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.
Nicotine Tob Res ; 22(8): 1310-1315, 2020 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-31930296

RESUMO

INTRODUCTION: Smoking is a leading cause of death, and genetic variation contributes to smoking behaviors. Identifying genes and sets of genes that contribute to risk for addiction is necessary to prioritize targets for functional characterization and for personalized medicine. METHODS: We performed a gene set-based association and heritable enrichment study of two addiction-related gene sets, those on the Smokescreen Genotyping Array and the nicotinic acetylcholine receptors, using the largest available GWAS summary statistics. We assessed smoking initiation, cigarettes per day, smoking cessation, and age of smoking initiation. RESULTS: Individual genes within each gene set were significantly associated with smoking behaviors. Both sets of genes were significantly associated with cigarettes per day, smoking initiation, and smoking cessation. Age of initiation was only associated with the Smokescreen gene set. Although both sets of genes were enriched for trait heritability, each accounts for only a small proportion of the single nucleotide polymorphism-based heritability (2%-12%). CONCLUSIONS: These two gene sets are associated with smoking behaviors, but collectively account for a limited amount of the genetic and phenotypic variation of these complex traits, consistent with high polygenicity. IMPLICATIONS: We evaluated evidence for the association and heritable contribution of expert-curated and bioinformatically identified sets of genes related to smoking. Although they impact smoking behaviors, these specifically targeted genes do not account for much of the heritability in smoking and will be of limited use for predictive purposes. Advanced genome-wide approaches and integration of other 'omics data will be needed to fully account for the genetic variation in smoking phenotypes.


Assuntos
Comportamento Aditivo/genética , Marcadores Genéticos , Estudo de Associação Genômica Ampla , Polimorfismo de Nucleotídeo Único , Receptores Nicotínicos/genética , Fumar/genética , Idade de Início , Comportamento Aditivo/epidemiologia , Comportamento Aditivo/psicologia , Colorado/epidemiologia , Humanos , Fenótipo , Fumar/epidemiologia , Fumar/psicologia
2.
Elife ; 62017 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-29173281

RESUMO

AKT is a kinase regulating numerous cellular processes in the brain, and mutations in AKT are known to affect brain function. AKT is indirectly implicated in synaptic plasticity, but its direct role has not been studied. Moreover, three highly related AKT isoforms are expressed in the brain, but their individual roles are poorly understood. We find in Mus musculus, each AKT isoform has a unique expression pattern in the hippocampus, with AKT1 and AKT3 primarily in neurons but displaying local differences, while AKT2 is in astrocytes. We also find isoform-specific roles for AKT in multiple paradigms of hippocampal synaptic plasticity in area CA1. AKT1, but not AKT2 or AKT3, is required for L-LTP through regulating activity-induced protein synthesis. Interestingly, AKT activity inhibits mGluR-LTD, with overlapping functions for AKT1 and AKT3. In summary, our studies identify distinct expression patterns and roles in synaptic plasticity for AKT isoforms in the hippocampus.


Assuntos
Expressão Gênica , Hipocampo/fisiologia , Potenciação de Longa Duração , Plasticidade Neuronal , Proteínas Proto-Oncogênicas c-akt/biossíntese , Animais , Astrócitos/enzimologia , Astrócitos/fisiologia , Perfilação da Expressão Gênica , Hipocampo/enzimologia , Camundongos , Neurônios/enzimologia , Neurônios/fisiologia , Isoformas de Proteínas/biossíntese
3.
Nat Med ; 23(3): 347-354, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28112735

RESUMO

The prefrontal cortex (PFC) underlies higher cognitive processes that are modulated by nicotinic acetylcholine receptor (nAChR) activation by cholinergic inputs. PFC spontaneous default activity is altered in neuropsychiatric disorders, including schizophrenia-a disorder that can be accompanied by heavy smoking. Recently, genome-wide association studies (GWAS) identified single-nucleotide polymorphisms (SNPs) in the human CHRNA5 gene, encoding the α5 nAChR subunit, that increase the risks for both smoking and schizophrenia. Mice with altered nAChR gene function exhibit PFC-dependent behavioral deficits, but it is unknown how the corresponding human polymorphisms alter the cellular and circuit mechanisms underlying behavior. Here we show that mice expressing a human α5 SNP exhibit neurocognitive behavioral deficits in social interaction and sensorimotor gating tasks. Two-photon calcium imaging in awake mouse models showed that nicotine can differentially influence PFC pyramidal cell activity by nAChR modulation of layer II/III hierarchical inhibitory circuits. In α5-SNP-expressing and α5-knockout mice, lower activity of vasoactive intestinal polypeptide (VIP) interneurons resulted in an increased somatostatin (SOM) interneuron inhibitory drive over layer II/III pyramidal neurons. The decreased activity observed in α5-SNP-expressing mice resembles the hypofrontality observed in patients with psychiatric disorders, including schizophrenia and addiction. Chronic nicotine administration reversed this hypofrontality, suggesting that administration of nicotine may represent a therapeutic strategy for the treatment of schizophrenia, and a physiological basis for the tendency of patients with schizophrenia to self-medicate by smoking.


Assuntos
Comportamento Animal/efeitos dos fármacos , Inibição Neural/efeitos dos fármacos , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Comportamento Social , Animais , Sistemas CRISPR-Cas , Modelos Animais de Doenças , Imunofluorescência , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Polimorfismo de Nucleotídeo Único , Córtex Pré-Frontal/fisiopatologia , Inibição Pré-Pulso/efeitos dos fármacos , Receptores Adrenérgicos beta 2/genética , Receptores Nicotínicos/genética , Reflexo de Sobressalto/efeitos dos fármacos , Esquizofrenia/genética , Tabagismo/genética , Receptor Nicotínico de Acetilcolina alfa7/genética
4.
Sci Signal ; 7(308): ra4, 2014 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-24425786

RESUMO

Although antipsychotic drugs can reduce psychotic behavior within a few hours, full efficacy is not achieved for several weeks, implying that there may be rapid, short-term changes in neuronal function, which are consolidated into long-lasting changes. We showed that the antipsychotic drug haloperidol, a dopamine receptor type 2 (D2R) antagonist, stimulated the kinase Akt to activate the mRNA translation pathway mediated by the mammalian target of rapamycin complex 1 (mTORC1). In primary striatal D2R-positive neurons, haloperidol-mediated activation of mTORC1 resulted in increased phosphorylation of ribosomal protein S6 (S6) and eukaryotic translation initiation factor 4E-binding protein (4E-BP). Proteomic mass spectrometry revealed marked changes in the pattern of protein synthesis after acute exposure of cultured striatal neurons to haloperidol, including increased abundance of cytoskeletal proteins and proteins associated with translation machinery. These proteomic changes coincided with increased morphological complexity of neurons that was diminished by inhibition of downstream effectors of mTORC1, suggesting that mTORC1-dependent translation enhances neuronal complexity in response to haloperidol. In vivo, we observed rapid morphological changes with a concomitant increase in the abundance of cytoskeletal proteins in cortical neurons of haloperidol-injected mice. These results suggest a mechanism for both the acute and long-term actions of antipsychotics.


Assuntos
Antipsicóticos/farmacologia , Complexos Multiproteicos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Serina-Treonina Quinases TOR/efeitos dos fármacos , Animais , Células Cultivadas , Haloperidol/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Complexos Multiproteicos/metabolismo , Neurônios/citologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo
5.
J Neurosci ; 31(15): 5589-95, 2011 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-21490199

RESUMO

Generation of reactive oxygen species (ROS) causes cellular oxidative damage and has been implicated in the etiology of Alzheimer's disease (AD). In contrast, multiple lines of evidence indicate that ROS can normally modulate long-term potentiation (LTP), a cellular model for memory formation. We recently showed that decreasing the level of superoxide through the overexpression of mitochondrial superoxide dismutase (SOD-2) prevents memory deficits in the Tg2576 mouse model of AD. In the current study, we explored whether AD-related LTP impairments could be prevented when ROS generation from mitochondria was diminished either pharmacologically or via genetic manipulation. In wild-type hippocampal slices treated with exogenous amyloid ß peptide (Aß1-42) and in slices from APP/PS1 mutant mice that model AD, LTP was impaired. The LTP impairments were prevented by MitoQ, a mitochondria-targeted antioxidant, and EUK134, an SOD and catalase mimetic. In contrast, inhibition of NADPH oxidase either by diphenyliodonium (DPI) or by genetically deleting gp91(phox), the key enzymatic component of NADPH oxidase, had no effect on Aß-induced LTP blockade. Moreover, live staining with MitoSOX Red, a mitochondrial superoxide indicator, combined with confocal microscopy, revealed that Aß-induced superoxide production could be blunted by MitoQ, but not DPI, in agreement with our electrophysiological findings. Finally, in transgenic mice overexpressing SOD-2, Aß-induced LTP impairments and superoxide generation were prevented. Our data suggest a causal relationship between mitochondrial ROS imbalance and Aß-induced impairments in hippocampal synaptic plasticity.


Assuntos
Peptídeos beta-Amiloides/fisiologia , Hipocampo/efeitos dos fármacos , Mitocôndrias/metabolismo , Plasticidade Neuronal/efeitos dos fármacos , Oxidantes/metabolismo , Superóxidos/metabolismo , Sinapses/efeitos dos fármacos , Peptídeos beta-Amiloides/genética , Animais , Antioxidantes/farmacologia , Compostos de Bifenilo/farmacologia , Fenômenos Eletrofisiológicos , Humanos , Técnicas In Vitro , Potenciação de Longa Duração/efeitos dos fármacos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , NADPH Oxidase 2 , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , NADPH Oxidases/fisiologia , Oniocompostos/farmacologia , Compostos Organofosforados/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/farmacologia
6.
J Neurosci ; 30(2): 694-702, 2010 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-20071534

RESUMO

Fragile X syndrome, the most common form of inherited mental retardation and leading genetic cause of autism, is caused by transcriptional silencing of the Fmr1 gene. The fragile X mental retardation protein (FMRP), the gene product of Fmr1, is an RNA binding protein that negatively regulates translation in neurons. The Fmr1 knock-out mouse, a model of fragile X syndrome, exhibits cognitive deficits and exaggerated metabotropic glutamate receptor (mGluR)-dependent long-term depression at CA1 synapses. However, the molecular mechanisms that link loss of function of FMRP to aberrant synaptic plasticity remain unclear. The mammalian target of rapamycin (mTOR) signaling cascade controls initiation of cap-dependent translation and is under control of mGluRs. Here we show that mTOR phosphorylation and activity are elevated in hippocampus of juvenile Fmr1 knock-out mice by four functional readouts: (1) association of mTOR with regulatory associated protein of mTOR; (2) mTOR kinase activity; (3) phosphorylation of mTOR downstream targets S6 kinase and 4E-binding protein; and (4) formation of eukaryotic initiation factor complex 4F, a critical first step in cap-dependent translation. Consistent with this, mGluR long-term depression at CA1 synapses of FMRP-deficient mice is exaggerated and rapamycin insensitive. We further show that the p110 subunit of the upstream kinase phosphatidylinositol 3-kinase (PI3K) and its upstream activator PI3K enhancer PIKE, predicted targets of FMRP, are upregulated in knock-out mice. Elevated mTOR signaling may provide a functional link between overactivation of group I mGluRs and aberrant synaptic plasticity in the fragile X mouse, mechanisms relevant to impaired cognition in fragile X syndrome.


Assuntos
Síndrome do Cromossomo X Frágil/complicações , Síndrome do Cromossomo X Frágil/metabolismo , Transdução de Sinais/fisiologia , Sirolimo/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Região CA1 Hipocampal/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Transtornos Cognitivos/etiologia , Modelos Animais de Doenças , Fator de Iniciação 4A em Eucariotos/genética , Fator de Iniciação 4A em Eucariotos/metabolismo , Fatores de Iniciação em Eucariotos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Imunoprecipitação/métodos , Técnicas In Vitro , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Depressão Sináptica de Longo Prazo/genética , Metoxi-Hidroxifenilglicol/análogos & derivados , Metoxi-Hidroxifenilglicol/farmacologia , Camundongos , Camundongos Knockout , Proteína Oncogênica v-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação/genética , Receptores de Glutamato Metabotrópico/metabolismo , Serina/metabolismo , Transdução de Sinais/genética
7.
Mol Cell Biol ; 26(15): 5908-20, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16847341

RESUMO

Reactive oxygen species (ROS) are required in a number of critical cellular signaling events, including those underlying hippocampal synaptic plasticity and hippocampus-dependent memory; however, the source of ROS is unknown. We previously have shown that NADPH oxidase is required for N-methyl-D-aspartate (NMDA) receptor-dependent signal transduction in the hippocampus, suggesting that NADPH oxidase may be required for NMDA receptor-dependent long-term potentiation (LTP) and hippocampus-dependent memory. Herein we present the first evidence that NADPH oxidase is involved in hippocampal synaptic plasticity and memory. We have found that pharmacological inhibitors of NADPH oxidase block LTP. Moreover, mice that lack the NADPH oxidase proteins gp91(phox) and p47(phox), both of which are mouse models of human chronic granulomatous disease (CGD), also lack LTP. We also found that the gp91(phox) and p47(phox) mutant mice have mild impairments in hippocampus-dependent memory. The gp91(phox) mutant mice exhibited a spatial memory deficit in the Morris water maze, and the p47(phox) mutant mice exhibited impaired context-dependent fear memory. Taken together, our results are consistent with NADPH oxidase being required for hippocampal synaptic plasticity and memory and are consistent with reports of cognitive dysfunction in patients with CGD.


Assuntos
Doença Granulomatosa Crônica/fisiopatologia , Potenciação de Longa Duração/fisiologia , Memória/fisiologia , Plasticidade Neuronal/fisiologia , Sinapses/fisiologia , Animais , Condicionamento Operante , Modelos Animais de Doenças , Medo , Feminino , Hipocampo/anatomia & histologia , Hipocampo/fisiologia , Humanos , Masculino , Aprendizagem em Labirinto , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Oxidase 2 , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
8.
Nature ; 416(6883): 870-4, 2002 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-11976688

RESUMO

Activity-regulated gene expression mediates many aspects of neural plasticity, including long-term memory. In the prevailing view, patterned synaptic activity causes kinase-mediated activation of the transcription factor cyclic AMP response-element-binding protein, CREB. Together with appropriate cofactors, CREB then transcriptionally induces a group of 'immediate early' transcription factors and, eventually, effector proteins that establish or consolidate synaptic change. Here, using a Drosophila model synapse, we analyse cellular functions and regulation of the best known immediate early transcription factor, AP-1; a heterodimer of the basic leucine zipper proteins Fos and Jun. We observe that AP-1 positively regulates both synaptic strength and synapse number, thus showing a greater range of influence than CREB. Observations from genetic epistasis and RNA quantification experiments indicate that AP-1 acts upstream of CREB, regulates levels of CREB messenger RNA, and functions at the top of the hierarchy of transcription factors known to regulate long-term plasticity. A Jun-kinase signalling module provides a CREB-independent route for neuronal AP-1 activation; thus, CREB regulation of AP-1 expression may, in some neurons, constitute a positive feedback loop rather than the primary step in AP-1 activation.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Plasticidade Neuronal , Sinapses/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Contagem de Células , AMP Cíclico/metabolismo , Dimerização , Drosophila melanogaster/crescimento & desenvolvimento , Eletrofisiologia , Epistasia Genética , Proteínas Quinases JNK Ativadas por Mitógeno , Larva/genética , Larva/crescimento & desenvolvimento , Larva/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Junção Neuromuscular/citologia , Junção Neuromuscular/enzimologia , Junção Neuromuscular/crescimento & desenvolvimento , Junção Neuromuscular/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Sinapses/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA