Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 236
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 182(3): 754-769.e18, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32610082

RESUMEN

To discover regulatory elements driving the specificity of gene expression in different cell types and regions of the developing human brain, we generated an atlas of open chromatin from nine dissected regions of the mid-gestation human telencephalon, as well as microdissected upper and deep layers of the prefrontal cortex. We identified a subset of open chromatin regions (OCRs), termed predicted regulatory elements (pREs), that are likely to function as developmental brain enhancers. pREs showed temporal, regional, and laminar differences in chromatin accessibility and were correlated with gene expression differences across regions and gestational ages. We identified two functional de novo variants in a pRE for autism risk gene SLC6A1, and using CRISPRa, demonstrated that this pRE regulates SCL6A1. Additionally, mouse transgenic experiments validated enhancer activity for pREs proximal to FEZF2 and BCL11A. Thus, this atlas serves as a resource for decoding neurodevelopmental gene regulation in health and disease.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica/genética , Corteza Prefrontal/embriología , Telencéfalo/embriología , Animales , Trastorno Autístico/genética , Línea Celular , Secuenciación de Inmunoprecipitación de Cromatina , Eucromatina/genética , Proteínas Transportadoras de GABA en la Membrana Plasmática/genética , Ontología de Genes , Predisposición Genética a la Enfermedad , Edad Gestacional , Humanos , Ratones , Ratones Transgénicos , Motivos de Nucleótidos , Mutación Puntual , Corteza Prefrontal/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Análisis Espacio-Temporal , Telencéfalo/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
Cell ; 172(3): 491-499.e15, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29358049

RESUMEN

Non-coding "ultraconserved" regions containing hundreds of consecutive bases of perfect sequence conservation across mammalian genomes can function as distant-acting enhancers. However, initial deletion studies in mice revealed that loss of such extraordinarily constrained sequences had no immediate impact on viability. Here, we show that ultraconserved enhancers are required for normal development. Focusing on some of the longest ultraconserved sites genome wide, located near the essential neuronal transcription factor Arx, we used genome editing to create an expanded series of knockout mice lacking individual or combinations of ultraconserved enhancers. Mice with single or pairwise deletions of ultraconserved enhancers were viable and fertile but in nearly all cases showed neurological or growth abnormalities, including substantial alterations of neuron populations and structural brain defects. Our results demonstrate the functional importance of ultraconserved enhancers and indicate that remarkably strong sequence conservation likely results from fitness deficits that appear subtle in a laboratory setting.


Asunto(s)
Secuencia Conservada , Desarrollo Embrionario/genética , Elementos de Facilitación Genéticos , Animales , Encéfalo/anomalías , Encéfalo/embriología , Encéfalo/metabolismo , Femenino , Eliminación de Gen , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Cell ; 155(7): 1521-31, 2013 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-24360275

RESUMEN

Enhancers are distal regulatory elements that can activate tissue-specific gene expression and are abundant throughout mammalian genomes. Although substantial progress has been made toward genome-wide annotation of mammalian enhancers, their temporal activity patterns and global contributions in the context of developmental in vivo processes remain poorly explored. Here we used epigenomic profiling for H3K27ac, a mark of active enhancers, coupled to transgenic mouse assays to examine the genome-wide utilization of enhancers in three different mouse tissues across seven developmental stages. The majority of the ∼90,000 enhancers identified exhibited tightly temporally restricted predicted activity windows and were associated with stage-specific biological functions and regulatory pathways in individual tissues. Comparative genomic analysis revealed that evolutionary conservation of enhancers decreases following midgestation across all tissues examined. The dynamic enhancer activities uncovered in this study illuminate rapid and pervasive temporal in vivo changes in enhancer usage that underlie processes central to development and disease.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Estudio de Asociación del Genoma Completo , Acetilación , Animales , Epigénesis Genética , Evolución Molecular , Histonas/metabolismo , Ratones , Ratones Transgénicos , Especificidad de Órganos
4.
Cell ; 152(4): 895-908, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23375746

RESUMEN

The mammalian telencephalon plays critical roles in cognition, motor function, and emotion. Though many of the genes required for its development have been identified, the distant-acting regulatory sequences orchestrating their in vivo expression are mostly unknown. Here, we describe a digital atlas of in vivo enhancers active in subregions of the developing telencephalon. We identified more than 4,600 candidate embryonic forebrain enhancers and studied the in vivo activity of 329 of these sequences in transgenic mouse embryos. We generated serial sets of histological brain sections for 145 reproducible forebrain enhancers, resulting in a publicly accessible web-based data collection comprising more than 32,000 sections. We also used epigenomic analysis of human and mouse cortex tissue to directly compare the genome-wide enhancer architecture in these species. These data provide a primary resource for investigating gene regulatory mechanisms of telencephalon development and enable studies of the role of distant-acting enhancers in neurodevelopmental disorders.


Asunto(s)
Elementos de Facilitación Genéticos , Telencéfalo/metabolismo , Animales , Embrión de Mamíferos/metabolismo , Feto/metabolismo , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Telencéfalo/embriología , Transcriptoma , Factores de Transcripción p300-CBP/metabolismo
5.
Development ; 151(11)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38819455

RESUMEN

The vertebrate Dlx gene family encode homeobox transcription factors that are related to the Drosophila Distal-less (Dll) gene and are crucial for development. Over the last ∼35 years detailed information has accrued about the redundant and unique expression and function of the six mammalian Dlx family genes. DLX proteins interact with general transcriptional regulators, and co-bind with other transcription factors to enhancer elements with highly specific activity in the developing forebrain. Integration of the genetic and biochemical data has yielded a foundation for a gene regulatory network governing the differentiation of forebrain GABAergic neurons. In this Primer, we describe the discovery of vertebrate Dlx genes and their crucial roles in embryonic development. We largely focus on the role of Dlx family genes in mammalian forebrain development revealed through studies in mice. Finally, we highlight questions that remain unanswered regarding vertebrate Dlx genes despite over 30 years of research.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio , Prosencéfalo , Factores de Transcripción , Animales , Prosencéfalo/metabolismo , Prosencéfalo/embriología , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Humanos , Mamíferos/genética , Ratones
6.
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
7.
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
8.
Proc Natl Acad Sci U S A ; 119(15): e2108760119, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35377797

RESUMEN

Enhancers integrate transcription factor signaling pathways that drive cell fate specification in the developing brain. We paired enhancer labeling and single-cell RNA-sequencing (scRNA-seq) to delineate and distinguish specification of neuronal lineages in mouse medial, lateral, and caudal ganglionic eminences (MGE, LGE, and CGE) at embryonic day (E)11.5. We show that scRNA-seq clustering using transcription factors improves resolution of regional and developmental populations, and that enhancer activities identify specific and overlapping GE-derived neuronal populations. First, we mapped the activities of seven evolutionarily conserved brain enhancers at single-cell resolution in vivo, finding that the selected enhancers had diverse activities in specific progenitor and neuronal populations across the GEs. We then applied enhancer-based labeling, scRNA-seq, and analysis of in situ hybridization data to distinguish transcriptionally distinct and spatially defined subtypes of MGE-derived GABAergic and cholinergic projection neurons and interneurons. Our results map developmental origins and specification paths underlying neurogenesis in the embryonic basal ganglia and showcase the power of scRNA-seq combined with enhancer-based labeling to resolve the complex paths of neuronal specification underlying mouse brain development.


Asunto(s)
Ganglios Basales , Neuronas Colinérgicas , Elementos de Facilitación Genéticos , Neuronas GABAérgicas , Neurogénesis , Animales , Ganglios Basales/citología , Ganglios Basales/embriología , Linaje de la Célula/genética , Neuronas Colinérgicas/metabolismo , Neuronas GABAérgicas/metabolismo , Ratones , Neurogénesis/genética , RNA-Seq , Análisis de la Célula Individual , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Dev Biol ; 503: 10-24, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37532091

RESUMEN

The external globus pallidus (GPe) is an essential component of the basal ganglia, a group of subcortical nuclei that are involved in control of action. Changes in the firing of GPe neurons are associated with both passive and active body movements. Aberrant activity of GPe neurons has been linked to motor symptoms of a variety of movement disorders, such as Parkinson's Disease, Huntington's disease and dystonia. Recent studies have helped delineate functionally distinct subtypes of GABAergic GPe projection neurons. However, not much is known about specific molecular mechanisms underlying the development of GPe neuronal subtypes. We show that the transcriptional regulator Lmo3 is required for the development of medial ganglionic eminence derived Nkx2.1+ and PV+ GPe neurons, but not lateral ganglionic eminence derived FoxP2+ neurons. As a consequence of the reduction in PV+ neurons, Lmo3-null mice have a reduced GPe input to the subthalamic nucleus.


Asunto(s)
Neuronas GABAérgicas , Globo Pálido , Proteínas con Dominio LIM , Movimiento , Animales , Ratones , Neuronas GABAérgicas/metabolismo , Globo Pálido/metabolismo , Ratones Noqueados , Movimiento/fisiología , Trastornos del Movimiento/genética , Trastornos del Movimiento/metabolismo , Trastornos del Movimiento/fisiopatología , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo
10.
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
11.
Cell ; 135(3): 396-400, 2008 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-18984148

RESUMEN

Genetic studies are refining our understanding of neurodevelopmental mechanisms in autism. Some autism-related mutations appear to disrupt genes regulated by neuronal activity, which are especially important in development of the postnatal nervous system. Gene replacement studies in mice indicate that the developmental window to ameliorate symptoms may be wider than previously anticipated.


Asunto(s)
Trastorno Autístico/fisiopatología , Encéfalo/crecimiento & desarrollo , Animales , Trastorno Autístico/genética , Encéfalo/fisiología , Femenino , Humanos , Masculino , Sinapsis/fisiología
12.
Proc Natl Acad Sci U S A ; 117(11): 6189-6195, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32123116

RESUMEN

Neurofibromatosis 1 (NF1) is caused by mutations in the NF1 gene, which encodes the protein, neurofibromin, an inhibitor of Ras activity. Cortical GABAergic interneurons (CINs) are implicated in NF1 pathology, but the cellular and molecular changes to CINs are unknown. We deleted mouse Nf1 from the medial ganglionic eminence, which gives rise to both oligodendrocytes and CINs that express somatostatin and parvalbumin. Nf1 loss led to a persistence of immature oligodendrocytes that prevented later-generated oligodendrocytes from occupying the cortex. Moreover, molecular and cellular properties of parvalbumin (PV)-positive CINs were altered by the loss of Nf1, without changes in somatostatin (SST)-positive CINs. We discovered that loss of Nf1 results in a dose-dependent decrease in Lhx6 expression, the transcription factor necessary to establish SST+ and PV+ CINs, which was rescued by the MEK inhibitor SL327, revealing a mechanism whereby a neurofibromin/Ras/MEK pathway regulates a critical CIN developmental milestone.


Asunto(s)
Corteza Cerebral/patología , Neuronas GABAérgicas/patología , Interneuronas/patología , Proteínas con Homeodominio LIM/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurofibromatosis 1/patología , Neurofibromina 1/genética , Factores de Transcripción/metabolismo , Aminoacetonitrilo/administración & dosificación , Aminoacetonitrilo/análogos & derivados , Animales , Células Cultivadas , Corteza Cerebral/citología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Femenino , Neuronas GABAérgicas/metabolismo , Humanos , Interneuronas/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Eminencia Media/citología , Ratones , Ratones Noqueados , Neurofibromatosis 1/genética , Neurofibromina 1/metabolismo , Neuroglía/citología , Parvalbúminas/metabolismo , Cultivo Primario de Células , Somatostatina/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo
13.
J Neurosci ; 40(11): 2215-2227, 2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-31988060

RESUMEN

Manipulations that enhance GABAergic inhibition have been associated with improved behavioral phenotypes in autism models, suggesting that autism may be treated by correcting underlying deficits of inhibition. Interneuron transplantation is a method for increasing recipient synaptic inhibition, and it has been considered a prospective therapy for conditions marked by deficient inhibition, including neuropsychiatric disorders. It is unknown, however, whether interneuron transplantation may be therapeutically effective only for conditions marked by reduced inhibition, and it is also unclear whether transplantation improves behavioral phenotypes solely by normalizing underlying circuit defects. To address these questions, we studied the effects of interneuron transplantation in male and female mice lacking the autism-associated gene, Pten, in GABAergic interneurons. Pten mutant mice exhibit social behavior deficits, elevated synaptic inhibition in prefrontal cortex, abnormal baseline and social interaction-evoked electroencephalogram (EEG) signals, and an altered composition of cortical interneuron subtypes. Transplantation of wild-type embryonic interneurons from the medial ganglionic eminence into the prefrontal cortex of neonatal Pten mutants rescued social behavior despite exacerbating excessive levels of synaptic inhibition. Furthermore, transplantation did not normalize recipient EEG signals measured during baseline states. Interneuron transplantation can thus correct behavioral deficits even when those deficits are associated with elevated synaptic inhibition. Moreover, transplantation does not exert therapeutic effects solely by restoring wild-type circuit states. Our findings indicate that interneuron transplantation could offer a novel cell-based approach to autism treatment while challenging assumptions that effective therapies must reverse underlying circuit defects.SIGNIFICANCE STATEMENT Imbalances between neural excitation and inhibition are hypothesized to contribute to the pathophysiology of autism. Interneuron transplantation is a method for altering recipient inhibition, and it has been considered a prospective therapy for neuropsychiatric disorders, including autism. Here we examined the behavioral and physiological effects of interneuron transplantation in a mouse genetic model of autism. They demonstrate that transplantation rescues recipient social interaction deficits without correcting a common measure of recipient inhibition, or circuit-level physiological measures. These findings demonstrate that interneuron transplantation can exert therapeutic behavioral effects without necessarily restoring wild-type circuit states, while highlighting the potential of interneuron transplantation as an autism therapy.


Asunto(s)
Trastorno Autístico/cirugía , Trasplante de Tejido Encefálico , Trasplante de Tejido Fetal , Neuronas GABAérgicas/fisiología , Interneuronas/trasplante , Inhibición Neural/fisiología , Fosfohidrolasa PTEN/deficiencia , Conducta Social , Animales , Trastorno Autístico/fisiopatología , Trastorno Autístico/psicología , Modelos Animales de Enfermedad , Electroencefalografía , Conducta Exploratoria , Femenino , Masculino , Aprendizaje por Laberinto , Eminencia Media/citología , Eminencia Media/embriología , Ratones , Ratones Noqueados , Fosfohidrolasa PTEN/fisiología , Técnicas de Placa-Clamp , Fenotipo , Corteza Prefrontal/fisiopatología , Distribución Aleatoria , Sinapsis/fisiología
14.
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
15.
Development ; 144(21): 3867-3878, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29089360

RESUMEN

Cortical interneurons are a diverse group of neurons that project locally and are crucial for regulating information processing and flow throughout the cortex. Recent studies in mice have advanced our understanding of how these neurons are specified, migrate and mature. Here, we evaluate new findings that provide insights into the development of cortical interneurons and that shed light on when their fate is determined, on the influence that regional domains have on their development, and on the role that key transcription factors and other crucial regulatory genes play in these events. We focus on cortical interneurons that are derived from the medial ganglionic eminence, as most studies have examined this interneuron population. We also assess how these data inform our understanding of neuropsychiatric disease and discuss the potential role of cortical interneurons in cell-based therapies.


Asunto(s)
Corteza Cerebral/citología , Interneuronas/citología , Animales , Linaje de la Célula , Modelos Biológicos , Factores de Tiempo , Transcripción Genética
16.
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
17.
Nat Methods ; 14(6): 621-628, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28504679

RESUMEN

Approaches to differentiating pluripotent stem cells (PSCs) into neurons currently face two major challenges-(i) generated cells are immature, with limited functional properties; and (ii) cultures exhibit heterogeneous neuronal subtypes and maturation stages. Using lineage-determining transcription factors, we previously developed a single-step method to generate glutamatergic neurons from human PSCs. Here, we show that transient expression of the transcription factors Ascl1 and Dlx2 (AD) induces the generation of exclusively GABAergic neurons from human PSCs with a high degree of synaptic maturation. These AD-induced neuronal (iN) cells represent largely nonoverlapping populations of GABAergic neurons that express various subtype-specific markers. We further used AD-iN cells to establish that human collybistin, the loss of gene function of which causes severe encephalopathy, is required for inhibitory synaptic function. The generation of defined populations of functionally mature human GABAergic neurons represents an important step toward enabling the study of diseases affecting inhibitory synaptic transmission.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/fisiología , Proteínas de Homeodominio/genética , Células Madre Pluripotentes/fisiología , Factores de Transcripción/genética , Animales , Ingeniería Celular , Células Cultivadas , Humanos , Ratones , Células Madre Pluripotentes/citología
18.
Mol Psychiatry ; 24(9): 1248-1257, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31089192

RESUMEN

In 2003 Rubenstein and Merzenich hypothesized that some forms of Autism (ASD) might be caused by a reduction in signal-to-noise in key neural circuits, which could be the result of changes in excitatory-inhibitory (E-I) balance. Here, we have clarified the concept of E-I balance, and updated the original hypothesis in light of the field's increasingly sophisticated understanding of neuronal circuits. We discuss how specific developmental mechanisms, which reduce inhibition, affect cortical and hippocampal functions. After describing how mutations of some ASD genes disrupt inhibition in mice, we close by suggesting that E-I balance represents an organizing framework for understanding findings related to pathophysiology and for identifying appropriate treatments.


Asunto(s)
Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/fisiopatología , Red Nerviosa/fisiología , Animales , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Hipocampo/fisiopatología , Humanos , Inhibición Psicológica , Trastornos Mentales/fisiopatología , Ratones , Neuronas/fisiología
19.
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
20.
Cereb Cortex ; 29(11): 4831-4849, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-30796806

RESUMEN

Generation of olfactory bulb (OB) interneurons requires neural stem/progenitor cell specification, proliferation, differentiation, and young interneuron migration and maturation. Here, we show that the homeobox transcription factors Dlx1/2 are central and essential components in the transcriptional code for generating OB interneurons. In Dlx1/2 constitutive null mutants, the differentiation of GSX2+ and ASCL1+ neural stem/progenitor cells in the dorsal lateral ganglionic eminence is blocked, resulting in a failure of OB interneuron generation. In Dlx1/2 conditional mutants (hGFAP-Cre; Dlx1/2F/- mice), GSX2+ and ASCL1+ neural stem/progenitor cells in the postnatal subventricular zone also fail to differentiate into OB interneurons. In contrast, overexpression of Dlx1&2 in embryonic mouse cortex led to ectopic production of OB-like interneurons that expressed Gad1, Sp8, Sp9, Arx, Pbx3, Etv1, Tshz1, and Prokr2. Pax6 mutants generate cortical ectopia with OB-like interneurons, but do not do so in compound Pax6; Dlx1/2 mutants. We propose that DLX1/2 promote OB interneuron development mainly through activating the expression of Sp8/9, which further promote Tshz1 and Prokr2 expression. Based on this study, in combination with earlier ones, we propose a transcriptional network for the process of OB interneuron development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Interneuronas/metabolismo , Células-Madre Neurales/metabolismo , Bulbo Olfatorio/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Neocórtex/embriología , Neocórtex/metabolismo , Bulbo Olfatorio/embriología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA