Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Neurosci Lett ; 724: 134880, 2020 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-32135163

RESUMO

Focal cortical dysplasia (FCD) is one of the main causes of medically intractable epilepsy. Some studies have reported that transient receptor potential canonical channel 3 (TRPC3) may play an important role in the occurrence of seizures. In this study, we investigated the expression patterns of TRPC3 in different types of FCD. Forty-five FCD specimens and 12 control samples from autopsies were used in our study. Western blotting, immunohistochemistry, and immunofluorescence staining were employed to detect protein expression and distribution. The amount of TRPC3 protein was markedly elevated in the FCD group. The immunohistochemistry results revealed that TRPC3 staining was strong in the malformed cells and microcolumns. Most of the TRPC3-positive cells were colabeled with glutamatergic and GABAergic markers. The overexpression and altered cellular distribution of TRPC3 in the FCD samples suggest that TRPC3 may be related to epileptogenesis in FCD.


Assuntos
Córtex Cerebral/metabolismo , Malformações do Desenvolvimento Cortical/genética , Malformações do Desenvolvimento Cortical/metabolismo , Canais de Cátion TRPC/biossíntese , Canais de Cátion TRPC/genética , Adolescente , Criança , Pré-Escolar , Feminino , Expressão Gênica , Humanos , Masculino , Malformações do Desenvolvimento Cortical/diagnóstico , Adulto Jovem
2.
J Neuropathol Exp Neurol ; 75(8): 718-730, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27288906

RESUMO

Focal cortical dysplasia (FCD) likely results from abnormal migration of neural progenitor cells originating from the subventricular zone. To elucidate the roles in molecules that are involved in neural migration pathway abnormalities in FCDs, we investigated the expression patterns of transient receptor potential canonical channel 6 (TRPC6) and brain-derived neurotrophic factor (BDNF) in cortical lesions from FCD patients and in samples of normal control cortex. TRPC6 and BDNF mRNA and protein levels were increased in FCD lesions. By immunohistochemistry, they were strongly expressed in microcolumns, heterotopic neurons, dysmorphic neurons, and balloon cells (BCs). Colocalization assays revealed that most of the misshapen TRPC6-positive or heterotopic cells had a neuronal lineage with the exception of TRPC6-positive FCDiib patient BCs, which had both neuronal and glial features. Most TRPC6-positive cells were glutamatergic neurons. There was also greater expression of calmodulin-dependent kinase IV (CaMKIV), the downstream factor of TRPC6, in FCD lesions, suggesting that TRPC6 expression promoted dendritic growth and the development of dendritic spines and excitatory synapses via the CaMKIV-CREB pathway in FCD. Thus, overexpression of BDNF and TRPC6 and activation of the TRPC6 signal transduction pathway in cortical lesions of FCD patients may contribute to FC pathogenesis and epileptogenesis.

3.
J Neuropathol Exp Neurol ; 75(1): 61-8, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26671983

RESUMO

Focal cortical dysplasias (FCDs) are major brain malformations that commonly lead to medically intractable epilepsy. The purinergic ionotropic P2X7 receptor (P2X7R) is an atypical P2X subtype that gates calcium and sodium ions. Previous animal studies have suggested that P2X7R is a contributing factor in epileptogenesis. This study aimed to define the distribution and expression of P2X7R in 35 FCD patient-surgical-resection specimens relative to autopsy control samples (n = 8). Immunohistochemical colocalization assays revealed that P2X7R was primarily expressed in neurons, astrocytes, and microglia. In FCD samples, P2X7R protein levels were increased in abnormal cell types such as dysmorphic neurons and balloon cells, which are characteristic of FCD. By real-time PCR and Western blotting, P2X7R mRNA and protein expression levels were elevated in FCD patient samples vs control samples; P2X7R expression was also higher in FCDII vs FCDIa patient samples. Because interleukin-1ß is a downstream factor of the P2X7R signaling pathway, we determined that there was also moderate-to-strong interleukin-1ß expression in the dysmorphic neurons, balloon cells, and microglia in FCD patient lesions. These results indicate that increasing P2X7R levels may contribute to the pathogenesis of human FCD and that P2X7R represents a potential anti-epileptogenic target.


Assuntos
Córtex Cerebral/química , Córtex Cerebral/metabolismo , Malformações do Desenvolvimento Cortical/metabolismo , Receptores Purinérgicos P2X7/análise , Receptores Purinérgicos P2X7/biossíntese , Adolescente , Córtex Cerebral/patologia , Criança , Pré-Escolar , Feminino , Regulação da Expressão Gênica , Humanos , Lactente , Masculino , Malformações do Desenvolvimento Cortical/diagnóstico , Malformações do Desenvolvimento Cortical/genética , Receptores Purinérgicos P2X7/genética , Adulto Jovem
4.
J Mol Neurosci ; 55(3): 561-9, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25085710

RESUMO

Focal cortical dysplasias (FCDs) are frequently associated with the medical refractory epilepsy in both children and adults. Transient receptor potential canonical channel 5 (TRPC5), a receptor-operated cation channel, has been well recognized as a regulator in the central nervous system. Here, we examined the expression and cellular distribution of TRPC5 in the specimens from patients with FCDIa (n = 14), FCDIIa (n = 12), and FCDIIb (n = 12) compared with the age-matched control cortex (CTX). TRPC5 mRNA and protein levels were significantly higher in FCDs compared with CTX. Immunohistochemical data showed that TRPC5 was strongly expressed in the misshapen cells, particularly in neuronal microcolumns, dysmorphic neurons, and balloon cells. Moreover, the double-label immunofluorescence analyses demonstrated that TRPC5 localized on NeuN-positive neurons. In addition, its co-localization with glutamate and gamma-aminobutyric acid (GABA) indicated that TRPC5 was distributed on both glutamatergic and GABAergic neurons. Taken together, these results suggested that increased expression of TRPC5 in FCDs and the cell-specific distribution patterns of TRPC5 in the misshapen neurons in FCDs could potentially contribute to the epileptogenesis of FCDs.


Assuntos
Córtex Cerebral/metabolismo , Malformações do Desenvolvimento Cortical/metabolismo , Canais de Cátion TRPC/metabolismo , Estudos de Casos e Controles , Córtex Cerebral/patologia , Criança , Pré-Escolar , Feminino , Neurônios GABAérgicos/metabolismo , Neurônios GABAérgicos/patologia , Ácido Glutâmico/metabolismo , Humanos , Masculino , Malformações do Desenvolvimento Cortical/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Canais de Cátion TRPC/genética , Ácido gama-Aminobutírico/metabolismo
5.
J Mol Neurosci ; 52(4): 586-97, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24057922

RESUMO

Microgyria is associated with epilepsy and due to developmental disruption of neuronal migration. However, the role of endogenous subventricular zone-derived neural progenitors (SDNPs) in formation and hyperexcitability has not been fully elucidated. Here, we establish a neonatal cortex freeze-lesion (FL) model, which was considered as a model for focal microgyria, and simultaneously label SDNPs by CM-DiI. Morphological investigation showed that SDNPs migrated into FL and differentiated into neuronal and glia cell types, suggesting the involvement of endogenous SDNPs in the formation of FL-induced microgyria. Patch-clamp recordings in CM-DiI positive (CM-DiI(+)) pyramidal neurons within FL indicated an increase in frequency of spontaneous action potentials, while the resting membrane potential did not differ from the controls. We also found that spontaneous excitatory postsynaptic currents (EPSCs) increased in frequency but not in amplitude compared with controls. The evoked EPSCs showed a significant increase of 10-90% in rise time and decay time in the CM-DiI(+) neurons. Moreover, paired-pulse facilitation was dramatically larger in CM-DiI(+) pyramidal neurons. Western blotting data showed that AMPA and NMDA receptors were increased to some extent in the FL cortex compared with controls, and the NMDA/AMPA ratio of eEPSCs at CM-DiI(+) pyramidal neurons was significantly increased. Taken together, our findings provide novel evidence for the contribution of endogenous SDNPs in the formation and epileptogenicity of FL-induced focal microgyria.


Assuntos
Malformações do Desenvolvimento Cortical/patologia , Malformações do Desenvolvimento Cortical/fisiopatologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Nicho de Células-Tronco/fisiologia , Potenciais de Ação/fisiologia , Animais , Animais Recém-Nascidos , Carbocianinas/toxicidade , Movimento Celular/fisiologia , Modelos Animais de Doenças , Potenciais Pós-Sinápticos Excitadores/fisiologia , Injeções Intraventriculares , Malformações do Desenvolvimento Cortical/induzido quimicamente , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Células Piramidais/patologia , Células Piramidais/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato/fisiologia
6.
J Neuropathol Exp Neurol ; 73(3): 206-22, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24487795

RESUMO

Focal cortical dysplasia (FCD) is a well-known cause of medically intractable epilepsy. To understand the potential role of the inflammatory cytokine interleukin 2 (IL-2) in the pathogenesis of FCD, we investigated the expression patterns of IL-2 and its receptors (IL-2Rs) in FCD and control samples that included epileptic neocortex from mesial temporal lobe epilepsy patients and nonepileptic normal cortex (CTX). Greater mRNA and protein levels of IL-2 and IL-2Rs were observed in FCD versus CTX samples. Moreover, the expression of IL-2 and IL-2Rs was significantly higher in FCD II than FCD I. In situ hybridization and immunohistochemistry results indicated that IL-2 and IL-2Rs were strongly expressed in hypertrophic neurons and neuronal microcolumns in FCD I and highly expressed in malformed cells in FCD II. In addition, the protein levels of Janus kinase 1, Janus kinase 3, phosphorylated signal transducer and activator of transcription 5, which are important downstream factors in the IL-2 signaling pathway, were increased in FCD lesions. Soluble IL-2R was decreased in FCD compared with that in CTX samples. These results suggest that upregulation of IL-2 and IL-2Rs combined with activation of IL-2-dependent signaling pathways may contribute to the pathogenesis of FCD.


Assuntos
Córtex Cerebral/metabolismo , Interleucina-2/metabolismo , Malformações do Desenvolvimento Cortical/patologia , Receptores de Interleucina-2/metabolismo , Transdução de Sinais/fisiologia , Adolescente , Contagem de Células , Córtex Cerebral/patologia , Criança , Pré-Escolar , Ensaio de Imunoadsorção Enzimática , Epilepsia do Lobo Temporal/etiologia , Epilepsia do Lobo Temporal/patologia , Feminino , Regulação da Expressão Gênica , Humanos , Lactente , Interleucina-2/genética , Janus Quinases/genética , Janus Quinases/metabolismo , Masculino , Malformações do Desenvolvimento Cortical/complicações , RNA Mensageiro/metabolismo , Receptores de Interleucina-2/genética , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo
7.
J Mol Neurosci ; 49(1): 182-93, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22936245

RESUMO

Transient receptor potential vanilloid type-1 (TRPV1) is a ligand-gated nonselective cation channel that has been well characterized in peripheral pain pathway. Recent evidence from animal models of temporal lobe epilepsy (TLE) has supported the important role of TRPV1 in epileptogenesis. In this study, we investigated the expression and cellular distribution of TRPV1 in the temporal cortex (CTX) and hippocampus (HPC) from 26 patients with mesial TLE (MTLE) compared with 12 histologically normal samples. Reverse transcription-PCR and Western blotting revealed up-regulated mRNA and protein levels of TRPV1 in the MTLE group versus the control group. Immunohistochemistry data demonstrated that TRPV1 was mainly distributed in the cell bodies and dendrites of neurons. Double-labeled immunofluorescence further revealed that TRPV1 was localized on NeuN-positive neurons and GFAP-positive astrocytes, but not on HLA-positive microglia. In addition, its co-localization with glutamate and gamma-aminobutyric acid (GABA) indicated that TRPV1 was distributed on both glutamatergic and GABAergic neurons. Moreover, nerve growth factor, a sensitizing factor for TRPV1, was showed a higher expression pattern in MTLE patients. Taken together, our findings suggest that the overexpression and distribution patterns of TRPV1 might be involved in the pathogenesis and epileptogenesis of human MTLE.


Assuntos
Córtex Cerebral/metabolismo , Epilepsia do Lobo Temporal/congênito , Hipocampo/metabolismo , Canais de Cátion TRPV/genética , Regulação para Cima , Adolescente , Adulto , Astrócitos/metabolismo , Estudos de Casos e Controles , Córtex Cerebral/patologia , Dendritos/metabolismo , Epilepsia do Lobo Temporal/diagnóstico , Epilepsia do Lobo Temporal/genética , Feminino , Neurônios GABAérgicos/metabolismo , Ácido Glutâmico/metabolismo , Hipocampo/patologia , Humanos , Masculino , Microglia/metabolismo , Fator de Crescimento Neural/genética , Fator de Crescimento Neural/metabolismo , RNA Mensageiro/biossíntese , Canais de Cátion TRPV/metabolismo , Ácido gama-Aminobutírico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA