Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(8): 114508, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39018244

RESUMEN

Lissencephaly is a rare brain malformation for which our understanding remains limited due to the absence of suitable animal models that accurately represent human phenotypes. Here, we establish doublecortin (DCX) knockout ferrets as a model that faithfully replicates key features of the disorder. We reveal the critical roles of DCX in neural progenitor cell proliferation and radial glial fiber extension, processes essential for normal cortical development. Utilizing single-nucleus RNA sequencing (snRNA-seq) and spatial transcriptomics, we provide a detailed atlas of the lissencephalic cortex, illustrating disrupted neuronal lamination and the specific interactions between inhibitory and excitatory neurons. These findings enhance our understanding of the cellular and molecular mechanisms underlying lissencephaly and highlight the potential of DCX knockout ferrets as a valuable tool for neurodevelopmental research, offering insights into both the pathology of lissencephaly and the general principles of brain development.

2.
Cell Res ; 34(3): 193-213, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38177242

RESUMEN

The spinal cord is a crucial component of the central nervous system that facilitates sensory processing and motor performance. Despite its importance, the spatiotemporal codes underlying human spinal cord development have remained elusive. In this study, we have introduced an image-based single-cell transcription factor (TF) expression decoding spatial transcriptome method (TF-seqFISH) to investigate the spatial expression and regulation of TFs during human spinal cord development. By combining spatial transcriptomic data from TF-seqFISH and single-cell RNA-sequencing data, we uncovered the spatial distribution of neural progenitor cells characterized by combinatorial TFs along the dorsoventral axis, as well as the molecular and spatial features governing neuronal generation, migration, and differentiation along the mediolateral axis. Notably, we observed a sandwich-like organization of excitatory and inhibitory interneurons transiently appearing in the dorsal horns of the developing human spinal cord. In addition, we integrated data from 10× Visium to identify early and late waves of neurogenesis in the dorsal horn, revealing the formation of laminas in the dorsal horns. Our study also illuminated the spatial differences and molecular cues underlying motor neuron (MN) diversification, and the enrichment of Amyotrophic Lateral Sclerosis (ALS) risk genes in MNs and microglia. Interestingly, we detected disease-associated microglia (DAM)-like microglia groups in the developing human spinal cord, which are predicted to be vulnerable to ALS and engaged in the TYROBP causal network and response to unfolded proteins. These findings provide spatiotemporal transcriptomic resources on the developing human spinal cord and potential strategies for spinal cord injury repair and ALS treatment.


Asunto(s)
Esclerosis Amiotrófica Lateral , Factores de Transcripción , Animales , Humanos , Factores de Transcripción/genética , Neurogénesis , Sistema Nervioso Central
3.
Nat Commun ; 14(1): 7613, 2023 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-37993461

RESUMEN

Human cerebellar development is orchestrated by molecular regulatory networks to achieve cytoarchitecture and coordinate motor and cognitive functions. Here, we combined single-cell transcriptomics, spatial transcriptomics and single cell chromatin accessibility states to systematically depict an integrative spatiotemporal landscape of human fetal cerebellar development. We revealed that combinations of transcription factors and cis-regulatory elements (CREs) play roles in governing progenitor differentiation and cell fate determination along trajectories in a hierarchical manner, providing a gene expression regulatory map of cell fate and spatial information for these cells. We also illustrated that granule cells located in different regions of the cerebellar cortex showed distinct molecular signatures regulated by different signals during development. Finally, we mapped single-nucleotide polymorphisms (SNPs) of disorders related to cerebellar dysfunction and discovered that several disorder-associated genes showed spatiotemporal and cell type-specific expression patterns only in humans, indicating the cellular basis and possible mechanisms of the pathogenesis of neuropsychiatric disorders.


Asunto(s)
Epigenómica , Transcriptoma , Humanos , Cromatina/genética , Regulación de la Expresión Génica , Secuencias Reguladoras de Ácidos Nucleicos , Análisis de la Célula Individual
4.
Cell Stem Cell ; 30(6): 851-866.e7, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37192616

RESUMEN

The emergence of the three germ layers and the lineage-specific precursor cells orchestrating organogenesis represent fundamental milestones during early embryonic development. We analyzed the transcriptional profiles of over 400,000 cells from 14 human samples collected from post-conceptional weeks (PCW) 3 to 12 to delineate the dynamic molecular and cellular landscape of early gastrulation and nervous system development. We described the diversification of cell types, the spatial patterning of neural tube cells, and the signaling pathways likely involved in transforming epiblast cells into neuroepithelial cells and then into radial glia. We resolved 24 clusters of radial glial cells along the neural tube and outlined differentiation trajectories for the main classes of neurons. Lastly, we identified conserved and distinctive features across species by comparing early embryonic single-cell transcriptomic profiles between humans and mice. This comprehensive atlas sheds light on the molecular mechanisms underlying gastrulation and early human brain development.


Asunto(s)
Gastrulación , Estratos Germinativos , Humanos , Ratones , Animales , Gastrulación/genética , Diferenciación Celular , Organogénesis , Encéfalo
5.
Sci China Life Sci ; 66(8): 1841-1857, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36929272

RESUMEN

Iron is important for life, and iron deficiency impairs development, but whether the iron level regulates neural differentiation remains elusive. In this study, with iron-regulatory proteins (IRPs) knockout embryonic stem cells (ESCs) that showed severe iron deficiency, we found that the Pax6- and Sox2-positive neuronal precursor cells and Tuj1 fibers in IRP1-/-IRP2-/- ESCs were significantly decreased after inducing neural differentiation. Consistently, in vivo study showed that the knockdown of IRP1 in IRP2-/- fetal mice remarkably affected the differentiation of neuronal precursors and the migration of neurons. These findings suggest that low intracellular iron status significantly inhibits neurodifferentiation. When supplementing IRP1-/-IRP2-/- ESCs with iron, these ESCs could differentiate normally. Further investigations revealed that the underlying mechanism was associated with an increase in reactive oxygen species (ROS) production caused by the substantially low level of iron and the down-regulation of iron-sulfur cluster protein ISCU, which, in turn, affected the proliferation and differentiation of stem cells. Thus, the appropriate amount of iron is crucial for maintaining normal neural differentiation that is termed ferrodifferentiation.


Asunto(s)
Deficiencias de Hierro , Proteínas Hierro-Azufre , Especies Reactivas de Oxígeno , Animales , Ratones , Hierro/metabolismo , Proteína 1 Reguladora de Hierro/metabolismo , Proteína 2 Reguladora de Hierro/metabolismo , Proteínas Hierro-Azufre/metabolismo , Especies Reactivas de Oxígeno/metabolismo
6.
Nat Commun ; 13(1): 3883, 2022 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-35794099

RESUMEN

Epigenetic information regulates gene expression and development. However, our understanding of the evolution of epigenetic regulation on brain development in primates is limited. Here, we compared chromatin accessibility landscapes and transcriptomes during fetal prefrontal cortex (PFC) development between rhesus macaques and humans. A total of 304,761 divergent DNase I-hypersensitive sites (DHSs) are identified between rhesus macaques and humans, although many of these sites share conserved DNA sequences. Interestingly, most of the cis-elements linked to orthologous genes with dynamic expression are divergent DHSs. Orthologous genes expressed at earlier stages tend to have conserved cis-elements, whereas orthologous genes specifically expressed at later stages seldom have conserved cis-elements. These genes are enriched in synapse organization, learning and memory. Notably, DHSs in the PFC at early stages are linked to human educational attainment and cognitive performance. Collectively, the comparison of the chromatin epigenetic landscape between rhesus macaques and humans suggests a potential role for regulatory elements in the evolution of differences in cognitive ability between non-human primates and humans.


Asunto(s)
Cromatina , Epigénesis Genética , Animales , Cromatina/genética , Desoxirribonucleasa I/metabolismo , Humanos , Macaca mulatta/genética , Macaca mulatta/metabolismo , Corteza Prefrontal/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos
7.
Cell Stem Cell ; 29(2): 328-343.e5, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-34879244

RESUMEN

The hypothalamus comprises various nuclei and neuronal subpopulations that control fundamental homeostasis and behaviors. However, spatiotemporal molecular characterization of hypothalamus development in humans is largely unexplored. Here, we revealed spatiotemporal transcriptome profiles and cell-type characteristics of human hypothalamus development and illustrated the molecular diversity of neural progenitors and the cell-fate decision, which is programmed by a combination of transcription factors. Different neuronal and glial fates are sequentially produced and showed spatial developmental asynchrony. Moreover, human hypothalamic gliogenesis occurs at an earlier stage of gestation and displays distinctive transcription profiles compared with those in mouse. Notably, early oligodendrocyte cells in humans exhibit different gene patterns and interact with neuronal cells to regulate neuronal maturation by Wnt, Hippo, and integrin signals. Overall, our study provides a comprehensive molecular landscape of human hypothalamus development at early- and mid-embryonic stages and a foundation for understanding its spatial and functional complexity.


Asunto(s)
Hipotálamo , Neurogénesis , Animales , Humanos , Ratones , Neurogénesis/genética , Neuroglía , Neuronas/fisiología , Oligodendroglía
8.
Science ; 374(6573): eabj6641, 2021 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-34882453

RESUMEN

Genetic variation confers susceptibility to neurodevelopmental disorders by affecting the development of specific cell types. Changes in cortical and striatal γ-aminobutyric acid­expressing (GABAergic) neurons are common in autism and schizophrenia. In this study, we used single-cell RNA sequencing to characterize the emergence of cell diversity in the human ganglionic eminences, the transitory structures of the human fetal brain where striatal and cortical GABAergic neurons are generated. We identified regional and temporal diversity among progenitor cells underlying the generation of a variety of projection neurons and interneurons. We found that these cells are specified within the human ganglionic eminences by transcriptional programs similar to those previously identified in rodents. Our findings reveal an evolutionarily conserved regulatory logic controlling the specification, migration, and differentiation of GABAergic neurons in the human telencephalon.


Asunto(s)
Interneuronas/fisiología , Neurogénesis , Telencéfalo/embriología , Transcriptoma , Animales , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Células-Madre Neurales/fisiología , RNA-Seq , Análisis de la Célula Individual , Telencéfalo/citología , Ácido gamma-Aminobutírico/metabolismo
9.
Natl Sci Rev ; 8(4): nwaa179, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34691611

RESUMEN

The human retina is a complex neural tissue that detects light and sends visual information to the brain. However, the molecular and cellular processes that underlie aging primate retina remain unclear. Here, we provide a comprehensive transcriptomic atlas based on 119 520 single cells of the foveal and peripheral retina of humans and macaques covering different ages. The molecular features of retinal cells differed between the two species, suggesting distinct regional and species specializations of the human and macaque retinae. In addition, human retinal aging occurred in a region- and cell-type-specific manner. Aging of human retina exhibited a foveal to peripheral gradient. MYO9A- rods and a horizontal cell subtype were greatly reduced in aging retina, indicating their vulnerability to aging. Moreover, we generated a dataset showing the cell-type- and region-specific gene expression associated with 55 types of human retinal disease, which provides a foundation to understanding of the molecular and cellular mechanisms underlying human retinal diseases. Such datasets are valuable to understanding of the molecular characteristics of primate retina, as well as molecular regulation of aging progression and related diseases.

10.
Front Immunol ; 12: 707404, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34276703

RESUMEN

Thymic blood vessels at the perivascular space (PVS) are the critical site for both homing of hematopoietic progenitor cells (HPCs) and egress of mature thymocytes. It has been intriguing how different opposite migrations can happen in the same place. A subset of specialized thymic portal endothelial cells (TPECs) associated with PVS has been identified to function as the entry site for HPCs. However, the cellular basis and mechanism underlying egress of mature thymocytes has not been well defined. In this study, using various conventional and conditional gene-deficient mouse models, we first confirmed the role of endothelial lymphotoxin beta receptor (LTßR) for thymic egress and ruled out the role of LTßR from epithelial cells or dendritic cells. In addition, we found that T cell-derived ligands lymphotoxin (LT) and LIGHT are required for thymic egress, suggesting a crosstalk between T cells and endothelial cells (ECs) for thymic egress control. Furthermore, immunofluorescence staining analysis interestingly showed that TPECs are also the exit site for mature thymocytes. Single-cell transcriptomic analysis of thymic endothelial cells suggested that TPECs are heterogeneous and can be further divided into two subsets depending on BST-1 expression level. Importantly, BST-1hi population is associated with thymic egressing thymocytes while BST-1lo/- population is associated with HPC settling. Thus, we have defined a LT/LIGHT-LTßR signaling-mediated cellular crosstalk regulating thymic egress and uncovered distinct subsets of TPECs controlling thymic homing and egress, respectively.


Asunto(s)
Movimiento Celular/fisiología , Células Endoteliales/metabolismo , Receptor beta de Linfotoxina/metabolismo , Timocitos/metabolismo , Timo/metabolismo , Animales , Linfotoxina-alfa/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/inmunología , Linfocitos T/metabolismo , Timo/citología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
11.
Sci China Life Sci ; 64(3): 419-433, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32803714

RESUMEN

Cenpj is a centrosomal protein located at the centrosomes and the base of cilia, it plays essential roles in regulating neurogenesis and cerebral cortex development. Although centrosomal and cilium dysfunction are one of the causes of obesity, insulin resistance, and type 2 diabetes, the role that Cenpj plays in the regulation of body weight remains unclear. Here, we deleted Cenpj by crossing Cenpjflox/flox mice with Nkx2.1-Cre mice. Loss of the centrosomal protein Cenpj in Nkx2.1-expressing cells causes morbid obesity in mice at approximately 4 months of age with expended brain ventricles but no change of brain size. We found that hypothalamic cells exhibited reduced proliferation and increased apoptosis upon Cenpj depletion at the embryonic stages, resulting in a dramatic decrease in the number of Proopiomelanocortin (POMC) neurons and electrophysiological dysfunction of NPY neurons in the arcuate nucleus (ARC) in adults. Furthermore, depletion of Cenpj also reduced the neuronal projection from the ARC to the paraventricular nucleus (PVN), with decreased melanocortin-4 receptors (MC4R) expression in PVN neurons. The study defines the roles that Cenpj plays in regulating hypothalamus development and body weight, providing a foundation for further understanding of the pathological mechanisms of related diseases.


Asunto(s)
Técnicas de Silenciamiento del Gen , Hipotálamo/fisiopatología , Proteínas Asociadas a Microtúbulos/genética , Obesidad Mórbida/fisiopatología , Animales , Apoptosis , Línea Celular , Proliferación Celular , Hipotálamo/embriología , Hipotálamo/metabolismo , Ratones , Neuronas/metabolismo , Obesidad Mórbida/genética , Proopiomelanocortina/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Factor Nuclear Tiroideo 1/genética
12.
Nat Commun ; 11(1): 4063, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32792525

RESUMEN

The neuroendocrine hypothalamus is the central regulator of vital physiological homeostasis and behavior. However, the cellular and molecular properties of hypothalamic neural progenitors remain unexplored. Here, hypothalamic radial glial (hRG) and hypothalamic mantle zone radial glial (hmRG) cells are found to be neural progenitors in the developing mammalian hypothalamus. The hmRG cells originate from hRG cells and produce neurons. During the early development of hypothalamus, neurogenesis occurs in radial columns and is initiated from hRG cells. The radial glial fibers are oriented toward the locations of hypothalamic subregions which act as a scaffold for neuronal migration. Furthermore, we use single-cell RNA sequencing to reveal progenitor subtypes in human developing hypothalamus and characterize specific progenitor genes, such as TTYH1, HMGA2, and FAM107A. We also demonstrate that HMGA2 is involved in E2F1 pathway, regulating the proliferation of progenitor cells by targeting on the downstream MYBL2. Different neuronal subtypes start to differentiate and express specific genes of hypothalamic nucleus at gestational week 10. Finally, we reveal the developmental conservation of nuclear structures and marker genes in mouse and human hypothalamus. Our identification of cellular and molecular properties of neural progenitors provides a basic understanding of neurogenesis and regional formation of the non-laminated hypothalamus.


Asunto(s)
Hipotálamo/citología , Hipotálamo/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Animales , Análisis por Conglomerados , Femenino , Genes Supresores de Tumor , Proteína HMGA2/genética , Proteína HMGA2/metabolismo , Humanos , Hibridación in Situ , Mamíferos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Neurogénesis/genética , Neurogénesis/fisiología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Embarazo
13.
PLoS Biol ; 18(5): e3000705, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32401820

RESUMEN

Modeling the processes of neuronal progenitor proliferation and differentiation to produce mature cortical neuron subtypes is essential for the study of human brain development and the search for potential cell therapies. We demonstrated a novel paradigm for the generation of vascularized organoids (vOrganoids) consisting of typical human cortical cell types and a vascular structure for over 200 days as a vascularized and functional brain organoid model. The observation of spontaneous excitatory postsynaptic currents (sEPSCs), spontaneous inhibitory postsynaptic currents (sIPSCs), and bidirectional electrical transmission indicated the presence of chemical and electrical synapses in vOrganoids. More importantly, single-cell RNA-sequencing analysis illustrated that vOrganoids exhibited robust neurogenesis and that cells of vOrganoids differentially expressed genes (DEGs) related to blood vessel morphogenesis. The transplantation of vOrganoids into the mouse S1 cortex resulted in the construction of functional human-mouse blood vessels in the grafts that promoted cell survival in the grafts. This vOrganoid culture method could not only serve as a model to study human cortical development and explore brain disease pathology but also provide potential prospects for new cell therapies for nervous system disorders and injury.


Asunto(s)
Técnicas de Cultivo de Célula , Neurogénesis , Organoides/irrigación sanguínea , Telencéfalo/embriología , Animales , Células Madre Embrionarias , Células Endoteliales de la Vena Umbilical Humana , Humanos , Células Madre Pluripotentes Inducidas , Ratones Endogámicos NOD , Ratones SCID , Organoides/metabolismo , Organoides/trasplante
14.
Dev Cell ; 53(4): 473-491.e9, 2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32386599

RESUMEN

The development of single-cell RNA sequencing (scRNA-seq) has allowed high-resolution analysis of cell-type diversity and transcriptional networks controlling cell-fate specification. To identify the transcriptional networks governing human retinal development, we performed scRNA-seq analysis on 16 time points from developing retina as well as four early stages of retinal organoid differentiation. We identified evolutionarily conserved patterns of gene expression during retinal progenitor maturation and specification of all seven major retinal cell types. Furthermore, we identified gene-expression differences between developing macula and periphery and between distinct populations of horizontal cells. We also identified species-specific patterns of gene expression during human and mouse retinal development. Finally, we identified an unexpected role for ATOH7 expression in regulation of photoreceptor specification during late retinogenesis. These results provide a roadmap to future studies of human retinal development and may help guide the design of cell-based therapies for treating retinal dystrophies.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Evolución Biológica , Regulación del Desarrollo de la Expresión Génica , Organogénesis , Retina/citología , Células Fotorreceptoras Retinianas Conos/metabolismo , Análisis de la Célula Individual/métodos , Anciano de 80 o más Años , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular , Femenino , Humanos , Ratones , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/citología , Especificidad de la Especie
15.
Sci Adv ; 6(6): eaay5247, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32083182

RESUMEN

Retinal organoids (ROs) derived from human induced pluripotent stem cells (hiPSCs) provide potential opportunities for studying human retinal development and disorders; however, to what extent ROs recapitulate the epigenetic features of human retinal development is unknown. In this study, we systematically profiled chromatin accessibility and transcriptional dynamics over long-term human retinal and RO development. Our results showed that ROs recapitulated the human retinogenesis to a great extent, but divergent chromatin features were also discovered. We further reconstructed the transcriptional regulatory network governing human and RO retinogenesis in vivo. Notably, NFIB and THRA were identified as regulators in human retinal development. The chromatin modifications between developing human and mouse retina were also cross-analyzed. Notably, we revealed an enriched bivalent modification of H3K4me3 and H3K27me3 in human but not in murine retinogenesis, suggesting a more dedicated epigenetic regulation on human genome.


Asunto(s)
Ensamble y Desensamble de Cromatina , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Organogénesis , Organoides , Retina/citología , Retina/metabolismo , Animales , Biomarcadores , Diferenciación Celular/genética , Cromatina/genética , Epigénesis Genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Inmunohistoquímica , Ratones , Transducción de Señal
16.
Nature ; 577(7791): 531-536, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31942070

RESUMEN

The hippocampus is an important part of the limbic system in the human brain that has essential roles in spatial navigation and the consolidation of information from short-term memory to long-term memory1,2. Here we use single-cell RNA sequencing and assay for transposase-accessible chromatin using sequencing (ATAC-seq) analysis to illustrate the cell types, cell linage, molecular features and transcriptional regulation of the developing human hippocampus. Using the transcriptomes of 30,416 cells from the human hippocampus at gestational weeks 16-27, we identify 47 cell subtypes and their developmental trajectories. We also identify the migrating paths and cell lineages of PAX6+ and HOPX+ hippocampal progenitors, and regional markers of CA1, CA3 and dentate gyrus neurons. Multiomic data have uncovered transcriptional regulatory networks of the dentate gyrus marker PROX1. We also illustrate spatially specific gene expression in the developing human prefrontal cortex and hippocampus. The molecular features of the human hippocampus at gestational weeks 16-20 are similar to those of the mouse at postnatal days 0-5 and reveal gene expression differences between the two species. Transient expression of the primate-specific gene NBPF1 leads to a marked increase in PROX1+ cells in the mouse hippocampus. These data provides a blueprint for understanding human hippocampal development and a tool for investigating related diseases.


Asunto(s)
Linaje de la Célula , Regulación del Desarrollo de la Expresión Génica/genética , Hipocampo/citología , Hipocampo/embriología , Animales , Proteínas Portadoras/metabolismo , Giro Dentado/citología , Giro Dentado/embriología , Giro Dentado/metabolismo , Evolución Molecular , Femenino , Hipocampo/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis , Neuronas/citología , Neuronas/metabolismo , Factor de Transcripción PAX6/metabolismo , Corteza Prefrontal/citología , Corteza Prefrontal/embriología , Corteza Prefrontal/metabolismo , Especificidad de la Especie , Transcriptoma/genética , Proteínas Supresoras de Tumor/metabolismo
17.
Cell Res ; 28(7): 730-745, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29867213

RESUMEN

The cellular complexity of human brain development has been intensively investigated, although a regional characterization of the entire human cerebral cortex based on single-cell transcriptome analysis has not been reported. Here, we performed RNA-seq on over 4,000 individual cells from 22 brain regions of human mid-gestation embryos. We identified 29 cell sub-clusters, which showed different proportions in each region and the pons showed especially high percentage of astrocytes. Embryonic neurons were not as diverse as adult neurons, although they possessed important features of their destinies in adults. Neuron development was unsynchronized in the cerebral cortex, as dorsal regions appeared to be more mature than ventral regions at this stage. Region-specific genes were comprehensively identified in each neuronal sub-cluster, and a large proportion of these genes were neural disease related. Our results present a systematic landscape of the regionalized gene expression and neuron maturation of the human cerebral cortex.


Asunto(s)
Corteza Cerebral/embriología , Desarrollo Embrionario/genética , Enfermedades del Sistema Nervioso/genética , Neurogénesis/genética , Análisis de la Célula Individual/métodos , Astrocitos/citología , Secuencia de Bases , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Masculino , Neuronas/citología , Embarazo , Segundo Trimestre del Embarazo , Análisis de Secuencia de ARN/métodos
18.
Nature ; 555(7697): 524-528, 2018 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-29539641

RESUMEN

The mammalian prefrontal cortex comprises a set of highly specialized brain areas containing billions of cells and serves as the centre of the highest-order cognitive functions, such as memory, cognitive ability, decision-making and social behaviour. Although neural circuits are formed in the late stages of human embryonic development and even after birth, diverse classes of functional cells are generated and migrate to the appropriate locations earlier in development. Dysfunction of the prefrontal cortex contributes to cognitive deficits and the majority of neurodevelopmental disorders; there is therefore a need for detailed knowledge of the development of the prefrontal cortex. However, it is still difficult to identify cell types in the developing human prefrontal cortex and to distinguish their developmental features. Here we analyse more than 2,300 single cells in the developing human prefrontal cortex from gestational weeks 8 to 26 using RNA sequencing. We identify 35 subtypes of cells in six main classes and trace the developmental trajectories of these cells. Detailed analysis of neural progenitor cells highlights new marker genes and unique developmental features of intermediate progenitor cells. We also map the timeline of neurogenesis of excitatory neurons in the prefrontal cortex and detect the presence of interneuron progenitors in early developing prefrontal cortex. Moreover, we reveal the intrinsic development-dependent signals that regulate neuron generation and circuit formation using single-cell transcriptomic data analysis. Our screening and characterization approach provides a blueprint for understanding the development of the human prefrontal cortex in the early and mid-gestational stages in order to systematically dissect the cellular basis and molecular regulation of prefrontal cortex function in humans.


Asunto(s)
Diferenciación Celular/genética , Corteza Prefrontal/citología , Corteza Prefrontal/embriología , ARN/análisis , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Humanos , Interneuronas/citología , Interneuronas/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Neuronas/clasificación , Neuronas/citología , Neuronas/metabolismo , ARN/genética , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA