Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
bioRxiv ; 2023 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-37425940

RESUMEN

Transcription factors (TFs) bind combinatorially to genomic cis-regulatory elements (cREs), orchestrating transcription programs. While studies of chromatin state and chromosomal interactions have revealed dynamic neurodevelopmental cRE landscapes, parallel understanding of the underlying TF binding lags. To elucidate the combinatorial TF-cRE interactions driving mouse basal ganglia development, we integrated ChIP-seq for twelve TFs, H3K4me3-associated enhancer-promoter interactions, chromatin and transcriptional state, and transgenic enhancer assays. We identified TF-cREs modules with distinct chromatin features and enhancer activity that have complementary roles driving GABAergic neurogenesis and suppressing other developmental fates. While the majority of distal cREs were bound by one or two TFs, a small proportion were extensively bound, and these enhancers also exhibited exceptional evolutionary conservation, motif density, and complex chromosomal interactions. Our results provide new insights into how modules of combinatorial TF-cRE interactions activate and repress developmental expression programs and demonstrate the value of TF binding data in modeling gene regulatory wiring.

2.
Development ; 149(11)2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35695185

RESUMEN

In the developing subpallium, the fate decision between neurons and glia is driven by expression of Dlx1/2 or Olig1/2, respectively, two sets of transcription factors with a mutually repressive relationship. The mechanism by which Dlx1/2 repress progenitor and oligodendrocyte fate, while promoting transcription of genes needed for differentiation, is not fully understood. We identified a motif within DLX1 that binds RBBP4, a NuRD complex subunit. ChIP-seq studies of genomic occupancy of DLX1 and six different members of the NuRD complex show that DLX1 and NuRD colocalize to putative regulatory elements enriched near other transcription factor genes. Loss of Dlx1/2 leads to dysregulation of genome accessibility at putative regulatory elements near genes repressed by Dlx1/2, including Olig2. Consequently, heterozygosity of Dlx1/2 and Rbbp4 leads to an increase in the production of OLIG2+ cells. These findings highlight the importance of the interplay between transcription factors and chromatin remodelers in regulating cell-fate decisions.


Asunto(s)
Proteínas de Homeodominio , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2 , Diferenciación Celular/genética , Genes Homeobox , Proteínas de Homeodominio/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Development ; 149(4)2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35156680

RESUMEN

The striatum is a central regulator of behavior and motor function through the actions of D1 and D2 medium-sized spiny neurons (MSNs), which arise from a common lateral ganglionic eminence (LGE) progenitor. The molecular mechanisms of cell fate specification of these two neuronal subtypes are incompletely understood. Here, we found that deletion of murine Meis2, which is highly expressed in the LGE and derivatives, led to a large reduction in striatal MSNs due to a block in their differentiation. Meis2 directly binds to the Zfp503 and Six3 promoters and is required for their expression and specification of D1 and D2 MSNs, respectively. Finally, Meis2 expression is regulated by Dlx1/2 at least partially through the enhancer hs599 in the LGE subventricular zone. Overall, our findings define a pathway in the LGE whereby Dlx1/2 drives expression of Meis2, which subsequently promotes the fate determination of striatal D1 and D2 MSNs via Zfp503 and Six3.


Asunto(s)
Cuerpo Estriado/metabolismo , Proteínas de Homeodominio/metabolismo , Neuronas/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Proteínas de Homeodominio/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ventrículos Laterales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis , Neuronas/citología , Bulbo Olfatorio/crecimiento & desarrollo , Bulbo Olfatorio/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Factores de Transcripción/genética , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Proteína Homeobox SIX3
4.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34921112

RESUMEN

We uncovered a transcription factor (TF) network that regulates cortical regional patterning in radial glial stem cells. Screening the expression of hundreds of TFs in the developing mouse cortex identified 38 TFs that are expressed in gradients in the ventricular zone (VZ). We tested whether their cortical expression was altered in mutant mice with known patterning defects (Emx2, Nr2f1, and Pax6), which enabled us to define a cortical regionalization TF network (CRTFN). To identify genomic programming underlying this network, we performed TF ChIP-seq and chromatin-looping conformation to identify enhancer-gene interactions. To map enhancers involved in regional patterning of cortical progenitors, we performed assays for epigenomic marks and DNA accessibility in VZ cells purified from wild-type and patterning mutant mice. This integrated approach has identified a CRTFN and VZ enhancers involved in cortical regional patterning in the mouse.


Asunto(s)
Corteza Cerebral/embriología , Redes Reguladoras de Genes , Elementos Reguladores de la Transcripción , Factores de Transcripción/metabolismo , Animales , Factor de Transcripción COUP I/metabolismo , Corteza Cerebral/metabolismo , Epigenoma , Proteínas de Homeodominio/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Ratones , Factor de Transcripción PAX6/metabolismo , Factor de Transcripción 1 de la Leucemia de Células Pre-B/metabolismo , Factores de Transcripción/genética
5.
Commun Biol ; 4(1): 95, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33479483

RESUMEN

GABAergic neurons of the hypothalamus regulate many innate behaviors, but little is known about the mechanisms that control their development. We previously identified hypothalamic neurons that express the LIM homeodomain transcription factor Lhx6, a master regulator of cortical interneuron development, as sleep-promoting. In contrast to telencephalic interneurons, hypothalamic Lhx6 neurons do not undergo long-distance tangential migration and do not express cortical interneuronal markers such as Pvalb. Here, we show that Lhx6 is necessary for the survival of hypothalamic neurons. Dlx1/2, Nkx2-2, and Nkx2-1 are each required for specification of spatially distinct subsets of hypothalamic Lhx6 neurons, and that Nkx2-2+/Lhx6+ neurons of the zona incerta are responsive to sleep pressure. We further identify multiple neuropeptides that are enriched in spatially segregated subsets of hypothalamic Lhx6 neurons, and that are distinct from those seen in cortical neurons. These findings identify common and divergent molecular mechanisms by which Lhx6 controls the development of GABAergic neurons in the hypothalamus.


Asunto(s)
Diferenciación Celular , Neuronas GABAérgicas/fisiología , Redes Reguladoras de Genes , Hipotálamo/citología , Proteínas con Homeodominio LIM/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Factores de Transcripción/metabolismo , Animales , Supervivencia Celular , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/metabolismo , Hipotálamo/metabolismo , Ratones , Proteínas Nucleares , Sueño/fisiología
6.
eNeuro ; 7(6)2020.
Artículo en Inglés | MEDLINE | ID: mdl-33199411

RESUMEN

Cortical interneuron (CIN) dysfunction is thought to play a major role in neuropsychiatric conditions like epilepsy, schizophrenia and autism. It is therefore essential to understand how the development, physiology, and functions of CINs influence cortical circuit activity and behavior in model organisms such as mice and primates. While transgenic driver lines are powerful tools for studying CINs in mice, this technology is limited in other species. An alternative approach is to use viral vectors such as AAV, which can be used in multiple species including primates and also have potential for therapeutic use in humans. Thus, we sought to discover gene regulatory enhancer elements (REs) that can be used in viral vectors to drive expression in specific cell types. The present study describes the systematic genome-wide identification of putative REs (pREs) that are preferentially active in immature CINs by histone modification chromatin immunoprecipitation and sequencing (ChIP-seq). We evaluated two novel pREs in AAV vectors, alongside the well-established Dlx I12b enhancer, and found that they drove CIN-specific reporter expression in adult mice. We also showed that the identified Arl4d pRE could drive sufficient expression of channelrhodopsin for optogenetic rescue of behavioral deficits in the Dlx5/6+/- mouse model of fast-spiking CIN dysfunction.


Asunto(s)
Trastorno Autístico , Interneuronas , Elementos Reguladores de la Transcripción , Esquizofrenia , Animales , Animales Modificados Genéticamente , Dependovirus , Vectores Genéticos , Ratones , Factores de Transcripción
7.
Elife ; 92020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32452758

RESUMEN

​Maf (c-Maf) and Mafb transcription factors (TFs) have compensatory roles in repressing somatostatin (SST+) interneuron (IN) production in medial ganglionic eminence (MGE) secondary progenitors in mice. Maf and Mafb conditional deletion (cDKO) decreases the survival of MGE-derived cortical interneurons (CINs) and changes their physiological properties. Herein, we show that (1) Mef2c and Snap25 are positively regulated by Maf and Mafb to drive IN morphological maturation; (2) Maf and Mafb promote Mef2c expression which specifies parvalbumin (PV+) INs; (3) Elmo1, Igfbp4 and Mef2c are candidate markers of immature PV+ hippocampal INs (HIN). Furthermore, Maf/Mafb neonatal cDKOs have decreased CINs and increased HINs, that express Pnoc, an HIN specific marker. Our findings not only elucidate key gene targets of Maf and Mafb that control IN development, but also identify for the first time TFs that differentially regulate CIN vs. HIN production.


Asunto(s)
Regulación de la Expresión Génica , Interneuronas/metabolismo , Factor de Transcripción MafB/fisiología , Proteínas Proto-Oncogénicas c-maf/fisiología , Animales , Femenino , Factores de Transcripción MEF2/metabolismo , Ratones , Enfermedades del Sistema Nervioso/etiología , Embarazo , Precursores de Proteínas/genética , Receptores CXCR4/metabolismo , Receptores Opioides/genética , Análisis de la Célula Individual , Proteína 25 Asociada a Sinaptosomas/metabolismo , Transcriptoma
8.
Cell Rep ; 28(8): 2048-2063.e8, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31433982

RESUMEN

DLX transcription factors (TFs) are master regulators of the developing vertebrate brain, driving forebrain GABAergic neuronal differentiation. Ablation of Dlx1&2 alters expression of genes that are critical for forebrain GABAergic development. We integrated epigenomic and transcriptomic analyses, complemented with in situ hybridization (ISH), and in vivo and in vitro studies of regulatory element (RE) function. This revealed the DLX-organized gene regulatory network at genomic, cellular, and spatial levels in mouse embryonic basal ganglia. DLX TFs perform dual activating and repressing functions; the consequences of their binding were determined by the sequence and genomic context of target loci. Our results reveal and, in part, explain the paradox of widespread DLX binding contrasted with a limited subset of target loci that are sensitive at the epigenomic and transcriptomic level to Dlx1&2 ablation. The regulatory properties identified here for DLX TFs suggest general mechanisms by which TFs orchestrate dynamic expression programs underlying neurodevelopment.


Asunto(s)
Neuronas GABAérgicas/metabolismo , Redes Reguladoras de Genes , Genoma , Proteínas de Homeodominio/metabolismo , Prosencéfalo/embriología , Factores de Transcripción/metabolismo , Transcripción Genética , Animales , Secuencia de Bases , Cromatina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Sitios Genéticos , Ratones , Modelos Genéticos , Regiones Promotoras Genéticas/genética , Unión Proteica , Reproducibilidad de los Resultados
9.
Cereb Cortex ; 29(6): 2653-2667, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29878134

RESUMEN

Immature neurons generated by the subpallial MGE tangentially migrate to the cortex where they become parvalbumin-expressing (PV+) and somatostatin (SST+) interneurons. Here, we show that the Sp9 transcription factor controls the development of MGE-derived cortical interneurons. SP9 is expressed in the MGE subventricular zone and in MGE-derived migrating interneurons. Sp9 null and conditional mutant mice have approximately 50% reduction of MGE-derived cortical interneurons, an ectopic aggregation of MGE-derived neurons in the embryonic ventral telencephalon, and an increased ratio of SST+/PV+ cortical interneurons. RNA-Seq and SP9 ChIP-Seq reveal that SP9 regulates MGE-derived cortical interneuron development through controlling the expression of key transcription factors Arx, Lhx6, Lhx8, Nkx2-1, and Zeb2 involved in interneuron development, as well as genes implicated in regulating interneuron migration Ackr3, Epha3, and St18. Thus, Sp9 has a central transcriptional role in MGE-derived cortical interneuron development.


Asunto(s)
Corteza Cerebral/citología , Interneuronas/citología , Eminencia Media/embriología , Neurogénesis/fisiología , Proteínas de Unión al ARN/metabolismo , Animales , Movimiento Celular/fisiología , Corteza Cerebral/embriología , Interneuronas/metabolismo , Eminencia Media/citología , Ratones , Factores de Transcripción/metabolismo
10.
Front Neurosci ; 12: 571, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30186101

RESUMEN

Epigenetic factors (EFs) regulate multiple aspects of cerebral cortex development, including proliferation, differentiation, laminar fate, and regional identity. The same neurodevelopmental processes are also regulated by transcription factors (TFs), notably the Pax6→ Tbr2→ Tbr1 cascade expressed sequentially in radial glial progenitors (RGPs), intermediate progenitors, and postmitotic projection neurons, respectively. Here, we studied the EF landscape and its regulation in embryonic mouse neocortex. Microarray and in situ hybridization assays revealed that many EF genes are expressed in specific cortical cell types, such as intermediate progenitors, or in rostrocaudal gradients. Furthermore, many EF genes are directly bound and transcriptionally regulated by Pax6, Tbr2, or Tbr1, as determined by chromatin immunoprecipitation-sequencing and gene expression analysis of TF mutant cortices. Our analysis demonstrated that Pax6, Tbr2, and Tbr1 form a direct feedforward genetic cascade, with direct feedback repression. Results also revealed that each TF regulates multiple EF genes that control DNA methylation, histone marks, chromatin remodeling, and non-coding RNA. For example, Tbr1 activates Rybp and Auts2 to promote the formation of non-canonical Polycomb repressive complex 1 (PRC1). Also, Pax6, Tbr2, and Tbr1 collectively drive massive changes in the subunit isoform composition of BAF chromatin remodeling complexes during differentiation: for example, a novel switch from Bcl7c (Baf40c) to Bcl7a (Baf40a), the latter directly activated by Tbr2. Of 11 subunits predominantly in neuronal BAF, 7 were transcriptionally activated by Pax6, Tbr2, or Tbr1. Using EFs, Pax6→ Tbr2→ Tbr1 effect persistent changes of gene expression in cell lineages, to propagate features such as regional and laminar identity from progenitors to neurons.

11.
Development ; 145(14)2018 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-29967281

RESUMEN

Dopamine receptor DRD1-expressing medium spiny neurons (D1 MSNs) and dopamine receptor DRD2-expressing medium spiny neurons (D2 MSNs) are the principal projection neurons in the striatum, which is divided into dorsal striatum (caudate nucleus and putamen) and ventral striatum (nucleus accumbens and olfactory tubercle). Progenitors of these neurons arise in the lateral ganglionic eminence (LGE). Using conditional deletion, we show that mice lacking the transcription factor genes Sp8 and Sp9 lose virtually all D2 MSNs as a result of reduced neurogenesis in the LGE, whereas D1 MSNs are largely unaffected. SP8 and SP9 together drive expression of the transcription factor Six3 in a spatially restricted domain of the LGE subventricular zone. Conditional deletion of Six3 also prevents the formation of most D2 MSNs, phenocopying the Sp8/9 mutants. Finally, ChIP-Seq reveals that SP9 directly binds to the promoter and a putative enhancer of Six3 Thus, this study defines components of a transcription pathway in a regionally restricted LGE progenitor domain that selectively drives the generation of D2 MSNs.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas del Ojo/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Unión al ADN/genética , Proteínas del Ojo/genética , Proteínas de Homeodominio/genética , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/citología , Neuronas/citología , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Factores de Transcripción/genética , Proteína Homeobox SIX3
12.
Cereb Cortex ; 28(11): 3797-3815, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29028947

RESUMEN

The postnatal functions of the Dlx1&2 transcription factors in cortical interneurons (CINs) are unknown. Here, using conditional Dlx1, Dlx2, and Dlx1&2 knockouts (CKOs), we defined their roles in specific CINs. The CKOs had dendritic, synaptic, and survival defects, affecting even PV+ CINs. We provide evidence that DLX2 directly drives Gad1, Gad2, and Vgat expression, and show that mutants had reduced mIPSC amplitude. In addition, the mutants formed fewer GABAergic synapses on excitatory neurons and had reduced mIPSC frequency. Furthermore, Dlx1/2 CKO had hypoplastic dendrites, fewer excitatory synapses, and reduced excitatory input. We provide evidence that some of these phenotypes were due to reduced expression of GRIN2B (a subunit of the NMDA receptor), a high confidence Autism gene. Thus, Dlx1&2 coordinate key components of CIN postnatal development by promoting their excitability, inhibitory output, and survival.


Asunto(s)
Corteza Cerebral/crecimiento & desarrollo , Neuronas GABAérgicas/fisiología , Proteínas de Homeodominio/fisiología , Interneuronas/fisiología , Sinapsis/fisiología , Factores de Transcripción/fisiología , Ácido gamma-Aminobutírico/biosíntesis , Animales , Corteza Cerebral/citología , Femenino , Neuronas GABAérgicas/citología , Regulación del Desarrollo de la Expresión Génica , Glutamato Descarboxilasa/metabolismo , Proteínas de Homeodominio/genética , Interneuronas/citología , Masculino , Ratones Noqueados , Potenciales Postsinápticos Miniatura , Factores de Transcripción/genética , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo
13.
Development ; 144(15): 2837-2851, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28694260

RESUMEN

Distinct cortical interneuron (CIN) subtypes have unique circuit functions; dysfunction in specific subtypes is implicated in neuropsychiatric disorders. Somatostatin- and parvalbumin-expressing (SST+ and PV+) interneurons are the two major subtypes generated by medial ganglionic eminence (MGE) progenitors. Spatial and temporal mechanisms governing their cell-fate specification and differential integration into cortical layers are largely unknown. We provide evidence that Coup-TF1 and Coup-TF2 (Nr2f1 and Nr2f2) transcription factor expression in an arc-shaped progenitor domain within the MGE promotes time-dependent survival of this neuroepithelium and the time-dependent specification of layer V SST+ CINs. Coup-TF1 and Coup-TF2 autonomously repress PV+ fate in MGE progenitors, in part through directly driving Sox6 expression. These results have identified, in mouse, a transcriptional pathway that controls SST-PV fate.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Factor de Transcripción COUP I/metabolismo , Interneuronas/metabolismo , Neocórtex/citología , Animales , Factor de Transcripción COUP I/genética , Factor de Transcripción COUP II/genética , Células Cultivadas , Inmunoprecipitación de Cromatina , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Parvalbúminas/genética , Parvalbúminas/metabolismo , Factores de Transcripción SOXD/genética , Factores de Transcripción SOXD/metabolismo , Somatostatina/genética , Somatostatina/metabolismo
14.
Neuron ; 92(1): 59-74, 2016 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-27710791

RESUMEN

Elucidating the transcriptional circuitry controlling forebrain development requires an understanding of enhancer activity and regulation. We generated stable transgenic mouse lines that express CreERT2 and GFP from ten different enhancer elements with activity in distinct domains within the embryonic basal ganglia. We used these unique tools to generate a comprehensive regional fate map of the mouse subpallium, including sources for specific subtypes of amygdala neurons. We then focused on deciphering transcriptional mechanisms that control enhancer activity. Using machine-learning computations, in vivo chromosomal occupancy of 13 transcription factors that regulate subpallial patterning and differentiation and analysis of enhancer activity in Dlx1/2 and Lhx6 mutants, we elucidated novel molecular mechanisms that regulate region-specific enhancer activity in the developing brain. Thus, these subpallial enhancer transgenic lines are data and tool resources to study transcriptional regulation of GABAergic cell fate.


Asunto(s)
Diferenciación Celular/genética , Elementos de Facilitación Genéticos/genética , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Animales , Ganglios Basales/crecimiento & desarrollo , Proteínas de Homeodominio/genética , Proteínas con Homeodominio LIM/genética , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
Neuron ; 88(6): 1192-1207, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26671461

RESUMEN

We demonstrate using conditional mutagenesis that Pbx1, with and without Pbx2(+/-) sensitization, regulates regional identity and laminar patterning of the developing mouse neocortex in cortical progenitors (Emx1-Cre) and in newly generated neurons (Nex1-Cre). Pbx1/2 mutants have three salient molecular phenotypes of cortical regional and laminar organization: hypoplasia of the frontal cortex, ventral expansion of the dorsomedial cortex, and ventral expansion of Reelin expression in the cortical plate of the frontal cortex, concomitant with an inversion of cortical layering in the rostral cortex. Molecular analyses, including PBX ChIP-seq, provide evidence that PBX promotes frontal cortex identity by repressing genes that promote dorsocaudal fate.


Asunto(s)
Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Proteínas de Homeodominio/fisiología , Células-Madre Neurales/fisiología , Neuronas/fisiología , Factores de Transcripción/fisiología , Animales , Animales Recién Nacidos , Ratones , Ratones Transgénicos , Mitosis/fisiología , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Proteína Reelina , Células Madre/fisiología
16.
Cell Rep ; 12(3): 482-94, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26166575

RESUMEN

The Otx2 homeodomain transcription factor is essential for gastrulation and early neural development. We generated Otx2 conditional knockout (cKO) mice to investigate its roles in telencephalon development after neurulation (approximately embryonic day 9.0). We conducted transcriptional profiling and in situ hybridization to identify genes de-regulated in Otx2 cKO ventral forebrain. In parallel, we used chromatin immunoprecipitation sequencing to identify enhancer elements, the OTX2 binding motif, and de-regulated genes that are likely direct targets of OTX2 transcriptional regulation. We found that Otx2 was essential in septum specification, regulation of Fgf signaling in the rostral telencephalon, and medial ganglionic eminence (MGE) patterning, neurogenesis, and oligodendrogenesis. Within the MGE, Otx2 was required for ventral, but not dorsal, identity, thus controlling the production of specific MGE derivatives.


Asunto(s)
Corteza Cerebral/embriología , Factores de Transcripción Otx/genética , Factores de Transcripción Otx/metabolismo , Animales , Corteza Cerebral/citología , Femenino , Expresión Génica , Ratones
17.
Neuron ; 84(5): 940-53, 2014 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-25467980

RESUMEN

Little is known about genetic mechanisms that regulate the ratio of cortical excitatory and inhibitory neurons. We show that NPAS1 and NPAS3 transcription factors (TFs) are expressed in progenitor domains of the mouse basal ganglia (subpallium, MGE, and CGE). NPAS1(-/-) mutants had increased proliferation, ERK signaling, and expression of Arx in the MGE and CGE. NPAS1(-/-) mutants also had increased neocortical inhibition (sIPSC and mIPSC) and generated an excess of somatostatin(+) (SST) (MGE-derived) and vasoactive intestinal polypeptide(+) (VIP) (CGE-derived) neocortical interneurons, but had a normal density of parvalbumin(+) (PV) (MGE-derived) interneurons. In contrast, NPAS3(-/-) mutants showed decreased proliferation and ERK signaling in progenitors of the ganglionic eminences and had fewer SST(+) and VIP(+) interneurons. NPAS1 repressed activity of an Arx enhancer, and Arx overexpression resulted in increased proliferation of CGE progenitors. These results provide insights into genetic regulation of cortical interneuron numbers and cortical inhibitory tone.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Corteza Cerebral/citología , Regulación del Desarrollo de la Expresión Génica/fisiología , Interneuronas/clasificación , Interneuronas/fisiología , Proteínas del Tejido Nervioso/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Trastorno Autístico/genética , Trastorno Autístico/patología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proliferación Celular/genética , Células Cultivadas , Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Humanos , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Polimorfismo de Nucleótido Simple/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
18.
Neuron ; 82(5): 989-1003, 2014 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-24814534

RESUMEN

Elucidating the genetic control of cerebral cortical (pallial) development is essential for understanding function, evolution, and disorders of the brain. Transcription factors (TFs) that embryonically regulate pallial regionalization are expressed in gradients, raising the question of how discrete domains are generated. We provide evidence that small enhancer elements active in protodomains integrate broad transcriptional information. CreER(T2) and GFP expression from 14 different enhancer elements in stable transgenic mice allowed us to define a comprehensive regional fate map of the pallium. We explored transcriptional mechanisms that control the activity of the enhancers using informatics, in vivo occupancy by TFs that regulate cortical patterning (CoupTFI, Pax6, and Pbx1), and analysis of enhancer activity in Pax6 mutants. Overall, the results provide insights into how broadly expressed patterning TFs regulate the activity of small enhancer elements that drive gene expression in pallial protodomains that fate map to distinct cortical regions.


Asunto(s)
Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Transcripción Genética , Animales , Sitios de Unión , Factor de Transcripción COUP I/metabolismo , Proteínas del Ojo/metabolismo , Hipocampo/embriología , Hipocampo/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Ratones Transgénicos , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/metabolismo , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo
19.
Neuron ; 77(1): 83-98, 2013 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-23312518

RESUMEN

Mammalian pallial (cortical and hippocampal) and striatal interneurons are both generated in the embryonic subpallium, including the medial ganglionic eminence (MGE). Herein we demonstrate that the Zfhx1b (Sip1, Zeb2) zinc finger homeobox gene is required in the MGE, directly downstream of Dlx1&2, to generate cortical interneurons that express Cxcr7, MafB, and cMaf. In its absence, Nkx2-1 expression is not repressed, and cells that ordinarily would become cortical interneurons appear to transform toward a subtype of GABAergic striatal interneurons. These results show that Zfhx1b is required to generate cortical interneurons, and suggest a mechanism for the epilepsy observed in humans with Zfhx1b mutations (Mowat-Wilson syndrome).


Asunto(s)
Corteza Cerebral/embriología , Cuerpo Estriado/embriología , Proteínas de Homeodominio/biosíntesis , Interneuronas/fisiología , Proteínas Represoras/biosíntesis , Factores de Transcripción/biosíntesis , Animales , Animales Recién Nacidos , Secuencia de Bases , Células Cultivadas , Corteza Cerebral/crecimiento & desarrollo , Cuerpo Estriado/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Genes Homeobox/genética , Proteínas de Homeodominio/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Neurogénesis/fisiología , Proteínas Represoras/genética , Factores de Transcripción/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc
20.
Nucleic Acids Res ; 37(21): 7151-62, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19786495

RESUMEN

The conserved mRNA export receptor NXF1 (Mex67 in yeast) assembles with messenger ribonucleoproteins (mRNP) in the nucleus and guides them through the nuclear pore complex into the cytoplasm. The DEAD family RNA helicase Dbp5 is essential for nuclear export of mRNA and is thought to dissociate Mex67 from mRNP upon translocation, thereby generating directional passage. However, the molecular mechanism by which Dbp5 recognizes Mex67-containing mRNP is not clear. Here we report that the human NXF1-binding protein RBM15 binds specifically to human DBP5 and facilitates its direct contact with mRNA in vivo. We found that RBM15 is targeted to the nuclear envelope, where it colocalizes extensively with DBP5 and NXF1. Gene silencing of RBM15 leads to cytoplasmic depletion and nuclear accumulation of general mRNA as well as individual endogenous transcripts, indicating that RBM15 is required for efficient mRNA export. We propose a model in which RBM15 acts locally at the nuclear pore complex, by facilitating the recognition of NXF1-mRNP complexes by DBP5 during translocation, thereby contributing to efficient mRNA export.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Transporte de ARN , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Línea Celular , Humanos , Membrana Nuclear/metabolismo , Interferencia de ARN , Empalme del ARN , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Ribonucleoproteínas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA