Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 175
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Genes Dev ; 37(5-6): 218-242, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36931659

RESUMEN

Pioneer transcription factors are thought to play pivotal roles in developmental processes by binding nucleosomal DNA to activate gene expression, though mechanisms through which pioneer transcription factors remodel chromatin remain unclear. Here, using single-cell transcriptomics, we show that endogenous expression of neurogenic transcription factor ASCL1, considered a classical pioneer factor, defines a transient population of progenitors in human neural differentiation. Testing ASCL1's pioneer function using a knockout model to define the unbound state, we found that endogenous expression of ASCL1 drives progenitor differentiation by cis-regulation both as a classical pioneer factor and as a nonpioneer remodeler, where ASCL1 binds permissive chromatin to induce chromatin conformation changes. ASCL1 interacts with BAF SWI/SNF chromatin remodeling complexes, primarily at targets where it acts as a nonpioneer factor, and we provide evidence for codependent DNA binding and remodeling at a subset of ASCL1 and SWI/SNF cotargets. Our findings provide new insights into ASCL1 function regulating activation of long-range regulatory elements in human neurogenesis and uncover a novel mechanism of its chromatin remodeling function codependent on partner ATPase activity.


Asunto(s)
Regulación de la Expresión Génica , Factores de Transcripción , Humanos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Diferenciación Celular/genética , Ensamble y Desensamble de Cromatina , Cromatina , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo
2.
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
3.
Genes Dev ; 33(9-10): 479-481, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31043492

RESUMEN

Adult neural stem cells are mostly quiescent and only rarely enter the cell cycle to self-renew and generate neuronal or glial progenies. The Notch signaling pathway is essential for both the quiescent and proliferative states of neural stem cells. However, these are mutually exclusive cellular states; thus, how Notch promotes both of these programs within adult neural stem cells has remained unclear. In this issue of Genes & Development, Sueda and colleagues (pp. 511-523) use an extensive repertoire of mouse genetic tools and techniques to demonstrate that it is the levels and dynamic expression of the Notch transcriptional effector Hairy and Enhancer of Split 1 that enables this dual role.


Asunto(s)
Células Madre Adultas/citología , Células-Madre Neurales/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Ciclo Celular , Ratones , Sistema Nervioso , Transducción de Señal , Factor de Transcripción HES-1
4.
Development ; 148(20)2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34557919

RESUMEN

Adult mouse hippocampal neural stem cells (NSCs) generate new neurons that integrate into existing hippocampal networks and modulate mood and memory. These NSCs are largely quiescent and are stimulated by niche signals to activate and produce neurons. Wnt/ß-catenin signalling acts at different steps along the hippocampal neurogenic lineage, but whether it has a direct role in the regulation of NSCs remains unclear. Here, we used Wnt/ß-catenin reporters and transcriptomic data from in vivo and in vitro models to show that adult NSCs respond to Wnt/ß-catenin signalling. Wnt/ß-catenin stimulation instructed the neuronal differentiation of proliferating NSCs and promoted the activation or differentiation of quiescent NSCs in a dose-dependent manner. However, deletion of ß-catenin in NSCs did not affect either their activation or maintenance of their stem cell characteristics. Together, these results indicate that, although NSCs do respond to Wnt/ß-catenin stimulation in a dose-dependent and state-specific manner, Wnt/ß-catenin signalling is not cell-autonomously required to maintain NSC homeostasis, which reconciles some of the contradictions in the literature as to the role of Wnt/ß-catenin signalling in adult hippocampal NSCs.


Asunto(s)
Homeostasis/fisiología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/fisiología , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo , Células Madre Adultas/metabolismo , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Femenino , Hipocampo/metabolismo , Masculino , Ratones , Neurogénesis/fisiología , Neuronas/metabolismo
5.
Brain ; 146(10): 4247-4261, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37082944

RESUMEN

Although the Na-K-Cl cotransporter (NKCC1) inhibitor bumetanide has prominent positive effects on the pathophysiology of many neurological disorders, the mechanism of action is obscure. Attention paid to elucidating the role of Nkcc1 has mainly been focused on neurons, but recent single cell mRNA sequencing analysis has demonstrated that the major cellular populations expressing NKCC1 in the cortex are non-neuronal. We used a combination of conditional transgenic animals, in vivo electrophysiology, two-photon imaging, cognitive behavioural tests and flow cytometry to investigate the role of Nkcc1 inhibition by bumetanide in a mouse model of controlled cortical impact (CCI). Here, we found that bumetanide rescues parvalbumin-positive interneurons by increasing interneuron-microglia contacts shortly after injury. The longitudinal phenotypic changes in microglia were significantly modified by bumetanide, including an increase in the expression of microglial-derived BDNF. These effects were accompanied by the prevention of CCI-induced decrease in hippocampal neurogenesis. Treatment with bumetanide during the first week post-CCI resulted in significant recovery of working and episodic memory as well as changes in theta band oscillations 1 month later. These results disclose a novel mechanism for the neuroprotective action of bumetanide mediated by an acceleration of microglial activation dynamics that leads to an increase in parvalbumin interneuron survival following CCI, possibly resulting from increased microglial BDNF expression and contact with interneurons. Salvage of interneurons may normalize ambient GABA, resulting in the preservation of adult neurogenesis processes as well as contributing to bumetanide-mediated improvement of cognitive performance.


Asunto(s)
Bumetanida , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico , Ratones , Animales , Bumetanida/farmacología , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/farmacología , Microglía/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Parvalbúminas/metabolismo , Parvalbúminas/farmacología , Miembro 2 de la Familia de Transportadores de Soluto 12 , Interneuronas/metabolismo , Neurogénesis
6.
Development ; 147(12)2020 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-32467237

RESUMEN

Thymus function depends on the epithelial compartment of the thymic stroma. Cortical thymic epithelial cells (cTECs) regulate T cell lineage commitment and positive selection, while medullary (m) TECs impose central tolerance on the T cell repertoire. During thymus organogenesis, these functionally distinct sub-lineages are thought to arise from a common thymic epithelial progenitor cell (TEPC). However, the mechanisms controlling cTEC and mTEC production from the common TEPC are not understood. Here, we show that emergence of the earliest mTEC lineage-restricted progenitors requires active NOTCH signaling in progenitor TEC and that, once specified, further mTEC development is NOTCH independent. In addition, we demonstrate that persistent NOTCH activity favors maintenance of undifferentiated TEPCs at the expense of cTEC differentiation. Finally, we uncover a cross-regulatory relationship between NOTCH and FOXN1, a master regulator of TEC differentiation. These data establish NOTCH as a potent regulator of TEPC and mTEC fate during fetal thymus development, and are thus of high relevance to strategies aimed at generating/regenerating functional thymic tissue in vitro and in vivo.


Asunto(s)
Desarrollo Embrionario/genética , Receptores Notch/metabolismo , Timo/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Mutación con Ganancia de Función , Regulación del Desarrollo de la Expresión Génica , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/deficiencia , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Organogénesis , Receptores Notch/genética , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Timo/citología , Timo/crecimiento & desarrollo
7.
Mol Psychiatry ; 26(12): 7280-7295, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34561615

RESUMEN

Despite the central role of Rho GTPases in neuronal development, their functions in adult hippocampal neurogenesis remain poorly explored. Here, by using a retrovirus-based loss-of-function approach in vivo, we show that the atypical Rho GTPase Rnd2 is crucial for survival, positioning, somatodendritic morphogenesis, and functional maturation of adult-born dentate granule neurons. Interestingly, most of these functions are specific to granule neurons generated during adulthood since the deletion of Rnd2 in neonatally-born granule neurons only affects dendritogenesis. In addition, suppression of Rnd2 in adult-born dentate granule neurons increases anxiety-like behavior whereas its deletion in pups has no such effect, a finding supporting the adult neurogenesis hypothesis of anxiety disorders. Thus, our results are in line with the view that adult neurogenesis is not a simple continuation of earlier processes from development, and establish a causal relationship between Rnd2 expression and anxiety.


Asunto(s)
Ansiedad , Giro Dentado , Neurogénesis , Proteínas de Unión al GTP rho/metabolismo , Animales , Ansiedad/genética , Giro Dentado/metabolismo , Ratones , Neuronas/metabolismo , Proteínas de Unión al GTP rho/genética
8.
Proc Natl Acad Sci U S A ; 116(9): 3817-3826, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30808767

RESUMEN

Midbrain dopamine neurons, which can be regulated by neuropeptides and hormones, play a fundamental role in controlling cognitive processes, reward mechanisms, and motor functions. The hormonal actions of insulin-like growth factor 1 (IGF-1) produced by the liver have been well described, but the role of neuronally derived IGF-1 remains largely unexplored. We discovered that dopamine neurons secrete IGF-1 from the cell bodies following depolarization, and that IGF-1 controls release of dopamine in the ventral midbrain. In addition, conditional deletion of dopamine neuron-derived IGF-1 in adult mice leads to decrease of dopamine content in the striatum and deficits in dopamine neuron firing and causes reduced spontaneous locomotion and impairments in explorative and learning behaviors. These data identify that dopamine neuron-derived IGF-1 acts as a regulator of dopamine neurons and regulates dopamine-mediated behaviors.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Locomoción/genética , Mesencéfalo/fisiología , Animales , Cognición/fisiología , Cuerpo Estriado/metabolismo , Cuerpo Estriado/fisiología , Neuronas Dopaminérgicas/patología , Neuronas Dopaminérgicas/fisiología , Conducta Exploratoria/fisiología , Hormonas/metabolismo , Factor I del Crecimiento Similar a la Insulina/biosíntesis , Aprendizaje/fisiología , Locomoción/fisiología , Mesencéfalo/metabolismo , Ratones , Neuropéptidos/genética
9.
Development ; 145(19)2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30201687

RESUMEN

Neural progenitors undergo temporal identity transitions to sequentially generate the neuronal and glial cells that make up the mature brain. Proneural genes have well-characterised roles in promoting neural cell differentiation and subtype specification, but they also regulate the timing of identity transitions through poorly understood mechanisms. Here, we investigated how the highly related proneural genes Neurog1 and Neurog2 interact to control the timing of neocortical neurogenesis. We found that Neurog1 acts in an atypical fashion as it is required to suppress rather than promote neuronal differentiation in early corticogenesis. In Neurog1-/- neocortices, early born neurons differentiate in excess, whereas, in vitro, Neurog1-/- progenitors have a decreased propensity to proliferate and form neurospheres. Instead, Neurog1-/- progenitors preferentially generate neurons, a phenotype restricted to the Neurog2+ progenitor pool. Mechanistically, Neurog1 and Neurog2 heterodimerise, and while Neurog1 and Neurog2 individually promote neurogenesis, misexpression together blocks this effect. Finally, Neurog1 is also required to induce the expression of neurogenic factors (Dll1 and Hes5) and to repress the expression of neuronal differentiation genes (Fezf2 and Neurod6). Neurog1 thus employs different mechanisms to temper the pace of early neocortical neurogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neocórtex/embriología , Neocórtex/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis , Neuronas/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Proliferación Celular/genética , Autorrenovación de las Células/genética , Embrión de Mamíferos/citología , Regulación del Desarrollo de la Expresión Génica , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Unión Proteica , Factores de Tiempo , Transcripción Genética
10.
Neuropathol Appl Neurobiol ; 47(6): 781-795, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33797808

RESUMEN

AIMS: We understand little of the pathogenesis of developmental cortical lesions, because we understand little of the diversity of the cell types that contribute to the diseases or how those cells interact. We tested the hypothesis that cellular diversity and cell-cell interactions play an important role in these disorders by investigating the signalling molecules in the commonest cortical malformations that lead to childhood epilepsy, focal cortical dysplasia (FCD) and tuberous sclerosis (TS). METHODS: Transcriptional profiling clustered cases into molecularly distinct groups. Using gene expression data, we identified the secretory signalling molecules in FCD/TS and characterised the cell types expressing these molecules. We developed a functional model using organotypic cultures. RESULTS: We identified 113 up-regulated secretory molecules in FCDIIB/TS. The top 12 differentially expressed genes (DEGs) were validated by immunohistochemistry. This highlighted two molecules, Chitinase 3-like protein 1 (CHI3L1) and C-C motif chemokine ligand 2 (CCL2) (MCP1) that were expressed in a unique population of small cells in close proximity to balloon cells (BC). We then characterised these cells and developed a functional model in organotypic slice cultures. We found that the number of CHI3L1 and CCL2 expressing cells decreased following inhibition of mTOR, the main aberrant signalling pathway in TS and FCD. CONCLUSIONS: Our findings highlight previously uncharacterised small cell populations in FCD and TS which express specific signalling molecules. These findings indicate a new level of diversity and cellular interactions in cortical malformations and provide a generalisable approach to understanding cell-cell interactions and cellular heterogeneity in developmental neuropathology.


Asunto(s)
Encéfalo/metabolismo , Discapacidades del Desarrollo/metabolismo , Malformaciones del Desarrollo Cortical/patología , Transducción de Señal/fisiología , Esclerosis Tuberosa/metabolismo , Encéfalo/patología , Discapacidades del Desarrollo/patología , Humanos , Inmunohistoquímica , Malformaciones del Desarrollo Cortical/metabolismo , Malformaciones del Desarrollo Cortical de Grupo I/metabolismo , Esclerosis Tuberosa/genética , Esclerosis Tuberosa/patología
11.
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
12.
Mol Psychiatry ; 23(11): 2209-2226, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29712998

RESUMEN

Perturbation of CaMKIIß expression has been associated with multiple neuropsychiatric diseases, highlighting CaMKIIß as a gene of interest. Yet, in contrast to CaMKIIα, the specific functions of CaMKIIß in the brain remain poorly explored. Here, we reveal a novel function for this CaMKII isoform in vivo during neuronal development. By using in utero electroporation, we show that CaMKIIß is an important regulator of radial migration of projection neurons during cerebral cortex development. Knockdown of CaMKIIß causes accelerated migration of nascent pyramidal neurons, whereas overexpression of CaMKIIß inhibits migration, demonstrating that precise regulation of CaMKIIß expression is required for correct neuronal migration. More precisely, CaMKIIß controls the multipolar-bipolar transition in the intermediate zone and locomotion in the cortical plate through its actin-binding and -bundling activities. In addition, our data indicate that a fine-tuned balance between CaMKIIß and cofilin activities is necessary to ensure proper migration of cortical neurons. Thus, our findings define a novel isoform-specific function for CaMKIIß, demonstrating that CaMKIIß has a major biological function in the developing brain.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Movimiento Celular/fisiología , Corteza Cerebral/fisiología , Neurogénesis/fisiología , Animales , Encéfalo/embriología , Encéfalo/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Corteza Cerebral/metabolismo , Embrión de Mamíferos/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Ratones , Proteínas de Microfilamentos/genética , Trastornos del Neurodesarrollo/genética , Neurogénesis/genética , Neuronas/metabolismo , Cultivo Primario de Células , Isoformas de Proteínas/metabolismo , Células Piramidales/metabolismo
13.
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
14.
J Neurosci ; 37(9): 2305-2316, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28130357

RESUMEN

Midbrain dopaminergic neurons are highly heterogeneous. They differ in their connectivity and firing patterns and, therefore, in their functional properties. The molecular underpinnings of this heterogeneity are largely unknown, and there is a paucity of markers that distinguish these functional subsets. In this paper, we report the identification and characterization of a novel subset of midbrain dopaminergic neurons located in the ventral tegmental area that expresses the basic helix-loop-helix transcription factor, Neurogenic Differentiation Factor-6 (NEUROD6). Retrograde fluorogold tracing experiments demonstrate that Neurod6+ midbrain dopaminergic neurons neurons project to two distinct septal regions: the dorsal and intermediate region of the lateral septum. Loss-of-function studies in mice demonstrate that Neurod6 and the closely related family member Neurod1 are both specifically required for the survival of this lateral-septum projecting neuronal subset during development. Our findings underscore the complex organization of midbrain dopaminergic neurons and provide an entry point for future studies of the functions of the Neurod6+ subset of midbrain dopaminergic neurons.SIGNIFICANCE STATEMENT Midbrain dopaminergic neurons regulate diverse brain functions, including voluntary movement and cognitive and emotive behaviors. These neurons are heterogeneous, and distinct subsets are thought to regulate different behaviors. However, we currently lack the means to identify and modify gene function in specific subsets of midbrain dopaminergic neurons. In this study, we identify the transcription factor NEUROD6 as a specific marker for a novel subset of midbrain dopaminergic neurons in the ventral midbrain that project to the lateral septum, and we reveal essential roles for Neurod1 and Neurod6 in the survival of these neurons during development. Our findings highlight the molecular and anatomical heterogeneity of midbrain dopaminergic neurons and contribute to a better understanding of this functionally complex group of neurons.


Asunto(s)
Apoptosis/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neuronas Dopaminérgicas/fisiología , Proteínas del Tejido Nervioso/metabolismo , Núcleos Septales/citología , Área Tegmental Ventral/citología , Aldehído Deshidrogenasa/metabolismo , Familia de Aldehído Deshidrogenasa 1 , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Biotina/análogos & derivados , Biotina/metabolismo , Calbindinas/metabolismo , Recuento de Células , Dextranos/metabolismo , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Transgénicos , Mutación/genética , Proteínas del Tejido Nervioso/genética , Vías Nerviosas/fisiología , Factores de Transcripción Otx/genética , Factores de Transcripción Otx/metabolismo , Retinal-Deshidrogenasa , Núcleos Septales/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Área Tegmental Ventral/embriología , Área Tegmental Ventral/crecimiento & desarrollo
15.
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
16.
Cereb Cortex ; 27(6): 3378-3396, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27600842

RESUMEN

The T-box containing Tbr2 gene encodes for a transcription factor essential for the specification of the intermediate neural progenitors (INPs) originating the excitatory neurons of the cerebral cortex. However, its overall mechanism of action, direct target genes and cofactors remain unknown. Herein, we carried out global gene expression profiling combined with genome-wide binding site identification to determine the molecular pathways regulated by TBR2 in INPs. This analysis led to the identification of novel protein-protein interactions that control multiple features of INPs including cell-type identity, morphology, proliferation and migration dynamics. In particular, NEUROG2 and JMJD3 were found to associate with TBR2 revealing unexplored TBR2-dependent mechanisms. These interactions can explain, at least in part, the role of this transcription factor in the implementation of the molecular program controlling developmental milestones during corticogenesis. These data identify TBR2 as a major determinant of the INP-specific traits by regulating both genetic and epigenetic pathways.


Asunto(s)
Diferenciación Celular/genética , Corteza Cerebral/citología , Regulación del Desarrollo de la Expresión Génica/genética , Células-Madre Neurales/fisiología , Neuronas/fisiología , Proteínas de Dominio T Box/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Ciclo Celular/genética , Movimiento Celular/genética , Polaridad Celular/genética , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/fisiología , Redes Reguladoras de Genes/genética , Hipocampo/citología , Histona Demetilasas con Dominio de Jumonji/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis por Micromatrices , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/metabolismo
17.
Cereb Cortex ; 27(11): 5303-5317, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28334226

RESUMEN

Neuronal migration is a fundamental process of brain development, and its disruption underlies devastating neurodevelopmental disorders. The transcriptional programs governing this process are relatively well characterized. However, how environmental cues instruct neuronal migration remains poorly understood. Here, we demonstrate that the cannabinoid CB1 receptor is strictly required for appropriate pyramidal neuron migration in the developing cortex. Acute silencing of the CB1 receptor alters neuronal morphology and impairs radial migration. Consequently, CB1 siRNA-electroporated mice display cortical malformations mimicking subcortical band heterotopias and increased seizure susceptibility in adulthood. Importantly, rescuing the CB1 deficiency-induced radial migration arrest by knockdown of the GTPase protein RhoA restored the hyperexcitable neuronal network and seizure susceptibility. Our findings show that CB1 receptor/RhoA signaling regulates pyramidal neuron migration, and that deficient CB1 receptor signaling may contribute to cortical development malformations leading to refractory epilepsy independently of its canonical neuromodulatory role in the adult brain.


Asunto(s)
Movimiento Celular/fisiología , Corteza Cerebral/anomalías , Corteza Cerebral/metabolismo , Células Piramidales/metabolismo , Receptor Cannabinoide CB1/deficiencia , Convulsiones/metabolismo , Animales , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/metabolismo , Susceptibilidad a Enfermedades/patología , Electroporación , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Hibridación in Situ , Ratones Transgénicos , Microscopía Confocal , Pentilenotetrazol , Células Piramidales/patología , ARN Interferente Pequeño , Receptor Cannabinoide CB1/genética , Convulsiones/patología , Técnicas de Cultivo de Tejidos , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
18.
J Neurosci ; 36(15): 4339-50, 2016 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-27076429

RESUMEN

The enteric nervous system (ENS) is organized into neural circuits within the gastrointestinal wall where it controls the peristaltic movements, secretion, and blood flow. Although proper gut function relies on the complex neuronal composition of the ENS, little is known about the transcriptional networks that regulate the diversification into different classes of enteric neurons and glia during development. Here we redefine the role of Ascl1 (Mash1), one of the few regulatory transcription factors described during ENS development. We show that enteric glia and all enteric neuronal subtypes appear to be derived from Ascl1-expressing progenitor cells. In the gut of Ascl1(-/-) mutant mice, neurogenesis is delayed and reduced, and posterior gliogenesis impaired. The ratio of neurons expressing Calbindin, TH, and VIP is selectively decreased while, for instance, 5-HT(+) neurons, which previously were believed to be Ascl1-dependent, are formed in normal numbers. Essentially the same differentiation defects are observed in Ascl1(KINgn2) transgenic mutants, where the proneural activity of Ngn2 replaces Ascl1, demonstrating that Ascl1 is required for the acquisition of specific enteric neuronal subtype features independent of its role in neurogenesis. In this study, we provide novel insights into the expression and function of Ascl1 in the differentiation process of specific neuronal subtypes during ENS development. SIGNIFICANCE STATEMENT: The molecular mechanisms underlying the generation of different neuronal subtypes during development of the enteric nervous system are poorly understood despite its pivotal function in gut motility and involvement in gastrointestinal pathology. This report identifies novel roles for the transcription factor Ascl1 in enteric gliogenesis and neurogenesis. Moreover, independent of its proneurogenic activity, Ascl1 is required for the normal expression of specific enteric neuronal subtype characteristics. Distinct enteric neuronal subtypes are formed in a temporally defined order, and we observe that the early-born 5-HT(+) neurons are generated in Ascl1(-/-) mutants, despite the delayed neurogenesis. Enteric nervous system progenitor cells may therefore possess strong intrinsic control over their specification at the initial waves of neurogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Sistema Nervioso Entérico/crecimiento & desarrollo , Neuronas/fisiología , Animales , Calbindinas/metabolismo , Diferenciación Celular/genética , Femenino , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación/genética , Células-Madre Neurales/fisiología , Neurogénesis/genética , Neurogénesis/fisiología , Neuroglía/fisiología , Embarazo , Neuronas Serotoninérgicas/fisiología , Tirosina 3-Monooxigenasa/metabolismo , Péptido Intestinal Vasoactivo/metabolismo
19.
Development ; 141(19): 3721-31, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25249462

RESUMEN

Glia constitute the majority of cells in the mammalian central nervous system and are crucial for neurological function. However, there is an incomplete understanding of the molecular control of glial cell development. We find that the transcription factor Ascl1 (Mash1), which is best known for its role in neurogenesis, also functions in both astrocyte and oligodendrocyte lineages arising in the mouse spinal cord at late embryonic stages. Clonal fate mapping in vivo reveals heterogeneity in Ascl1-expressing glial progenitors and shows that Ascl1 defines cells that are restricted to either gray matter (GM) or white matter (WM) as astrocytes or oligodendrocytes. Conditional deletion of Ascl1 post-neurogenesis shows that Ascl1 is required during oligodendrogenesis for generating the correct numbers of WM but not GM oligodendrocyte precursor cells, whereas during astrocytogenesis Ascl1 functions in balancing the number of dorsal GM protoplasmic astrocytes with dorsal WM fibrous astrocytes. Thus, in addition to its function in neurogenesis, Ascl1 marks glial progenitors and controls the number and distribution of astrocytes and oligodendrocytes in the GM and WM of the spinal cord.


Asunto(s)
Astrocitos/citología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Linaje de la Célula/fisiología , Oligodendroglía/citología , Médula Espinal/citología , Médula Espinal/embriología , Animales , Astrocitos/metabolismo , Diferenciación Celular/fisiología , Técnica del Anticuerpo Fluorescente , Ratones , Oligodendroglía/metabolismo
20.
Development ; 141(14): 2803-12, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24924197

RESUMEN

The proper balance of excitatory and inhibitory neurons is crucial for normal processing of somatosensory information in the dorsal spinal cord. Two neural basic helix-loop-helix transcription factors (TFs), Ascl1 and Ptf1a, have contrasting functions in specifying these neurons. To understand how Ascl1 and Ptf1a function in this process, we identified their direct transcriptional targets genome-wide in the embryonic mouse neural tube using ChIP-Seq and RNA-Seq. We show that Ascl1 and Ptf1a directly regulate distinct homeodomain TFs that specify excitatory or inhibitory neuronal fates. In addition, Ascl1 directly regulates genes with roles in several steps of the neurogenic program, including Notch signaling, neuronal differentiation, axon guidance and synapse formation. By contrast, Ptf1a directly regulates genes encoding components of the neurotransmitter machinery in inhibitory neurons, and other later aspects of neural development distinct from those regulated by Ascl1. Moreover, Ptf1a represses the excitatory neuronal fate by directly repressing several targets of Ascl1. Ascl1 and Ptf1a bind sequences primarily enriched for a specific E-Box motif (CAGCTG) and for secondary motifs used by Sox, Rfx, Pou and homeodomain factors. Ptf1a also binds sequences uniquely enriched in the CAGATG E-box and in the binding motif for its co-factor Rbpj, providing two factors that influence the specificity of Ptf1a binding. The direct transcriptional targets identified for Ascl1 and Ptf1a provide a molecular understanding of how these DNA-binding proteins function in neuronal development, particularly as key regulators of homeodomain TFs required for neuronal subtype specification.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Redes Reguladoras de Genes , Inhibición Neural , Neuronas/metabolismo , Médula Espinal/citología , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Tipificación del Cuerpo/genética , Pollos , Cromatina/metabolismo , Elementos E-Box/genética , Neuronas GABAérgicas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genoma/genética , Glutamatos/metabolismo , Ratones , Datos de Secuencia Molecular , Tubo Neural/citología , Tubo Neural/embriología , Tubo Neural/metabolismo , Neurogénesis/genética , Neuronas/citología , Motivos de Nucleótidos/genética , Unión Proteica , Médula Espinal/embriología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA