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
País de afiliación
Intervalo de año de publicación
1.
Nature ; 582(7811): 246-252, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32499648

RESUMEN

A wealth of specialized neuroendocrine command systems intercalated within the hypothalamus control the most fundamental physiological needs in vertebrates1,2. Nevertheless, we lack a developmental blueprint that integrates the molecular determinants of neuronal and glial diversity along temporal and spatial scales of hypothalamus development3. Here we combine single-cell RNA sequencing of 51,199 mouse cells of ectodermal origin, gene regulatory network (GRN) screens in conjunction with genome-wide association study-based disease phenotyping, and genetic lineage reconstruction to show that nine glial and thirty-three neuronal subtypes are generated by mid-gestation under the control of distinct GRNs. Combinatorial molecular codes that arise from neurotransmitters, neuropeptides and transcription factors are minimally required to decode the taxonomical hierarchy of hypothalamic neurons. The differentiation of γ-aminobutyric acid (GABA) and dopamine neurons, but not glutamate neurons, relies on quasi-stable intermediate states, with a pool of GABA progenitors giving rise to dopamine cells4. We found an unexpected abundance of chemotropic proliferation and guidance cues that are commonly implicated in dorsal (cortical) patterning5 in the hypothalamus. In particular, loss of SLIT-ROBO signalling impaired both the production and positioning of periventricular dopamine neurons. Overall, we identify molecular principles that shape the developmental architecture of the hypothalamus and show how neuronal heterogeneity is transformed into a multimodal neural unit to provide virtually infinite adaptive potential throughout life.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Hipotálamo/citología , Hipotálamo/embriología , Morfogénesis , Animales , Diferenciación Celular , Linaje de la Célula , Dopamina/metabolismo , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Ectodermo/citología , Ectodermo/metabolismo , Femenino , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/metabolismo , Redes Reguladoras de Genes , Estudio de Asociación del Genoma Completo , Ácido Glutámico/metabolismo , Hipotálamo/metabolismo , Masculino , Ratones , Morfogénesis/genética , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Neuropéptidos/metabolismo , Neurotransmisores/metabolismo , Receptores Inmunológicos/metabolismo , Regulón/genética , Transducción de Señal , Factores de Transcripción/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Proteínas Roundabout
2.
Mol Psychiatry ; 25(1): 22-36, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31735910

RESUMEN

The evolution of human diets led to preferences toward polyunsaturated fatty acid (PUFA) content with 'Western' diets enriched in ω-6 PUFAs. Mounting evidence points to ω-6 PUFA excess limiting metabolic and cognitive processes that define longevity in humans. When chosen during pregnancy, ω-6 PUFA-enriched 'Western' diets can reprogram maternal bodily metabolism with maternal nutrient supply precipitating the body-wide imprinting of molecular and cellular adaptations at the level of long-range intercellular signaling networks in the unborn fetus. Even though unfavorable neurological outcomes are amongst the most common complications of intrauterine ω-6 PUFA excess, cellular underpinnings of life-long modifications to brain architecture remain unknown. Here, we show that nutritional ω-6 PUFA-derived endocannabinoids desensitize CB1 cannabinoid receptors, thus inducing epigenetic repression of transcriptional regulatory networks controlling neuronal differentiation. We found that cortical neurons lose their positional identity and axonal selectivity when mouse fetuses are exposed to excess ω-6 PUFAs in utero. Conversion of ω-6 PUFAs into endocannabinoids disrupted the temporal precision of signaling at neuronal CB1 cannabinoid receptors, chiefly deregulating Stat3-dependent transcriptional cascades otherwise required to execute neuronal differentiation programs. Global proteomics identified the immunoglobulin family of cell adhesion molecules (IgCAMs) as direct substrates, with DNA methylation and chromatin accessibility profiling uncovering epigenetic reprogramming at >1400 sites in neurons after prolonged cannabinoid exposure. We found anxiety and depression-like behavioral traits to manifest in adult offspring, which is consistent with genetic models of reduced IgCAM expression, to suggest causality for cortical wiring defects. Overall, our data uncover a regulatory mechanism whose disruption by maternal food choices could limit an offspring's brain function for life.


Asunto(s)
Encéfalo/efectos de los fármacos , Dieta Occidental/efectos adversos , Epigénesis Genética/efectos de los fármacos , Animales , Ansiedad , Encéfalo/metabolismo , Metilación de ADN/efectos de los fármacos , Depresión , Dieta , Suplementos Dietéticos , Endocannabinoides/metabolismo , Epigénesis Genética/genética , Epigenómica/métodos , Ácidos Grasos Omega-3/metabolismo , Ácidos Grasos Omega-6/metabolismo , Ácidos Grasos Insaturados/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Embarazo , Receptor Cannabinoide CB1/efectos de los fármacos
3.
Nat Genet ; 51(6): 990-998, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31133746

RESUMEN

The histone acetyl reader bromodomain-containing protein 4 (BRD4) is an important regulator of chromatin structure and transcription, yet factors modulating its activity have remained elusive. Here we describe two complementary screens for genetic and physical interactors of BRD4, which converge on the folate pathway enzyme MTHFD1 (methylenetetrahydrofolate dehydrogenase, cyclohydrolase and formyltetrahydrofolate synthetase 1). We show that a fraction of MTHFD1 resides in the nucleus, where it is recruited to distinct genomic loci by direct interaction with BRD4. Inhibition of either BRD4 or MTHFD1 results in similar changes in nuclear metabolite composition and gene expression; pharmacological inhibitors of the two pathways synergize to impair cancer cell viability in vitro and in vivo. Our finding that MTHFD1 and other metabolic enzymes are chromatin associated suggests a direct role for nuclear metabolism in the control of gene expression.


Asunto(s)
Ácido Fólico/metabolismo , Regulación de la Expresión Génica , Metilenotetrahidrofolato Deshidrogenasa (NADP)/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Cromatina/genética , Técnicas de Inactivación de Genes , Humanos , Mutación con Pérdida de Función , Unión Proteica , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Transporte de Proteínas , Transducción de Señal , Transcripción Genética
4.
Nat Neurosci ; 20(2): 176-188, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27991900

RESUMEN

The hypothalamus contains the highest diversity of neurons in the brain. Many of these neurons can co-release neurotransmitters and neuropeptides in a use-dependent manner. Investigators have hitherto relied on candidate protein-based tools to correlate behavioral, endocrine and gender traits with hypothalamic neuron identity. Here we map neuronal identities in the hypothalamus by single-cell RNA sequencing. We distinguished 62 neuronal subtypes producing glutamatergic, dopaminergic or GABAergic markers for synaptic neurotransmission and harboring the ability to engage in task-dependent neurotransmitter switching. We identified dopamine neurons that uniquely coexpress the Onecut3 and Nmur2 genes, and placed these in the periventricular nucleus with many synaptic afferents arising from neuromedin S+ neurons of the suprachiasmatic nucleus. These neuroendocrine dopamine cells may contribute to the dopaminergic inhibition of prolactin secretion diurnally, as their neuromedin S+ inputs originate from neurons expressing Per2 and Per3 and their tyrosine hydroxylase phosphorylation is regulated in a circadian fashion. Overall, our catalog of neuronal subclasses provides new understanding of hypothalamic organization and function.


Asunto(s)
Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Hipotálamo/metabolismo , Neuropéptidos/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Animales , Inmunohistoquímica/métodos , Ratones Endogámicos C57BL , Ratones Transgénicos , Neurotransmisores/fisiología , Núcleo Supraquiasmático/metabolismo , Transmisión Sináptica/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA