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

Base de dados
Tipo de documento
Assunto da revista
País de afiliação
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33443211

RESUMO

Hippocampal synaptic plasticity is important for learning and memory formation. Homeostatic synaptic plasticity is a specific form of synaptic plasticity that is induced upon prolonged changes in neuronal activity to maintain network homeostasis. While astrocytes are important regulators of synaptic transmission and plasticity, it is largely unclear how they interact with neurons to regulate synaptic plasticity at the circuit level. Here, we show that neuronal activity blockade selectively increases the expression and secretion of IL-33 (interleukin-33) by astrocytes in the hippocampal cornu ammonis 1 (CA1) subregion. This IL-33 stimulates an increase in excitatory synapses and neurotransmission through the activation of neuronal IL-33 receptor complex and synaptic recruitment of the scaffold protein PSD-95. We found that acute administration of tetrodotoxin in hippocampal slices or inhibition of hippocampal CA1 excitatory neurons by optogenetic manipulation increases IL-33 expression in CA1 astrocytes. Furthermore, IL-33 administration in vivo promotes the formation of functional excitatory synapses in hippocampal CA1 neurons, whereas conditional knockout of IL-33 in CA1 astrocytes decreases the number of excitatory synapses therein. Importantly, blockade of IL-33 and its receptor signaling in vivo by intracerebroventricular administration of its decoy receptor inhibits homeostatic synaptic plasticity in CA1 pyramidal neurons and impairs spatial memory formation in mice. These results collectively reveal an important role of astrocytic IL-33 in mediating the negative-feedback signaling mechanism in homeostatic synaptic plasticity, providing insights into how astrocytes maintain hippocampal network homeostasis.


Assuntos
Astrócitos/metabolismo , Região CA1 Hipocampal/metabolismo , Interleucina-33/metabolismo , Plasticidade Neuronal , Transdução de Sinais/efeitos dos fármacos , Memória Espacial/efeitos dos fármacos , Animais , Astrócitos/efeitos dos fármacos , Proteína 4 Homóloga a Disks-Large/metabolismo , Técnicas de Inativação de Genes , Hipocampo/metabolismo , Homeostase , Interleucina-33/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Ratos , Sinapses/efeitos dos fármacos , Sinapses/genética , Sinapses/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Tetrodotoxina/farmacologia
2.
Proc Natl Acad Sci U S A ; 113(19): E2705-13, 2016 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-27091974

RESUMO

Alzheimer's disease (AD) is a devastating condition with no known effective treatment. AD is characterized by memory loss as well as impaired locomotor ability, reasoning, and judgment. Emerging evidence suggests that the innate immune response plays a major role in the pathogenesis of AD. In AD, the accumulation of ß-amyloid (Aß) in the brain perturbs physiological functions of the brain, including synaptic and neuronal dysfunction, microglial activation, and neuronal loss. Serum levels of soluble ST2 (sST2), a decoy receptor for interleukin (IL)-33, increase in patients with mild cognitive impairment, suggesting that impaired IL-33/ST2 signaling may contribute to the pathogenesis of AD. Therefore, we investigated the potential therapeutic role of IL-33 in AD, using transgenic mouse models. Here we report that IL-33 administration reverses synaptic plasticity impairment and memory deficits in APP/PS1 mice. IL-33 administration reduces soluble Aß levels and amyloid plaque deposition by promoting the recruitment and Aß phagocytic activity of microglia; this is mediated by ST2/p38 signaling activation. Furthermore, IL-33 injection modulates the innate immune response by polarizing microglia/macrophages toward an antiinflammatory phenotype and reducing the expression of proinflammatory genes, including IL-1ß, IL-6, and NLRP3, in the cortices of APP/PS1 mice. Collectively, our results demonstrate a potential therapeutic role for IL-33 in AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/fisiopatologia , Encéfalo/fisiopatologia , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/fisiopatologia , Interleucina-33/administração & dosagem , Doença de Alzheimer/diagnóstico , Animais , Encéfalo/efeitos dos fármacos , Transtornos Cognitivos/diagnóstico , Citocinas/metabolismo , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Fármacos Neuroprotetores/administração & dosagem , Resultado do Tratamento
3.
Proc Natl Acad Sci U S A ; 111(27): 9959-64, 2014 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-24958880

RESUMO

Alzheimer's disease (AD), characterized by cognitive decline, has emerged as a disease of synaptic failure. The present study reveals an unanticipated role of erythropoietin-producing hepatocellular A4 (EphA4) in mediating hippocampal synaptic dysfunctions in AD and demonstrates that blockade of the ligand-binding domain of EphA4 reverses synaptic impairment in AD mouse models. Enhanced EphA4 signaling was observed in the hippocampus of amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic mouse model of AD, whereas soluble amyloid-ß oligomers (Aß), which contribute to synaptic loss in AD, induced EphA4 activation in rat hippocampal slices. EphA4 depletion in the CA1 region or interference with EphA4 function reversed the suppression of hippocampal long-term potentiation in APP/PS1 transgenic mice, suggesting that the postsynaptic EphA4 is responsible for mediating synaptic plasticity impairment in AD. Importantly, we identified a small-molecule rhynchophylline as a novel EphA4 inhibitor based on molecular docking studies. Rhynchophylline effectively blocked the EphA4-dependent signaling in hippocampal neurons, and oral administration of rhynchophylline reduced the EphA4 activity effectively in the hippocampus of APP/PS1 transgenic mice. More importantly, rhynchophylline administration restored the impaired long-term potentiation in transgenic mouse models of AD. These findings reveal a previously unidentified role of EphA4 in mediating AD-associated synaptic dysfunctions, suggesting that it is a new therapeutic target for this disease.


Assuntos
Doença de Alzheimer/fisiopatologia , Modelos Animais de Doenças , Hipocampo/fisiopatologia , Receptor EphA4/metabolismo , Sinapses/fisiologia , Doença de Alzheimer/metabolismo , Animais , Hipocampo/metabolismo , Camundongos , Camundongos Transgênicos , Receptor EphA4/genética , Sinapses/metabolismo
4.
Neurosignals ; 21(1-2): 55-60, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22398430

RESUMO

Cyclin-dependent kinase 5 (Cdk5), a member of the cyclin-dependent kinase family, is critical for regulating neural development and neuronal survival. Dysregulation of Cdk5 is associated with abnormal expression of cell cycle-related proteins during neuronal apoptosis. We have previously found that p35, a Cdk5 activator, interacts with mSds3, an integral component of the histone deacetylase complex in vitro, suggesting a functional role of Cdk5 in gene regulation through modulation of chromatin integrity. In this study, we further demonstrate that Cdk5-dependent phosphorylation of mSds3 at Ser228 occurs in mouse brain nuclei. The expression of mSds3 protein and its interaction with Cdk5 activators is developmentally regulated in the mouse brain. Importantly, our findings suggest that the ability of Cdk5 to regulate activity deprivation-induced apoptosis of cerebellar granule neurons is likely mediated by the regulation of histone acetylation. Suppression of Cdk5 not only attenuates the induction of histone H3 acetylation and the aberrant upregulation of cyclin proteins in neurons after activity deprivation, but also results in protection of neurons against apoptotic cell death. Taken together, our findings suggest that Cdk5 regulates neuronal survival by precise epigenetic control through modulation of histone acetylation.


Assuntos
Quinase 5 Dependente de Ciclina/metabolismo , Histona Desacetilases/metabolismo , Histonas/metabolismo , Neurônios/metabolismo , Acetilação , Animais , Morte Celular/fisiologia , Células Cultivadas , Cerebelo/metabolismo , Células HEK293 , Humanos , Camundongos , Fosforilação/fisiologia
5.
ACS Chem Neurosci ; 12(22): 4249-4256, 2021 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-34738783

RESUMO

Alzheimer's disease (AD), the most common neurodegenerative disease, has limited treatment options. As such, extensive studies have been conducted to identify novel therapeutic approaches. We previously reported that rhynchophylline (Rhy), a small molecule EphA4 inhibitor, rescues impaired hippocampal synaptic plasticity and cognitive dysfunctions in APP/PS1 mice, an AD transgenic mouse model. To assess whether Rhy can be developed as an alternative treatment for AD, it is important to examine its pharmacokinetics and effects on other disease-associated pathologies. Here, we show that Rhy ameliorates amyloid plaque burden and reduces inflammation in APP/PS1 mice. Transcriptome analysis revealed that Rhy regulates various molecular pathways in APP/PS1 mouse brains associated with amyloid metabolism and inflammation, specifically the ubiquitin proteasome system, angiogenesis, and microglial functional states. These results show that Rhy, which is blood-brain barrier permeable, is beneficial to amyloid pathology and regulates multiple molecular pathways.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides , Precursor de Proteína beta-Amiloide/genética , Animais , Modelos Animais de Doenças , Inflamação/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oxindóis , Placa Amiloide/tratamento farmacológico , Presenilina-1/genética
6.
Nat Neurosci ; 14(2): 181-9, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21186356

RESUMO

Homeostatic plasticity is crucial for maintaining neuronal output by counteracting unrestrained changes in synaptic strength. Chronic elevation of synaptic activity by bicuculline reduces the amplitude of miniature excitatory postsynaptic currents (mEPSCs), but the underlying mechanisms of this effect remain unclear. We found that activation of EphA4 resulted in a decrease in synaptic and surface GluR1 and attenuated mEPSC amplitude through a degradation pathway that requires the ubiquitin proteasome system (UPS). Elevated synaptic activity resulted in increased tyrosine phosphorylation of EphA4, which associated with the ubiquitin ligase anaphase-promoting complex (APC) and its activator Cdh1 in neurons in a ligand-dependent manner. APC(Cdh1) interacted with and targeted GluR1 for proteasomal degradation in vitro, whereas depletion of Cdh1 in neurons abolished the EphA4-dependent downregulation of GluR1. Knockdown of EphA4 or Cdh1 prevented the reduction in mEPSC amplitude in neurons that was a result of chronic elevated activity. Our results define a mechanism by which EphA4 regulates homeostatic plasticity through an APC(Cdh1)-dependent degradation pathway.


Assuntos
Regulação para Baixo/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Receptor EphA4/metabolismo , Receptores de AMPA/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Análise de Variância , Ciclossomo-Complexo Promotor de Anáfase , Animais , Bicuculina/farmacologia , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Eletrofisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Antagonistas de Receptores de GABA-A/farmacologia , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Potenciais Pós-Sinápticos em Miniatura/efeitos dos fármacos , Potenciais Pós-Sinápticos em Miniatura/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , RNA Interferente Pequeno , Receptores de AMPA/genética , Estatísticas não Paramétricas , Sinapses/efeitos dos fármacos , Sinapses/fisiologia , Complexos Ubiquitina-Proteína Ligase/genética , Ubiquitinação/efeitos dos fármacos , Ubiquitinação/fisiologia
7.
Proc Natl Acad Sci U S A ; 104(41): 16347-52, 2007 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-17911252

RESUMO

EphA4-dependent growth cone collapse requires reorganization of actin cytoskeleton through coordinated activation of Rho family GTPases. Whereas various guanine exchange factors have recently been identified to be involved in EphA4-mediated regulation of Rho GTPases and growth cone collapse, the functional roles of GTPase-activating proteins in the process are largely unknown. Here we report that EphA4 interacts with alpha2-chimaerin through its Src homology 2 domain. Activated EphA4 induces a rapid increase of tyrosine phosphorylation of alpha2-chimaerin and enhances its GTPase-activating protein activity toward Rac1. More importantly, alpha2-chimaerin regulates the action of EphA4 in growth cone collapse through modulation of Rac1 activity. Our findings have therefore identified a new alpha2-chimaerin-dependent signaling mechanism through which EphA4 transduces its signals to the actin cytoskeleton and modulates growth cone morphology.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Quimerina 1/metabolismo , Cones de Crescimento/metabolismo , Receptor EphA4/metabolismo , Actinas/metabolismo , Animais , Linhagem Celular , Quimerina 1/química , Cones de Crescimento/ultraestrutura , Humanos , Neurônios/metabolismo , Fosforilação , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Tirosina/química , Proteínas rac1 de Ligação ao GTP/metabolismo
8.
J Biol Chem ; 279(52): 54438-44, 2004 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-15489224

RESUMO

Cyclin-dependent kinase 5 (Cdk5), a serine/threonine kinase that displays kinase activity predominantly in neurons, is activated by two non-cyclin activators, p35 or p39. Here, we report a physical and functional interaction between the Cdk5/p35 complex and mouse Sds3 (mSds3), an essential component of mSin3-histone deacetylase (HDAC) co-repressor complex. mSds3 binds to p35 both in vitro and in vivo, enabling active Cdk5 to phosphorylate mSds3 at serine 228. A mSds3 S228A mutant retained mSin3 binding activity, but its dimerization was not greatly enhanced by p35 when compared with wild type. Notably, p35 overexpression augmented mSds3-mediated transcriptional repression in vitro. Interestingly, mutational studies revealed that the ability of exogenous mSds3 to rescue cell growth and viability in mSds3 null cells correlates with its ability to be phosphorylated by Cdk5. The identification of mSds3 as a substrate of the Cdk5/p35 complex reveals a new regulatory mechanism in controlling the mSin3-HDAC transcriptional repressor activity and provides a new potential therapeutic means to inhibit specific HDAC activities in disease.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/metabolismo , Animais , Northern Blotting , Química Encefálica , Células COS , Linhagem Celular , Linhagem Celular Transformada , Quinase 5 Dependente de Ciclina , Dimerização , Glutationa Transferase/genética , Histona Desacetilases/metabolismo , Camundongos , Músculos/química , Mutagênese , Células NIH 3T3 , Fosforilação , RNA Mensageiro/análise , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/genética , Serina/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato , Transfecção , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA