Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Elife ; 122023 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-36718990

RESUMEN

The tuberal hypothalamus controls life-supporting homeostatic processes, but despite its fundamental role, the cells and signalling pathways that specify this unique region of the central nervous system in embryogenesis are poorly characterised. Here, we combine experimental and bioinformatic approaches in the embryonic chick to show that the tuberal hypothalamus is progressively generated from hypothalamic floor plate-like cells. Fate-mapping studies show that a stream of tuberal progenitors develops in the anterior-ventral neural tube as a wave of neuroepithelial-derived BMP signalling sweeps from anterior to posterior through the hypothalamic floor plate. As later-specified posterior tuberal progenitors are generated, early specified anterior tuberal progenitors become progressively more distant from these BMP signals and differentiate into tuberal neurogenic cells. Gain- and loss-of-function experiments in vivo and ex vivo show that BMP signalling initiates tuberal progenitor specification, but must be eliminated for these to progress to anterior neurogenic progenitors. scRNA-Seq profiling shows that tuberal progenitors that are specified after the major period of anterior tuberal specification begin to upregulate genes that characterise radial glial cells. This study provides an integrated account of the development of the tuberal hypothalamus.


Asunto(s)
Hipotálamo , Neurogénesis , Animales , Hipotálamo/metabolismo , Neurogénesis/fisiología , Transducción de Señal , Pollos
2.
Cell Rep ; 38(3): 110251, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35045288

RESUMEN

The hypothalamus regulates many innate behaviors, but its development remains poorly understood. Here, we used single-cell RNA sequencing (RNA-seq) and hybridization chain reaction (HCR) to profile multiple stages of early hypothalamic development in the chick. Hypothalamic neuroepithelial cells are initially induced from prethalamic-like cells. Two distinct hypothalamic progenitor populations then emerge and give rise to tuberal and mammillary/paraventricular hypothalamic cells. At later stages, the regional organization of the chick and mouse hypothalamus is highly similar. We identify selective markers for major subdivisions of the developing chick hypothalamus and many previously uncharacterized candidate regulators of hypothalamic induction, regionalization, and neurogenesis. As proof of concept for the power of the dataset, we demonstrate that prethalamus-derived follistatin inhibits hypothalamic induction. This study clarifies the organization of the nascent hypothalamus and identifies molecular mechanisms that may control its induction and subsequent development.


Asunto(s)
Hipotálamo/embriología , Células-Madre Neurales/citología , Neurogénesis/fisiología , Animales , Embrión de Pollo , RNA-Seq , Análisis de la Célula Individual
3.
Commun Biol ; 4(1): 95, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33479483

RESUMEN

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


Asunto(s)
Diferenciación Celular , Neuronas GABAérgicas/fisiología , Redes Reguladoras de Genes , Hipotálamo/citología , Proteínas con Homeodominio LIM/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Factores de Transcripción/metabolismo , Animales , Supervivencia Celular , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/metabolismo , Hipotálamo/metabolismo , Ratones , Proteínas Nucleares , Sueño/fisiología
4.
Nat Commun ; 11(1): 4360, 2020 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-32868762

RESUMEN

The hypothalamus is a central regulator of many innate behaviors essential for survival, but the molecular mechanisms controlling hypothalamic patterning and cell fate specification are poorly understood. To identify genes that control hypothalamic development, we have used single-cell RNA sequencing (scRNA-Seq) to profile mouse hypothalamic gene expression across 12 developmental time points between embryonic day 10 and postnatal day 45. This identified genes that delineated clear developmental trajectories for all major hypothalamic cell types, and readily distinguished major regional subdivisions of the developing hypothalamus. By using our developmental dataset, we were able to rapidly annotate previously unidentified clusters from existing scRNA-Seq datasets collected during development and to identify the developmental origins of major neuronal populations of the ventromedial hypothalamus. We further show that our approach can rapidly and comprehensively characterize mutants that have altered hypothalamic patterning, identifying Nkx2.1 as a negative regulator of prethalamic identity. These data serve as a resource for further studies of hypothalamic development, physiology, and dysfunction.


Asunto(s)
Diferenciación Celular , Hipotálamo , Neuronas/metabolismo , Factor Nuclear Tiroideo 1/metabolismo , Animales , Secuencia de Bases , Tipificación del Cuerpo , Regulación del Desarrollo de la Expresión Génica , Hipotálamo/citología , Hipotálamo/embriología , Hipotálamo/crecimiento & desarrollo , Hipotálamo/metabolismo , Ratones , Mutación , Análisis de la Célula Individual , Factor Nuclear Tiroideo 1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA