Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Neurosci ; 37(36): 8816-8829, 2017 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-28821666

RESUMO

GABA is the key inhibitory neurotransmitter in the cortex but regulation of its synthesis during forebrain development is poorly understood. In the telencephalon, members of the distal-less (Dlx) homeobox gene family are expressed in, and regulate the development of, the basal ganglia primodia from which many GABAergic neurons originate and migrate to other forebrain regions. The Dlx1/Dlx2 double knock-out mice die at birth with abnormal cortical development, including loss of tangential migration of GABAergic inhibitory interneurons to the neocortex (Anderson et al., 1997a). We have discovered that specific promoter regulatory elements of glutamic acid decarboxylase isoforms (Gad1 and Gad2), which regulate GABA synthesis from the excitatory neurotransmitter glutamate, are direct transcriptional targets of both DLX1 and DLX2 homeoproteins in vivo Further gain- and loss-of-function studies in vitro and in vivo demonstrated that both DLX1 and DLX2 are necessary and sufficient for Gad gene expression. DLX1 and/or DLX2 activated the transcription of both Gad genes, and defects in Dlx function disrupted the differentiation of GABAergic interneurons with global reduction in GABA levels in the forebrains of the Dlx1/Dlx2 double knock-out mouse in vivo Identification of Gad genes as direct Dlx transcriptional targets is significant; it extends our understanding of Dlx gene function in the developing forebrain beyond the regulation of tangential interneuron migration to the differentiation of GABAergic interneurons arising from the basal telencephalon, and may help to unravel the pathogenesis of several developmental brain disorders.SIGNIFICANCE STATEMENT GABA is the major inhibitory neurotransmitter in the brain. We show that Dlx1/Dlx2 homeobox genes regulate GABA synthesis during forebrain development through direct activation of glutamic acid decarboxylase enzyme isoforms that convert glutamate to GABA. This discovery helps explain how Dlx mutations result in abnormal forebrain development, due to defective differentiation, in addition to the loss of tangential migration of GABAergic inhibitory interneurons to the neocortex. Reduced numbers or function of cortical GABAergic neurons may lead to hyperactivity states such as seizures (Cobos et al., 2005) or contribute to the pathogenesis of some autism spectrum disorders. GABAergic dysfunction in the basal ganglia could disrupt the learning and development of complex motor and cognitive behaviors (Rubenstein and Merzenich, 2003).


Assuntos
Prosencéfalo Basal/fisiologia , Diferenciação Celular/fisiologia , Neurônios GABAérgicos/fisiologia , Glutamato Descarboxilase/metabolismo , Proteínas de Homeodomínio/metabolismo , Interneurônios/fisiologia , Fatores de Transcrição/metabolismo , Animais , Prosencéfalo Basal/citologia , Movimento Celular/fisiologia , Células Cultivadas , Feminino , Neurônios GABAérgicos/citologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Interneurônios/citologia , Masculino , Camundongos , Camundongos Knockout , Ácido gama-Aminobutírico/metabolismo
2.
Nucleic Acids Res ; 32(3): 884-92, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14769946

RESUMO

Understanding homeobox gene specificity and function has been hampered by the lack of proven direct transcriptional targets during development. Dlx genes are expressed in the developing forebrain, retina, craniofacial structures and limbs. Dlx1/Dlx2 double knockout mice die at birth with multiple defects including abnormal forebrain development and decreased Dlx5 and Dlx6 expression. We have successfully applied chromatin immunoprecipitation (ChIP) to identify a direct transcriptional target of DLX homeoproteins from embryonic tissues in vivo. We optimized cross-linking conditions to enrich for protein-DNA complexes, then using specific high affinity DLX antibodies captured immunoenriched DLX genomic DNA transcriptional targets. DLX homeobox proteins bind differentially to the Dlx5/Dlx6 intergenic enhancer in newborn retina (DLX2) and embryonic striatum (DLX1, DLX2) in situ. Reporter gene assays demonstrated the functional significance of the binding of DLX proteins to this regulatory element, confirmed in vitro by electrophoretic mobility shift assays, using tissue extracts or recombinant DLX proteins. ChIP provides the best approach to identify direct Dlx homeoprotein targets from developing tissues in situ. The use of this technology will advance our understanding of Dlx gene function in the vertebrate in vivo and can be applied to examine targets of other homeobox genes and other classes of transcription factors.


Assuntos
Proteínas de Homeodomínio/metabolismo , Prosencéfalo/embriologia , Retina/embriologia , Fatores de Transcrição/metabolismo , Animais , Sítios de Ligação , Cromatina/metabolismo , Cisplatino/química , Reagentes de Ligações Cruzadas , DNA/metabolismo , DNA Intergênico/genética , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/imunologia , Camundongos , Neostriado/metabolismo , Proteínas Nucleares/metabolismo , Testes de Precipitina , Prosencéfalo/metabolismo , RNA Mensageiro/metabolismo , Retina/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Ativação Transcricional
3.
J Comp Neurol ; 461(2): 187-204, 2003 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-12724837

RESUMO

Distal-less homeobox genes are expressed in the developing forebrain. We assessed Dlx gene expression in the developing and adult mouse retina. Dlx1 and Dlx2 are detected in retinal neuroprogenitors by embryonic day (E) 12.5 (Eisenstat et al. [1999] J. Comp. Neurol. 217-237). At E13.5, the expression of four homeodomain proteins, DLX2, BRN3b, PAX6, and CHX10, define distinct yet overlapping domains in the retinal neuroepithelium. By postnatal day (P) 0, DLX2 is expressed in the neuroblastic layer and the ganglion cell layer (GCL) consisting of ganglion and displaced amacrine cells. DLX1 expression resembles DLX2 to P0 but decreases postnatally. In the adult, DLX2 is localized to ganglion, amacrine, and horizontal cells as determined by coexpression with retinal cell-specific markers. There is coincident expression of DLX2 with gamma-aminobutyric acid (GABA), glutamic acid decarboxylase (GAD)65, and GAD67 in the inner nuclear layer (INL) and GCL. In the adult, DLX2 is coexpressed with BRN3b in ganglion cells; PAX6 in amacrine, horizontal, and ganglion cells; and Chx10 in some bipolar cells. We predict that a combinatorial code of these homeobox genes and others specify retinal cell fate. Our results support a possible role for Dlx1 and Dlx2 in inner retinal development and in the terminal differentiation and/or maintenance of INL interneurons and ganglion cells in the adult. The correlation of DLX2 with GABA expression in the mouse retina closely mirrors the relationship of DLX2 to GABAergic neuronal differentiation in the embryonic forebrain, including neocortex, olfactory bulb and hippocampus, signifying a conservation of function of Dlx genes in the developing central nervous system.


Assuntos
Diferenciação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Homeodomínio/metabolismo , Retina/embriologia , Células Ganglionares da Retina/metabolismo , Células Amácrinas/citologia , Células Amácrinas/metabolismo , Animais , Linhagem da Célula/genética , Proteínas do Citoesqueleto , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dopamina/metabolismo , Proteínas do Olho , Feto , Glutamato Descarboxilase/metabolismo , Proteínas de Homeodomínio/genética , Imuno-Histoquímica , Interneurônios/citologia , Interneurônios/metabolismo , Isoenzimas/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA , Proteínas Repressoras , Retina/citologia , Retina/metabolismo , Células Ganglionares da Retina/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Fator de Transcrição Brn-3 , Fator de Transcrição Brn-3B , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ácido gama-Aminobutírico/metabolismo
4.
J Biol Chem ; 282(26): 19071-81, 2007 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-17259176

RESUMO

Dlx homeobox genes play an important role in vertebrate forebrain development. Dlx1/Dlx2 null mice die at birth with an abnormal cortical phenotype, including impaired differentiation and migration of GABAergic interneurons to the neocortex. However, the molecular basis for these defects downstream of loss of Dlx1/Dlx2 function is unknown. Neuropilin-2 (NRP-2) is a receptor for Class III semaphorins, which inhibit neuronal migration. Herein, we show that Neuropilin-2 is a specific DLX1 and DLX2 transcriptional target by applying chromatin immunoprecipitation to embryonic forebrain tissues. Both homeobox proteins repress Nrp-2 expression in vitro, confirming the functional significance of DLX binding. Furthermore, the homeodomain of DLX1 and DLX2 is necessary for DNA binding and this binding is essential for Dlx repression of Nrp-2 expression. Of importance, there is up-regulated and aberrant expression of NRP-2 in the forebrains of Dlx1/Dlx2 null mice. This is the first report that DLX1 or DLX2 can function as transcriptional repressors. Our data show that DLX proteins specifically mediate the repression of Neuropilin-2 in the developing forebrain. As well, our results support the hypothesis that down-regulation of Neuropilin-2 expression may facilitate tangential interneuron migration from the basal forebrain.


Assuntos
Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Interneurônios/patologia , Neuropilina-2/genética , Prosencéfalo/anormalidades , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Movimento Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Interneurônios/fisiologia , Camundongos , Camundongos Mutantes Neurológicos , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/fisiologia , Prosencéfalo/patologia
5.
Development ; 132(2): 311-22, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15604100

RESUMO

Dlx homeobox genes, the vertebrate homologs of Distal-less, play important roles in the development of the vertebrate forebrain, craniofacial structures and limbs. Members of the Dlx gene family are also expressed in retinal ganglion cells (RGC), amacrine and horizontal cells of the developing and postnatal retina. Expression begins at embryonic day 12.5 and is maintained until late embryogenesis for Dlx1, while Dlx2 expression extends to adulthood. We have assessed the retinal phenotype of the Dlx1/Dlx2 double knockout mouse, which dies at birth. The Dlx1/2 null retina displays a reduced ganglion cell layer (GCL), with loss of differentiated RGCs due to increased apoptosis, and corresponding thinning of the optic nerve. Ectopic expression of Crx, the cone and rod photoreceptor homeobox gene, in the GCL and neuroblastic layers of the mutants may signify altered cell fate of uncommitted RGC progenitors. However, amacrine and horizontal cell differentiation is relatively unaffected in the Dlx1/2 null retina. Herein, we propose a model whereby early-born RGCs are Dlx1 and Dlx2 independent, but Dlx function is necessary for terminal differentiation of late-born RGC progenitors.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/fisiologia , Retina/embriologia , Células Ganglionares da Retina/metabolismo , Animais , Apoptose , Encéfalo/metabolismo , Bromodesoxiuridina/farmacologia , Diferenciação Celular , Proteínas de Homeodomínio/genética , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Modelos Biológicos , Mutação , Fenótipo , Retina/metabolismo , Fatores de Tempo , Fatores de Transcrição
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA