Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cereb Cortex ; 29(4): 1644-1658, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29912395

RESUMO

The delicate balance of excitation and inhibition is crucial for proper function of the cerebral cortex, relying on the accurate number and subtype composition of inhibitory gamma-aminobutyric (GABA)-expressing interneurons. Various intrinsic and extrinsic factors precisely orchestrate their multifaceted development including the long-range migration from the basal telencephalon to cortical targets as well as interneuron survival throughout the developmental period. Particularly expressed guidance receptors were described to channel the migration of cortical interneurons deriving from the medial ganglionic eminence (MGE) and the preoptic area (POA) along distinct routes. Hence, unveiling the regulatory genetic networks controlling subtype-specific gene expression profiles is key to understand interneuron-specific developmental programs and to reveal causes for associated disorders. In contrast to MGE-derived interneurons, little is known about the transcriptional networks in interneurons born in the POA. Here, we provide first evidence for the LIM-homeobox transcription factor LHX1 as a crucial key player in the post-mitotic development of POA-derived cortical interneurons. By transcriptional regulation of related genes, LHX1 modulates their survival as well as the subtype-specific expression of guidance receptors of the Eph/ephrin family, thereby affecting directional migration and layer distribution in the adult cortex.


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Interneurônios/fisiologia , Proteínas com Homeodomínio LIM/fisiologia , Área Pré-Óptica/crescimento & desenvolvimento , Fatores de Transcrição/fisiologia , Animais , Movimento Celular , Sobrevivência Celular , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Efrina-B3/genética , Efrina-B3/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Interneurônios/citologia , Interneurônios/metabolismo , Proteínas com Homeodomínio LIM/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Área Pré-Óptica/citologia , Área Pré-Óptica/metabolismo , Receptor EphA4/genética , Receptor EphA4/fisiologia , Fatores de Transcrição/genética
2.
J Exp Neurosci ; 12: 1179069518760783, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29551912

RESUMO

The correct establishment of inhibitory circuits is crucial for cortical functionality and defects during the development of γ-aminobutyric acid-expressing cortical interneurons contribute to the pathophysiology of psychiatric disorders. A critical developmental step is the migration of cortical interneurons from their site of origin within the subpallium to the cerebral cortex, orchestrated by intrinsic and extrinsic signals. In addition to genetic networks, epigenetic mechanisms such as DNA methylation by DNA methyltransferases (DNMTs) are suggested to drive stage-specific gene expression underlying developmental processes. The mosaic structure of the interneuron generating domains producing a variety of interneurons for diverse destinations complicates research on regulatory instances of cortical interneuron migration. To this end, we performed single-cell transcriptome analysis revealing Dnmt1 expression in subsets of migrating interneurons. We found that DNMT1 preserves the migratory morphology in part through transcriptional control over Pak6 that promotes neurite complexity in postmigratory cells. In addition, we identified Ccdc184, a gene of unknown function, to be highly expressed in postmitotic interneurons. Single-cell mRNA sequencing revealed a positive correlation of Ccdc184 with cell adhesion-associated genes pointing to potential implications of CCDC184 in processes relying on cell-cell adhesion-like migration or morphological differentiation of interneurons that deserves further investigations.

3.
Neural Regen Res ; 12(11): 1768-1775, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29239313

RESUMO

The limited regenerative capacity of neuronal cells requires tight orchestration of cell death and survival regulation in the context of longevity, age-associated diseases as well as during the development of the nervous system. Subordinate to genetic networks epigenetic mechanisms like DNA methylation and histone modifications are involved in the regulation of neuronal development, function and aging. DNA methylation by DNA methyltransferases (DNMTs), mostly correlated with gene silencing, is a dynamic and reversible process. In addition to their canonical actions performing cytosine methylation, DNMTs influence gene expression by interactions with histone modifying enzymes or complexes increasing the complexity of epigenetic transcriptional networks. DNMTs are expressed in neuronal progenitors, post-mitotic as well as adult neurons. In this review, we discuss the role and mode of actions of DNMTs including downstream networks in the regulation of neuronal survival in the developing and aging nervous system and its relevance for associated disorders.

4.
Cereb Cortex ; 27(12): 5696-5714, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29117290

RESUMO

The proliferative niches in the subpallium generate a rich cellular variety fated for diverse telencephalic regions. The embryonic preoptic area (POA) represents one of these domains giving rise to the pool of cortical GABAergic interneurons and glial cells, in addition to striatal and residual POA cells. The migration from sites of origin within the subpallium to the distant targets like the cerebral cortex, accomplished by the adoption and maintenance of a particular migratory morphology, is a critical step during interneuron development. To identify factors orchestrating this process, we performed single-cell transcriptome analysis and detected Dnmt1 expression in murine migratory GABAergic POA-derived cells. Deletion of Dnmt1 in postmitotic immature cells of the POA caused defective migration and severely diminished adult cortical interneuron numbers. We found that DNA methyltransferase 1 (DNMT1) preserves the migratory shape in part through negative regulation of Pak6, which stimulates neuritogenesis at postmigratory stages. Our data underline the importance of DNMT1 for the migration of POA-derived cells including cortical interneurons.


Assuntos
Movimento Celular/fisiologia , Córtex Cerebral/embriologia , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Interneurônios/enzimologia , Células-Tronco Neurais/enzimologia , Área Pré-Óptica/embriologia , Animais , Animais Recém-Nascidos , Contagem de Células , Sobrevivência Celular/fisiologia , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/enzimologia , Metilação de DNA , Neurônios GABAérgicos/citologia , Neurônios GABAérgicos/enzimologia , Interneurônios/citologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Crescimento Neuronal/fisiologia , Área Pré-Óptica/citologia , Área Pré-Óptica/enzimologia , Técnicas de Cultura de Tecidos , Transcriptoma , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo
6.
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
7.
Front Cell Neurosci ; 8: 185, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25100946

RESUMO

During embryonic development the preoptic area (POA) gives rise to two populations of neurons which are generated at the same time, cortical interneurons and striatal cells. POA-derived cortical interneurons take a superficial path and avoid the developing striatum (Str) when they migrate to their target region. We found that EphB1, which is expressed in the striatal anlage, prevents cortical interneurons from entering the Str via ephrin-B3 reverse signaling. In contrast, for striatal neurons which also express ephrin-B3, EphB1 acts as a stop signal. This dual role of EphB1 is due to differences in ephrin-B3 reverse signaling cascades. For striatal neurons, binding of EphB1 to ephrin-B3 reduces endogenously high levels of pSrc and pFAK, which then causes the cells to stop migration. In contrast, in cortical interneurons EphB1-ephrin-B3 reverse signaling leads to phosphorylation of Src and focal adhesion kinase (FAK) which then mediates repulsion. Consistent with these in vitro findings, in an ephrin-B3 knockout mouse line, we discovered misrouted cortical interneurons in the Str and an over-migration of striatal neurons in their target region. Thus, EphB1/ephrin-B3 reverse signaling has a different impact on two sets of neurons which are generated at the same time and place: it can act as a repulsive cue for migrating neurons or it can terminate neuronal migration, a novel role of the Eph/ephrin system.

8.
Development ; 141(2): 460-71, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24381199

RESUMO

Inhibitory interneurons control the flow of information and synchronization in the cerebral cortex at the circuit level. During embryonic development, multiple subtypes of cortical interneurons are generated in different regions of the ventral telencephalon, such as the medial and caudal ganglionic eminence (MGE and CGE), as well as the preoptic area (POA). These neurons then migrate over long distances towards their cortical target areas. Diverse families of diffusible and cell-bound signaling molecules, including the Eph/ephrin system, regulate and orchestrate interneuron migration. Ephrin A3 and A5, for instance, are expressed at the borders of the pathway of MGE-derived interneurons and prevent these cells from entering inappropriate regions via EphA4 forward signaling. We found that MGE-derived interneurons, in addition to EphA4, also express ephrin A and B ligands, suggesting Eph/ephrin forward and reverse signaling in the same cell. In vitro and in vivo approaches showed that EphA4-induced reverse signaling in MGE-derived interneurons promotes their migration and that this effect is mediated by ephrin A2 ligands. In EphA4 mutant mice, as well as after ephrin A2 knockdown using in utero electroporation, we found delayed interneuron migration at embryonic stages. Thus, besides functions in guiding MGE-derived interneurons to the cortex through forward signaling, here we describe a novel role of the ephrins in driving these neurons to their target via reverse signaling.


Assuntos
Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Efrina-A2/metabolismo , Interneurônios/fisiologia , Receptor EphA4/metabolismo , Telencéfalo/embriologia , Telencéfalo/metabolismo , Animais , Movimento Celular/fisiologia , Córtex Cerebral/citologia , Efrina-A2/antagonistas & inibidores , Efrina-A2/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Proteínas Motores Moleculares/genética , Proteínas Motores Moleculares/metabolismo , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor EphA4/genética , Transdução de Sinais , Telencéfalo/citologia
9.
PLoS Genet ; 9(12): e1003988, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24367272

RESUMO

Hereditary spastic paraplegias (HSPs) are characterized by progressive weakness and spasticity of the legs because of the degeneration of cortical motoneuron axons. SPG15 is a recessively inherited HSP variant caused by mutations in the ZFYVE26 gene and is additionally characterized by cerebellar ataxia, mental decline, and progressive thinning of the corpus callosum. ZFYVE26 encodes the FYVE domain-containing protein ZFYVE26/SPASTIZIN, which has been suggested to be associated with the newly discovered adaptor protein 5 (AP5) complex. We show that Zfyve26 is broadly expressed in neurons, associates with intracellular vesicles immunopositive for the early endosomal marker EEA1, and co-fractionates with a component of the AP5 complex. As the function of ZFYVE26 in neurons was largely unknown, we disrupted Zfyve26 in mice. Zfyve26 knockout mice do not show developmental defects but develop late-onset spastic paraplegia with cerebellar ataxia confirming that SPG15 is caused by ZFYVE26 deficiency. The morphological analysis reveals axon degeneration and progressive loss of both cortical motoneurons and Purkinje cells in the cerebellum. Importantly, neuron loss is preceded by accumulation of large intraneuronal deposits of membrane-surrounded material, which co-stains with the lysosomal marker Lamp1. A density gradient analysis of brain lysates shows an increase of Lamp1-positive membrane compartments with higher densities in Zfyve26 knockout mice. Increased levels of lysosomal enzymes in brains of aged knockout mice further support an alteration of the lysosomal compartment upon disruption of Zfyve26. We propose that SPG15 is caused by an endolysosomal membrane trafficking defect, which results in endolysosomal dysfunction. This appears to be particularly relevant in neurons with highly specialized neurites such as cortical motoneurons and Purkinje cells.


Assuntos
Proteínas de Transporte/genética , Endossomos/metabolismo , Lisossomos/metabolismo , Degeneração Retiniana/genética , Paraplegia Espástica Hereditária/genética , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas de Transporte/metabolismo , Corpo Caloso/metabolismo , Corpo Caloso/patologia , Modelos Animais de Doenças , Endossomos/patologia , Humanos , Lisossomos/genética , Camundongos , Camundongos Knockout , Neurônios Motores/metabolismo , Mutação , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia , Paraplegia Espástica Hereditária/metabolismo , Paraplegia Espástica Hereditária/patologia
10.
J Clin Invest ; 123(10): 4273-82, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24051375

RESUMO

Axonopathies are a group of clinically diverse disorders characterized by the progressive degeneration of the axons of specific neurons. In hereditary spastic paraplegia (HSP), the axons of cortical motor neurons degenerate and cause a spastic movement disorder. HSP is linked to mutations in several loci known collectively as the spastic paraplegia genes (SPGs). We identified a heterozygous receptor accessory protein 1 (REEP1) exon 2 deletion in a patient suffering from the autosomal dominantly inherited HSP variant SPG31. We generated the corresponding mouse model to study the underlying cellular pathology. Mice with heterozygous deletion of exon 2 in Reep1 displayed a gait disorder closely resembling SPG31 in humans. Homozygous exon 2 deletion resulted in the complete loss of REEP1 and a more severe phenotype with earlier onset. At the molecular level, we demonstrated that REEP1 is a neuron-specific, membrane-binding, and membrane curvature-inducing protein that resides in the ER. We further show that Reep1 expression was prominent in cortical motor neurons. In REEP1-deficient mice, these neurons showed reduced complexity of the peripheral ER upon ultrastructural analysis. Our study connects proper neuronal ER architecture to long-term axon survival.


Assuntos
Retículo Endoplasmático/metabolismo , Proteínas de Membrana Transportadoras/genética , Neurônios Motores/metabolismo , Paraplegia Espástica Hereditária/genética , Animais , Sequência de Bases , Membrana Celular/química , Membrana Celular/metabolismo , Retículo Endoplasmático/patologia , Éxons , Marcha , Humanos , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Dados de Sequência Molecular , Neurônios Motores/patologia , Deleção de Sequência , Paraplegia Espástica Hereditária/patologia , Medula Espinal/patologia
11.
Cereb Cortex ; 23(3): 604-14, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22368082

RESUMO

Previous work demonstrated that members of the semaphorin family, Sema3A and Sema3C, act as repulsive and attractive guidance signals, respectively, for cortical axons. During the development of corticofugal projections, these semaphorins are expressed in adjacent cortical zones, but there is a considerable overlap between Sema3A and Sema3C expression in the subventricular zone. We used different in vitro assays to examine the response of cortical axons exposed to defined mixtures of these opposing guidance cues. Results showed that even at very low concentrations, Sema3A overrides the effects of Sema3C. Moreover, experiments with function-blocking antibodies directed against neuropilin provided insights into how cortical axons integrate disparate guidance signals at the receptor level. These in vitro data suggest that the pathway of corticofugal axons is defined by an attractive cue in the intermediate zone, where Sema3C is expressed alone. To directly test this hypothesis in vivo, we performed axon-tracing experiments in Sema3C-deficient mice. Compared with wild-type animals, corticofugal axons take a more superficial route in Sema3C(-/-) mice, and the corticofugal pathway is more compacted. This phenotype is expected when an attractive cue for cortical axons, Sema3C, is eliminated and a repulsive cue, Sema3A, becomes predominant.


Assuntos
Axônios/metabolismo , Córtex Cerebral/crescimento & desenvolvimento , Neurogênese/fisiologia , Semaforinas/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/metabolismo , Técnicas de Cocultura , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
12.
J Neurosci ; 31(50): 18364-80, 2011 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-22171039

RESUMO

The integration of interneuron subtypes into specific microcircuits is essential for proper cortical function. Understanding to what extent interneuron diversity is regulated and maintained during development might help to reveal the principles that govern their role as synchronizing elements as well as causes for dysfunction. Particular interneuron subtypes are generated in a temporally regulated manner in the medial ganglionic eminence (MGE), the caudal ganglionic eminence, and the preoptic area (POA) of the basal telencephalon. Long-range tangential migration from their site of origin to cortical targets is orchestrated by a variety of attractive, repulsive, membrane-bound, and secreted signaling molecules, to establish the critical balance of inhibition and excitation. It remains unknown whether interneurons deriving from distinct domains are predetermined to migrate in particular routes and whether this process underlies cell type-specific regulation. We found that POA- and MGE-derived cortical interneurons migrate within spatially segregated corridors. EphrinB3, expressed in POA-derived interneurons traversing the superficial route, acts as a repellent signal for deeply migrating interneurons born in the MGE, which is mediated by EphA4 forward signaling. In contrast, EphA4 induces repulsive ephrinB3 reverse signaling in interneurons generated in the POA, restricting this population to the superficial path. Perturbation of this bidirectional ephrinB3/EphA4 signaling in vitro and in vivo leads to a partial intermingling of cells in these segregated migratory pathways. Thus, we conclude that cell contact-mediated bidirectional ephrinB3/EphA4 signaling mediates the sorting of MGE- and POA-derived interneurons in the deep and superficial migratory stream.


Assuntos
Movimento Celular/fisiologia , Efrina-B3/metabolismo , Interneurônios/metabolismo , Área Pré-Óptica/metabolismo , Receptor EphA4/metabolismo , Transdução de Sinais/fisiologia , Telencéfalo/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Camundongos , Área Pré-Óptica/embriologia , Telencéfalo/embriologia
13.
Cell Adh Migr ; 4(3): 400-8, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20473036

RESUMO

Cortical interneurons are born in the proliferative zones of the ganglionic eminences in the subpallium and migrate to the developing cortex along well-defined tangential routes. The mechanisms regulating interneuron migration are not completely understood. Here we examine the role of class-A members of the Eph/ephrin system in directing the migration of interneurons. In situ hybridizations demonstrated that ephrin-A3 is expressed in the developing striatum, an area that is strictly avoided by migrating cortical interneurons in vivo, which express the EphA4 receptor. We then examined interneuron migration in grafting experiments, where explants of the medial ganglionic eminence (MGE) from enhanced green fluorescent protein-expressing transgenic mice were homotopically grafted into host slices from wildtype littermate embryos. After blocking ephrin-A ligands, many interneurons invaded the striatal anlage. Moreover, stripe assay experiments revealed that ephrin-A3 acts as a repellent cue for neurons from the medial ganglionic eminence. Downregulation of the EphA4 receptor via siRNA transfection reduced the repulsive effect of ephrin-A3, indicating that EphA4 mediates at least in part the repulsive effect of ephrin-A3 on these cells. Together, these results suggest that ephrin-A3 acts as a repulsive cue that restricts cortical interneurons from entering inappropriate regions and thus contributes to define the migratory route of cortical interneurons.


Assuntos
Movimento Celular , Córtex Cerebral/citologia , Efrina-A3/metabolismo , Efrina-A4/metabolismo , Interneurônios/citologia , Telencéfalo/citologia , Animais , Bioensaio , Regulação para Baixo , Humanos , Interneurônios/transplante , Ligantes , Eminência Mediana/citologia , Camundongos , Modelos Biológicos , Neostriado/metabolismo , Neostriado/patologia , Telencéfalo/metabolismo
14.
Cereb Cortex ; 20(10): 2411-22, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20071458

RESUMO

Chondroitin sulfate (CS) carrying proteoglycans (PGs) are widely expressed in the nervous system, and there is increasing evidence that they regulate developmental mechanisms like neurite outgrowth, axonal guidance and neuronal migration. Moreover, they can also act indirectly by organizing and/or modulating growth factors and guidance molecules. We found that chondroitin-4-sulfate is coexpressed with semaphorin 3A (Sema 3A) in the striatal mantle zone (SMZ), a nontarget region of neuropilin (Nrp)-1-expressing cortical interneurons flanking their migratory route in the subpallium. Using in vitro assays, we showed that CS PGs exert a repulsive effect on cortical interneurons, independently of Sema 3A, due to the CS side chains. We further showed that extracellular Sema 3A binds to CS. Disrupting Sema 3A-Nrp-1 signaling led migrating medial ganglionic eminence neurons to inappropriately invade the SMZ and even more so after removal of the CS side chains. Moreover, we found that soluble Sema 3A enhances the CS-induced repulsion in vitro. We concluded that CS acts as a repellent for cortical interneurons and that, in addition, CS restricts secreted Sema 3A within SMZ. Thus, both molecules act in concert to repel cortical interneurons from the SMZ during tangential migration toward the cerebral cortex.


Assuntos
Movimento Celular/fisiologia , Córtex Cerebral/citologia , Sulfatos de Condroitina/metabolismo , Corpo Estriado/metabolismo , Interneurônios/fisiologia , Semaforina-3A/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Condroitina ABC Liase/farmacologia , Proteoglicanas de Sulfatos de Condroitina/genética , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Fluorescência Verde/genética , Interneurônios/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal/métodos , Neuropilina-1/metabolismo , Técnicas de Cultura de Órgãos , Semaforina-3A/genética
15.
Eur J Neurosci ; 28(1): 62-73, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18662335

RESUMO

Cortical interneurons are born in the germinative zones of the ganglionic eminences in the subpallium, and migrate tangentially in spatially and temporally well-defined corridors into the neocortex. Because ephrin-A5 is expressed in the ventricular zone (VZ) of the ganglionic eminences at these developmental stages, we examined the possible effects of this molecule on interneuron migration. Double-immunocytochemistry of dissociated neurons from the medial ganglionic eminences (MGE) revealed that calbindin-positive cells express the EphA4-receptor. In situ, EphA4 is strongly expressed in the subventricular zone of the ganglionic eminences. Using different in vitro assays, we found that ephrin-A5 acts as a repellent cue for MGE neurons. We then examined interneuron migration in slice overlay experiments, where MGE-derived explants from enhanced green fluorescent protein-expressing transgenic mice were homotopically grafted into host slices from wild-type littermate embryos. In these in vitro preparations, interneurons recapitulated in vivo cell migration in several respects. However, interneurons in brain slices also migrated in the VZ of the ganglionic eminences, a region that is strictly avoided in vivo. In situ hybridizations revealed that ephrin-A5 became downregulated in the VZ in vitro. When recombinant ephrin-A5-Fc was added to the slices, it preferentially bound to the VZ, and migrating MGE neurons avoided the VZ as in vivo. The restoration of the normal migration pathway in slices required ephrin-A5 clustering and signalling of Src family kinases. Together, these experiments suggest that ephrin-A5 acts as an inhibitory flank that contributes to define the pathway of migrating interneurons.


Assuntos
Movimento Celular/fisiologia , Efrina-A5/metabolismo , Interneurônios/fisiologia , Animais , Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Técnicas de Cocultura , Sinais (Psicologia) , Efrina-A5/genética , Feminino , Fibroblastos/citologia , Fibroblastos/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Interneurônios/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células NIH 3T3 , Receptor EphA4/metabolismo
16.
J Neurosci ; 27(21): 5643-53, 2007 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-17522309

RESUMO

The Eph receptor tyrosine kinases and their membrane-bound ligands, the ephrins, are involved in a variety of developmental processes such as axonal guidance, cell migration, cell adhesion, proliferation, and differentiation. In addition to repulsive effects, ephrins can also induce attractive responses. Up to now, little was known about the underlying signaling mechanisms that regulate attractive versus repulsive effects. In this study, we show that ephrin-A5 enhances the motility of cortical neurons that is dependent on the activity of Src-family kinases (SFKs). Ephrin-A5 further changes the adhesive properties of neurons by inducing the formation of cell aggregates. Using the stripe assay, we found that the motogenic effect of ephrin-A5 is the result of repulsive ephrin-A interactions. Blocking SFK function leads to a conversion of repulsion into adhesion, suggesting that SFKs can act as a biological switch for the response of EphA receptors. Finally, we discovered a ligand-induced release of membrane particles containing EphA receptors, suggesting membrane ripping as a novel mechanism to overcome the "ephrin paradox" of repulsion after high-affinity receptor-ligand binding.


Assuntos
Efrina-A5/fisiologia , Neurônios/enzimologia , Quinases da Família src/fisiologia , Animais , Agregação Celular/fisiologia , Movimento Celular/fisiologia , Humanos , Camundongos , Células NIH 3T3 , Neurônios/citologia , Neurônios/metabolismo , Receptor EphA5/antagonistas & inibidores , Receptor EphA5/metabolismo , Receptor EphA5/fisiologia , Quinases da Família src/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...