Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Psychiatry ; 27(5): 2414-2424, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35449295

RESUMO

The lysine-63 deubiquitinase cylindromatosis (CYLD) is long recognized as a tumor suppressor in immunity and inflammation, and its loss-of-function mutations lead to familial cylindromatosis. However, recent studies reveal that CYLD is enriched in mammalian brain postsynaptic densities, and a gain-of-function mutation causes frontotemporal dementia (FTD), suggesting critical roles at excitatory synapses. Here we report that CYLD drives synapse elimination and weakening by acting on the Akt-mTOR-autophagy axis. Mice lacking CYLD display abnormal sociability, anxiety- and depression-like behaviors, and cognitive inflexibility. These behavioral impairments are accompanied by excessive synapse numbers, increased postsynaptic efficacy, augmented synaptic summation, and impaired NMDA receptor-dependent hippocampal long-term depression (LTD). Exogenous expression of CYLD results in removal of established dendritic spines from mature neurons in a deubiquitinase activity-dependent manner. In search of underlying molecular mechanisms, we find that CYLD knockout mice display marked overactivation of Akt and mTOR and reduced autophagic flux, and conversely, CYLD overexpression potently suppresses Akt and mTOR activity and promotes autophagy. Consequently, abrogating the Akt-mTOR-autophagy signaling pathway abolishes CYLD-induced spine loss, whereas enhancing autophagy in vivo by the mTOR inhibitor rapamycin rescues the synaptic pruning and LTD deficits in mutant mice. Our findings establish CYLD, via Akt-mTOR signaling, as a synaptic autophagy activator that exerts critical modulations on synapse maintenance, function, and plasticity.


Assuntos
Macroautofagia , Proteínas Proto-Oncogênicas c-akt , Animais , Enzimas Desubiquitinantes/metabolismo , Mamíferos/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Sinapses/metabolismo , Serina-Treonina Quinases TOR/metabolismo
2.
J Int Med Res ; 48(5): 300060520925325, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32459108

RESUMO

OBJECTIVE: We investigated the "BURP" maneuver's effect on the association between difficult laryngoscopy and difficult intubation, and predictors of a difficult airway. METHODS: Adult patients who underwent general anesthesia and tracheal intubation from September 2016 to May 2018 were included. The "BURP" maneuver was performed when glottic exposure was classified as Cormack-Lehane grade 3 or 4, suggesting difficult laryngoscopy. The thyromental distance, modified Mallampati score, and interincisor distance were assessed before anesthesia. RESULTS: Among this study's 2028 patients, the "BURP" maneuver decreased difficult laryngoscopies from 428 (21.1%) to 124 (6.1%) cases and increased the difficult intubation to difficult laryngoscopy ratio from 53/428 (12.4%) to 52/124 (41.9%). For laryngoscopies classified as difficult without the "BURP" maneuver, the area under the curve (AUC) of the thyromental distance, modified Mallampati score, and interincisor distance was 0.60, 0.57, and 0.66, respectively. In difficult laryngoscopies using the "BURP" maneuver, the AUC of the thyromental distance, modified Mallampati score, and interincisor distance was 0.71, 0.67, and 0.76, respectively. CONCLUSIONS: The "BURP" maneuver improves the laryngoscopic view and assists in difficult laryngoscopies. Compared with difficult laryngoscopies without the "BURP" maneuver, those with the "BURP" maneuver are more closely associated with difficult intubations and are more predictable. Trial registration: www.chictr.org.cn identifier: ChiCTR-ROC- 16009050.


Assuntos
Anestesia Geral/métodos , Glote/diagnóstico por imagem , Intubação Intratraqueal/métodos , Laringoscopia/métodos , Adulto , Idoso , Idoso de 80 Anos ou mais , Anestesia Geral/instrumentação , Feminino , Humanos , Intubação Intratraqueal/instrumentação , Laringoscopia/instrumentação , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Resultado do Tratamento , Adulto Jovem
3.
Brain Res ; 1727: 146569, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31783001

RESUMO

Ubiquitination and its reverse process, deubiquitination, play essential roles in neural development, function, and plasticity. A20, a ubiquitin editing enzyme that can remove K63-polyubiquitin chains from substrates and attach K48-polyubiquitin chains to them, is a critical component in the NF-κB signaling pathway in the immune system. This dual ubiquitin enzyme is also present in mammalian brains, but its potential role in neurons and synapses is unknown. We show that A20 in pyramidal neurons potently regulates dendritic arborization, spine morphogenesis, and synaptic transmission through an NF-κB-dependent mechanism. In cultured hippocampal neurons, overexpression of A20 reduced dendritic complexity and spine size and density, whereas A20 knockdown increased spine size and density, as well as clustering of the postsynaptic scaffold PSD-95 and glutamate receptor subunit GluA1. A20 effects in vitro were recapitulated in vivo where increasing or decreasing A20 expression in mouse brains reduced and enhanced spine density, respectively. Functionally, A20 knockdown significantly increased the amplitude, but not frequency of miniature excitatory postsynaptic currents, suggesting a role in postsynaptic efficacy. A20 negatively regulated NF-κB activation in neurons and A20 mutants deficient in either the deubiquitinase or the ubiquitin ligase activity failed to suppress NF-κB activation or reduce spine morphogenesis. Finally, selective inhibition of NF-κB abolished A20 knockdown-elicited spine formation, suggesting that A20 exerts its modulation on synapses through NF-κB signaling. Together, our study reveals a previously unknown role for A20, the only known ubiquitin editing enzyme with both deubiquitinase and ubiquitin ligase activity, in dendritic arborization, spine remodeling, and synaptic plasticity.


Assuntos
Células Piramidais/fisiologia , Sinapses/fisiologia , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/fisiologia , Animais , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Células Piramidais/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/administração & dosagem
4.
Proc Natl Acad Sci U S A ; 114(41): E8760-E8769, 2017 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-28973854

RESUMO

Ubiquitination-directed proteasomal degradation of synaptic proteins, presumably mediated by lysine 48 (K48) of ubiquitin, is a key mechanism in synapse and neural circuit remodeling. However, more than half of polyubiquitin (polyUb) species in the mammalian brain are estimated to be non-K48; among them, the most abundant is Lys 63 (K63)-linked polyUb chains that do not tag substrates for degradation but rather modify their properties and activity. Virtually nothing is known about the role of these nonproteolytic polyUb chains at the synapse. Here we report that K63-polyUb chains play a significant role in postsynaptic protein scaffolding and synaptic strength and plasticity. We found that the postsynaptic scaffold PSD-95 (postsynaptic density protein 95) undergoes K63 polyubiquitination, which markedly modifies PSD-95's scaffolding potentials, enables its synaptic targeting, and promotes synapse maturation and efficacy. TNF receptor-associated factor 6 (TRAF6) is identified as a direct E3 ligase for PSD-95, which, together with the E2 complex Ubc13/Uev1a, assembles K63-chains on PSD-95. In contrast, CYLD (cylindromatosis tumor-suppressor protein), a K63-specific deubiquitinase enriched in postsynaptic densities, cleaves K63-chains from PSD-95. We found that neuronal activity exerts potent control of global and synaptic K63-polyUb levels and, through NMDA receptors, drives rapid, CYLD-mediated PSD-95 deubiquitination, mobilizing and depleting PSD-95 from synapses. Silencing CYLD in hippocampal neurons abolishes NMDA-induced chemical long-term depression. Our results unveil a previously unsuspected role for nonproteolytic polyUb chains in the synapse and illustrate a mechanism by which a PSD-associated K63-linkage-specific ubiquitin machinery acts on a major postsynaptic scaffold to regulate synapse organization, function, and plasticity.


Assuntos
Proteína 4 Homóloga a Disks-Large/fisiologia , Hipocampo/fisiologia , Neurônios/fisiologia , Poliubiquitina/metabolismo , Densidade Pós-Sináptica , Complexo de Endopeptidases do Proteassoma/metabolismo , Sinapses/fisiologia , Animais , Hipocampo/citologia , Lisina , Camundongos , Camundongos Knockout , Neurônios/citologia , Ubiquitinação
5.
PLoS One ; 12(9): e0183854, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28886095

RESUMO

We previously reported a 84-Kb hemi-deletion copy number variant at the SLC1A1 gene locus that reduces its expression and appeared causally linked to schizophrenia. In this report, we characterize the in vivo and in vitro consequences of reduced expression of Slc1a1 in mice. Heterozygous (HET) Slc1a1+/- mice, which more closely model the hemi-deletion we found in human subjects, were examined in a series of behavioral, anatomical and biochemical assays. Knockout (KO) mice were also included in the behavioral studies for comparative purposes. Both HET and KO mice exhibited evidence of increased anxiety-like behavior, impaired working memory, decreased exploratory activity and impaired sensorimotor gating, but no changes in overall locomotor activity. The magnitude of changes was approximately equivalent in the HET and KO mice suggesting a dominant effect of the haploinsufficiency. Behavioral changes in the HET mice were accompanied by reduced thickness of the dorsomedial prefrontal cortex. Whole transcriptome RNA-Seq analysis detected expression changes of genes and pathways involved in cytokine signaling and synaptic functions in both brain and blood. Moreover, the brains of Slc1a1+/- mice displayed elevated levels of oxidized glutathione, a trend for increased oxidative DNA damage, and significantly increased levels of cytokines. This latter finding was further supported by SLC1A1 knockdown and overexpression studies in differentiated human neuroblastoma cells, which led to decreased or increased cytokine expression, respectively. Taken together, our results suggest that partial loss of the Slc1a1 gene in mice causes haploinsufficiency associated with behavioral, histological and biochemical changes that reflect an altered redox state and may promote the expression of behavioral features and inflammatory states consistent with those observed in schizophrenia.


Assuntos
Cognição , Transportador 3 de Aminoácido Excitatório/genética , Regulação da Expressão Gênica , Inflamação/genética , Esquizofrenia/genética , Psicologia do Esquizofrênico , Filtro Sensorial/genética , Animais , Ansiedade/genética , Apoptose , Comportamento Animal , Encéfalo/metabolismo , Encéfalo/patologia , Citocinas/metabolismo , Dano ao DNA , Modelos Animais de Doenças , Feminino , Redes Reguladoras de Genes , Genótipo , Glutationa/metabolismo , Haploinsuficiência/genética , Hipocampo/metabolismo , Hipocampo/patologia , Inflamação/imunologia , Inflamação/metabolismo , Locomoção/genética , Masculino , Camundongos , Camundongos Knockout , Oxirredução , Estresse Oxidativo , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/patologia , Esquizofrenia/imunologia , Esquizofrenia/metabolismo , Sinapses/metabolismo
6.
Neuropsychopharmacology ; 41(13): 3103-3113, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27485686

RESUMO

Lysine (K) methyltransferase 2a (Kmt2a) and other regulators of H3 lysine 4 methylation, a histone modification enriched at promoters and enhancers, are widely expressed throughout the brain, but molecular and cellular phenotypes in subcortical areas remain poorly explored. We report that Kmt2a conditional deletion in postnatal forebrain is associated with excessive nocturnal activity and with absent or blunted responses to stimulant and dopaminergic agonist drugs, in conjunction with near-complete loss of spike-timing-dependent long-term potentiation in medium spiny neurons (MSNs). Selective ablation of Kmt2a, but not the ortholog Kmt2b, in adult ventral striatum/nucleus accumbens neurons markedly increased anxiety scores in multiple behavioral paradigms. Striatal transcriptome sequencing in adult mutants identified 262 Kmt2a-sensitive genes, mostly downregulated in Kmt2a-deficient mice. Transcriptional repression includes the 5-Htr2a serotonin receptor, strongly associated with anxiety- and depression-related disorders in human and animal models. Consistent with the role of Kmt2a in promoting gene expression, the transcriptional regulators Bahcc1, Isl1, and Sp9 were downregulated and affected by H3K4 promoter hypomethylation. Therefore, Kmt2a regulates synaptic plasticity in striatal neurons and provides an epigenetic drug target for anxiety and dopamine-mediated behaviors.


Assuntos
Potenciais de Ação/genética , Ansiedade , Dopaminérgicos/farmacologia , Histona-Lisina N-Metiltransferase/deficiência , Proteína de Leucina Linfoide-Mieloide/deficiência , Plasticidade Neuronal/genética , Neurônios/fisiologia , Estriado Ventral/citologia , Potenciais de Ação/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Ansiedade/tratamento farmacológico , Ansiedade/genética , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Ritmo Circadiano/efeitos dos fármacos , Ritmo Circadiano/genética , Modelos Animais de Doenças , Feminino , Histona-Lisina N-Metiltransferase/genética , Locomoção/efeitos dos fármacos , Locomoção/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Análise de Sequência com Séries de Oligonucleotídeos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
7.
J Neurosci ; 35(13): 5097-108, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25834037

RESUMO

Neuronal histone H3-lysine 4 methylation landscapes are defined by sharp peaks at gene promoters and other cis-regulatory sequences, but molecular and cellular phenotypes after neuron-specific deletion of H3K4 methyl-regulators remain largely unexplored. We report that neuronal ablation of the H3K4-specific methyltransferase, Kmt2a/Mixed-lineage leukemia 1 (Mll1), in mouse postnatal forebrain and adult prefrontal cortex (PFC) is associated with increased anxiety and robust cognitive deficits without locomotor dysfunction. In contrast, only mild behavioral phenotypes were observed after ablation of the Mll1 ortholog Kmt2b/Mll2 in PFC. Impaired working memory after Kmt2a/Mll1 ablation in PFC neurons was associated with loss of training-induced transient waves of Arc immediate early gene expression critical for synaptic plasticity. Medial prefrontal layer V pyramidal neurons, a major output relay of the cortex, demonstrated severely impaired synaptic facilitation and temporal summation, two forms of short-term plasticity essential for working memory. Chromatin immunoprecipitation followed by deep sequencing in Mll1-deficient cortical neurons revealed downregulated expression and loss of the transcriptional mark, trimethyl-H3K4, at <50 loci, including the homeodomain transcription factor Meis2. Small RNA-mediated Meis2 knockdown in PFC was associated with working memory defects similar to those elicited by Mll1 deletion. Therefore, mature prefrontal neurons critically depend on maintenance of Mll1-regulated H3K4 methylation at a subset of genes with an essential role in cognition and emotion.


Assuntos
Histona-Lisina N-Metiltransferase/metabolismo , Memória de Curto Prazo/fisiologia , Proteína de Leucina Linfoide-Mieloide/metabolismo , Plasticidade Neuronal/fisiologia , Córtex Pré-Frontal/fisiologia , Animais , Comportamento Animal/fisiologia , Proteínas do Citoesqueleto/metabolismo , Expressão Gênica , Técnicas de Silenciamento de Genes , Proteínas de Homeodomínio/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Masculino , Metilação , Camundongos , Camundongos Transgênicos , Mutação , Proteínas do Tecido Nervoso/metabolismo , Prosencéfalo/fisiologia , Células Piramidais/fisiologia
8.
J Neurogenet ; 28(1-2): 98-111, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24702501

RESUMO

Excessive activation of the N-methyl-d-aspartate (NMDA) receptor and the neurotransmitter dopamine (DA) mediate neurotoxicity and neurodegeneration under many neurological conditions, including Huntington's disease (HD), an autosomal dominant neurodegenerative disease characterized by the preferential loss of medium spiny projection neurons (MSNs) in the striatum. PSD-95 is a major scaffolding protein in the postsynaptic density (PSD) of dendritic spines, where a classical role for PSD-95 is to stabilize glutamate receptors at sites of synaptic transmission. Our recent studies indicate that PSD-95 also interacts with the D1 DA receptor localized in spines and negatively regulates spine D1 signaling. Moreover, PSD-95 forms ternary protein complexes with D1 and NMDA receptors, and plays a role in limiting the reciprocal potentiation between both receptors from being escalated. These studies suggest a neuroprotective role for PSD-95. Here we show that mice lacking PSD-95, resulting from genetic deletion of the GK domain of PSD-95 (PSD-95-ΔGK mice), sporadically develop progressive neurological impairments characterized by hypolocomotion, limb clasping, and loss of DARPP-32-positive MSNs. Electrophysiological experiments indicated that NMDA receptors in mutant MSNs were overactive, suggested by larger, NMDA receptor-mediated miniature excitatory postsynaptic currents (EPSCs) and higher ratios of NMDA- to AMPA-mediated corticostriatal synaptic transmission. In addition, NMDA receptor currents in mutant cortical neurons were more sensitive to potentiation by the D1 receptor agonist SKF81297. Finally, repeated administration of the psychostimulant cocaine at a dose regimen not producing overt toxicity-related phenotypes in normal mice reliably converted asymptomatic mutant mice to clasping symptomatic mice. These results support the hypothesis that deletion of PSD-95 in mutant mice produces concomitant overactivation of both D1 and NMDA receptors that makes neurons more susceptible to NMDA excitotoxicity, causing neuronal damage and neurological impairments. Understanding PSD-95-dependent neuroprotective mechanisms may help elucidate processes underlying neurodegeneration in HD and other neurological disorders.


Assuntos
Corpo Estriado/patologia , Dopamina/metabolismo , Ácido Glutâmico/metabolismo , Guanilato Quinases/deficiência , Proteínas de Membrana/deficiência , Transtornos dos Movimentos/genética , Doenças Neurodegenerativas/genética , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Fatores Etários , Animais , Benzazepinas/farmacologia , Contagem de Células , Proteína 4 Homóloga a Disks-Large , Agonistas de Dopamina/farmacologia , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , Regulação da Expressão Gênica/genética , Guanilato Quinases/genética , Magnésio/farmacologia , Potenciais da Membrana/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Atividade Motora/genética , Doenças Neurodegenerativas/patologia , Neurônios/fisiologia
9.
J Neurogenet ; 27(1-2): 43-58, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23527882

RESUMO

Postsynaptic membrane rafts are believed to play important roles in synaptic signaling, plasticity, and maintenance. We recently demonstrated the presence, at the electron microscopic level, of complexes consisting of membrane rafts and postsynaptic densities (PSDs) in detergent-resistant membranes (DRMs) prepared from synaptic plasma membranes (SPMs) ( Suzuki et al., 2011 , J Neurochem, 119, 64-77). To further explore these complexes, here we investigated the nature of the binding between purified SPM-DRMs and PSDs in vitro. In binding experiments, we used SPM-DRMs prepared after treating SPMs with n-octyl-ß-d-glucoside, because at concentrations of 1.0% or higher it completely separates SPM-DRMs and PSDs, providing substantially PSD-free unique SPM-DRMs as well as DRM-free PSDs. PSD binding to PSD-free DRMs was identified by mass spectrometry, Western blotting, and electron microscopy. PSD proteins were not incorporated into SPMs, and significantly less PSD proteins were incorporated into DRMs prepared from liver membranes, providing in vitro evidence that binding of PSDs to DRMs is specific and suggestion of the presence of specific interacting molecules. These specific interactions may have important roles in synaptic development, function, and plasticity in vivo. In addition, the binding system we developed may be a good tool to search for binding molecules and binding mechanisms between PSDs and rafts.


Assuntos
Microdomínios da Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Densidade Pós-Sináptica/metabolismo , Membranas Sinápticas/metabolismo , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Encéfalo/citologia , Encéfalo/ultraestrutura , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Colesterol/metabolismo , Gangliosídeo G(M1)/metabolismo , Masculino , Microdomínios da Membrana/ultraestrutura , Microscopia Eletrônica de Transmissão , Densidade Pós-Sináptica/ultraestrutura , Ratos , Ratos Wistar , Receptores de AMPA/metabolismo , Membranas Sinápticas/ultraestrutura
12.
J Neurosci ; 29(22): 7124-36, 2009 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-19494135

RESUMO

Here, we report that postsynaptic density protein of 95 kDa (PSD-95), a postsynaptic density scaffolding protein, classically conceptualized as being essential for the regulation of ionotropic glutamatergic signaling at the postsynaptic membrane, plays an unanticipated and essential role in mediating the actions of hallucinogens and atypical antipsychotic drugs at 5-HT(2A) and 5-HT(2C) serotonergic G-protein-coupled receptors. We show that PSD-95 is crucial for normal 5-HT(2A) and 5-HT(2C) expression in vivo and that PSD-95 maintains normal receptor expression by promoting apical dendritic targeting and stabilizing receptor turnover in vivo. Significantly, 5-HT(2A)- and 5-HT(2C)-mediated downstream signaling is impaired in PSD-95(null) mice, and the 5-HT(2A)-mediated head-twitch response is abnormal. Furthermore, the ability of 5-HT(2A) inverse agonists to normalize behavioral changes induced by glutamate receptor antagonists is abolished in the absence of PSD-95 in vivo. These results demonstrate that PSD-95, in addition to the well known role it plays in scaffolding macromolecular glutamatergic signaling complexes, profoundly modulates metabotropic 5-HT(2A) and 5-HT(2C) receptor function.


Assuntos
Antipsicóticos/farmacologia , Alucinógenos/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas de Membrana/fisiologia , Neurônios/efeitos dos fármacos , Receptores de Serotonina/metabolismo , 8-Hidroxi-2-(di-n-propilamino)tetralina/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Proteína 4 Homóloga a Disks-Large , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Guanilato Quinases , Hipotermia/induzido quimicamente , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Proteínas de Membrana/deficiência , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Ligação Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Pirazinas/farmacologia , RNA Mensageiro/metabolismo , Receptor 5-HT2A de Serotonina/genética , Receptor 5-HT2A de Serotonina/metabolismo , Receptor 5-HT2C de Serotonina/genética , Receptor 5-HT2C de Serotonina/metabolismo , Receptores de Serotonina/efeitos dos fármacos , Receptores de Serotonina/genética , Antagonistas da Serotonina/metabolismo , Antagonistas da Serotonina/farmacologia , Agonistas do Receptor de Serotonina/farmacologia , Transdução Genética
13.
J Neurosci ; 29(9): 2948-60, 2009 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-19261890

RESUMO

Classical dopaminergic signaling paradigms and emerging studies on direct physical interactions between the D(1) dopamine (DA) receptor and the NMDA glutamate receptor predict a reciprocally facilitating, positive feedback loop. This loop, if not controlled, may cause concomitant overactivation of both D(1) and NMDA receptors, triggering neurotoxicity. Endogenous protective mechanisms must exist. Here, we report that PSD-95, a prototypical structural and signaling scaffold in the postsynaptic density, inhibits D(1)-NMDA receptor subunit 1 (NR1) NMDA receptor association and uncouples NMDA receptor-dependent enhancement of D(1) signaling. This uncoupling is achieved, at least in part, via a disinhibition mechanism by which PSD-95 abolishes NMDA receptor-dependent inhibition of D(1) internalization. Knockdown of PSD-95 immobilizes D(1) receptors on the cell surface and escalates NMDA receptor-dependent D(1) cAMP signaling in neurons. Thus, in addition to its role in receptor stabilization and synaptic plasticity, PSD-95 acts as a brake on the D(1)-NMDA receptor complex and dampens the interaction between them.


Assuntos
Dopamina/fisiologia , Ácido Glutâmico/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas de Membrana/fisiologia , Receptores de Dopamina D1/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Animais , Western Blotting , Linhagem Celular , AMP Cíclico/fisiologia , Proteína 4 Homóloga a Disks-Large , Imunofluorescência , Guanilato Quinases , Humanos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Infecções por Lentivirus/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Microscopia Confocal , Plasmídeos , Ensaio Radioligante , Transdução de Sinais/fisiologia , Transfecção
18.
J Biol Chem ; 282(21): 15778-89, 2007 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-17369255

RESUMO

Dopamine D1 receptors play an important role in movement, reward, and learning and are implicated in a number of neurological and psychiatric disorders. These receptors are concentrated in dendritic spines of neurons, including the spine head and the postsynaptic density. D1 within spines is thought to modulate the local channels and receptors to control the excitability and synaptic properties of spines. The molecular mechanisms mediating D1 trafficking, anchorage, and function in spines remain elusive. Here we show that the synaptic scaffolding protein PSD-95 thought to play a role in stabilizing glutamate receptors in the postsynaptic density, interacts with D1 and regulates its trafficking and function. Interestingly, the D1-PSD-95 interaction does not require the well characterized domains of PSD-95 but is mediated by the carboxyl-terminal tail of D1 and the NH(2) terminus of PSD-95, a region that is recognized only recently to participate in protein-protein interaction. Co-expression of PSD-95 with D1 in mammalian cells inhibits the D1-mediated cAMP accumulation without altering the total expression level or the agonist binding properties of the receptor. The diminished D1 signaling is mediated by reduced D1 expression at the cell surface as a consequence of an enhanced constitutive, dynamin-dependent endocytosis. In addition, genetically engineered mice lacking PSD-95 show a heightened behavioral response to either a D1 agonist or the psychostimulant amphetamine. These studies demonstrate a role for a glutamatergic scaffold in dopamine receptor signaling and trafficking and identify a new potential target for the modulation of abnormal dopaminergic function.


Assuntos
Espinhas Dendríticas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Receptores de Dopamina D1/metabolismo , Transdução de Sinais/fisiologia , Anfetamina/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , AMP Cíclico/metabolismo , Proteína 4 Homóloga a Disks-Large , Dopaminérgicos/farmacologia , Dinaminas/metabolismo , Endocitose/efeitos dos fármacos , Endocitose/genética , Expressão Gênica , Guanilato Quinases , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Aprendizagem/efeitos dos fármacos , Locomoção/efeitos dos fármacos , Locomoção/genética , Proteínas de Membrana/genética , Transtornos Mentais/genética , Transtornos Mentais/metabolismo , Camundongos , Camundongos Knockout , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Estrutura Terciária de Proteína/genética , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/antagonistas & inibidores , Receptores de Dopamina D1/genética , Receptores de Glutamato/metabolismo
19.
J Pharmacol Exp Ther ; 321(1): 116-27, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17234900

RESUMO

Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor that directly responds to endogenous monoamines as well as amphetamine-related psychostimulants, including methamphetamine. In the present study, we demonstrate TAAR1 mRNA and protein expression in rhesus monkey brain regions associated with monoaminergic systems, variable cellular distribution of TAAR1 in rhesus monkey brain, and TAAR1 coexpression with the dopamine transporter (DAT) in a subset of dopamine neurons in both rhesus monkey and mouse substantia nigra. On this basis, we evaluated rhesus monkey TAAR1 activation by different compounds and its functional relation with monoamine transporters and the dopamine D2 receptor (D2) short isoform (D2s) autoreceptor in vitro using a cAMP response element-luciferase assay. TAAR1 activation by monoamines and amphetamine-related compounds was greatly enhanced by coexpression of dopamine, norepinephrine, or serotonin transporters, and the activation enhancement was blocked by monoamine transporter inhibitors. This enhancement did not occur in control experiments in which the dopamine D1 receptor (D1) was substituted for TAAR1. Furthermore, activation of TAAR1 by dopamine was completely inhibited by D2s when coexpressed with TAAR1, and this inhibition was blocked by the D2 antagonist raclopride. Last, dopamine activation of TAAR1 could induce c-FOS-luciferase expression but only in the presence of DAT, whereas dopamine activation of D1 resulted in equivalent c-FOS expression in the presence or absence of DAT. Together, these data reveal a broad agonist spectrum for TAAR1, a functional relation of TAAR1 with monoamine transporters and D2s, and a mechanism by which D2 receptor drugs can influence brain monoaminergic function and have efficacy through affecting TAAR1 signaling.


Assuntos
Autorreceptores/fisiologia , Proteínas da Membrana Plasmática de Transporte de Dopamina/fisiologia , Receptores de Dopamina D2/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Transdução de Sinais/fisiologia , Anfetamina/farmacologia , Animais , Western Blotting , Células Cultivadas , Estimulantes do Sistema Nervoso Central/farmacologia , Eletroforese em Gel de Poliacrilamida , Imunofluorescência , Genes Reporter/fisiologia , Imuno-Histoquímica , Luciferases/metabolismo , Macaca mulatta , Neurônios/fisiologia , Proteínas Proto-Oncogênicas c-fos/fisiologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ensaio Radioligante , Receptores de Dopamina D1/fisiologia , Receptores Acoplados a Proteínas G/biossíntese , Receptores Acoplados a Proteínas G/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Substância Negra/citologia , Substância Negra/fisiologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA