Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Cell ; 155(3): 621-35, 2013 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-24243019

RESUMEN

Direct lineage reprogramming is a promising approach for human disease modeling and regenerative medicine, with poorly understood mechanisms. Here, we reveal a hierarchical mechanism in the direct conversion of fibroblasts into induced neuronal (iN) cells mediated by the transcription factors Ascl1, Brn2, and Myt1l. Ascl1 acts as an "on-target" pioneer factor by immediately occupying most cognate genomic sites in fibroblasts. In contrast, Brn2 and Myt1l do not access fibroblast chromatin productively on their own; instead, Ascl1 recruits Brn2 to Ascl1 sites genome wide. A unique trivalent chromatin signature in the host cells predicts the permissiveness for Ascl1 pioneering activity among different cell types. Finally, we identified Zfp238 as a key Ascl1 target gene that can partially substitute for Ascl1 during iN cell reprogramming. Thus, a precise match between pioneer factors and the chromatin context at key target genes is determinative for transdifferentiation to neurons and likely other cell types.


Asunto(s)
Reprogramación Celular , Embrión de Mamíferos/citología , Fibroblastos/citología , Redes Reguladoras de Genes , Neuronas/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Cromatina/metabolismo , Fibroblastos/metabolismo , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Factores del Dominio POU/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo
2.
Genes Dev ; 27(16): 1769-86, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23964093

RESUMEN

The majority of neural stem cells (NSCs) in the adult brain are quiescent, and this fraction increases with aging. Although signaling pathways that promote NSC quiescence have been identified, the transcriptional mechanisms involved are mostly unknown, largely due to lack of a cell culture model. In this study, we first demonstrate that NSC cultures (NS cells) exposed to BMP4 acquire cellular and transcriptional characteristics of quiescent cells. We then use epigenomic profiling to identify enhancers associated with the quiescent NS cell state. Motif enrichment analysis of these enhancers predicts a major role for the nuclear factor one (NFI) family in the gene regulatory network controlling NS cell quiescence. Interestingly, we found that the family member NFIX is robustly induced when NS cells enter quiescence. Using genome-wide location analysis and overexpression and silencing experiments, we demonstrate that NFIX has a major role in the induction of quiescence in cultured NSCs. Transcript profiling of NS cells overexpressing or silenced for Nfix and the phenotypic analysis of the hippocampus of Nfix mutant mice suggest that NFIX controls the quiescent state by regulating the interactions of NSCs with their microenvironment.


Asunto(s)
Epigénesis Genética , Factores de Transcripción NFI/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Animales , Proteína Morfogenética Ósea 4/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Elementos de Facilitación Genéticos , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Ratones , Factores de Transcripción NFI/genética , Células-Madre Neurales/efectos de los fármacos , Unión Proteica
3.
Genes Dev ; 25(9): 930-45, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21536733

RESUMEN

Proneural genes such as Ascl1 are known to promote cell cycle exit and neuronal differentiation when expressed in neural progenitor cells. The mechanisms by which proneural genes activate neurogenesis--and, in particular, the genes that they regulate--however, are mostly unknown. We performed a genome-wide characterization of the transcriptional targets of Ascl1 in the embryonic brain and in neural stem cell cultures by location analysis and expression profiling of embryos overexpressing or mutant for Ascl1. The wide range of molecular and cellular functions represented among these targets suggests that Ascl1 directly controls the specification of neural progenitors as well as the later steps of neuronal differentiation and neurite outgrowth. Surprisingly, Ascl1 also regulates the expression of a large number of genes involved in cell cycle progression, including canonical cell cycle regulators and oncogenic transcription factors. Mutational analysis in the embryonic brain and manipulation of Ascl1 activity in neural stem cell cultures revealed that Ascl1 is indeed required for normal proliferation of neural progenitors. This study identified a novel and unexpected activity of the proneural gene Ascl1, and revealed a direct molecular link between the phase of expansion of neural progenitors and the subsequent phases of cell cycle exit and neuronal differentiation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis , Telencéfalo/citología , Telencéfalo/embriología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular , Línea Celular , Proliferación Celular , Células Cultivadas , Femenino , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Estudio de Asociación del Genoma Completo , Ratones , Embarazo
4.
Genome Res ; 25(1): 41-56, 2015 01.
Artículo en Inglés | MEDLINE | ID: mdl-25294244

RESUMEN

The gene regulatory network (GRN) that supports neural stem cell (NS cell) self-renewal has so far been poorly characterized. Knowledge of the central transcription factors (TFs), the noncoding gene regulatory regions that they bind to, and the genes whose expression they modulate will be crucial in unlocking the full therapeutic potential of these cells. Here, we use DNase-seq in combination with analysis of histone modifications to identify multiple classes of epigenetically and functionally distinct cis-regulatory elements (CREs). Through motif analysis and ChIP-seq, we identify several of the crucial TF regulators of NS cells. At the core of the network are TFs of the basic helix-loop-helix (bHLH), nuclear factor I (NFI), SOX, and FOX families, with CREs often densely bound by several of these different TFs. We use machine learning to highlight several crucial regulatory features of the network that underpin NS cell self-renewal and multipotency. We validate our predictions by functional analysis of the bHLH TF OLIG2. This TF makes an important contribution to NS cell self-renewal by concurrently activating pro-proliferation genes and preventing the untimely activation of genes promoting neuronal differentiation and stem cell quiescence.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular , Células Cultivadas , Análisis por Conglomerados , Epigenómica , Modelos Logísticos , Ratones , Análisis por Micromatrices , Modelos Teóricos , Factores de Transcripción NFI/genética , Factores de Transcripción NFI/metabolismo , Proteínas del Tejido Nervioso/genética , Factor de Transcripción 2 de los Oligodendrocitos , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción SOX/genética , Factores de Transcripción SOX/metabolismo , Análisis de Secuencia de ADN
5.
EMBO Rep ; 16(9): 1177-91, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26265007

RESUMEN

Embryonic stem cell (ESC) identity is orchestrated by co-operativity between the transcription factors (TFs) Sox2 and the class V POU-TF Oct4 at composite Sox/Oct motifs. Neural stem cells (NSCs) lack Oct4 but express Sox2 and class III POU-TFs Oct6, Brn1 and Brn2. This raises the question of how Sox2 interacts with POU-TFs to transcriptionally specify ESCs versus NSCs. Here, we show that Oct4 alone binds the Sox/Oct motif and the octamer-containing palindromic MORE equally well. Sox2 binding selectively increases the affinity of Oct4 for the Sox/Oct motif. In contrast, Oct6 binds preferentially to MORE and is unaffected by Sox2. ChIP-Seq in NSCs shows the MORE to be the most enriched motif for class III POU-TFs, including MORE subtypes, and that the Sox/Oct motif is not enriched. These results suggest that in NSCs, co-operativity between Sox2 and class III POU-TFs may not occur and that POU-TF-driven transcription uses predominantly the MORE cis architecture. Thus, distinct interactions between Sox2 and POU-TF subclasses distinguish pluripotent ESCs from multipotent NSCs, providing molecular insight into how Oct4 alone can convert NSCs to pluripotency.


Asunto(s)
Células Madre Embrionarias/metabolismo , Células-Madre Neurales/metabolismo , Factores del Dominio POU/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Animales , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Ratones , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Factores del Dominio POU/genética , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Transcripción Genética
6.
Cereb Cortex ; 25(10): 3758-78, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25331604

RESUMEN

Transcription factors of the nuclear factor one (NFI) family play a pivotal role in the development of the nervous system. One member, NFIX, regulates the development of the neocortex, hippocampus, and cerebellum. Postnatal Nfix(-/-) mice also display abnormalities within the subventricular zone (SVZ) lining the lateral ventricles, a region of the brain comprising a neurogenic niche that provides ongoing neurogenesis throughout life. Specifically, Nfix(-/-) mice exhibit more PAX6-expressing progenitor cells within the SVZ. However, the mechanism underlying the development of this phenotype remains undefined. Here, we reveal that NFIX contributes to multiple facets of SVZ development. Postnatal Nfix(-/-) mice exhibit increased levels of proliferation within the SVZ, both in vivo and in vitro as assessed by a neurosphere assay. Furthermore, we show that the migration of SVZ-derived neuroblasts to the olfactory bulb is impaired, and that the olfactory bulbs of postnatal Nfix(-/-) mice are smaller. We also demonstrate that gliogenesis within the rostral migratory stream is delayed in the absence of Nfix, and reveal that Gdnf (glial-derived neurotrophic factor), a known attractant for SVZ-derived neuroblasts, is a target for transcriptional activation by NFIX. Collectively, these findings suggest that NFIX regulates both proliferation and migration during the development of the SVZ neurogenic niche.


Asunto(s)
Movimiento Celular , Proliferación Celular , Ventrículos Laterales/embriología , Factores de Transcripción NFI/fisiología , Células-Madre Neurales/fisiología , Neurogénesis , Animales , Femenino , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Interneuronas/fisiología , Ventrículos Laterales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción NFI/genética , Factores de Transcripción NFI/metabolismo , Neuroglía/fisiología , Bulbo Olfatorio/embriología , Bulbo Olfatorio/metabolismo , Nicho de Células Madre
8.
Cereb Cortex ; 24(11): 2822-34, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23720416

RESUMEN

We aimed to identify cis-regulatory elements that control gene expression in progenitors of the cerebral cortex. A list of 975 putative enhancers were retrieved from a ChIP-Seq experiment performed in NS5 mouse stem cells with antibodies to Sox2, Brn2/Pou3f2, or Brn1/Pou3f3. Through a selection pipeline including gene ontology and expression pattern, we reduced the number of candidate enhancer sequences to 20. Ex vivo electroporation of green fluorescent pProtein (GFP) reporter constructs in the telencephalon of mouse embryos showed that 35% of the 20 selected candidate sequences displayed enhancer activity in the developing cortex at E13.5. In silico transcription factor binding site (TFBS) searches and mutagenesis experiments showed that enhancer activity is related to the presence of Sox/Pou TFBS pairs in the sequence. Comparative genomic analyses showed that enhancer activity is not related to the evolutionary conservation of the sequence. Finally, the combination of in utero electroporation of GFP reporter constructs with immunostaining for Tbr2 (basal progenitor marker) and phospho-histoneH3 (mitotic activity marker) demonstrated that each enhancer is specifically active in precise subpopulations of progenitors in the cortical germinal zone, highlighting the heterogeneity of these progenitors in terms of cis-regulation.


Asunto(s)
Corteza Cerebral/citología , Corteza Cerebral/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Factores del Dominio POU/metabolismo , Factores de Transcripción SOXB1/metabolismo , Células Madre/fisiología , Animales , Sitios de Unión/genética , Evolución Biológica , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular , Embrión de Mamíferos , Femenino , Histonas/genética , Histonas/metabolismo , Técnicas In Vitro , Ratones , Ratones Transgénicos , Mutagénesis/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Técnicas de Cultivo de Órganos , Factores del Dominio POU/genética , Embarazo , Factores de Transcripción SOXB1/genética , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo
10.
Development ; 137(3): 487-97, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20081193

RESUMEN

Foxg1 is required for development of the ventral telencephalon in the embryonic mammalian forebrain. Although one existing hypothesis suggests that failed ventral telencephalic development in the absence of Foxg1 is due to reduced production of the morphogens sonic hedgehog (Shh) and fibroblast growth factor 8 (Fgf8), the possibility that telencephalic cells lacking Foxg1 are intrinsically incompetent to generate the ventral telencephalon has remained untested. We examined the ability of Foxg1(-/-) telencephalic cells to respond to Shh and Fgf8 by examining the expression of genes whose activation requires Shh or Fgf8 in vivo and by testing their responses to Shh and Fgf8 in culture. We found that many elements of the Shh and Fgf8 signalling pathways continue to function in the absence of Foxg1 but, nevertheless, we were unable to elicit normal responses of key ventral telencephalic marker genes in Foxg1(-/-) telencephalic tissue following a range of in vivo and in vitro manipulations. We explored the development of Foxg1(-/-) cells in Foxg1(-/-) Foxg1(+/+) chimeric embryos that contained ventral telencephalon created by normally patterned wild-type cells. We found that Foxg1(-/-) cells contributed to the chimeric ventral telencephalon, but that they retained abnormal specification, expressing dorsal rather than ventral telencephalic markers. These findings indicate that, in addition to regulating the production of ventralising signals, Foxg1 acts cell-autonomously in the telencephalon to ensure that cells develop the competence to adopt ventral identities.


Asunto(s)
Factores de Transcripción Forkhead/fisiología , Proteínas del Tejido Nervioso/fisiología , Telencéfalo/citología , Animales , Células Cultivadas , Factor 8 de Crecimiento de Fibroblastos/fisiología , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/fisiología , Factores de Transcripción de Tipo Kruppel , Ratones , Ratones Noqueados , Telencéfalo/embriología , Telencéfalo/crecimiento & desarrollo , Factores de Transcripción , Proteína Gli3 con Dedos de Zinc
11.
Cell Mol Life Sci ; 68(10): 1769-83, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-20981563

RESUMEN

Neural stem (NS) cells are a self-renewing population of symmetrically dividing multipotent radial glia-like stem cells, characterized by homogeneous expansion in monolayer. Here we report that fetal NS cells isolated from different regions of the developing mouse nervous system behave in a similar manner with respect to self-renewal and neuropotency, but exhibit distinct positional identities. For example, NS cells from the neocortex maintain the expression of anterior transcription factors, including Otx2 and Foxg1, while Hoxb4 and Hoxb9 are uniquely found in spinal cord-derived NS cells. This molecular signature was stable for over 20 passages and was strictly linked to the developmental stage of the donor, because only NS cells derived from E14.5 cortex, and not those derived from E12.5 cortex, carried a consistent transcription factor profile. We also showed that traits of this positional code are maintained during neuronal differentiation, leading to the generation of electrophysiologically active neurons, even if they do not acquire a complete neurochemical identity.


Asunto(s)
Feto/citología , Células-Madre Neurales/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Neocórtex/citología , Neocórtex/embriología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factores de Transcripción Otx/genética , Factores de Transcripción Otx/metabolismo , Técnicas de Placa-Clamp , Médula Espinal/citología , Médula Espinal/embriología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
iScience ; 15: 257-273, 2019 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-31082736

RESUMEN

Visual system development involves the formation of neuronal projections connecting the retina to the thalamic dorso-lateral geniculate nucleus (dLGN) and the thalamus to the visual cerebral cortex. Patients carrying mutations in the SOX2 transcription factor gene present severe visual defects, thought to be linked to SOX2 functions in the retina. We show that Sox2 is strongly expressed in mouse postmitotic thalamic projection neurons. Cre-mediated deletion of Sox2 in these neurons causes reduction of the dLGN, abnormal distribution of retino-thalamic and thalamo-cortical projections, and secondary defects in cortical patterning. Reduced expression, in mutants, of Sox2 target genes encoding ephrin-A5 and the serotonin transport molecules SERT and vMAT2 (important for establishment of thalamic connectivity) likely provides a molecular contribution to these defects. These findings unveil thalamic SOX2 function as a novel regulator of visual system development and a plausible additional cause of brain-linked genetic blindness in humans.

14.
Cell Stem Cell ; 24(3): 462-476.e6, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30849367

RESUMEN

The SOX2 transcription factor is critical for neural stem cell (NSC) maintenance and brain development. Through chromatin immunoprecipitation (ChIP) and chromatin interaction analysis (ChIA-PET), we determined genome-wide SOX2-bound regions and Pol II-mediated long-range chromatin interactions in brain-derived NSCs. SOX2-bound DNA was highly enriched in distal chromatin regions interacting with promoters and carrying epigenetic enhancer marks. Sox2 deletion caused widespread reduction of Pol II-mediated long-range interactions and decreased gene expression. Genes showing reduced expression in Sox2-deleted cells were significantly enriched in interactions between promoters and SOX2-bound distal enhancers. Expression of one such gene, Suppressor of Cytokine Signaling 3 (Socs3), rescued the self-renewal defect of Sox2-ablated NSCs. Our work identifies SOX2 as a major regulator of gene expression through connections to the enhancer network in NSCs. Through the definition of such a connectivity network, our study shows the way to the identification of genes and enhancers involved in NSC maintenance and neurodevelopmental disorders.


Asunto(s)
Cromatina/metabolismo , Células-Madre Neurales/metabolismo , Factores de Transcripción SOXB1/metabolismo , Animales , Células Cultivadas , Redes Reguladoras de Genes/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación , Factores de Transcripción SOXB1/deficiencia , Factores de Transcripción SOXB1/genética , Pez Cebra
15.
Neuron ; 93(2): 348-361, 2017 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-28041881

RESUMEN

Mutations in NIPBL are the most frequent cause of Cornelia de Lange syndrome (CdLS), a developmental disorder encompassing several neurological defects, including intellectual disability and seizures. How NIPBL mutations affect brain development is not understood. Here we identify Nipbl as a functional interaction partner of the neural transcription factor Zfp609 in brain development. Depletion of Zfp609 or Nipbl from cortical neural progenitors in vivo is detrimental to neuronal migration. Zfp609 and Nipbl overlap at genomic binding sites independently of cohesin and regulate genes that control cortical neuron migration. We find that Zfp609 and Nipbl interact with the Integrator complex, which functions in RNA polymerase 2 pause release. Indeed, Zfp609 and Nipbl co-localize at gene promoters containing paused RNA polymerase 2, and Integrator similarly regulates neuronal migration. Our data provide a rationale and mechanistic insights for the role of Nipbl in the neurological defects associated with CdLS.


Asunto(s)
Movimiento Celular/genética , Corteza Cerebral/crecimiento & desarrollo , Síndrome de Cornelia de Lange/genética , Regulación del Desarrollo de la Expresión Génica , Células-Madre Neurales/citología , Neuronas/citología , Transactivadores/genética , Factores de Transcripción/genética , Animales , Proteínas de Ciclo Celular/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Ratones , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Regiones Promotoras Genéticas , ARN Polimerasa II/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Cohesinas
16.
Stem Cells Dev ; 24(18): 2114-26, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26083238

RESUMEN

Murine postnatal neural stem cells (NSCs) give rise to neurons, astrocytes, or oligodendrocytes (OLs); however, our knowledge of the genes that control this lineage specification is incomplete. In this study, we show that nuclear factor I X (NFIX), a transcription factor known to regulate NSC quiescence, also suppresses oligodendrogenesis (ODG) from NSCs. Immunostaining reveals little or no expression of NFIX in OL lineage cells both in vivo and in vitro. Loss of NFIX from subventricular zone (SVZ) NSCs results in enhanced ODG both in vivo and in vitro, while forced expression of NFIX blocks NSC differentiation into OLs in vitro. RNA-seq analysis shows that genes previously shown to be differentially expressed in OL progenitors are significantly enriched in RNA from Nfix(-/-) versus wild-type NSCs. These data indicate that NFIX influences the lineage specification of postnatal SVZ NSCs, specifically suppressing ODG.


Asunto(s)
Ventrículos Laterales/embriología , Factores de Transcripción NFI/genética , Células-Madre Neurales/citología , Neurogénesis/fisiología , Oligodendroglía/citología , Animales , Astrocitos/citología , Linaje de la Célula , Células Cultivadas , Ventrículos Laterales/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología , Factores de Transcripción SOXE/metabolismo
17.
Neuron ; 83(5): 1085-97, 2014 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-25189209

RESUMEN

The activity of adult stem cells is regulated by signals emanating from the surrounding tissue. Many niche signals have been identified, but it is unclear how they influence the choice of stem cells to remain quiescent or divide. Here we show that when stem cells of the adult hippocampus receive activating signals, they first induce the expression of the transcription factor Ascl1 and only subsequently exit quiescence. Moreover, lowering Ascl1 expression reduces the proliferation rate of hippocampal stem cells, and inactivating Ascl1 blocks quiescence exit completely, rendering them unresponsive to activating stimuli. Ascl1 promotes the proliferation of hippocampal stem cells by directly regulating the expression of cell-cycle regulatory genes. Ascl1 is similarly required for stem cell activation in the adult subventricular zone. Our results support a model whereby Ascl1 integrates inputs from both stimulatory and inhibitory signals and converts them into a transcriptional program activating adult neural stem cells.


Asunto(s)
Células Madre Adultas/citología , Regulación del Desarrollo de la Expresión Génica/genética , Hipocampo/citología , Neurogénesis/genética , Células Madre Adultas/metabolismo , Factores de Edad , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proliferación Celular , Ventrículos Cerebrales/citología , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 1 de Aminoácidos Excitadores/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/deficiencia , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Ácido Kaínico/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción p300-CBP/metabolismo
18.
Cell Rep ; 4(3): 477-91, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23891001

RESUMEN

FOXO transcription factors are central regulators of longevity from worms to humans. FOXO3, the FOXO isoform associated with exceptional human longevity, preserves adult neural stem cell pools. Here, we identify FOXO3 direct targets genome-wide in primary cultures of adult neural progenitor cells (NPCs). Interestingly, FOXO3-bound sites are enriched for motifs for bHLH transcription factors, and FOXO3 shares common targets with the proneuronal bHLH transcription factor ASCL1/MASH1 in NPCs. Analysis of the chromatin landscape reveals that FOXO3 and ASCL1 are particularly enriched at the enhancers of genes involved in neurogenic pathways. Intriguingly, FOXO3 inhibits ASCL1-dependent neurogenesis in NPCs and direct neuronal conversion in fibroblasts. FOXO3 also restrains neurogenesis in vivo. Our study identifies a genome-wide interaction between the prolongevity transcription factor FOXO3 and the cell-fate determinant ASCL1 and raises the possibility that FOXO3's ability to restrain ASCL1-dependent neurogenesis may help preserve the neural stem cell pool.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Animales , Sitios de Unión , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Genoma , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo
19.
Neuron ; 74(2): 209-11, 2012 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-22542173

RESUMEN

A crucial event in the birth of a neuron is the detachment of its apical process from the neuroepithelium. In this issue of Neuron, Rousso et al. (2012) show that repression of N-cadherin by Foxp transcription factors disrupts apical adherens junctions and triggers neurogenesis.

20.
Artículo en Inglés | MEDLINE | ID: mdl-23028117

RESUMEN

The mammalian nervous system is the most complex organ of any living organism. How this complexity is generated during neural development is just beginning to be elucidated. This article discusses the signaling, transcriptional, and epigenetic mechanisms that are involved in neural development. The first part focuses on molecules that control neuronal numbers through regulation of the timing of onset of neurogenesis, the timing of the neuronal-to-glial switch, and the rate of progenitor proliferation. The second part focuses on molecules that control neuronal diversity by generating spatially or temporally distinct populations of neuronal progenitors. Most of the studies discussed in this article are focused on the developing mammalian cerebral cortex, because this is one of the main model systems for neural developmental studies and many of the mechanisms identified in this tissue also operate elsewhere in the developing brain and spinal cord.


Asunto(s)
Corteza Cerebral/crecimiento & desarrollo , Epigénesis Genética , Mamíferos/crecimiento & desarrollo , Neurogénesis/fisiología , Transducción de Señal , Animales , Diferenciación Celular/genética , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Regulación del Desarrollo de la Expresión Génica , Mamíferos/genética , Mamíferos/metabolismo , Modelos Biológicos , Placa Neural/embriología , Placa Neural/metabolismo , Placa Neural/fisiología , Neurogénesis/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA