Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell ; 187(13): 3236-3248.e21, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38772369

RESUMEN

Leveraging AAVs' versatile tropism and labeling capacity, we expanded the scale of in vivo CRISPR screening with single-cell transcriptomic phenotyping across embryonic to adult brains and peripheral nervous systems. Through extensive tests of 86 vectors across AAV serotypes combined with a transposon system, we substantially amplified labeling efficacy and accelerated in vivo gene delivery from weeks to days. Our proof-of-principle in utero screen identified the pleiotropic effects of Foxg1, highlighting its tight regulation of distinct networks essential for cell fate specification of Layer 6 corticothalamic neurons. Notably, our platform can label >6% of cerebral cells, surpassing the current state-of-the-art efficacy at <0.1% by lentivirus, to achieve analysis of over 30,000 cells in one experiment and enable massively parallel in vivo Perturb-seq. Compatible with various phenotypic measurements (single-cell or spatial multi-omics), it presents a flexible approach to interrogate gene function across cell types in vivo, translating gene variants to their causal function.


Asunto(s)
Redes Reguladoras de Genes , Análisis de la Célula Individual , Animales , Ratones , Análisis de la Célula Individual/métodos , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Corteza Cerebral/metabolismo , Corteza Cerebral/citología , Neuronas/metabolismo , Neuronas/citología , Femenino , Dependovirus/genética , Humanos , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Sistemas CRISPR-Cas/genética , Vectores Genéticos/metabolismo , Ratones Endogámicos C57BL , Transcriptoma/genética
2.
bioRxiv ; 2023 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-37790302

RESUMEN

Systematic analysis of gene function across diverse cell types in vivo is hindered by two challenges: obtaining sufficient cells from live tissues and accurately identifying each cell's perturbation in high-throughput single-cell assays. Leveraging AAV's versatile cell type tropism and high labeling capacity, we expanded the resolution and scale of in vivo CRISPR screens: allowing phenotypic analysis at single-cell resolution across a multitude of cell types in the embryonic brain, adult brain, and peripheral nervous system. We undertook extensive tests of 86 AAV serotypes, combined with a transposon system, to substantially amplify labeling and accelerate in vivo gene delivery from weeks to days. Using this platform, we performed an in utero genetic screen as proof-of-principle and identified pleiotropic regulatory networks of Foxg1 in cortical development, including Layer 6 corticothalamic neurons where it tightly controls distinct networks essential for cell fate specification. Notably, our platform can label >6% of cerebral cells, surpassing the current state-of-the-art efficacy at <0.1% (mediated by lentivirus), and achieve analysis of over 30,000 cells in one experiment, thus enabling massively parallel in vivo Perturb-seq. Compatible with various perturbation techniques (CRISPRa/i) and phenotypic measurements (single-cell or spatial multi-omics), our platform presents a flexible, modular approach to interrogate gene function across diverse cell types in vivo, connecting gene variants to their causal functions.

3.
Neuron ; 111(21): 3378-3396.e9, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37657442

RESUMEN

A genetically valid animal model could transform our understanding of schizophrenia (SCZ) disease mechanisms. Rare heterozygous loss-of-function (LoF) mutations in GRIN2A, encoding a subunit of the NMDA receptor, greatly increase the risk of SCZ. By transcriptomic, proteomic, and behavioral analyses, we report that heterozygous Grin2a mutant mice show (1) large-scale gene expression changes across multiple brain regions and in neuronal (excitatory and inhibitory) and non-neuronal cells (astrocytes and oligodendrocytes), (2) evidence of hypoactivity in the prefrontal cortex (PFC) and hyperactivity in the hippocampus and striatum, (3) an elevated dopamine signaling in the striatum and hypersensitivity to amphetamine-induced hyperlocomotion (AIH), (4) altered cholesterol biosynthesis in astrocytes, (5) a reduction in glutamatergic receptor signaling proteins in the synapse, and (6) an aberrant locomotor pattern opposite of that induced by antipsychotic drugs. These findings reveal potential pathophysiologic mechanisms, provide support for both the "hypo-glutamate" and "hyper-dopamine" hypotheses of SCZ, and underscore the utility of Grin2a-deficient mice as a genetic model of SCZ.


Asunto(s)
Dopamina , Proteómica , Receptores de N-Metil-D-Aspartato , Animales , Ratones , Encéfalo/metabolismo , Dopamina/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Corteza Prefrontal/metabolismo , Modelos Animales de Enfermedad , Receptores de N-Metil-D-Aspartato/genética
4.
Nat Aging ; 3(3): 327-345, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-37118429

RESUMEN

Aging is a complex process involving transcriptomic changes associated with deterioration across multiple tissues and organs, including the brain. Recent studies using heterochronic parabiosis have shown that various aspects of aging-associated decline are modifiable or even reversible. To better understand how this occurs, we performed single-cell transcriptomic profiling of young and old mouse brains after parabiosis. For each cell type, we cataloged alterations in gene expression, molecular pathways, transcriptional networks, ligand-receptor interactions and senescence status. Our analyses identified gene signatures, demonstrating that heterochronic parabiosis regulates several hallmarks of aging in a cell-type-specific manner. Brain endothelial cells were found to be especially malleable to this intervention, exhibiting dynamic transcriptional changes that affect vascular structure and function. These findings suggest new strategies for slowing deterioration and driving regeneration in the aging brain through approaches that do not rely on disease-specific mechanisms or actions of individual circulating factors.


Asunto(s)
Células Endoteliales , Transcriptoma , Animales , Ratones , Transcriptoma/genética , Envejecimiento/genética , Parabiosis , Encéfalo
5.
Nat Biotechnol ; 41(2): 204-211, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36109685

RESUMEN

Here we introduce a mostly natural sequencing-by-synthesis (mnSBS) method for single-cell RNA sequencing (scRNA-seq), adapted to the Ultima genomics platform, and systematically benchmark it against current scRNA-seq technology. mnSBS uses mostly natural, unmodified nucleotides and only a low fraction of fluorescently labeled nucleotides, which allows for high polymerase processivity and lower costs. We demonstrate successful application in four scRNA-seq case studies of different technical and biological types, including 5' and 3' scRNA-seq, human peripheral blood mononuclear cells from a single individual and in multiplex, as well as Perturb-Seq. Benchmarking shows that results from mnSBS-based scRNA-seq are very similar to those using Illumina sequencing, with minor differences in results related to the position of reads relative to annotated gene boundaries, owing to single-end reads of Ultima being closer to gene ends than reads from Illumina. The method is thus compatible with state-of-the-art scRNA-seq libraries independent of the sequencing technology. We expect mnSBS to be of particular utility for cost-effective large-scale scRNA-seq projects.


Asunto(s)
Perfilación de la Expresión Génica , Leucocitos Mononucleares , Humanos , Perfilación de la Expresión Génica/métodos , Análisis de Secuencia de ARN/métodos , Análisis de Expresión Génica de una Sola Célula , Análisis de la Célula Individual/métodos , Nucleótidos
6.
Nature ; 602(7896): 268-273, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35110736

RESUMEN

Genetic risk for autism spectrum disorder (ASD) is associated with hundreds of genes spanning a wide range of biological functions1-6. The alterations in the human brain resulting from mutations in these genes remain unclear. Furthermore, their phenotypic manifestation varies across individuals7,8. Here we used organoid models of the human cerebral cortex to identify cell-type-specific developmental abnormalities that result from haploinsufficiency in three ASD risk genes-SUV420H1 (also known as KMT5B), ARID1B and CHD8-in multiple cell lines from different donors, using single-cell RNA-sequencing (scRNA-seq) analysis of more than 745,000 cells and proteomic analysis of individual organoids, to identify phenotypic convergence. Each of the three mutations confers asynchronous development of two main cortical neuronal lineages-γ-aminobutyric-acid-releasing (GABAergic) neurons and deep-layer excitatory projection neurons-but acts through largely distinct molecular pathways. Although these phenotypes are consistent across cell lines, their expressivity is influenced by the individual genomic context, in a manner that is dependent on both the risk gene and the developmental defect. Calcium imaging in intact organoids shows that these early-stage developmental changes are followed by abnormal circuit activity. This research uncovers cell-type-specific neurodevelopmental abnormalities that are shared across ASD risk genes and are finely modulated by human genomic context, finding convergence in the neurobiological basis of how different risk genes contribute to ASD pathology.


Asunto(s)
Trastorno del Espectro Autista , Predisposición Genética a la Enfermedad , Neuronas , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/patología , Corteza Cerebral/citología , Proteínas de Unión al ADN/genética , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/patología , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Neuronas/clasificación , Neuronas/metabolismo , Neuronas/patología , Organoides/citología , Proteómica , RNA-Seq , Análisis de la Célula Individual , Factores de Transcripción/genética
7.
Genome Biol ; 22(1): 73, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33663567

RESUMEN

BACKGROUND: Many neurodegenerative diseases develop only later in life, when cells in the nervous system lose their structure or function. In many forms of neurodegenerative diseases, this late-onset phenomenon remains largely unexplained. RESULTS: Analyzing single-cell RNA sequencing from Alzheimer's disease (AD) and Huntington's disease (HD) patients, we find increased transcriptional heterogeneity in disease-state neurons. We hypothesize that transcriptional heterogeneity precedes neurodegenerative disease pathologies. To test this idea experimentally, we use juvenile forms (72Q; 180Q) of HD iPSCs, differentiate them into committed neuronal progenitors, and obtain single-cell expression profiles. We show a global increase in gene expression variability in HD. Autophagy genes become more stable, while energy and actin-related genes become more variable in the mutant cells. Knocking down several differentially variable genes results in increased aggregate formation, a pathology associated with HD. We further validate the increased transcriptional heterogeneity in CHD8+/- cells, a model for autism spectrum disorder. CONCLUSIONS: Overall, our results suggest that although neurodegenerative diseases develop over time, transcriptional regulation imbalance is present already at very early developmental stages. Therefore, an intervention aimed at this early phenotype may be of high diagnostic value.


Asunto(s)
Regulación de la Expresión Génica , Heterogeneidad Genética , Predisposición Genética a la Enfermedad , Modelos Biológicos , Enfermedades Neurodegenerativas/etiología , Células Madre Pluripotentes/metabolismo , Adulto , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Antecedentes Genéticos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mutación , RNA-Seq , Análisis de la Célula Individual/métodos
8.
Science ; 370(6520)2020 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-33243861

RESUMEN

The number of disease risk genes and loci identified through human genetic studies far outstrips the capacity to systematically study their functions. We applied a scalable genetic screening approach, in vivo Perturb-Seq, to functionally evaluate 35 autism spectrum disorder/neurodevelopmental delay (ASD/ND) de novo loss-of-function risk genes. Using CRISPR-Cas9, we introduced frameshift mutations in these risk genes in pools, within the developing mouse brain in utero, followed by single-cell RNA-sequencing of perturbed cells in the postnatal brain. We identified cell type-specific and evolutionarily conserved gene modules from both neuronal and glial cell classes. Recurrent gene modules and cell types are affected across this cohort of perturbations, representing key cellular effects across sets of ASD/ND risk genes. In vivo Perturb-Seq allows us to investigate how diverse mutations affect cell types and states in the developing organism.


Asunto(s)
Trastorno Autístico/genética , Trastorno Autístico/patología , Encéfalo/anomalías , Neuroglía/patología , Neuronas/patología , Animales , Ancirinas/genética , Ancirinas/metabolismo , Sistemas CRISPR-Cas , Proteínas de Unión al ADN/genética , Mutación del Sistema de Lectura , Perfilación de la Expresión Génica , Sitios Genéticos , Humanos , Ratones , Neuroglía/metabolismo , Neuronas/metabolismo , Proteínas Represoras/genética , Riesgo , Factores de Transcripción/genética
9.
Nature ; 583(7818): 819-824, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32699411

RESUMEN

The thalamic reticular nucleus (TRN), the major source of thalamic inhibition, regulates thalamocortical interactions that are critical for sensory processing, attention and cognition1-5. TRN dysfunction has been linked to sensory abnormality, attention deficit and sleep disturbance across multiple neurodevelopmental disorders6-9. However, little is known about the organizational principles that underlie its divergent functions. Here we performed an integrative study linking single-cell molecular and electrophysiological features of the mouse TRN to connectivity and systems-level function. We found that cellular heterogeneity in the TRN is characterized by a transcriptomic gradient of two negatively correlated gene-expression profiles, each containing hundreds of genes. Neurons in the extremes of this transcriptomic gradient express mutually exclusive markers, exhibit core or shell-like anatomical structure and have distinct electrophysiological properties. The two TRN subpopulations make differential connections with the functionally distinct first-order and higher-order thalamic nuclei to form molecularly defined TRN-thalamus subnetworks. Selective perturbation of the two subnetworks in vivo revealed their differential role in regulating sleep. In sum, our study provides a comprehensive atlas of TRN neurons at single-cell resolution and links molecularly defined subnetworks to the functional organization of thalamocortical circuits.


Asunto(s)
Redes Reguladoras de Genes , Núcleos Talámicos/citología , Núcleos Talámicos/metabolismo , Animales , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Hibridación Fluorescente in Situ , Metaloendopeptidasas/metabolismo , Ratones , Vías Nerviosas , Neuronas/metabolismo , Osteopontina/metabolismo , Técnicas de Placa-Clamp , RNA-Seq , Análisis de la Célula Individual , Sueño/genética , Sueño/fisiología , Núcleos Talámicos/fisiología , Transcriptoma
10.
Nat Biotechnol ; 38(6): 737-746, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32341560

RESUMEN

The scale and capabilities of single-cell RNA-sequencing methods have expanded rapidly in recent years, enabling major discoveries and large-scale cell mapping efforts. However, these methods have not been systematically and comprehensively benchmarked. Here, we directly compare seven methods for single-cell and/or single-nucleus profiling-selecting representative methods based on their usage and our expertise and resources to prepare libraries-including two low-throughput and five high-throughput methods. We tested the methods on three types of samples: cell lines, peripheral blood mononuclear cells and brain tissue, generating 36 libraries in six separate experiments in a single center. To directly compare the methods and avoid processing differences introduced by the existing pipelines, we developed scumi, a flexible computational pipeline that can be used with any single-cell RNA-sequencing method. We evaluated the methods for both basic performance, such as the structure and alignment of reads, sensitivity and extent of multiplets, and for their ability to recover known biological information in the samples.


Asunto(s)
Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Programas Informáticos , Animales , Encéfalo/citología , Células Cultivadas , Humanos , Leucocitos Mononucleares/citología , Ratones
12.
Nat Neurosci ; 22(10): 1696-1708, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31551601

RESUMEN

The mammalian brain is complex, with multiple cell types performing a variety of diverse functions, but exactly how each cell type is affected in aging remains largely unknown. Here we performed a single-cell transcriptomic analysis of young and old mouse brains. We provide comprehensive datasets of aging-related genes, pathways and ligand-receptor interactions in nearly all brain cell types. Our analysis identified gene signatures that vary in a coordinated manner across cell types and gene sets that are regulated in a cell-type specific manner, even at times in opposite directions. These data reveal that aging, rather than inducing a universal program, drives a distinct transcriptional course in each cell population, and they highlight key molecular processes, including ribosome biogenesis, underlying brain aging. Overall, these large-scale datasets (accessible online at https://portals.broadinstitute.org/single_cell/study/aging-mouse-brain ) provide a resource for the neuroscience community that will facilitate additional discoveries directed towards understanding and modifying the aging process.


Asunto(s)
Envejecimiento/genética , Encéfalo/crecimiento & desarrollo , Neuronas/fisiología , Análisis de la Célula Individual , Transcriptoma/genética , Animales , Encéfalo/citología , Comunicación Celular/genética , Linaje de la Célula/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Masculino , Ratones , Ratones Endogámicos C57BL , Ribosomas/genética
13.
Nature ; 570(7762): 523-527, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31168097

RESUMEN

Experimental models of the human brain are needed for basic understanding of its development and disease1. Human brain organoids hold unprecedented promise for this purpose; however, they are plagued by high organoid-to-organoid variability2,3. This has raised doubts as to whether developmental processes of the human brain can occur outside the context of embryogenesis with a degree of reproducibility that is comparable to the endogenous tissue. Here we show that an organoid model of the dorsal forebrain can reliably generate a rich diversity of cell types appropriate for the human cerebral cortex. We performed single-cell RNA-sequencing analysis of 166,242 cells isolated from 21 individual organoids, finding that 95% of the organoids generate a virtually indistinguishable compendium of cell types, following similar developmental trajectories and with a degree of organoid-to-organoid variability comparable to that of individual endogenous brains. Furthermore, organoids derived from different stem cell lines show consistent reproducibility in the cell types produced. The data demonstrate that reproducible development of the complex cellular diversity of the central nervous system does not require the context of the embryo, and that establishment of terminal cell identity is a highly constrained process that can emerge from diverse stem cell origins and growth environments.


Asunto(s)
Corteza Cerebral/citología , Organoides/citología , Técnicas de Cultivo de Tejidos , Línea Celular , Corteza Cerebral/metabolismo , Femenino , Feto/citología , Feto/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Masculino , Organoides/metabolismo , Prosencéfalo/citología , Prosencéfalo/metabolismo , RNA-Seq , Reproducibilidad de los Resultados , Análisis de la Célula Individual , Factores de Tiempo , Técnicas de Cultivo de Tejidos/normas , Transcriptoma/genética
14.
Nat Methods ; 15(12): 1126, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30459407

RESUMEN

The original version of this paper contained an incorrect primer sequence. In the Methods subsection "Rampage libraries," the text for modification 3 stated that the reverse primer used for library indexing was 5'-CAAGCAGAAGACGGCATACGAGATXXXXXXXXGTGACTGGAGT-3'. The correct sequence of the oligonucleotide used is 5'-CAAGCAGAAGACGGCATACGAGATXXXXXXXXGTGACTGGAGTTCAGACGTGTGCTCTTCCGATCT-3'. This error has been corrected in the PDF and HTML versions of the paper.

15.
Nat Methods ; 15(7): 505-511, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29867192

RESUMEN

Specialized RNA-seq methods are required to identify the 5' ends of transcripts, which are critical for studies of gene regulation, but these methods have not been systematically benchmarked. We directly compared six such methods, including the performance of five methods on a single human cellular RNA sample and a new spike-in RNA assay that helps circumvent challenges resulting from uncertainties in annotation and RNA processing. We found that the 'cap analysis of gene expression' (CAGE) method performed best for mRNA and that most of its unannotated peaks were supported by evidence from other genomic methods. We applied CAGE to eight brain-related samples and determined sample-specific transcription start site (TSS) usage, as well as a transcriptome-wide shift in TSS usage between fetal and adult brain.


Asunto(s)
ARN/química , Análisis de Secuencia de ARN/métodos , Secuencia de Bases , Encéfalo , Células Madre Embrionarias , Biblioteca de Genes , Humanos , ARN/genética , ARN/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...