Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 10(6)2021 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-34198584

RESUMO

MAGI1 is a cytoplasmic scaffolding protein initially identified as a component of cell-to-cell contacts stabilizing cadherin-mediated cell-cell adhesion in epithelial and endothelial cells. Clinical-pathological and experimental evidence indicates that MAGI1 expression is decreased in some inflammatory diseases, and also in several cancers, including hepatocellular carcinoma, colorectal, cervical, breast, brain, and gastric cancers and appears to act as a tumor suppressor, modulating the activity of oncogenic pathways such as the PI3K/AKT and the Wnt/ß-catenin pathways. Genomic mutations and other mechanisms such as mechanical stress or inflammation have been described to regulate MAGI1 expression. Intriguingly, in breast and colorectal cancers, MAGI1 expression is induced by non-steroidal anti-inflammatory drugs (NSAIDs), suggesting a role in mediating the tumor suppressive activity of NSAIDs. More recently, MAGI1 was found to localize at mature focal adhesion and to regulate integrin-mediated adhesion and signaling in endothelial cells. Here, we review MAGI1's role as scaffolding protein, recent developments in the understanding of MAGI1 function as tumor suppressor gene, its role in endothelial cells and its implication in cancer and vascular biology. We also discuss outstanding questions about its regulation and potential translational implications in oncology.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Moléculas de Adesão Celular , Células Endoteliais/metabolismo , Regulação Neoplásica da Expressão Gênica , Guanilato Quinases , Mutação , Neoplasias , Proteínas Supressoras de Tumor , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Adesão Celular/genética , Moléculas de Adesão Celular/biossíntese , Moléculas de Adesão Celular/genética , Adesões Focais/genética , Adesões Focais/metabolismo , Guanilato Quinases/biossíntese , Guanilato Quinases/genética , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Transdução de Sinais/genética , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/genética
2.
Arch Biochem Biophys ; 701: 108789, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33548214

RESUMO

Calcium/calmodulin-dependent serine protein kinase (CASK), a member of membrane-associated guanylate kinase (MAGUK) super-family, is implicated in regulating cell proliferation, cytoskeletal remodeling, and cell metastasis. Our study aimed to investigate the effect of CASK on the malignant behaviors of pancreatic cancer cells and to determine the signaling pathway involved. CASK expression in pancreatic cancer tissues based on the TCGA database was analyzed using GEPIA online tool. The overall survival (OS) and disease-free survival (DFS) in patients with pancreatic cancer based on CASK expression was also analyzed using GEPIA. KEGG pathway enrichment analysis was used to show the association of 1522 CASK-related genes and signaling pathways. The expression of CASK, Notch1 and Hey1 was detected by Western blot. Cell proliferation, colony number, invasion, and apoptosis were detected by CCK-8, colony formation assay, Transwell invasion assay, and flow cytometry analysis, respectively. Results showed that CASK was upregulated in pancreatic cancer tissues and cells. Pancreatic cancer patients with high CASK expression showed shorter OS and DFS than patients with low CASK expression. KEGG pathway enrichment analysis proved that CASK and 1522 CASK-associated genes were primarily associated with the Notch pathway. CASK silencing inhibited cell proliferation, colony formation ability, and invasion and elicited apoptosis in pancreatic cancer cells. Additionally, we confirmed that CASK silencing inhibited the Notch pathway in pancreatic cancer cells. Overexpression of Notch1 resisted the anti-tumor functions of CASK knockdown in pancreatic cancer cells. In conclusion, CASK knockdown suppressed the malignant behaviors of pancreatic cancer cells by inactivating the Notch pathway.


Assuntos
Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Guanilato Quinases/biossíntese , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogênicas/biossíntese , Receptor Notch1/metabolismo , Transdução de Sinais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Guanilato Quinases/genética , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas/genética , Receptor Notch1/genética
3.
J Neurosci ; 36(15): 4296-312, 2016 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-27076426

RESUMO

The number of AMPA-type glutamate receptors (AMPARs) at synapses is the major determinant of synaptic strength and varies from synapse to synapse. To clarify the underlying molecular mechanisms, the density of AMPARs, PSD-95, and transmembrane AMPAR regulatory proteins (TARPs) were compared at Schaffer collateral/commissural (SCC) synapses in the adult mouse hippocampal CA1 by quantitative immunogold electron microscopy using serial sections. We examined four types of SCC synapses: perforated and nonperforated synapses on pyramidal cells and axodendritic synapses on parvalbumin-positive (PV synapse) and pravalbumin-negative interneurons (non-PV synapse). SCC synapses were categorized into those expressing high-density (perforated and PV synapses) or low-density (nonperforated and non-PV synapses) AMPARs. Although the density of PSD-95 labeling was fairly constant, the density and composition of TARP isoforms was highly variable depending on the synapse type. Of the three TARPs expressed in hippocampal neurons, the disparity in TARP γ-2 labeling was closely related to that of AMPAR labeling. Importantly, AMPAR density was significantly reduced at perforated and PV synapses in TARP γ-2-knock-out (KO) mice, resulting in a virtual loss of AMPAR disparity among SCC synapses. In comparison, TARP γ-8 was the only TARP expressed at nonperforated synapses, where AMPAR labeling further decreased to a background level in TARP γ-8-KO mice. These results show that synaptic inclusion of TARP γ-2 potently increases AMPAR expression and transforms low-density synapses into high-density ones, whereas TARP γ-8 is essential for low-density or basal expression of AMPARs at nonperforated synapses. Therefore, these TARPs are critically involved in AMPAR density control at SCC synapses. SIGNIFICANCE STATEMENT: Although converging evidence implicates the importance of transmembrane AMPA-type glutamate receptor (AMPAR) regulatory proteins (TARPs) in AMPAR stabilization during basal transmission and synaptic plasticity, how they control large disparities in AMPAR numbers or densities across central synapses remains largely unknown. We compared the density of AMPARs with that of TARPs among four types of Schaffer collateral/commissural (SCC) hippocampal synapses in wild-type and TARP-knock-out mice. We show that the density of AMPARs correlates with that of TARP γ-2 across SCC synapses and its high expression is linked to high-density AMPAR expression at perforated type of pyramidal cell synapses and synapses on parvalbumin-positive interneurons. In comparison, TARP γ-8 is the only TARP expressed at nonperforated type of pyramidal cell synapses, playing an essential role in low-density or basal AMPAR expression.


Assuntos
Região CA1 Hipocampal/fisiologia , Canais de Cálcio/fisiologia , Receptores de AMPA/metabolismo , Sinapses/fisiologia , Animais , Canais de Cálcio/genética , Proteína 4 Homóloga a Disks-Large , Potenciais Pós-Sinápticos Excitadores , Guanilato Quinases/biossíntese , Guanilato Quinases/genética , Masculino , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Piramidais/fisiologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Transmissão Sináptica
4.
Pharmacol Biochem Behav ; 139(Pt A): 15-26, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26476132

RESUMO

Cholinesterase inhibitors are first-line therapy for Alzheimer's disease (AD). DL0410 is an AChE/BuChE dual inhibitor with a novel new structural scaffold. It has been demonstrated that DL0410 could improve memory deficits in both Aß1-42-induced and scopolamine-induced amnesia in mice. In the present study, the therapeutic effect of DL0410 and its action mechanism were investigated in APP/PS1 transgenic mice. Six-month old APP/PS1 transgenic mice were orally administered with DL0410 (3, 10, 30 mg/kg/day). After 60 days, several behavioural tests, including the Morris water maze and step-down tests, were used to investigate the effects of DL0410 on mice behaviours. All the behavioural experimental results showed that DL0410 significantly ameliorated memory deficits. Meanwhile, DL0410 attenuated neural cell damage and reduced senile plaques significantly in the hippocampus of APP/PS1 transgenic mice. In addition, DL0410 significantly decreased Aß plaques, while increasing the number of synapses and the thickness of PSD in the hippocampus. We also found DL0410 decreased the expression of APP, NMDAR1B and the phosphorylation level of NMDAR2B, and increased the phosphorylation level of CAMKII and the expression of PSD-95. In this study, the results of behavioural tests demonstrated for the first time that DL0410 could improve learning and memory dysfunction in APP/PS1 transgenic mice. The mechanism of its beneficial effects might be related to cholinesterase inhibition, Aß plaques inhibition, improvement of synapse loss by regulating of expression of proteins related to synapses. As a result, DL0410 could be considered as a candidate drug for the therapy of AD.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/patologia , Placa Amiloide/tratamento farmacológico , Presenilina-1/genética , Sinapses/efeitos dos fármacos , Sinapses/patologia , Acetilcolinesterase/sangue , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/biossíntese , Animais , Comportamento Animal/efeitos dos fármacos , Butirilcolinesterase/sangue , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Córtex Cerebral/metabolismo , Inibidores da Colinesterase/farmacologia , Inibidores da Colinesterase/uso terapêutico , Modelos Animais de Doenças , Proteína 4 Homóloga a Disks-Large , Relação Dose-Resposta a Droga , Guanilato Quinases/biossíntese , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/ultraestrutura , Masculino , Proteínas de Membrana/biossíntese , Camundongos , Camundongos Transgênicos , Fosforilação/efeitos dos fármacos , Placa Amiloide/metabolismo , Densidade Pós-Sináptica/efeitos dos fármacos , Densidade Pós-Sináptica/ultraestrutura , Receptores de N-Metil-D-Aspartato/biossíntese , Receptores de N-Metil-D-Aspartato/metabolismo
5.
J Neurosci ; 35(25): 9402-8, 2015 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-26109663

RESUMO

In brain, specific RNA-binding proteins (RBPs) associate with localized mRNAs and function as regulators of protein synthesis at synapses exerting an indirect control on neuronal activity. Thus, the Fragile X Mental Retardation protein (FMRP) regulates expression of the scaffolding postsynaptic density protein PSD95, but the mode of control appears to be different from other FMRP target mRNAs. Here, we show that the fragile X mental retardation-related protein 2 (FXR2P) cooperates with FMRP in binding to the 3'-UTR of mouse PSD95/Dlg4 mRNA. Absence of FXR2P leads to decreased translation of PSD95/Dlg4 mRNA in the hippocampus, implying a role for FXR2P as translation activator. Remarkably, mGluR-dependent increase of PSD95 synthesis is abolished in neurons lacking Fxr2. Together, these findings show a coordinated regulation of PSD95/Dlg4 mRNA by FMRP and FXR2P that ultimately affects its fine-tuning during synaptic activity.


Assuntos
Regulação da Expressão Gênica/fisiologia , Guanilato Quinases/biossíntese , Proteínas de Membrana/biossíntese , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Western Blotting , Proteína 4 Homóloga a Disks-Large , Guanilato Quinases/genética , Imuno-Histoquímica , Imunoprecipitação , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Biossíntese de Proteínas/fisiologia , Proteínas de Ligação a RNA/genética
6.
J Neurosci ; 35(18): 7116-30, 2015 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-25948262

RESUMO

Fragile X syndrome (FXS) is caused by the loss of the fragile X mental retardation protein (FMRP), an RNA binding protein that regulates translation of numerous target mRNAs, some of which are dendritically localized. Our previous biochemical studies using synaptoneurosomes demonstrate a role for FMRP and miR-125a in regulating the translation of PSD-95 mRNA. However, the local translation of PSD-95 mRNA within dendrites and spines, as well as the roles of FMRP or miR-125a, have not been directly studied. Herein, local synthesis of a Venus-PSD-95 fusion protein was directly visualized in dendrites and spines using single-molecule imaging of a diffusion-restricted Venus-PSD-95 reporter under control of the PSD-95 3'UTR. The basal translation rates of Venus-PSD-95 mRNA was increased in cultured hippocampal neurons from Fmr1 KO mice compared with WT neurons, which correlated with a transient elevation of endogenous PSD-95 within dendrites. Following mGluR stimulation with (S)-3,5-dihydroxyphenylglycine, the rate of Venus-PSD-95 mRNA translation increased rapidly in dendrites of WT hippocampal neurons, but not in those of Fmr1 KO neurons or when the binding site of miR125a, previously shown to bind PSD-95 3'UTR, was mutated. This study provides direct support for the hypothesis that local translation within dendrites and spines is dysregulated in FXS. Impairments in the regulated local synthesis of PSD-95, a critical regulator of synaptic structure and function, may affect the spatiotemporal control of PSD-95 levels and affect dendritic spine development and synaptic plasticity in FXS.


Assuntos
Dendritos/metabolismo , Modelos Animais de Doenças , Síndrome do Cromossomo X Frágil/metabolismo , Guanilato Quinases/biossíntese , Proteínas de Membrana/biossíntese , Imagem Molecular/métodos , Biossíntese de Proteínas/fisiologia , Animais , Células Cultivadas , Dendritos/química , Proteína 4 Homóloga a Disks-Large , Guanilato Quinases/análise , Hipocampo/química , Hipocampo/metabolismo , Masculino , Proteínas de Membrana/análise , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
7.
Nat Commun ; 6: 5555, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25602919

RESUMO

CARMA1-mediated NF-κB activation controls lymphocyte activation through antigen receptors and survival of some malignant lymphomas. CARMA1 clusters are formed on physiological receptor-mediated activation or by its oncogenic mutation in activated B-cell-diffuse large B-cell lymphomas (ABC-DLBCLs) with constitutive NF-κB activation. However, regulatory mechanisms and relevance of CARMA1 clusters in the NF-κB pathway are unclear. Here we show that SH3 and GUK domain interactions of CARMA1 link CARMA1 clustering to signal activation. SH3 and GUK domains of CARMA1 interact by either intra- or intermolecular mechanisms, which are required for activation-induced assembly of CARMA1. Disruption of these interactions abolishes the formation of CARMA1 microclusters at the immunological synapse, CARMA-regulated signal activation following antigen receptor stimulation as well as spontaneous CARMA1 clustering and NF-κB activation by the oncogenic CARMA1 mutation in ABC-DLBCLs. Thus, the SH3-GUK interactions that regulate CARMA1 cluster formations are promising therapeutic targets for ABC-DLBCLs.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/biossíntese , Guanilato Ciclase/biossíntese , Subunidade p50 de NF-kappa B/metabolismo , Transdução de Sinais , Animais , Proteínas Adaptadoras de Sinalização CARD/química , Análise por Conglomerados , Cristalografia por Raios X , Proteína 4 Homóloga a Disks-Large , Feminino , Guanilato Ciclase/química , Guanilato Quinases/biossíntese , Guanilato Quinases/química , Humanos , Sistema Imunitário/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/química , Células Jurkat , Linfócitos/citologia , Masculino , Proteínas de Membrana/biossíntese , Proteínas de Membrana/química , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Mutação , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Ratos , Frações Subcelulares/metabolismo , Domínios de Homologia de src
8.
Anesth Analg ; 118(6): 1345-54, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24842180

RESUMO

BACKGROUND: N-methyl-D-aspartate receptor (NMDARs)-dependent central sensitization plays an important role in cancer pain. Binding of NMDAR subunit 2B (NR2B) by postsynaptic density protein-95 (PSD-95) can couple NMDAR activity to intracellular enzymes, such as neuronal nitric oxide synthase (nNOS), facilitate downstream signaling pathways, and modulate NMDAR stability, contributing to synaptic plasticity. In this study, we investigated whether perturbing the specific interaction between spinal NR2B-containing NMDAR and PSD-95, using a peptide-mimetic strategy, could attenuate bone cancer-related pain behaviors. METHODS: Osteosarcoma cells were implanted into the intramedullary space of the right femurs of C3H/HeJ mice to induce progressive bone cancer-related pain behaviors. Western blotting was applied to examine the expression of spinal phospho-Tyr1472 NR2B, nNOS, and PSD-95. We further investigated the effects of intrathecal injection of the mimetic peptide Myr-NR2B9c, which competitively disrupts the interaction between PSD-95 and NR2B, on nociceptive behaviors and on the upregulation of phospho-Tyr1472 NR2B, nNOS, and PSD-95 associated with bone cancer pain in the spinal cord. RESULTS: Inoculation of osteosarcoma cells induced progressive bone cancer pain and resulted in a significant upregulation of phospho-Tyr1472 NR2B, nNOS, and PSD-95. Intrathecal administration of Myr-NR2B9c attenuated bone cancer-evoked mechanical allodynia, thermal hyperalgesia, and reduced spinal phospho-Tyr1472 NR2B, nNOS, and PSD-95 expression. CONCLUSIONS: Intrathecal administration of Myr-NR2B9c reduced bone cancer pain. Internalization of spinal NR2B and dissociation NR2B-containing NMDARs activation from downstream nNOS signaling may contribute to the analgesic effects of Myr-NR2B9c. This approach may circumvent the negative consequences associated with blocking NMDARs, and may be a novel strategy for the treatment of bone cancer pain.


Assuntos
Neoplasias Ósseas/complicações , Guanilato Quinases/efeitos dos fármacos , Lipopeptídeos/uso terapêutico , Proteínas de Membrana/efeitos dos fármacos , Dor/tratamento farmacológico , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Western Blotting , Proteína 4 Homóloga a Disks-Large , Guanilato Quinases/biossíntese , Hiperalgesia/prevenção & controle , Imunoprecipitação , Injeções Espinhais , Lipopeptídeos/administração & dosagem , Masculino , Proteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos C3H , Transplante de Neoplasias , Óxido Nítrico Sintase Tipo I/biossíntese , Dor/etiologia , Dor/psicologia , Fosforilação , Ligação Proteica/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/biossíntese , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Células Tumorais Cultivadas
9.
Neuromolecular Med ; 15(3): 541-8, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23821337

RESUMO

Functional and ultrastructural investigations support the concept that altered brain connectivity, exhausted neural plasticity, and synaptic loss are the strongest correlates of cognitive decline in age-related neurodegenerative dementia of Alzheimer's type. We have previously demonstrated that in transgenic mice, expressing amyloid-ß precursor protein-Swedish mutation active caspase-3 accumulates in hippocampal postsynaptic compartments leading to altered postsynaptic density (PSD) composition, increased long-term depression (LTD), and dendritic spine loss. Furthermore, we found strong evidence that dendritic spine alteration is mediated by calcineurin activation, a calcium-dependent phosphatase involved in synapse signaling. In the present work, we analyzed the molecular mechanism linking alteration of synaptic plasticity to the increase of calcineurin activity. We found that acute treatment of young and plaque-free transgenic mice with the calcineurin inhibitor FK506 leads to a complete rescue of LTD and PSD composition. Our findings are in agreement with other results reporting that calcineurin inhibition improves memory function and restores dendritic spine density, confirming that calcineurin inhibition may be explored as a neuroprotective treatment to stop or slowdown synaptic alterations in Alzheimer's disease.


Assuntos
Doença de Alzheimer/prevenção & controle , Região CA1 Hipocampal/efeitos dos fármacos , Inibidores de Calcineurina , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Densidade Pós-Sináptica/efeitos dos fármacos , Tacrolimo/uso terapêutico , Doença de Alzheimer/fisiopatologia , Animais , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/fisiopatologia , Caspase 3/metabolismo , Dendritos/efeitos dos fármacos , Dendritos/ultraestrutura , Modelos Animais de Doenças , Proteína 4 Homóloga a Disks-Large , Avaliação Pré-Clínica de Medicamentos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Guanilato Quinases/biossíntese , Guanilato Quinases/genética , Masculino , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Metoxi-Hidroxifenilglicol/análogos & derivados , Metoxi-Hidroxifenilglicol/farmacologia , Camundongos , Camundongos Transgênicos , Fármacos Neuroprotetores/farmacologia , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Receptores de AMPA/metabolismo , Receptores de Glutamato Metabotrópico/agonistas , Tacrolimo/farmacologia
10.
Proc Natl Acad Sci U S A ; 110(31): 12822-7, 2013 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-23858442

RESUMO

Cranial irradiation is used routinely for the treatment of nearly all brain tumors, but may lead to progressive and debilitating impairments of cognitive function. Changes in synaptic plasticity underlie many neurodegenerative conditions that correlate to specific structural alterations in neurons that are believed to be morphologic determinants of learning and memory. To determine whether changes in dendritic architecture might underlie the neurocognitive sequelae found after irradiation, we investigated the impact of cranial irradiation (1 and 10 Gy) on a range of micromorphometric parameters in mice 10 and 30 d following exposure. Our data revealed significant reductions in dendritic complexity, where dendritic branching, length, and area were routinely reduced (>50%) in a dose-dependent manner. At these same doses and times we found significant reductions in the number (20-35%) and density (40-70%) of dendritic spines on hippocampal neurons of the dentate gyrus. Interestingly, immature filopodia showed the greatest sensitivity to irradiation compared with more mature spine morphologies, with reductions of 43% and 73% found 30 d after 1 and 10 Gy, respectively. Analysis of granule-cell neurons spanning the subfields of the dentate gyrus revealed significant reductions in synaptophysin expression at presynaptic sites in the dentate hilus, and significant increases in postsynaptic density protein (PSD-95) were found along dendrites in the granule cell and molecular layers. These findings are unique in demonstrating dose-responsive changes in dendritic complexity, synaptic protein levels, spine density and morphology, alterations induced in hippocampal neurons by irradiation that persist for at least 1 mo, and that resemble similar types of changes found in many neurodegenerative conditions.


Assuntos
Irradiação Craniana/efeitos adversos , Dendritos , Giro Denteado , Relação Dose-Resposta à Radiação , Raios gama/efeitos adversos , Lesões Experimentais por Radiação , Animais , Dendritos/metabolismo , Dendritos/patologia , Giro Denteado/metabolismo , Giro Denteado/patologia , Proteína 4 Homóloga a Disks-Large , Regulação da Expressão Gênica/efeitos da radiação , Guanilato Quinases/biossíntese , Proteínas de Membrana/biossíntese , Camundongos , Camundongos Transgênicos , Rede Nervosa/patologia , Rede Nervosa/efeitos da radiação , Lesões Experimentais por Radiação/metabolismo , Lesões Experimentais por Radiação/patologia , Sinaptofisina/biossíntese
11.
J Psychopharmacol ; 27(10): 930-9, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23863923

RESUMO

The putative antidepressant captodiamine is a 5-HT2c receptor antagonist and agonist at sigma-1 and D3 dopamine receptors, exerts an anti-immobility action in the forced swim paradigm, and enhances dopamine turnover in the frontal cortex. Captodiamine has also been found to ameliorate stress-induced anhedonia, reduce the associated elevations of hypothalamic corticotrophin-releasing factor (CRF) and restore the reductions in hypothalamic BDNF expression. Here we demonstrate chronic administration of captodiamine to have no significant effect on hypothalamic CRF expression through sigma-1 receptor agonism; however, both sigma-1 receptor agonism or 5-HT2c receptor antagonism were necessary to enhance BDNF expression. Regulation of BDNF expression by captodiamine was associated with increased phosphorylation of transcription factor CREB and mediated through sigma-1 receptor agonism but blocked by 5-HT2c receptor antagonism. The existence of two separate signalling pathways was confirmed by immunolocalisation of each receptor to distinct cell populations in the paraventricular nucleus of the hypothalamus. Increased BDNF induced by captodiamine was also associated with enhanced expression of synapsin, but not PSD-95, suggesting induction of long-term structural plasticity between hypothalamic synapses. These unique features of captodiamine may contribute to its ability to ameliorate stress-induced anhedonia as the hypothalamus plays a prominent role in regulating HPA axis activity.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/biossíntese , Etilaminas/farmacologia , Hipotálamo/metabolismo , Receptor 5-HT2C de Serotonina/metabolismo , Receptores sigma/agonistas , Antagonistas do Receptor 5-HT2 de Serotonina/farmacologia , Sulfetos/farmacologia , Animais , Antidepressivos/agonistas , Antidepressivos/antagonistas & inibidores , Antidepressivos/farmacologia , Proteína de Ligação a CREB/metabolismo , Carbazóis/farmacologia , Hormônio Liberador da Corticotropina/biossíntese , Proteína 4 Homóloga a Disks-Large , Interações Medicamentosas , Regulação da Expressão Gênica/efeitos dos fármacos , Guanilato Quinases/biossíntese , Hipotálamo/efeitos dos fármacos , Masculino , Proteínas de Membrana/biossíntese , Camundongos , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Fosforilação/efeitos dos fármacos , Ritanserina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sinapsinas/biossíntese , Receptor Sigma-1
12.
Biochim Biophys Acta ; 1832(6): 837-47, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23474306

RESUMO

Education and cognitive occupations are commonly associated to reduce risk of Alzheimer's disease (AD) or dementia. Animal studies have demonstrated that cognitive stimulation (CS) achieved by social/physical activities and/or enriched environments compensates for memory decline. We have elaborated a novel paradigm of CS that is devoid of physical/social activity and enriched environments. 4 month-old Tg2576 mice were cognitively trained for 8 weeks and, after a break of 8 months, long-lasting effects of CS on cognitive abilities and AD-like pathology were measured. Morris Water Maze (MWM) and Novel Object Recognition (NOR) tests showed that deficits in spatial and recognition memories were compensated by CS. These outcomes were accompanied by increased levels of hippocampal post-synaptic markers (PSD95 and NR1) and proteins involved in synaptic formation (Arc, ß-catenin). CS softened amyloid pathology in terms of reduced levels of Aß1-42 and the dodecameric assembly, referred as Aß*56. CS appeared to affect the APP processing since differences in levels of ADAM17, BACE1 and C99/C83 ratio were found. Tau hyper-phosphorylation and high activities of tau kinases were also reduced by CS. In contrast, CS did not induce any of these molecular changes in wild-type mice. The present findings suggest beneficial and long-lasting effects of CS early in life on cognitive decline and AD-like pathology.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/biossíntese , Comportamento Animal , Cognição , Aprendizagem em Labirinto , Antígeno 12E7 , Proteínas ADAM/biossíntese , Proteínas ADAM/genética , Proteína ADAM17 , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/biossíntese , Secretases da Proteína Precursora do Amiloide/genética , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Antígenos CD/biossíntese , Antígenos CD/genética , Ácido Aspártico Endopeptidases/biossíntese , Ácido Aspártico Endopeptidases/genética , Biomarcadores/metabolismo , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Modelos Animais de Doenças , Proteína 4 Homóloga a Disks-Large , Feminino , Guanilato Quinases/biossíntese , Guanilato Quinases/genética , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Humanos , Imunoglobulinas/biossíntese , Imunoglobulinas/genética , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Receptores de N-Metil-D-Aspartato , Antígeno CD83
13.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 4): 587-94, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23519667

RESUMO

PSD-93 (chapsyn-110, DLG2) is a member of the family of membrane-associated guanylate kinase (MAGUK) proteins. The MAGUK proteins are involved in receptor localization and signalling pathways. The best characterized MAGUK protein, PSD-95, is known to be involved in NMDA receptor signalling via its PDZ domains. The PDZ domains of PSD-95 and PSD-93 are structurally very similar, but relatively little is known about the function of PSD-93. PSD-93 has been suggested to interact with GluD2 from the family of ionotropic glutamate receptors. Here, the interactions of four residues (GTSI) representing the extreme C-terminus of GluD2 with PSD-93 PDZ1 have been investigated in the crystalline phase. Two different binding modes of these residues were observed, suggesting that the peptide is not tightly bound to PSD-93 PDZ1. In accordance, the two N-terminal PSD-93 PDZ domains show no appreciable binding affinity for a GluD2-derived C-terminal octapeptide, whereas micromolar affinity was observed for a GluN2B-derived C-terminal octapeptide. This indicates that if present, the interactions between GluD2 and PSD-93 involve more than the extreme terminus of the receptor. In contrast, the tumour-suppressor protein SCRIB PDZ3 shows low micromolar affinity towards the GluD2-derived octapeptide, which is in agreement with previous findings using high-throughput assays.


Assuntos
Guanilato Quinases/metabolismo , Mapeamento de Interação de Proteínas/métodos , Proteínas Supressoras de Tumor/metabolismo , Comunicação Celular/fisiologia , Cristalização , Cristalografia por Raios X , Polarização de Fluorescência , Guanilato Quinases/biossíntese , Guanilato Quinases/química , Humanos , Microscopia de Fluorescência por Excitação Multifotônica , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Estrutura Terciária de Proteína , Espectrometria de Fluorescência , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/química
14.
Int J Neuropsychopharmacol ; 16(1): 69-82, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22339950

RESUMO

Major depressive disorder (MDD) has been linked to changes in function and activity of the hippocampus, one of the central limbic regions involved in regulation of emotions and mood. The exact cellular and molecular mechanisms underlying hippocampal plasticity in response to stress are yet to be fully characterized. In this study, we examined the genetic profile of micro-dissected subfields of post-mortem hippocampus from subjects diagnosed with MDD and comparison subjects matched for sex, race and age. Gene expression profiles of the dentate gyrus and CA1 were assessed by 48K human HEEBO whole genome microarrays and a subgroup of identified genes was confirmed by real-time polymerase chain reaction (qPCR). Pathway analysis revealed altered expression of several gene families, including cytoskeletal proteins involved in rearrangement of neuronal processes. Based on this and evidence of hippocampal neuronal atrophy in MDD, we focused on the expression of cytoskeletal, synaptic and glutamate receptor genes. Our findings demonstrate significant dysregulation of synaptic function/structure related genes SNAP25, DLG2 (SAP93), and MAP1A, and 2-amino-3-(5-methyl-3-oxo-1,2-oxazol-4-yl)propanoic acid receptor subunit genes GLUR1 and GLUR3. Several of these human target genes were similarly dysregulated in a rat model of chronic unpredictable stress and the effects reversed by antidepressant treatment. Together, these studies provide new evidence that disruption of synaptic and glutamatergic signalling pathways contribute to the pathophysiology underlying MDD and provide interesting targets for novel therapeutic interventions.


Assuntos
Transtorno Depressivo Maior/genética , Hipocampo/metabolismo , Hipocampo/patologia , Proteínas do Tecido Nervoso/genética , Receptores de Glutamato/genética , Sinapses/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Transtorno Depressivo Maior/metabolismo , Transtorno Depressivo Maior/patologia , Feminino , Regulação da Expressão Gênica , Guanilato Quinases/biossíntese , Guanilato Quinases/genética , Humanos , Masculino , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Associadas aos Microtúbulos/genética , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/biossíntese , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/biossíntese , Receptores de AMPA/genética , Receptores de Glutamato/biossíntese , Sinapses/metabolismo , Proteína 25 Associada a Sinaptossoma/biossíntese , Proteína 25 Associada a Sinaptossoma/genética , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/genética
15.
Neurochem Int ; 62(1): 84-91, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23201339

RESUMO

Repetitive transcranial magnetic stimulation (rTMS) is a neuropsychiatric tool that can be used to investigate the neurobiology of learning and cognitive function. Few studies have examined the effects of low frequency (⩽1Hz) magnetic stimulation (MS) on structural synaptic plasticity of neurons in vitro, thus, the current study examined its effects on hippocampal neuron and synapse morphology, as well as synaptic protein markers and signaling pathways. Similarly, both intensities of low frequency magnetic stimulation (1Hz) activated brain-derived neurotrophic factor (BDNF) and tropomyosin-related kinase B (TrkB) pathways, including the pathways for mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) and for phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt). Specifically, low intensity magnetic stimulation (LIMS, 1.14Tesla, 1Hz) promoted more extensive dendritic and axonal arborization, as well as increasing synapses density, thickening PSD (post synaptic density) and upregulation of synaptophysin (SYN), growth associated protein 43 (GAP43) and post synaptic density 95 (PSD95). Conversely, high intensity magnetic stimulation (HIMS, 1.55Tesla, 1Hz) appeared to be detrimental, reducing dendritic and axonal arborization and causing apparent structural damage, including thinning of PSD, less synapses and disordered synaptic structure, as well as upregulation of GAP43 and PSD95, possibly for their ability to mitigate dysfunction. In conclusion, we infers that low frequency magnetic stimulation participates in regulating structural synaptic plasticity of hippocampal neurons via the activation of BDNF-TrkB signaling pathways.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Campos Eletromagnéticos , Hipocampo/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Receptor trkB/fisiologia , Transdução de Sinais/fisiologia , Actinas/biossíntese , Animais , Apoptose/fisiologia , Western Blotting , Sobrevivência Celular/fisiologia , Células Cultivadas , Proteína 4 Homóloga a Disks-Large , Proteína GAP-43/biossíntese , Guanilato Quinases/biossíntese , Imuno-Histoquímica , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas de Membrana/biossíntese , Camundongos , Fosfatidilinositol 3-Quinases/fisiologia , Sinapses/fisiologia , Sinaptofisina/biossíntese , Sinaptofisina/genética , Transcrição Gênica/fisiologia
16.
J Neurosci ; 32(49): 17800-12, 2012 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-23223299

RESUMO

Neonatal seizures can lead to later life epilepsy and neurobehavioral deficits, and there are no treatments to prevent these sequelae. We showed previously that hypoxia-induced seizures in a neonatal rat model induce rapid phosphorylation of serine-831 (S831) and Serine 845 (S845) sites of the AMPA receptor GluR1 subunit and later neuronal hyperexcitability and epilepsy, suggesting that seizure-induced posttranslational modifications may represent a novel therapeutic target. To unambiguously assess the contribution of these sites, we examined seizure susceptibility in wild-type mice versus transgenic knock-in mice with deficits in GluR1 S831 and S845 phosphorylation [GluR1 double-phosphomutant (GluR1 DPM) mice]. Phosphorylation of the GluR1 S831 and S845 sites was significantly increased in the hippocampus and cortex after a single episode of pentyleneterazol-induced seizures in postnatal day 7 (P7) wild-type mouse pups and that transgenic knock-in mice have a higher threshold and longer latencies to seizures. Like the rat, hypoxic seizures in P9 C57BL/6N wild-type mice resulted in transient increases in GluR1 S831 and GluR1 S845 phosphorylation in cortex and were associated with enhanced seizure susceptibility to later-life kainic-acid-induced seizures. In contrast, later-life seizure susceptibility after hypoxia-induced seizures was attenuated in GluR1 DPM mice, supporting a role for posttranslational modifications in seizure-induced network excitability. Finally, human hippocampal samples from neonatal seizure autopsy cases also showed an increase in GluR1 S831 and S845, supporting the validation of this potential therapeutic target in human tissue.


Assuntos
Suscetibilidade a Doenças/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiologia , Receptores de AMPA/metabolismo , Receptores de AMPA/fisiologia , Convulsões/metabolismo , Animais , Animais Recém-Nascidos , Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Proteína 4 Homóloga a Disks-Large , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Técnicas de Introdução de Genes/métodos , Guanilato Quinases/biossíntese , Humanos , Hipóxia , Ácido Caínico , Proteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Pentilenotetrazol , Fosforilação , Receptores de AMPA/genética , Convulsões/induzido quimicamente , Serina/metabolismo
17.
Exp Neurol ; 237(2): 335-45, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22776425

RESUMO

Brain-derived neurotrophic factor (BDNF) is a key player in learning and memory processes. However, little is known about brain area-specific functions of this neurotrophin. Here we investigated whether BDNF could differently affect motor neocortical and hippocampal-related cognitive and plastic morphologic changes in young (12-week-old) and middle-aged (30-week-old) BDNF heterozygous (BDNF⁺/⁻) and wild type (wt) mice. We found that at 30 weeks of age, BDNF⁺/⁻ mice showed impaired performance in accelerating rotarod and grasping tests while preserved spatial learning in a T-maze and recognition memory in an object recognition task compared with wt mice suggesting a specific neocortical dysfunction. Accordingly, a significant reduction of synaptic markers (PSD-95 and GluR1) and corresponding puncta was observed in motor neocortex but not in hippocampus of BDNF⁺/⁻ mice. Interestingly, 30-week-old BDNF⁺/⁻ mice displayed increased TrkB levels in the hippocampus but not in the motor neocortex, which suggests specific hippocampal compensatory mechanisms as a consequence of BDNF decrease. In conclusion, our data indicates that BDNF could differentially regulate the neuronal micro-structures and cognition in a region-specific and in an age-dependent manner.


Assuntos
Envelhecimento/fisiologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Encéfalo/metabolismo , Guanilato Quinases/biossíntese , Proteínas de Membrana/biossíntese , Receptor trkB/biossíntese , Animais , Western Blotting , Fator Neurotrófico Derivado do Encéfalo/genética , Espinhas Dendríticas/metabolismo , Proteína 4 Homóloga a Disks-Large , Ensaio de Imunoadsorção Enzimática , Heterozigoto , Aprendizagem/fisiologia , Memória/fisiologia , Camundongos , Microscopia Confocal , Plasticidade Neuronal/fisiologia
18.
Br J Pharmacol ; 163(8): 1707-20, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21391983

RESUMO

BACKGROUND AND PURPOSE We recently demonstrated that activation of the spinal sigma-1 receptor induces mechanical and thermal hypersensitivity via calcium-dependent second messenger cascades and phosphorylation of the spinal NMDA receptor GluN1 subunit (pGluN1). Here we examined the role of NO in this process, as it plays a critical role in PKC-mediated calcium signalling and the potentiation of NMDA receptor function. EXPERIMENTAL APPROACH The effects of intrathecal (i.t.) pretreatment with nNOS inhibitors on PRE084 (sigma-1 receptor agonist)-induced pain were assessed in mice by use of mechanical allodynia and thermal hyperalgesia tests. Western blot analysis, immunoprecipitation and immunohistochemical techniques were used to determine effects of these treatments on spinal pGluN1-immunoreactive (ir) cells, whether PRE084 induces a time-dependent modification of nNOS activity in the dorsal horn, and if any changes in nNOS activity can be blocked by sigma-1 receptor, calcineurin or soluble guanylyl cyclase (sGC) inhibitors. KEY RESULTS PRE084, injected i.t., induced mechanical and thermal hypersensitivity, and increased the number of PKC- and PKA-dependent pGluN1-ir cells in spinal cord. This PRE084-induced hypersensitivity and increase in PKC-dependent pGluN1 expression were blocked by pretreatment with N(G) -nitro-L-arginine methyl ester (L-NAME) or 7-nitroindazole (7-NI). PRE084 also time-dependently decreased the ratio of phosphorylated nNOS (pnNOS) to nNOS expression and the number of spinal pnNOS-ir cells. This decrease in pnNOS was prevented by BD1047, a sigma-1 receptor antagonist and cyclosporin A, a calcineurin inhibitor, but not by a sGC inhibitor. CONCLUSIONS AND IMPLICATIONS Spinal sigma-1 receptor-induced sensitization is mediated by an increase in nNOS activity, which is associated with an NO-induced increase in PKC-dependent pGluN1 expression.


Assuntos
Hiperalgesia/metabolismo , Óxido Nítrico Sintase Tipo I/fisiologia , Proteína Quinase C/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Receptores sigma/fisiologia , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/farmacologia , Animais , Grupos Controle , Proteína 4 Homóloga a Disks-Large , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/farmacologia , Guanilato Quinases/análise , Guanilato Quinases/biossíntese , Hiperalgesia/prevenção & controle , Injeções Espinhais , Masculino , Proteínas de Membrana/análise , Proteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos ICR , Morfolinas/administração & dosagem , Morfolinas/farmacologia , NG-Nitroarginina Metil Éster/administração & dosagem , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase Tipo I/análise , Dor/fisiopatologia , Medição da Dor , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/fisiopatologia , Receptor Sigma-1
19.
J Neurochem ; 117(5): 833-40, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21426345

RESUMO

The sodium-dependent glutamate transporter, glutamate transporter subtype 1 (GLT-1) is one of the main glutamate transporters in the brain. GLT-1 contains a COOH-terminal sequence similar to one in an isoform of Slo1 K(+) channel protein previously shown to bind MAGI-1 (membrane-associated guanylate kinase with inverted orientation protein-1). MAGI-1 is a scaffold protein which allows the formation of complexes between certain transmembrane proteins, actin-binding proteins, and other regulatory proteins. The glutathione S-transferase pull-down assay demonstrated that MAGI-1 was a binding partner of GLT-1. The interaction between MAGI-1 and GLT-1 was confirmed by co-immunoprecipitation. Immunofluorescence of MAGI-1 and GLT-1 demonstrated that the distribution of MAGI-1 and GLT-1 overlapped in astrocytes. Co-expression of MAGI-1 with GLT-1 in C6 Glioma cells resulted in a significant reduction in the surface expression of GLT-1, as assessed by cell-surface biotinylation. On the other hand, partial knockdown of endogenous MAGI-1 expression by small interfering RNA in differentiated cultured astrocytes increased glutamate uptake and the surface expression of endogenous GLT-1. Knockdown of MAGI-1 increased dihydrokainate-sensitive, Na(+) -dependent glutamate uptake, indicating that MAGI-1 regulates GLT-1 mediated glutamate uptake. These data suggest that MAGI-1 regulates surface expression of GLT-1 and the level of glutamate in the hippocampus.


Assuntos
Transportador 1 de Aminoácido Excitatório/biossíntese , Guanilato Quinases/biossíntese , Proteínas de Membrana/biossíntese , Animais , Astrócitos/metabolismo , Biotinilação , Linhagem Celular Tumoral , Células Cultivadas , Transportador 1 de Aminoácido Excitatório/genética , Transportador 3 de Aminoácido Excitatório/biossíntese , Transportador 3 de Aminoácido Excitatório/genética , Imunofluorescência , Regulação da Expressão Gênica , Glioma/metabolismo , Ácido Glutâmico/metabolismo , Glutationa Transferase/metabolismo , Guanilato Quinases/genética , Hipocampo/citologia , Hipocampo/metabolismo , Imunoprecipitação , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/biossíntese , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Proteínas de Membrana/genética , Plasmídeos/genética , RNA Interferente Pequeno/farmacologia , Ratos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Transfecção
20.
J Comp Neurol ; 518(24): 4842-54, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21031555

RESUMO

GABAergic interneurons are key elements regulating the activity of local circuits, and abnormal inhibitory circuits are implicated in certain psychiatric and neurodevelopmental diseases. The glutamatergic input that interneurons receive is a key determinant of their activity, yet its molecular structure and development, which are often distinct from those of glutamatergic input to pyramidal cells, are poorly defined. The membrane-associated guanylate kinase (MAGUK) homologs PSD-95/SAP90, PSD-93/chapsyn110, SAP97, and SAP102 are central organizers of the postsynaptic density at excitatory synapses on pyramidal neurons. We therefore studied the cell-type-specific and developmental expression of MAGUKs in the nonoverlapping parvalbumin (PV)- and somatostatin (SOM)-positive interneurons in the visual cortex. These interneuron subtypes account for the vast majority of interneurons in the cortex and have different functional properties and postsynaptic structures, being either axodendritic (PV(+)) or axospinous (SOM(+)). To study cell-type-specific MAGUK expression, we used DIG-labeled riboprobes against each MAGUK along with antibodies against either PV or SOM and examined tissue from juvenile (P15) and adult mice. Both PV(+) and SOM(+) interneurons express mRNA for PSD-95, PSD-93, and SAP102 in P15 and adult tissue. In contrast, these interneuron subtypes express SAP97 at P15, but for adult visual cortex we found that most PV(+) and SOM(+) interneurons show low or no expression of SAP97. Given the importance of SAP97 in regulating AMPA receptor GluA1 subunit and NMDA receptor subunits at glutamatergic synapses, these results suggest a developmental shift in glutamate receptor subunit composition and regulation of glutamatergic synapses on PV(+) and SOM(+) interneurons.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Guanilato Quinases/biossíntese , Proteínas de Membrana/biossíntese , Neuropeptídeos/biossíntese , Córtex Visual/enzimologia , Animais , Proteína 1 Homóloga a Discs-Large , Proteína 4 Homóloga a Disks-Large , Potenciais Pós-Sinápticos Excitadores/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Densidade Pós-Sináptica/fisiologia , Células Piramidais/citologia , Células Piramidais/enzimologia , Células Piramidais/crescimento & desenvolvimento , Homologia Estrutural de Proteína , Córtex Visual/citologia , Córtex Visual/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...