtex Cerebral/citologia , Agonistas de Aminoácidos Excitatórios/toxicidade , Ácido Caínico/antagonistas & inibidores , Neurônios/efeitos dos fármacos , Peptídeos/farmacologia , Fator de Transcrição AP-1/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Proteínas de Transporte/metabolismo , Morte Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Produtos do Gene tat/química , Produtos do Gene tat/farmacologia , Ácido Glutâmico/toxicidade , Ácido Caínico/toxicidade , Proteínas dos Microfilamentos/metabolismo , Dados de Sequência Molecular , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley
14.
Peptides ; 31(11): 2094-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20713105

RESUMO

Stanniocalcin 2 (STC2), the paralog of STC1, has been shown to act as a novel target of the mammalian unfolded protein response. We investigated the potential neuroprotective actions of STC2 against kainic acid toxicity in the hippocampus of ICR mice. STC2-treated mice experienced less neuronal cell loss in the CA3 area of the hippocampus. Also, microglial activation and heme oxygenase 1 expression were attenuated in the hippocampus of STC2-treated mice. To confirm whether STC2 regulates microglial activation directly, nitric oxide levels were measured in BV2 cells cultured with and without 10nM STC2. STC2 decreased the level of nitric oxide induced by lipopolysaccharide (LPS) treatment significantly. Also, STC2 pretreatment significantly decreased TNF-α and IL-1ß expression induced by LPS treatment. These observations demonstrated that STC2 exerts neuroprotective actions against excitotoxic insults through the inhibition of microglial activation.


Assuntos
Glicoproteínas/farmacologia , Ácido Caínico/antagonistas & inibidores , Fármacos Neuroprotetores/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Heme Oxigenase-1/biossíntese , Hipocampo/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Interleucina-1beta/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular , Lipopolissacarídeos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Microglia/efeitos dos fármacos , Microglia/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/biossíntese , Fator de Necrose Tumoral alfa/biossíntese
15.
Brain Behav Immun ; 24(8): 1354-61, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20599496

RESUMO

Excitotoxicity is a mechanism of neuronal cell death implicated in a range of neurodegenerative conditions. Systemic administration of the excitotoxin kainic acid (KA) induces inflammation and apoptosis in the hippocampus, resulting in neuronal loss. Evidence indicates that stimulation of glial ß(2)-adrenoceptors has anti-inflammatory and neurotrophic properties that could result in neuroprotection. Consequently, in this study we examined the effect of the ß(2)-adrenoceptor agonist clenbuterol on KA-induced inflammation, neurotrophic factor expression and apoptosis in the hippocampus. Clenbuterol (0.5mg/kg) was administered to rats one hour prior to KA (10mg/kg). Epileptic behaviour induced by KA was assessed for three hours following administration using the Racine scale. Twenty-four hours later TUNEL staining in the CA3 hippocampal subfield and hippocampal caspase-3 activity was assessed to measure KA-induced apoptosis. In addition, expression of inflammatory cytokines (IL-1ß and IFN-γ), inducible nitric oxide synthase (iNOS), kynurenine pathway enzymes indolamine 2,3-dioxygenase (IDO) and kynurenine monooxygenase (KMO), the microglial activation marker CD11b, and the neurotrophins BDNF and NGF were quantified in the hippocampus using real-time PCR. Whilst clenbuterol treatment did not significantly alter KA-induced epileptic behavior it ameliorated KA-induced apoptosis, and this neuroprotective effect was accompanied by reduced inflammatory cytokine expression, reduced expression of iNOS, IDO, KMO and CD11b, coupled with increased BDNF and NGF expression in KA-treated rats. In conclusion, the ß(2)-adrenoceptor agonist clenbuterol has anti-inflammatory and neurotrophic actions and elicits a neuroprotective effect in the KA model of neurodegeneration.


Assuntos
Agonistas de Receptores Adrenérgicos beta 3/farmacologia , Anti-Inflamatórios , Clembuterol/farmacologia , Agonistas de Aminoácidos Excitatórios/toxicidade , Ácido Caínico/antagonistas & inibidores , Ácido Caínico/toxicidade , Fatores de Crescimento Neural/metabolismo , Fármacos Neuroprotetores , Síndromes Neurotóxicas/prevenção & controle , Neurotoxinas/toxicidade , Animais , Apoptose/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Caspase 3/metabolismo , Hipocampo/patologia , Marcação In Situ das Extremidades Cortadas , Masculino , Síndromes Neurotóxicas/patologia , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Convulsões/induzido quimicamente , Convulsões/prevenção & controle
16.
BMC Neurosci ; 11: 42, 2010 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-20346117

RESUMO

BACKGROUND: Midkine (MK), a member of the heparin-binding growth factor family, which includes MK and pleiotrophin, is known to possess neurotrophic and neuroprotective properties in the central nervous system. Previous studies have shown that MK is an effective neuroprotective agent in reducing retinal degeneration caused by excessive light and decreasing hippocampal neuronal death in ischemic gerbil brain. The present study was undertaken to investigate whether MK acts as an anticonvulsant in kainic acid (KA)-induced seizure in mouse and blocks KA-mediated neuronal cell death in hippocampus. RESULTS: Increased expression of MK was found in hippocampus of mouse following seizures induced by intracerebroventricular injection of KA, and MK expression was found in glial fibrillary acidic protein (GFAP)-positive astrocytes. Concurrent injection of MK and KA attenuated KA-induced seizure activity and cell death of hippocampal neurons including pyramidal cells and glutamic acid decarboxylase 67 (GAD67)-positive GABAergic interneurons in the CA3 and hilar area. CONCLUSION: The results of the present study indicate that MK functions as an anticonvulsant and neuroprotective agent in hippocampus during KA-induced seizures.


Assuntos
Anticonvulsivantes/farmacologia , Citocinas/farmacologia , Epilepsia/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Degeneração Neural/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Animais , Anticonvulsivantes/uso terapêutico , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Biomarcadores/metabolismo , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Citocinas/uso terapêutico , Modelos Animais de Doenças , Epilepsia/induzido quimicamente , Epilepsia/fisiopatologia , Epilepsia do Lobo Temporal/induzido quimicamente , Epilepsia do Lobo Temporal/tratamento farmacológico , Epilepsia do Lobo Temporal/fisiopatologia , Proteína Glial Fibrilar Ácida/metabolismo , Glutamato Descarboxilase/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Injeções Intraventriculares , Interneurônios/efeitos dos fármacos , Interneurônios/metabolismo , Ácido Caínico/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Midkina , Degeneração Neural/fisiopatologia , Degeneração Neural/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Neurotoxinas/antagonistas & inibidores , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Ácido gama-Aminobutírico/metabolismo
17.
J Neurochem ; 113(3): 692-703, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20132465

RESUMO

The alpha-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate receptor (AMPAR) subunit GluR2, which regulates excitotoxicity and the inflammatory cytokine tumour necrosis factor alpha (TNFalpha) have both been implicated in motor neurone vulnerability in amyotrophic lateral sclerosis/motor neurone disease. TNFalpha has been reported to increase cell surface expression of AMPAR subunits to increase synaptic strength and enhance excitotoxicity, but whether this mechanism occurs in motor neurones is unknown. We used primary cultures of mouse motor neurones and cortical neurones to examine the interaction between TNFalpha receptor activation, GluR2 availability, AMPAR-mediated calcium entry and susceptibility to excitotoxicity. Short exposure to a physiologically relevant concentration of TNFalpha (10 ng/mL, 15 min) caused a marked redistribution of both GluR1 and GluR2 to the cell surface as determined by cell surface biotinylation and immunofluorescence. Using fura-2-acetoxymethyl ester microfluorimetry, we showed that exposure to TNFalpha caused a rapid reduction in the peak amplitude of AMPA-mediated calcium entry in a PI3-kinase and p38 kinase-dependent manner, consistent with increased insertion of GluR2-containing AMPAR into the plasma membrane. This resulted in a protection of motor neurones against kainate-induced cell death. Our data therefore, suggest that TNFalpha acts primarily as a physiological regulator of synaptic activity in motor neurones rather than a pathological drive in amyotrophic lateral sclerosis.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Neurônios Motores/efeitos dos fármacos , Degeneração Neural/patologia , Fármacos Neuroprotetores , Receptores de AMPA/antagonistas & inibidores , Receptores de Superfície Celular/biossíntese , Fator de Necrose Tumoral alfa/farmacologia , Animais , Biotinilação , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Agonistas de Aminoácidos Excitatórios/toxicidade , Feminino , Imunofluorescência , Ácido Caínico/antagonistas & inibidores , Ácido Caínico/toxicidade , Camundongos , Degeneração Neural/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Gravidez , Receptores de AMPA/biossíntese , Receptores de AMPA/genética , Receptores do Fator de Necrose Tumoral/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espectrometria de Fluorescência , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
18.
Brain Res ; 1307: 166-76, 2010 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-19840778

RESUMO

The pathophysiology of schizophrenia may involve reduced NMDA receptor function and experimental models of NMDA receptor hypofunction have proven useful for characterizing neurobiological abnormalities potentially relevant to schizophrenia. The present study assessed behavioral responses and induction of Fos after administration of kainic acid to wild type mice (NR1(+/+)) and mice with genetically reduced NMDA receptor expression (NR1(neo/neo)). At a dose of 20 mg/kg, kainic acid induced lethal seizures in 100% of the NR1(neo/neo) mice tested but produced no lethal seizures in the wild type mice. The NR1(neo/neo) mice also exhibited enhanced behavioral responses to kainic acid at a dose of 15 mg/kg but no lethal seizures were produced by this dose. A greater induction of Fos was observed in neocortical and limbic cortical regions of the NR1(neo/neo) compared to NR1(+/+) mice after administration of 15 mg/kg kainic acid. In contrast, there were no differences between the genotypes in kainic acid induced Fos in the amygdala, hippocampus, lateral septum, and nucleus accumbens. In order to determine if altered behavioral phenotypes of the NR1(neo/neo) mice could be related to increased sensitivity of kainate receptors to endogenous glutamate, effects of the highly selective kainate antagonist LY382884 were examined. The kainate antagonist reduced the exaggerated acoustic startle responses, deficits in prepulse inhibition of acoustic startle, and motor hyperactivity in the NR1(neo/neo) mice. These findings suggest that selective kainate receptor antagonists could be novel therapeutic candidates for schizophrenia.


Assuntos
Comportamento Animal/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/farmacologia , Ácido Caínico/farmacologia , Modelos Genéticos , Receptores de N-Metil-D-Aspartato/fisiologia , Estimulação Acústica/métodos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Modelos Animais de Doenças , Comportamento Exploratório/efeitos dos fármacos , Feminino , Ácido Glutâmico/metabolismo , Isoquinolinas/farmacologia , Ácido Caínico/antagonistas & inibidores , Masculino , Camundongos , Camundongos Transgênicos , Inibição Neural/efeitos dos fármacos , Inibição Neural/genética , Proteínas Oncogênicas v-fos/genética , Proteínas Oncogênicas v-fos/metabolismo , Receptores de N-Metil-D-Aspartato/deficiência , Reflexo de Sobressalto/efeitos dos fármacos , Reflexo de Sobressalto/genética , Convulsões/induzido quimicamente , Convulsões/genética
19.
J Neurosci Res ; 88(5): 1074-82, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19937811

RESUMO

Metallothioneins I and II (MTI/II) are metal-binding proteins overexpressed in response to brain injury. Recently, we have designed a peptide, termed EmtinB, which is modeled after the beta-domain of MT-II and mimics the biological effects of MTI/II in vitro. Here, we demonstrate the neuroprotective effect of EmtinB in the in vitro and in vivo models of kainic acid (KA)-induced neurotoxicity. We show that EmtinB passes the blood-brain barrier and is detectable in plasma for up to 24 hr. Treatment with EmtinB significantly attenuates seizures in C57BL/6J mice exposed to moderate (20 mg/kg) and high (30 mg/kg) KA doses and tends to decrease mortality induced by the high KA dose. Histopathological evaluation of hippocampal (CA3 and CA1) and cortical areas of mice treated with 20 mg/kg KA shows that EmtinB treatment reduces KA-induced neurodegeneration in the CA1 region. These findings establish EmtinB as a promising target for therapeutic development.


Assuntos
Ácido Caínico/antagonistas & inibidores , Metalotioneína/agonistas , Fármacos Neuroprotetores/farmacocinética , Neurotoxinas/antagonistas & inibidores , Peptídeos/farmacocinética , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Epilepsia do Lobo Temporal/tratamento farmacológico , Epilepsia do Lobo Temporal/metabolismo , Epilepsia do Lobo Temporal/fisiopatologia , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Hipocampo/fisiopatologia , Peptídeos e Proteínas de Sinalização Intercelular , Ácido Caínico/toxicidade , Masculino , Metalotioneína/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Degeneração Neural/induzido quimicamente , Degeneração Neural/tratamento farmacológico , Degeneração Neural/fisiopatologia , Fármacos Neuroprotetores/sangue , Fármacos Neuroprotetores/uso terapêutico , Neurotoxinas/toxicidade , Peptídeos/sangue , Peptídeos/uso terapêutico , Ratos , Ratos Wistar , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico , Convulsões/fisiopatologia
20.
Cell Mol Neurobiol ; 30(3): 347-60, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19777341

RESUMO

Insulin-like growth factor I (IGF-I) has been shown to act as a neuroprotectant both in in vitro studies and in in vivo animal models of ischemia, hypoxia, trauma in the brain or the spinal cord, multiple and amyotrophic lateral sclerosis, Alzheimer's and Parkinson's disease. In the present study, we investigated the neuroprotective potential of IGF-I in the "kainic acid-induced degeneration of the hippocampus" model of temporal lobe epilepsy. Increased cell death--as detected by FluoroJade B staining--and extensive cell loss--as determined by cresyl violet staining--were observed mainly in the CA3 and CA4 areas of the ipsilateral and contralateral hippocampus, 7 days following intrahippocampal administration of kainic acid. Kainic acid injection also resulted in intense astrogliosis--as assessed by the number of glial fibrillary acidic protein (GFAP) immunopositive cells--in both hemispheres, forming a clear astroglial scar ipsilaterally to the injection site. Heat-shock protein 70 (Hsp70) immunopositive cells were also observed in the ipsilateral dentate gyrus (DG) following kainic acid injection. When IGF-I was administered together with kainic acid, practically no signs of degeneration were detected in the contralateral hemisphere, while in the ipsilateral, there was a smaller degree of cell loss, reduced number of FluoroJade B-stained cells, decreased reactive gliosis and fewer Hsp70-positive cells. Our present results extend further the cases in which IGF-I is shown to exhibit neuroprotective properties in neurodegenerative processes in the CNS.


Assuntos
Epilepsia do Lobo Temporal/complicações , Hipocampo/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/farmacologia , Degeneração Neural/tratamento farmacológico , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Animais , Biomarcadores/análise , Biomarcadores/metabolismo , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Citoproteção/efeitos dos fármacos , Citoproteção/fisiologia , Giro Denteado/efeitos dos fármacos , Giro Denteado/patologia , Giro Denteado/fisiopatologia , Modelos Animais de Doenças , Epilepsia do Lobo Temporal/induzido quimicamente , Fluoresceínas , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/induzido quimicamente , Gliose/metabolismo , Gliose/fisiopatologia , Proteínas de Choque Térmico HSP72/efeitos dos fármacos , Proteínas de Choque Térmico HSP72/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Fator de Crescimento Insulin-Like I/uso terapêutico , Ácido Caínico/antagonistas & inibidores , Ácido Caínico/toxicidade , Masculino , Degeneração Neural/fisiopatologia , Degeneração Neural/prevenção & controle , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/uso terapêutico , Neurotoxinas/antagonistas & inibidores , Neurotoxinas/toxicidade , Compostos Orgânicos , Ratos , Ratos Wistar , Coloração e Rotulagem , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...