Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Genome Med ; 13(1): 135, 2021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34425903

RESUMEN

BACKGROUND: Genetic variants in the voltage-gated sodium channels SCN1A, SCN2A, SCN3A, and SCN8A are leading causes of epilepsy, developmental delay, and autism spectrum disorder. The mRNA splicing patterns of all four genes vary across development in the rodent brain, including mutually exclusive copies of the fifth protein-coding exon detected in the neonate (5N) and adult (5A). A second pair of mutually exclusive exons is reported in SCN8A only (18N and 18A). We aimed to quantify the expression of individual exons in the developing human brain. METHODS: RNA-seq data from 783 human brain samples across development were analyzed to estimate exon-level expression. Developmental changes in exon utilization were validated by assessing intron splicing. Exon expression was also estimated in RNA-seq data from 58 developing mouse neocortical samples. RESULTS: In the mature human neocortex, exon 5A is consistently expressed at least 4-fold higher than exon 5N in all four genes. For SCN2A, SCN3A, and SCN8A, a brain-wide synchronized 5N to 5A transition occurs between 24 post-conceptual weeks (2nd trimester) and 6 years of age. In mice, the equivalent 5N to 5A transition begins at or before embryonic day 15.5. In SCN8A, over 90% of transcripts in the mature human cortex include exon 18A. Early in fetal development, most transcripts include 18N or skip both 18N and 18A, with a transition to 18A inclusion occurring from 13 post-conceptual weeks to 6 months of age. No other protein-coding exons showed comparably dynamic developmental trajectories. CONCLUSIONS: Exon usage in SCN1A, SCN2A, SCN3A, and SCN8A changes dramatically during human brain development. These splice isoforms, which alter the biophysical properties of the encoded channels, may account for some of the observed phenotypic differences across development and between specific variants. Manipulation of the proportion of splicing isoforms at appropriate stages of development may act as a therapeutic strategy for specific mutations or even epilepsy in general.


Asunto(s)
Encéfalo/metabolismo , Regulación de la Expresión Génica , Canales de Sodio Activados por Voltaje/genética , Empalme Alternativo , Animales , Biomarcadores , Corteza Cerebral , Susceptibilidad a Enfermedades , Exones , Humanos , Intrones , Ratones , Familia de Multigenes , Sistemas de Lectura Abierta , Polimorfismo Genético , Unión Proteica , Sitios de Carácter Cuantitativo , Relación Estructura-Actividad , Canales de Sodio Activados por Voltaje/química , Canales de Sodio Activados por Voltaje/metabolismo
2.
Nat Neurosci ; 24(8): 1163-1175, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34140698

RESUMEN

The human neonatal cerebellum is one-fourth of its adult size yet contains the blueprint required to integrate environmental cues with developing motor, cognitive and emotional skills into adulthood. Although mature cerebellar neuroanatomy is well studied, understanding of its developmental origins is limited. In this study, we systematically mapped the molecular, cellular and spatial composition of human fetal cerebellum by combining laser capture microscopy and SPLiT-seq single-nucleus transcriptomics. We profiled functionally distinct regions and gene expression dynamics within cell types and across development. The resulting cell atlas demonstrates that the molecular organization of the cerebellar anlage recapitulates cytoarchitecturally distinct regions and developmentally transient cell types that are distinct from the mouse cerebellum. By mapping genes dominant for pediatric and adult neurological disorders onto our dataset, we identify relevant cell types underlying disease mechanisms. These data provide a resource for probing the cellular basis of human cerebellar development and disease.


Asunto(s)
Cerebelo/embriología , Neurogénesis , Feto , Humanos , Captura por Microdisección con Láser , Análisis de la Célula Individual , Transcriptoma
3.
Neuron ; 107(6): 1197-1211.e9, 2020 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-32707082

RESUMEN

Neural stem cells directly or indirectly generate all neurons and macroglial cells and guide migrating neurons by using a palisade-like scaffold made of their radial fibers. Here, we describe an unexpected role for the radial fiber scaffold in directing corticospinal and other axons at the junction between the striatum and globus pallidus. The maintenance of this scaffold, and consequently axon pathfinding, is dependent on the expression of an atypical RHO-GTPase, RND3/RHOE, together with its binding partner ARHGAP35/P190A, a RHO GTPase-activating protein, in the radial glia-like neural stem cells within the ventricular zone of the medial ganglionic eminence. This role is independent of RND3 and ARHGAP35 expression in corticospinal neurons, where they regulate dendritic spine formation, axon elongation, and pontine midline crossing in a FEZF2-dependent manner. The prevalence of neural stem cell scaffolds and their expression of RND3 and ARHGAP35 suggests that these observations might be broadly relevant for axon guidance and neural circuit formation.


Asunto(s)
Orientación del Axón , Células-Madre Neurales/citología , Neuroglía/citología , Animales , Axones/metabolismo , Cuerpo Estriado/citología , Cuerpo Estriado/crecimiento & desarrollo , Espinas Dendríticas/metabolismo , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Globo Pálido/citología , Globo Pálido/crecimiento & desarrollo , Humanos , Ratones , Células-Madre Neurales/metabolismo , Neuroglía/metabolismo , Tractos Piramidales/citología , Tractos Piramidales/crecimiento & desarrollo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
4.
Cell Rep ; 31(1): 107489, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32268104

RESUMEN

Gene expression levels vary across developmental stage, cell type, and region in the brain. Genomic variants also contribute to the variation in expression, and some neuropsychiatric disorder loci may exert their effects through this mechanism. To investigate these relationships, we present BrainVar, a unique resource of paired whole-genome and bulk tissue RNA sequencing from the dorsolateral prefrontal cortex of 176 individuals across prenatal and postnatal development. Here we identify common variants that alter gene expression (expression quantitative trait loci [eQTLs]) constantly across development or predominantly during prenatal or postnatal stages. Both "constant" and "temporal-predominant" eQTLs are enriched for loci associated with neuropsychiatric traits and disorders and colocalize with specific variants. Expression levels of more than 12,000 genes rise or fall in a concerted late-fetal transition, with the transitional genes enriched for cell-type-specific genes and neuropsychiatric risk loci, underscoring the importance of cataloging developmental trajectories in understanding cortical physiology and pathology.


Asunto(s)
Encéfalo/embriología , Biología Computacional/métodos , Corteza Prefrontal/metabolismo , Secuencia de Bases/genética , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Bases de Datos Genéticas , Predisposición Genética a la Enfermedad/genética , Variación Genética/genética , Estudio de Asociación del Genoma Completo/métodos , Genómica/métodos , Humanos , Fenotipo , Polimorfismo de Nucleótido Simple/genética , Sitios de Carácter Cuantitativo/genética , Análisis de Secuencia de ARN/métodos , Transcriptoma/genética , Secuenciación del Exoma/métodos , Secuenciación Completa del Genoma/métodos
5.
Science ; 362(6420)2018 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-30545855

RESUMEN

Human nervous system development is an intricate and protracted process that requires precise spatiotemporal transcriptional regulation. We generated tissue-level and single-cell transcriptomic data from up to 16 brain regions covering prenatal and postnatal rhesus macaque development. Integrative analysis with complementary human data revealed that global intraspecies (ontogenetic) and interspecies (phylogenetic) regional transcriptomic differences exhibit concerted cup-shaped patterns, with a late fetal-to-infancy (perinatal) convergence. Prenatal neocortical transcriptomic patterns revealed transient topographic gradients, whereas postnatal patterns largely reflected functional hierarchy. Genes exhibiting heterotopic and heterochronic divergence included those transiently enriched in the prenatal prefrontal cortex or linked to autism spectrum disorder and schizophrenia. Our findings shed light on transcriptomic programs underlying the evolution of human brain development and the pathogenesis of neuropsychiatric disorders.


Asunto(s)
Trastorno del Espectro Autista/genética , Encéfalo/embriología , Regulación del Desarrollo de la Expresión Génica , Neurogénesis/genética , Esquizofrenia/genética , Animales , Encéfalo/crecimiento & desarrollo , Humanos , Macaca mulatta , Corteza Prefrontal/enzimología , Corteza Prefrontal/crecimiento & desarrollo , Transcriptoma
6.
Science ; 362(6420)2018 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-30545854

RESUMEN

To broaden our understanding of human neurodevelopment, we profiled transcriptomic and epigenomic landscapes across brain regions and/or cell types for the entire span of prenatal and postnatal development. Integrative analysis revealed temporal, regional, sex, and cell type-specific dynamics. We observed a global transcriptomic cup-shaped pattern, characterized by a late fetal transition associated with sharply decreased regional differences and changes in cellular composition and maturation, followed by a reversal in childhood-adolescence, and accompanied by epigenomic reorganizations. Analysis of gene coexpression modules revealed relationships with epigenomic regulation and neurodevelopmental processes. Genes with genetic associations to brain-based traits and neuropsychiatric disorders (including MEF2C, SATB2, SOX5, TCF4, and TSHZ3) converged in a small number of modules and distinct cell types, revealing insights into neurodevelopment and the genomic basis of neuropsychiatric risks.


Asunto(s)
Encéfalo/embriología , Regulación del Desarrollo de la Expresión Génica , Trastornos Mentales/genética , Enfermedades del Sistema Nervioso/genética , Neurogénesis/genética , Encéfalo/crecimiento & desarrollo , Epigénesis Genética , Epigenómica , Redes Reguladoras de Genes , Humanos , Análisis de la Célula Individual , Transcriptoma
7.
Science ; 358(6366): 1027-1032, 2017 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-29170230

RESUMEN

To better understand the molecular and cellular differences in brain organization between human and nonhuman primates, we performed transcriptome sequencing of 16 regions of adult human, chimpanzee, and macaque brains. Integration with human single-cell transcriptomic data revealed global, regional, and cell-type-specific species expression differences in genes representing distinct functional categories. We validated and further characterized the human specificity of genes enriched in distinct cell types through histological and functional analyses, including rare subpallial-derived interneurons expressing dopamine biosynthesis genes enriched in the human striatum and absent in the nonhuman African ape neocortex. Our integrated analysis of the generated data revealed diverse molecular and cellular features of the phylogenetic reorganization of the human brain across multiple levels, with relevance for brain function and disease.


Asunto(s)
Macaca/genética , Neocórtex/crecimiento & desarrollo , Neocórtex/metabolismo , Vías Nerviosas/metabolismo , Pan troglodytes/genética , Transcriptoma , Animales , Perfilación de la Expresión Génica , Humanos , Interneuronas/metabolismo , Filogenia , Especificidad de la Especie
8.
Cell ; 170(2): 226-247, 2017 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-28708995

RESUMEN

The nervous system-in particular, the brain and its cognitive abilities-is among humans' most distinctive and impressive attributes. How the nervous system has changed in the human lineage and how it differs from that of closely related primates is not well understood. Here, we consider recent comparative analyses of extant species that are uncovering new evidence for evolutionary changes in the size and the number of neurons in the human nervous system, as well as the cellular and molecular reorganization of its neural circuits. We also discuss the developmental mechanisms and underlying genetic and molecular changes that generate these structural and functional differences. As relevant new information and tools materialize at an unprecedented pace, the field is now ripe for systematic and functionally relevant studies of the development and evolution of human nervous system specializations.


Asunto(s)
Evolución Biológica , Encéfalo/anatomía & histología , Encéfalo/fisiología , Sistema Nervioso/anatomía & histología , Sistema Nervioso/crecimiento & desarrollo , Animales , Encéfalo/citología , Regulación de la Expresión Génica , Lenguaje , Mutación , Proteínas del Tejido Nervioso/genética , Sistema Nervioso/citología , Fenómenos Fisiológicos del Sistema Nervioso , Primates/genética , Primates/fisiología , Especificidad de la Especie
9.
Cell Rep ; 16(10): 2576-2592, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27568284

RESUMEN

The mechanisms underlying Zika virus (ZIKV)-related microcephaly and other neurodevelopment defects remain poorly understood. Here, we describe the derivation and characterization, including single-cell RNA-seq, of neocortical and spinal cord neuroepithelial stem (NES) cells to model early human neurodevelopment and ZIKV-related neuropathogenesis. By analyzing human NES cells, organotypic fetal brain slices, and a ZIKV-infected micrencephalic brain, we show that ZIKV infects both neocortical and spinal NES cells as well as their fetal homolog, radial glial cells (RGCs), causing disrupted mitoses, supernumerary centrosomes, structural disorganization, and cell death. ZIKV infection of NES cells and RGCs causes centrosomal depletion and mitochondrial sequestration of phospho-TBK1 during mitosis. We also found that nucleoside analogs inhibit ZIKV replication in NES cells, protecting them from ZIKV-induced pTBK1 relocalization and cell death. We established a model system of human neural stem cells to reveal cellular and molecular mechanisms underlying neurodevelopmental defects associated with ZIKV infection and its potential treatment.


Asunto(s)
Mitosis , Células-Madre Neurales/enzimología , Células-Madre Neurales/virología , Células Neuroepiteliales/virología , Neuroglía/virología , Proteínas Serina-Treonina Quinasas/metabolismo , Virus Zika/patogenicidad , Encéfalo/embriología , Encéfalo/patología , Encéfalo/virología , Muerte Celular/efectos de los fármacos , Centrosoma/efectos de los fármacos , Centrosoma/metabolismo , Feto/virología , Perfilación de la Expresión Génica , Humanos , Inmunidad Innata/efectos de los fármacos , Microcefalia/patología , Microcefalia/virología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitosis/efectos de los fármacos , Neocórtex/patología , Células-Madre Neurales/inmunología , Células-Madre Neurales/ultraestructura , Células Neuroepiteliales/efectos de los fármacos , Células Neuroepiteliales/inmunología , Células Neuroepiteliales/ultraestructura , Neuroglía/patología , Neuroglía/ultraestructura , Neuronas/efectos de los fármacos , Neuronas/patología , Neuronas/virología , Fármacos Neuroprotectores/farmacología , Nucleósidos/farmacología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Médula Espinal/patología , Transcripción Genética/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Virus Zika/efectos de los fármacos , Virus Zika/fisiología , Virus Zika/ultraestructura , Infección por el Virus Zika/patología , Infección por el Virus Zika/virología , Tirosina Quinasa del Receptor Axl
10.
Trends Genet ; 31(2): 77-87, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25624274

RESUMEN

Transcriptional mechanisms mediated by the binding of transcription factors (TFs) to cis-acting regulatory elements (CREs) in DNA play crucial roles in directing gene expression. While TFs have been extensively studied, less effort has gone towards the identification and functional characterization of CREs and associated epigenetic modulation. However, owing to methodological and analytical advances, more comprehensive studies of regulatory elements and mechanisms are now possible. We summarize recent progress in integrative analyses of these regulatory components in the development of the cerebral neocortex, the part of the brain involved in cognition and complex behavior. These studies are uncovering not only the underlying transcriptional regulatory networks, but also how these networks are altered across species and in neurological and psychiatric disorders.


Asunto(s)
Regulación de la Expresión Génica , Redes Reguladoras de Genes , Neocórtex/embriología , Neocórtex/metabolismo , Elementos Reguladores de la Transcripción , Secuencias Reguladoras de Ácidos Nucleicos , Transcripción Genética , Animales , Diferenciación Celular/genética , Movimiento Celular/genética , Humanos , Trastornos Mentales/genética , Red Nerviosa , Enfermedades del Sistema Nervioso/genética , Neurogénesis/genética , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Neuronas/metabolismo
12.
Behav Genet ; 38(3): 277-91, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18363093

RESUMEN

We used short-term selection to produce outbred mouse lines with differences in contextual fear conditioning. Within two generations of selection all low selected mice were homozygous for the recessive tyrc allele and showed the corresponding albino coat color. Freezing differed in the high and low selected lines across a range of parameters. We identified several QTLs for the selection response, including a highly significant QTL at the tyr locus (p < 9.6(-10)). To determine whether the tyrc allele was directly responsible for the response to selection, we examined B6 mice that have a mutant tyr allele (tyr(c-2j-)) and an AJ congenic strain that has the wild-type B6 allele for tyr. These studies showed that the tyr allele had a small influence on fear learning. We used Affymetrix microarrays to identify many differentially expressed genes in the amygdala and hippocampus of the selected lines. We conclude that tyr is one of many alleles that influence fear conditioning.


Asunto(s)
Miedo , Perfilación de la Expresión Génica , Sitios de Carácter Cuantitativo , Selección Genética , Alelos , Amígdala del Cerebelo/metabolismo , Animales , Condicionamiento Psicológico , Genes Recesivos , Hipocampo/metabolismo , Homocigoto , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , Análisis de Secuencia por Matrices de Oligonucleótidos , Carácter Cuantitativo Heredable
13.
Brain Res ; 1140: 170-8, 2007 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-16884697

RESUMEN

The mouse Engrailed genes, En1 and En2, play an important role in the development of the cerebellum from its inception at the mid/hindbrain boundary in early embryonic development through cell type specification events and beyond. In the absence of En1, the cerebellum and caudal midbrain fail to develop normally--a phenotype that we have previously reported to be strain dependent. On the 129/S1 strain background, En1 null alleles lead to mid/hindbrain failure, whereas on the C57BL/6 background, En1 deficiency is compatible with near normal cerebellar development. We have pursued this dramatic effect of genetic background by performing a genetic modifier screen through F1 backcross and F1 intercross matings. The backcross has yielded two strong candidate intervals with suggestive linkage to a third region. Moreover, variations in rescue frequency among subgroups within the backcross indicate gender and parent of origin influences on rescue penetrance. The intercross data reveal locus heterogeneity of the En1 modifiers, with more than one compliment of C57BL/6 and 129/S1 alleles capable of mediating the rescue phenotype. These findings highlight the complexity and plasticity of gene networks involved in brain development.


Asunto(s)
Cerebelo/embriología , Cerebelo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Fenotipo , Animales , Mapeo Cromosómico/métodos , Femenino , Redes Reguladoras de Genes , Escala de Lod , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Embarazo , Rombencéfalo/embriología , Rombencéfalo/metabolismo
14.
Proc Natl Acad Sci U S A ; 100(14): 8401-6, 2003 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-12815099

RESUMEN

Langerhans cells (LCs) are suspected to be initial targets for HIV after sexual exposure (by becoming infected or by capturing virus). Here, productive R5 HIV infection of LC ex vivo and LC-mediated transmission of virus to CD4+ T cells were both found to depend on CCR5. By contrast, infection of monocyte-derived dendritic cells and transfer of infection from monocyte-derived dendritic cells to CD4+ T cells were mediated by CCR5-dependent as well as DC-specific ICAM-3-grabbing nonintegrin-dependent pathways. Furthermore, in 62 healthy individuals, R5 HIV infection levels in LCs ex vivo were associated with CCR5 genotype. Specifically, genotyping for ORF Delta 32 revealed that LCs isolated from ORF Delta 32/wt individuals were significantly less susceptible to HIV when compared with LCs isolated from ORFwt/wt individuals (P = 0.016). Strikingly, further genetic analyses of the A-2459G CCR5 promoter polymorphism in ORF Delta 32/wt heterozygous individuals revealed that LCs isolated from -2459A/G + ORF Delta 32/wt individuals were markedly less susceptible to HIV than were LCs from -2459A/A + ORF Delta 32/wt individuals (P = 0.012). Interestingly, these genetic susceptibility data in LCs parallel those of genetic susceptibility studies performed in cohorts of HIV-infected individuals. Thus, we suggest that CCR5-mediated infection of LCs, and not capture of virus by LCs, provides a biologic basis for understanding certain aspects of host genetic susceptibility to initial HIV infection.


Asunto(s)
Linfocitos T CD4-Positivos/virología , VIH/fisiología , Células de Langerhans/virología , Polimorfismo Genético , Receptores CCR5/fisiología , Anticuerpos Monoclonales/farmacología , Bencilaminas , Antagonistas de los Receptores CCR5 , Antígenos CD4/fisiología , Moléculas de Adhesión Celular/fisiología , Quimiocina CCL5/fisiología , Ciclamas , Células Dendríticas/virología , Endocitosis , Células Epiteliales/virología , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Infecciones por VIH/genética , Compuestos Heterocíclicos/farmacología , Humanos , Células de Langerhans/fisiología , Lectinas Tipo C/fisiología , Masculino , Sistemas de Lectura Abierta/genética , Receptores CCR5/genética , Receptores CXCR4/antagonistas & inhibidores , Receptores de Superficie Celular/fisiología , Eliminación de Secuencia , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...