Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Development ; 145(3)2018 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-29439133

RESUMO

Genetic factors underlying the human limb abnormality congenital talipes equinovarus ('clubfoot') remain incompletely understood. The spontaneous autosomal recessive mouse 'peroneal muscular atrophy' mutant (PMA) is a faithful morphological model of human clubfoot. In PMA mice, the dorsal (peroneal) branches of the sciatic nerves are absent. In this study, the primary developmental defect was identified as a reduced growth of sciatic nerve lateral motor column (LMC) neurons leading to failure to project to dorsal (peroneal) lower limb muscle blocks. The pma mutation was mapped and a candidate gene encoding LIM-domain kinase 1 (Limk1) identified, which is upregulated in mutant lateral LMC motor neurons. Genetic and molecular analyses showed that the mutation acts in the EphA4-Limk1-Cfl1/cofilin-actin pathway to modulate growth cone extension/collapse. In the chicken, both experimental upregulation of Limk1 by electroporation and pharmacological inhibition of actin turnover led to defects in hindlimb spinal motor neuron growth and pathfinding, and mimicked the clubfoot phenotype. The data support a neuromuscular aetiology for clubfoot and provide a mechanistic framework to understand clubfoot in humans.


Assuntos
Doença de Charcot-Marie-Tooth/embriologia , Pé Torto Equinovaro/embriologia , Pé Torto Equinovaro/genética , Quinases Lim/genética , Mutação , Animais , Axônios , Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/patologia , Embrião de Galinha , Mapeamento Cromossômico , Pé Torto Equinovaro/patologia , Modelos Animais de Doenças , Feminino , Membro Posterior/anormalidades , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Neurônios Motores/patologia , Músculo Esquelético/anormalidades , Músculo Esquelético/inervação , Nervo Fibular/anormalidades , Fenótipo , Gravidez , Receptor EphA4/deficiência , Receptor EphA4/genética , Nervo Isquiático/anormalidades , Regulação para Cima
2.
J Sleep Res ; 26(5): 539-546, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28488395

RESUMO

Slow waves occurring during non-rapid eye movement sleep have been associated with neurobehavioural performance and memory. In addition, the duration of previous wakefulness and sleep impacts characteristics of these slow waves. However, molecular mechanisms regulating the dynamics of slow-wave characteristics remain poorly understood. The EphA4 receptor regulates glutamatergic transmission and synaptic plasticity, which have both been linked to sleep slow waves. To investigate if EphA4 regulates slow-wave characteristics during non-rapid eye movement sleep, we compared individual parameters of slow waves between EphA4 knockout mice and wild-type littermates under baseline conditions and after a 6-h sleep deprivation. We observed that, compared with wild-type mice, knockout mice display a shorter duration of positive and negative phases of slow waves under baseline conditions and after sleep deprivation. However, the mutation did not change slow-wave density, amplitude and slope, and did not affect the sleep deprivation-dependent changes in slow-wave characteristics, suggesting that EphA4 is not involved in the response to elevated sleep pressure. Our present findings suggest a role for EphA4 in shaping cortical oscillations during sleep that is independent from sleep need.


Assuntos
Receptor EphA4/deficiência , Receptor EphA4/genética , Sono/fisiologia , Animais , Eletroencefalografia , Masculino , Camundongos , Camundongos Knockout , Receptor EphA4/metabolismo , Sono/genética , Privação do Sono/genética , Privação do Sono/fisiopatologia , Fatores de Tempo , Vigília/genética , Vigília/fisiologia
3.
Sleep ; 39(3): 613-24, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26612390

RESUMO

STUDY OBJECTIVES: Optimal sleep is ensured by the interaction of circadian and homeostatic processes. Although synaptic plasticity seems to contribute to both processes, the specific players involved are not well understood. The EphA4 tyrosine kinase receptor is a cell adhesion protein regulating synaptic plasticity. We investigated the role of EphA4 in sleep regulation using electrocorticography in mice lacking EphA4 and gene expression measurements. METHODS: EphA4 knockout (KO) mice, Clock(Δ19/Δ19) mutant mice and littermates, C57BL/6J and CD-1 mice, and Sprague-Dawley rats were studied under a 12 h light: 12 h dark cycle, under undisturbed conditions or 6 h sleep deprivation (SLD), and submitted to a 48 h electrophysiological recording and/or brain sampling at different time of day. RESULTS: EphA4 KO mice showed less rapid eye movement sleep (REMS), enhanced duration of individual bouts of wakefulness and nonrapid eye movement sleep (NREMS) during the light period, and a blunted daily rhythm of NREMS sigma activity. The NREMS delta activity response to SLD was unchanged in EphA4 KO mice. However, SLD increased EphA4 expression in the thalamic/hypothalamic region in C57BL/6J mice. We further show the presence of E-boxes in the promoter region of EphA4, a lower expression of EphA4 in Clock mutant mice, a rhythmic expression of EphA4 ligands in several brain areas, expression of EphA4 in the suprachiasmatic nuclei of the hypothalamus (SCN), and finally an unchanged number of cells expressing Vip, Grp and Avp in the SCN of EphA4 KO mice. CONCLUSIONS: Our results suggest that EphA4 is involved in circadian sleep regulation.


Assuntos
Ritmo Circadiano/fisiologia , Receptor EphA4/metabolismo , Privação do Sono/fisiopatologia , Sono/fisiologia , Animais , Proteínas CLOCK/genética , Ritmo Circadiano/genética , Escuridão , Eletrocorticografia , Fenômenos Eletrofisiológicos , Homeostase , Luz , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasticidade Neuronal , Regiões Promotoras Genéticas/genética , Ratos , Ratos Sprague-Dawley , Receptor EphA4/biossíntese , Receptor EphA4/deficiência , Receptor EphA4/genética , Sono/genética , Privação do Sono/genética , Sono REM/genética , Sono REM/fisiologia , Núcleo Supraquiasmático/metabolismo , Tálamo/metabolismo , Fatores de Tempo , Vigília/genética , Vigília/fisiologia
4.
Development ; 142(1): 140-50, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25480914

RESUMO

The phenotype of excitatory cerebral cortex neurons is specified at the progenitor level, orchestrated by various intrinsic and extrinsic factors. Here, we provide evidence for a subcortical contribution to cortical progenitor regulation by thalamic axons via ephrin A5-EphA4 interactions. Ephrin A5 is expressed by thalamic axons and represents a high-affinity ligand for EphA4 receptors detected in cortical precursors. Recombinant ephrin A5-Fc protein, as well as ephrin A ligand-expressing, thalamic axons affect the output of cortical progenitor division in vitro. Ephrin A5-deficient mice show an altered division mode of radial glial cells (RGCs) accompanied by increased numbers of intermediate progenitor cells (IPCs) and an elevated neuronal production for the deep cortical layers at E13.5. In turn, at E16.5 the pool of IPCs is diminished, accompanied by reduced rates of generated neurons destined for the upper cortical layers. This correlates with extended infragranular layers at the expense of superficial cortical layers in adult ephrin A5-deficient and EphA4-deficient mice. We suggest that ephrin A5 ligands imported by invading thalamic axons interact with EphA4-expressing RGCs, thereby contributing to the fine-tuning of IPC generation and thus the proper neuronal output for cortical layers.


Assuntos
Córtex Cerebral/citologia , Efrina-A5/metabolismo , Neurônios Aferentes/citologia , Neurônios Aferentes/metabolismo , Receptor EphA4/metabolismo , Células-Tronco/metabolismo , Tálamo/citologia , Animais , Axônios/metabolismo , Contagem de Células , Divisão Celular , Embrião de Mamíferos/citologia , Células Ependimogliais/citologia , Células Ependimogliais/metabolismo , Efrina-A5/deficiência , Ligantes , Camundongos Endogâmicos C57BL , Neurogênese , Receptor EphA4/deficiência , Transdução de Sinais , Células-Tronco/citologia , Tálamo/embriologia , Tálamo/metabolismo
5.
Birth Defects Res A Clin Mol Teratol ; 100(6): 507-11, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24753315

RESUMO

BACKGROUND: Interstitial 2q36 deletion is a rare event. Only two previously published cases of 2q36 deletions were characterized using array-CGH. This is the first case diagnosed prenatally. METHODS: We report on the prenatal diagnosis of a 2q36.1q36.3 interstitial deletion in a fetus with facial dysmorphism, spina bifida, and cleft palate. RESULTS: Array-CGH analysis revealed a 5.6 Mb interstitial deletion of the long arm of chromosome 2q36.1q36.3, including the PAX3 and EPHA4 genes. CONCLUSION: The present study reinforces the hypothesis that PAX3 haploinsufficiency may be associated with neural tube defects in humans and suggests that the EPHA4 gene might be implicated during palate development. This report also illustrates the added value of array-CGH to detect cryptic chromosomal imbalances in malformed fetuses and to improve genetic counseling prenatally.


Assuntos
Anormalidades Múltiplas/genética , Deleção Cromossômica , Cromossomos Humanos Par 2 , Fissura Palatina/genética , Fatores de Transcrição Box Pareados/genética , Receptor EphA4/genética , Disrafismo Espinal/genética , Anormalidades Múltiplas/diagnóstico , Anormalidades Múltiplas/patologia , Adulto , Fissura Palatina/diagnóstico , Fissura Palatina/patologia , Hibridização Genômica Comparativa , Feminino , Feto , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Cariotipagem , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/deficiência , Gravidez , Diagnóstico Pré-Natal , Receptor EphA4/deficiência , Disrafismo Espinal/diagnóstico , Disrafismo Espinal/patologia
6.
J Comp Neurol ; 520(8): 1702-20, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22121026

RESUMO

The guidance of axonal projections to ipsilateral and contralateral regions is essential for integration of bilateral sensory information and coordination of movement. In the development of olivocerebellar projections, newborn neurons of inferior olivary (IO) nuclei ventrally migrate from the hindbrain rhombic lip to the floor plate (FP). The cell bodies of IO neurons cannot cross the FP but their axons can, and thus IO neurons project their axons only to the contralateral cerebellar cortex. The molecular mechanisms determining the contralateral axonal projections of IO neurons, however, are obscure. The IO neurons and their axons express EphA4, whereas the FP expresses an EphA4 ligand, EphrinB3, from embryonic day 12.5. Therefore, we tested whether EphA4-deficient mice (EphA4(-/-) ) would show impairment in the development of olivocerebellar projections. We found that, in EphA4(-/-) embryos, some of the IO neurons projected their axons to the ipsilateral cerebellar cortex because the cell bodies of the IO neurons abnormally crossed the FP. Furthermore, even in adults, EphA4(-/-) cerebella were bilaterally innervated by unilateral IO subnuclei. These observations indicate that EphA4 is involved in the contralateral axonal projections of IO neurons by preventing their cell bodies from crossing the midline FP.


Assuntos
Movimento Celular/fisiologia , Lateralidade Funcional/fisiologia , Neurogênese/fisiologia , Neurônios/citologia , Núcleo Olivar/embriologia , Receptor EphA4/metabolismo , Animais , Axônios/metabolismo , Axônios/ultraestrutura , Padronização Corporal/fisiologia , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Neurônios/metabolismo , Núcleo Olivar/crescimento & desenvolvimento , Receptor EphA4/deficiência
7.
J Neurosci ; 30(47): 16015-24, 2010 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-21106840

RESUMO

There is growing evidence that astrocytes play critical roles in neuron-glial interactions at the synapse. Astrocytes are believed to regulate presynaptic and postsynaptic structures and functions, in part, by the release of gliotransmitters such as glutamate, ATP, and d-serine; however, little is known of how neurons and astrocytes communicate to regulate these processes. Here, we investigated a family of transmembrane proteins called ephrinBs and Eph receptors that are expressed in the synapse and are known to regulate synaptic transmission and plasticity. In addition to their presence on CA1 hippocampal neurons, we determined that ephrins and Eph receptors are also expressed on hippocampal astrocytes. Stimulation of hippocampal astrocytes with soluble ephrinB3, known to be expressed on CA1 postsynaptic dendrites, enhanced d-serine synthesis and release in culture. Conversely, ephrinB3 had no effect on d-serine release from astrocytes deficient in EphB3 and EphA4, which are the primary receptors for ephrinB3. Eph receptors mediate this response through interactions with PICK1 (protein interacting with C-kinase) and by dephosphorylating protein kinase C α to activate the conversion of l-serine to d-serine by serine racemase. These findings are supported in vivo, where reduced d-serine levels and synaptic transmissions are observed in the absence of EphB3 and EphA4. These data support a role for ephrins and Eph receptors in regulating astrocyte gliotransmitters, which may have important implications on synaptic transmission and plasticity.


Assuntos
Astrócitos/metabolismo , Efrina-B3/fisiologia , Serina/biossíntese , Serina/metabolismo , Animais , Células Cultivadas , Efrina-B3/deficiência , Hipocampo/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Plasticidade Neuronal/genética , Biossíntese de Proteínas/genética , Receptor EphA4/biossíntese , Receptor EphA4/deficiência , Receptor EphA4/fisiologia , Serina/análogos & derivados , Estereoisomerismo , Transmissão Sináptica/genética
8.
Nat Neurosci ; 12(10): 1285-92, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19734893

RESUMO

Astrocytes are critical participants in synapse development and function, but their role in synaptic plasticity is unclear. Eph receptors and their ephrin ligands have been suggested to regulate neuron-glia interactions, and EphA4-mediated ephrin reverse signaling is required for synaptic plasticity in the hippocampus. Here we show that long-term potentiation (LTP) at the CA3-CA1 synapse is modulated by EphA4 in the postsynaptic CA1 cell and by ephrin-A3, a ligand of EphA4 that is found in astrocytes. Lack of EphA4 increased the abundance of glial glutamate transporters, and ephrin-A3 modulated transporter currents in astrocytes. Pharmacological inhibition of glial glutamate transporters rescued the LTP defects in EphA4 (Epha4) and ephrin-A3 (Efna3) mutant mice. Transgenic overexpression of ephrin-A3 in astrocytes reduces glutamate transporter levels and produces focal dendritic swellings possibly caused by glutamate excitotoxicity. These results suggest that EphA4/ephrin-A3 signaling is a critical mechanism for astrocytes to regulate synaptic function and plasticity.


Assuntos
Efrina-A3/metabolismo , Ácido Glutâmico/metabolismo , Potenciação de Longa Duração/fisiologia , Neuroglia/fisiologia , Neurônios/fisiologia , Receptor EphA4/metabolismo , Animais , Animais Recém-Nascidos , Ácido Aspártico/análogos & derivados , Ácido Aspártico/farmacologia , Biofísica , Modelos Animais de Doenças , Estimulação Elétrica/métodos , Efrina-A3/genética , Antagonistas de Aminoácidos Excitatórios/farmacologia , Transportador 1 de Aminoácido Excitatório/metabolismo , Potenciais Pós-Sinápticos Excitadores/genética , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Fluorescência Verde/genética , Hipocampo/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp/métodos , Pentilenotetrazol , Receptor EphA4/deficiência , Convulsões/induzido quimicamente , Convulsões/genética , Convulsões/fisiopatologia , Transdução de Sinais/fisiologia , Sinapses/fisiologia , Regulação para Cima/genética
9.
Development ; 136(14): 2467-76, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19542359

RESUMO

Eph receptors are widely expressed during cerebral cortical development, yet a role for Eph signaling in the generation of cells during corticogenesis has not been shown. Cortical progenitor cells selectively express one receptor, EphA4, and reducing EphA4 signaling in cultured progenitors suppressed proliferation, decreasing cell number. In vivo, EphA4(-/-) cortex had a reduced area, fewer cells and less cell division compared with control cortex. To understand the effects of EphA4 signaling in corticogenesis, EphA4-mediated signaling was selectively depressed or elevated in cortical progenitors in vivo. Compared with control cells, cells with reduced EphA4 signaling were rare and mitotically inactive. Conversely, overexpression of EphA4 maintained cells in their progenitor states at the expense of subsequent maturation, enlarging the progenitor pool. These results support a role for EphA4 in the autonomous promotion of cell proliferation during corticogenesis. Although most ephrins were undetectable in cortical progenitors, ephrin B1 was highly expressed. Our analyses demonstrate that EphA4 and ephrin B1 bind to each other, thereby initiating signaling. Furthermore, overexpression of ephrin B1 stimulated cell division of neighboring cells, supporting the hypothesis that ephrin B1-initiated forward signaling of EphA4 promotes cortical cell division.


Assuntos
Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Receptor EphA4/metabolismo , Animais , Comunicação Celular , Proliferação de Células , Células Cultivadas , Córtex Cerebral/citologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Efrina-B1/genética , Efrina-B1/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Neurológicos , Gravidez , Receptor EphA4/deficiência , Receptor EphA4/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais
10.
Development ; 135(19): 3281-90, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18755772

RESUMO

Brain structures, whether mature or developing, display a wide diversity of pattern and shape, such as layers, nuclei or segments. The striatum in the mammalian forebrain displays a unique mosaic organization (subdivided into two morphologically and functionally defined neuronal compartments: the matrix and the striosomes) that underlies important functional features of the basal ganglia. Matrix and striosome neurons are generated sequentially during embryonic development, and segregate from each other to form a mosaic of distinct compartments. However, the molecular mechanisms that underlie this time-dependent process of neuronal segregation remain largely unknown. Using a novel organotypic assay, we identified ephrin/Eph family members as guidance cues that regulate matrix/striosome compartmentalization. We found that EphA4 and its ephrin ligands displayed specific temporal patterns of expression and function that play a significant role in the spatial segregation of matrix and striosome neurons. Analysis of the striatal patterning in ephrin A5/EphA4 mutant mice further revealed the requirement of EphA4 signalling for the proper sorting of matrix and striosome neuronal populations in vivo. These data constitute the first identification of genes involved in striatal compartmentalization, and reveal a novel mechanism by which the temporal control of guidance cues enables neuronal segregation, and thereby the generation of complex cellular patterns in the brain.


Assuntos
Padronização Corporal/fisiologia , Corpo Estriado/embriologia , Corpo Estriado/metabolismo , Efrina-A5/metabolismo , Receptor EphA4/metabolismo , Animais , Padronização Corporal/genética , Adesão Celular , Corpo Estriado/citologia , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Efrina-A5/deficiência , Efrina-A5/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Modelos Neurológicos , Gravidez , Receptor EphA4/deficiência , Receptor EphA4/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA