Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Neurochem Int ; 122: 8-18, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30336179

RESUMO

Medium spiny neurons (MSNs) expressing dopamine D1 receptor (D1R) or D2 receptor (D2R) are major components of the striatum. Stimulation of D1R activates protein kinase A (PKA) through Golf to increase neuronal activity, while D2R stimulation inhibits PKA through Gi. Adenosine A2A receptor (A2AR) coupled to Golf is highly expressed in D2R-MSNs within the striatum. However, how dopamine and adenosine co-operatively regulate PKA activity remains largely unknown. Here, we measured Rap1gap serine 563 phosphorylation to monitor PKA activity and examined dopamine and adenosine signals in MSNs. We found that a D1R agonist increased Rap1gap phosphorylation in striatal slices and in D1R-MSNs in vivo. A2AR agonist CGS21680 increased Rap1gap phosphorylation, and pretreatment with the D2R agonist quinpirole blocked this effect in striatal slices. D2R antagonist eticlopride increased Rap1gap phosphorylation in D2R-MSNs in vivo, and the effect of eticlopride was blocked by the pretreatment with the A2AR antagonist SCH58261. These results suggest that adenosine positively regulates PKA in D2R-MSNs through A2AR, while this effect is blocked by basal dopamine in vivo. Incorporating computational model analysis, we propose that the shift from D1R-MSNs to D2R-MSNs or vice versa appears to depend predominantly on a change in dopamine concentration.


Assuntos
Adenosina/metabolismo , Corpo Estriado/metabolismo , Dopamina/metabolismo , Transdução de Sinais , Animais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Agonistas de Dopamina/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Receptores de Dopamina D1/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo
2.
J Cell Biol ; 216(12): 4313-4330, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29089377

RESUMO

Throughout life, stem cells in the ventricular-subventricular zone generate neuroblasts that migrate via the rostral migratory stream (RMS) to the olfactory bulb, where they differentiate into local interneurons. Although progress has been made toward identifying extracellular factors that guide the migration of these cells, little is known about the intracellular mechanisms that govern the dynamic reshaping of the neuroblasts' morphology required for their migration along the RMS. In this study, we identify DOCK7, a member of the DOCK180-family, as a molecule essential for tangential neuroblast migration in the postnatal mouse forebrain. DOCK7 regulates the migration of these cells by controlling both leading process (LP) extension and somal translocation via distinct pathways. It controls LP stability/growth via a Rac-dependent pathway, likely by modulating microtubule networks while also regulating F-actin remodeling at the cell rear to promote somal translocation via a previously unrecognized myosin phosphatase-RhoA-interacting protein-dependent pathway. The coordinated action of both pathways is required to ensure efficient neuroblast migration along the RMS.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/genética , Fosfatase de Miosina-de-Cadeia-Leve/genética , Neurônios/metabolismo , Prosencéfalo/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas rho de Ligação ao GTP/genética , Actinas/genética , Actinas/metabolismo , Animais , Animais Recém-Nascidos , Diferenciação Celular , Linhagem Celular Tumoral , Movimento Celular , Embrião de Mamíferos , Proteínas Ativadoras de GTPase , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células HEK293 , Células HeLa , Humanos , Camundongos , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Neurônios/ultraestrutura , Cultura Primária de Células , Prosencéfalo/citologia , Prosencéfalo/crescimento & desenvolvimento , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP
3.
Neurosci Lett ; 630: 45-52, 2016 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-27424794

RESUMO

Developing cortical neurons undergo a number of sequential developmental events including neuronal survival/apoptosis, and the molecular mechanism underlying each characteristic process has been studied in detail. However, the survival pathway of cortical neurons at mature stages remains largely uninvestigated. We herein focused on mature corticostriatal neurons because of their important roles in various higher brain functions and the spectrum of neurological and neuropsychiatric disorders. The small GTPase Rho is known to control diverse and essential cellular functions through some effector molecules, including Rho-kinase, during neural development. In the present study, we investigated the role of Rho signaling through Rho-kinase in the survival of corticostriatal neurons. We performed the conditional expression of Clostridium botulinum C3 ADP-ribosyltransferase (C3 transferase) or dominant-negative form for Rho-kinase (Rho-K DN), a well-known inhibitor of Rho or Rho-kinase, respectively, in corticostriatal neurons using a dual viral vector approach combining a neuron-specific retrograde gene transfer lentiviral vector and an adeno-associated virus vector. C3 transferase markedly decreased the number of corticostriatal neurons, which was attributed to caspase-3-dependent enhanced apoptosis. In addition, Rho-K DN produced phenotypic defects similar to those caused by C3 transferase. These results indicate that the Rho/Rho-kinase signaling pathway plays a crucial role in the survival of corticostriatal neurons.


Assuntos
Apoptose , Córtex Cerebral/metabolismo , Corpo Estriado/metabolismo , Neurônios/metabolismo , Quinases Associadas a rho/metabolismo , ADP Ribose Transferases/genética , ADP Ribose Transferases/metabolismo , Animais , Toxinas Botulínicas/genética , Toxinas Botulínicas/metabolismo , Sobrevivência Celular , Dependovirus/fisiologia , Vetores Genéticos/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vias Neurais/metabolismo , Transdução de Sinais , Córtex Somatossensorial/metabolismo
4.
Cell Struct Funct ; 41(2): 105-20, 2016 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-27334702

RESUMO

Protein phosphorylation plays an important role in the physiological regulation of cardiac function. Myocardial contraction and pathogenesis of cardiac diseases have been reported to be associated with adaptive or maladaptive protein phosphorylation; however, phosphorylation signaling in the heart is not fully elucidated. We recently developed a novel kinase-interacting substrate screening (KISS) method for exhaustive screening of protein kinase substrates, using mass spectrometry and affinity chromatography. First, we examined protein phosphorylation by extracellular signal-regulated kinase (ERK) and protein kinase A (PKA), which has been relatively well studied in cardiomyocytes. The KISS method showed that ERK and PKA mediated the phosphorylation of known cardiac-substrates of each kinase such as Rps6ka1 and cTnI, respectively. Using this method, we found about 330 proteins as Rho-kinase-mediated substrates, whose substrate in cardiomyocytes is unknown. Among them, CARP/Ankrd1, a muscle ankyrin repeat protein, was confirmed as a novel Rho-kinase-mediated substrate. We also found that non-phosphorylatable form of CARP repressed cardiac hypertrophy-related gene Myosin light chain-2v (MLC-2v) promoter activity, and decreased cell size of heart derived H9c2 myoblasts more efficiently than wild type-CARP. Thus, focused proteomics enable us to reveal a novel signaling pathway in the heart.


Assuntos
Miocárdio/enzimologia , Proteômica , Transdução de Sinais , Quinases Associadas a rho/metabolismo , Proteínas 14-3-3/química , Proteínas 14-3-3/metabolismo , Animais , Encéfalo/metabolismo , Células Cultivadas , Cromatografia de Afinidade , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Imuno-Histoquímica , Espectrometria de Massas , Microscopia de Fluorescência , Proteínas Musculares/química , Proteínas Musculares/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fosforilação , Ligação Proteica , Ratos , Proteínas Repressoras/química , Proteínas Repressoras/metabolismo , Especificidade por Substrato , Quinases Associadas a rho/química
5.
Neuron ; 89(3): 550-65, 2016 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-26804993

RESUMO

Dopamine (DA) type 1 receptor (D1R) signaling in the striatum presumably regulates neuronal excitability and reward-related behaviors through PKA. However, whether and how D1Rs and PKA regulate neuronal excitability and behavior remain largely unknown. Here, we developed a phosphoproteomic analysis method to identify known and novel PKA substrates downstream of the D1R and obtained more than 100 candidate substrates, including Rap1 GEF (Rasgrp2). We found that PKA phosphorylation of Rasgrp2 activated its guanine nucleotide-exchange activity on Rap1. Cocaine exposure activated Rap1 in the nucleus accumbens in mice. The expression of constitutively active PKA or Rap1 in accumbal D1R-expressing medium spiny neurons (D1R-MSNs) enhanced neuronal firing rates and behavioral responses to cocaine exposure through MAPK. Knockout of Rap1 in the accumbal D1R-MSNs was sufficient to decrease these phenotypes. These findings demonstrate a novel DA-PKA-Rap1-MAPK intracellular signaling mechanism in D1R-MSNs that increases neuronal excitability to enhance reward-related behaviors.


Assuntos
Dopamina/metabolismo , Fosfoproteínas/metabolismo , Proteoma/metabolismo , Proteômica , Receptores de Dopamina D1/metabolismo , Recompensa , Transdução de Sinais , Proteínas rap1 de Ligação ao GTP/metabolismo , Potenciais de Ação/fisiologia , Animais , Benzazepinas/farmacologia , Cocaína/farmacologia , Colforsina/farmacologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dopamina/farmacologia , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Neurônios/fisiologia , Núcleo Accumbens/metabolismo , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteínas rap1 de Ligação ao GTP/genética
6.
Cell Struct Funct ; 40(1): 1-12, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25399539

RESUMO

Protein kinase A (PKA) is a serine/threonine kinase whose activity depends on the levels of cyclic AMP (cAMP). PKA plays essential roles in numerous cell types such as myocytes and neurons. Numerous substrate screens have been attempted to clarify the entire scope of the PKA signaling cascade, but it is still underway. Here, we performed a comprehensive screen that consisted of immunoprecipitation and mass spectrometry, with a focus on the identification of PKA substrates. The lysate of HeLa cells treated with Forskolin (FSK)/3-isobutyl methyl xanthine (IBMX) and/or H-89 was subjected to immunoprecipitation using anti-phospho-PKA substrate antibody. The identity of the phosophoproteins and phosphorylation sites in the precipitants was determined using liquid chromatography tandem mass spectrometry (LC/MS/MS). We obtained 112 proteins as candidate substrates and 65 candidate sites overall. Among the candidate substrates, Rho-kinase/ROCK2 was confirmed to be a novel substrate of PKA both in vitro and in vivo. In addition to Rho-kinase, we found more than a hundred of novel candidate substrates of PKA using this screen, and these discoveries provide us with new insights into PKA signaling.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteômica , 1-Metil-3-Isobutilxantina/farmacologia , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Colforsina/farmacologia , Células HeLa , Humanos , Imunoprecipitação , Isoquinolinas/farmacologia , Espectrometria de Massas , Fosforilação/efeitos dos fármacos , Ligação Proteica , Sulfonamidas/farmacologia , Quinases Associadas a rho/química , Quinases Associadas a rho/metabolismo
7.
J Cell Sci ; 125(Pt 9): 2198-211, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22344266

RESUMO

A latent process involving signal transduction and gene expression is needed as a preparation step for cellular function. We previously found that nerve growth factor (NGF)-induced cell differentiation has a latent process, which is dependent on ERK activity and gene expression and required for subsequent neurite extension. A latent process can be considered as a preparation step that decodes extracellular stimulus information into cellular functions; however, molecular mechanisms of this process remain unknown. We identified Metrnl, Dclk1 and Serpinb1a as genes that are induced during the latent process (LP) with distinct temporal expression profiles and are required for subsequent neurite extension in PC12 cells. The LP genes showed distinct dependency on the duration of ERK activity, and they were also induced during the latent process of PACAP- and forskolin-induced cell differentiation. Regardless of neurotrophic factors, expression levels of the LP genes during the latent process (0-12 hours), but not phosphorylation levels of ERK, always correlated with subsequent neurite extension length (12-24 hours). Overexpression of all LP genes together, but not of each gene separately, enhanced NGF-induced neurite extension. The LP gene products showed distinct spatial localization. Thus, the LP genes appear to be the common decoders for neurite extension length regardless of neurotrophic factors, and they might function in distinct temporal and spatial manners during the latent process. Our findings provide molecular insight into the physiological meaning of the latent process as the preparation step for decoding information for future phenotypic change.


Assuntos
Diferenciação Celular/genética , Expressão Gênica , Proteínas do Tecido Nervoso/genética , Neuritos/fisiologia , Proteínas Serina-Treonina Quinases/genética , Serpinas/genética , Animais , Diferenciação Celular/efeitos dos fármacos , Colforsina/farmacologia , Quinases Semelhantes a Duplacortina , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Expressão Gênica/efeitos dos fármacos , Fator de Crescimento Neural/farmacologia , Proteínas do Tecido Nervoso/metabolismo , Neuritos/efeitos dos fármacos , Células PC12 , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Serpinas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fatores de Tempo
8.
J Neurochem ; 111(2): 380-90, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19659462

RESUMO

The active transport of proteins and organelles is critical for cellular organization and function in eukaryotic cells. A substantial portion of long-distance transport depends on the opposite polarity of the kinesin and dynein family molecular motors to move cargo along microtubules. It is increasingly clear that many cargo molecules are moved bi-directionally by both sets of motors; however, the regulatory mechanism that determines the directionality of transport remains unclear. We previously reported that collapsin response mediator protein-2 (CRMP-2) played key roles in axon elongation and neuronal polarization. CRMP-2 was also found to associate with the anterograde motor protein Kinesin-1 and was transported with other cargoes toward the axon terminal. In this study, we investigated the association of CRMP-2 with a retrograde motor protein, cytoplasmic dynein. Immunoprecipitation assays showed that CRMP-2 interacted with cytoplasmic dynein heavy chain. Dynein heavy chain directly bound to the N-terminus of CRMP-2, which is the distinct side of CRMP-2's kinesin light chain-binding region. Furthermore, over-expression of the dynein-binding fragments of CRMP-2 prevented dynein-driven microtubule transport in COS-7 cells. Given that CRMP-2 is a key regulator of axon elongation, this interference with cytoplasmic dynein function by CRMP-2 might have an important role in axon formation, and neuronal development.


Assuntos
Axônios/metabolismo , Dineínas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Animais , Transporte Biológico Ativo/fisiologia , Células COS , Chlorocebus aethiops , Citoplasma/metabolismo , Dineínas/química , Cones de Crescimento/metabolismo , Hipocampo/citologia , Humanos , Microtúbulos/metabolismo , Neurônios/ultraestrutura , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA