Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Nature ; 573(7772): 61-68, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31435019

RESUMO

Elucidating the cellular architecture of the human cerebral cortex is central to understanding our cognitive abilities and susceptibility to disease. Here we used single-nucleus RNA-sequencing analysis to perform a comprehensive study of cell types in the middle temporal gyrus of human cortex. We identified a highly diverse set of excitatory and inhibitory neuron types that are mostly sparse, with excitatory types being less layer-restricted than expected. Comparison to similar mouse cortex single-cell RNA-sequencing datasets revealed a surprisingly well-conserved cellular architecture that enables matching of homologous types and predictions of properties of human cell types. Despite this general conservation, we also found extensive differences between homologous human and mouse cell types, including marked alterations in proportions, laminar distributions, gene expression and morphology. These species-specific features emphasize the importance of directly studying human brain.


Assuntos
Astrócitos/classificação , Evolução Biológica , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Neurônios/classificação , Adolescente , Adulto , Idoso , Animais , Astrócitos/citologia , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Inibição Neural , Neurônios/citologia , Análise de Componente Principal , RNA-Seq , Análise de Célula Única , Especificidade da Espécie , Transcriptoma/genética , Adulto Jovem
2.
Nature ; 563(7729): 72-78, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30382198

RESUMO

The neocortex contains a multitude of cell types that are segregated into layers and functionally distinct areas. To investigate the diversity of cell types across the mouse neocortex, here we analysed 23,822 cells from two areas at distant poles of the mouse neocortex: the primary visual cortex and the anterior lateral motor cortex. We define 133 transcriptomic cell types by deep, single-cell RNA sequencing. Nearly all types of GABA (γ-aminobutyric acid)-containing neurons are shared across both areas, whereas most types of glutamatergic neurons were found in one of the two areas. By combining single-cell RNA sequencing and retrograde labelling, we match transcriptomic types of glutamatergic neurons to their long-range projection specificity. Our study establishes a combined transcriptomic and projectional taxonomy of cortical cell types from functionally distinct areas of the adult mouse cortex.


Assuntos
Perfilação da Expressão Gênica , Neocórtex/citologia , Neocórtex/metabolismo , Animais , Biomarcadores/análise , Feminino , Neurônios GABAérgicos/metabolismo , Ácido Glutâmico/metabolismo , Masculino , Camundongos , Córtex Motor/anatomia & histologia , Córtex Motor/citologia , Córtex Motor/metabolismo , Neocórtex/anatomia & histologia , Especificidade de Órgãos , Análise de Sequência de RNA , Análise de Célula Única , Córtex Visual/anatomia & histologia , Córtex Visual/citologia , Córtex Visual/metabolismo
4.
Nature ; 505(7484): 555-8, 2014 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-24451543

RESUMO

Sexually dimorphic mammalian tissues, including sexual organs and the brain, contain stem cells that are directly or indirectly regulated by sex hormones. An important question is whether stem cells also exhibit sex differences in physiological function and hormonal regulation in tissues that do not show sex-specific morphological differences. The terminal differentiation and function of some haematopoietic cells are regulated by sex hormones, but haematopoietic stem-cell function is thought to be similar in both sexes. Here we show that mouse haematopoietic stem cells exhibit sex differences in cell-cycle regulation by oestrogen. Haematopoietic stem cells in female mice divide significantly more frequently than in male mice. This difference depends on the ovaries but not the testes. Administration of oestradiol, a hormone produced mainly in the ovaries, increased haematopoietic stem-cell division in males and females. Oestrogen levels increased during pregnancy, increasing haematopoietic stem-cell division, haematopoietic stem-cell frequency, cellularity, and erythropoiesis in the spleen. Haematopoietic stem cells expressed high levels of oestrogen receptor-α (ERα). Conditional deletion of ERα from haematopoietic stem cells reduced haematopoietic stem-cell division in female, but not male, mice and attenuated the increases in haematopoietic stem-cell division, haematopoietic stem-cell frequency, and erythropoiesis during pregnancy. Oestrogen/ERα signalling promotes haematopoietic stem-cell self-renewal, expanding splenic haematopoietic stem cells and erythropoiesis during pregnancy.


Assuntos
Estrogênios/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Animais , Contagem de Células , Divisão Celular/efeitos dos fármacos , Eritropoese , Receptor alfa de Estrogênio/metabolismo , Estrogênios/farmacologia , Feminino , Células-Tronco Hematopoéticas/efeitos dos fármacos , Masculino , Camundongos , Ovário/efeitos dos fármacos , Ovário/metabolismo , Gravidez , Caracteres Sexuais , Transdução de Sinais/efeitos dos fármacos , Baço/citologia
5.
Nat Methods ; 13(1): 87-93, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26524239

RESUMO

The diverse progenitors that give rise to the human neocortex have been difficult to characterize because progenitors, particularly radial glia (RG), are rare and are defined by a combination of intracellular markers, position and morphology. To circumvent these problems, we developed Fixed and Recovered Intact Single-cell RNA (FRISCR), a method for profiling the transcriptomes of individual fixed, stained and sorted cells. Using FRISCR, we profiled primary human RG that constitute only 1% of the midgestation cortex and classified them as ventricular zone-enriched RG (vRG) that express ANXA1 and CRYAB, and outer subventricular zone-localized RG (oRG) that express HOPX. Our study identified vRG and oRG markers and molecular profiles, an essential step for understanding human neocortical progenitor development. FRISCR allows targeted single-cell profiling of any tissues that lack live-cell markers.


Assuntos
Encéfalo/citologia , Neuroglia/citologia , Transcriptoma , Humanos , Análise de Célula Única
6.
PLoS Genet ; 7(7): e1002087, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21750678

RESUMO

Many signaling proteins and transcription factors that induce and pattern organs have been identified, but relatively few of the downstream effectors that execute morphogenesis programs. Because such morphogenesis genes may function in many organs and developmental processes, mutations in them are expected to be pleiotropic and hence ignored or discarded in most standard genetic screens. Here we describe a systematic screen designed to identify all Drosophila third chromosome genes (∼40% of the genome) that function in development of the tracheal system, a tubular respiratory organ that provides a paradigm for branching morphogenesis. To identify potentially pleiotropic morphogenesis genes, the screen included analysis of marked clones of homozygous mutant tracheal cells in heterozygous animals, plus a secondary screen to exclude mutations in general "house-keeping" genes. From a collection including more than 5,000 lethal mutations, we identified 133 mutations representing ∼70 or more genes that subdivide the tracheal terminal branching program into six genetically separable steps, a previously established cell specification step plus five major morphogenesis and maturation steps: branching, growth, tubulogenesis, gas-filling, and maintenance. Molecular identification of 14 of the 70 genes demonstrates that they include six previously known tracheal genes, each with a novel function revealed by clonal analysis, and two well-known growth suppressors that establish an integral role for cell growth control in branching morphogenesis. The rest are new tracheal genes that function in morphogenesis and maturation, many through cytoskeletal and secretory pathways. The results suggest systematic genetic screens that include clonal analysis can elucidate the full organogenesis program and that over 200 patterning and morphogenesis genes are required to build even a relatively simple organ such as the Drosophila tracheal system.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster , Genes de Insetos , Traqueia/crescimento & desenvolvimento , Fatores de Transcrição/genética , Animais , Animais Geneticamente Modificados , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Genes Letais , Heterozigoto , Homozigoto , Morfogênese , Mutação , Fatores de Transcrição/metabolismo
7.
bioRxiv ; 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38168178

RESUMO

Dravet syndrome (DS) is a devastating developmental epileptic encephalopathy marked by treatment-resistant seizures, developmental delay, intellectual disability, motor deficits, and a 10-20% rate of premature death. Most DS patients harbor loss-of-function mutations in one copy of SCN1A , which has been associated with inhibitory neuron dysfunction. Here we developed an interneuron-targeting AAV human SCN1A gene replacement therapy using cell class-specific enhancers. We generated a split-intein fusion form of SCN1A to circumvent AAV packaging limitations and deliver SCN1A via a dual vector approach using cell class-specific enhancers. These constructs produced full-length Na V 1.1 protein and functional sodium channels in HEK293 cells and in brain cells in vivo . After packaging these vectors into enhancer-AAVs and administering to mice, immunohistochemical analyses showed telencephalic GABAergic interneuron-specific and dose-dependent transgene biodistribution. These vectors conferred strong dose-dependent protection against postnatal mortality and seizures in two DS mouse models carrying independent loss-of-function alleles of Scn1a, at two independent research sites, supporting the robustness of this approach. No mortality or toxicity was observed in wild-type mice injected with single vectors expressing either the N-terminal or C-terminal halves of SCN1A , or the dual vector system targeting interneurons. In contrast, nonselective neuronal targeting of SCN1A conferred less rescue against mortality and presented substantial preweaning lethality. These findings demonstrate proof-of-concept that interneuron-specific AAV-mediated SCN1A gene replacement is sufficient for significant rescue in DS mouse models and suggest it could be an effective therapeutic approach for patients with DS.

8.
bioRxiv ; 2023 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-36711773

RESUMO

Delivering genes to and across the brain vasculature efficiently and specifically across species remains a critical challenge for addressing neurological diseases. We have evolved adeno-associated virus (AAV9) capsids into vectors that transduce brain endothelial cells specifically and efficiently following systemic administration in wild-type mice with diverse genetic backgrounds and rats. These AAVs also exhibit superior transduction of the CNS across non-human primates (marmosets and rhesus macaques), and ex vivo human brain slices although the endothelial tropism is not conserved across species. The capsid modifications translate from AAV9 to other serotypes such as AAV1 and AAV-DJ, enabling serotype switching for sequential AAV administration in mice. We demonstrate that the endothelial specific mouse capsids can be used to genetically engineer the blood-brain barrier by transforming the mouse brain vasculature into a functional biofactory. Vasculature-secreted Hevin (a synaptogenic protein) rescued synaptic deficits in a mouse model.

9.
Nat Nanotechnol ; 18(10): 1241-1251, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37430038

RESUMO

Crossing the blood-brain barrier in primates is a major obstacle for gene delivery to the brain. Adeno-associated viruses (AAVs) promise robust, non-invasive gene delivery from the bloodstream to the brain. However, unlike in rodents, few neurotropic AAVs efficiently cross the blood-brain barrier in non-human primates. Here we report on AAV.CAP-Mac, an engineered variant identified by screening in adult marmosets and newborn macaques, which has improved delivery efficiency in the brains of multiple non-human primate species: marmoset, rhesus macaque and green monkey. CAP-Mac is neuron biased in infant Old World primates, exhibits broad tropism in adult rhesus macaques and is vasculature biased in adult marmosets. We demonstrate applications of a single, intravenous dose of CAP-Mac to deliver functional GCaMP for ex vivo calcium imaging across multiple brain areas, or a cocktail of fluorescent reporters for Brainbow-like labelling throughout the macaque brain, circumventing the need for germline manipulations in Old World primates. As such, CAP-Mac is shown to have potential for non-invasive systemic gene transfer in the brains of non-human primates.


Assuntos
Encéfalo , Callithrix , Humanos , Animais , Recém-Nascido , Chlorocebus aethiops , Macaca mulatta/genética , Callithrix/genética , Encéfalo/fisiologia , Técnicas de Transferência de Genes , Neurônios , Vetores Genéticos/genética
10.
Elife ; 122023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37249212

RESUMO

Rodent studies have demonstrated that synaptic dynamics from excitatory to inhibitory neuron types are often dependent on the target cell type. However, these target cell-specific properties have not been well investigated in human cortex, where there are major technical challenges in reliably obtaining healthy tissue, conducting multiple patch-clamp recordings on inhibitory cell types, and identifying those cell types. Here, we take advantage of newly developed methods for human neurosurgical tissue analysis with multiple patch-clamp recordings, post-hoc fluorescent in situ hybridization (FISH), machine learning-based cell type classification and prospective GABAergic AAV-based labeling to investigate synaptic properties between pyramidal neurons and PVALB- vs. SST-positive interneurons. We find that there are robust molecular differences in synapse-associated genes between these neuron types, and that individual presynaptic pyramidal neurons evoke postsynaptic responses with heterogeneous synaptic dynamics in different postsynaptic cell types. Using molecular identification with FISH and classifiers based on transcriptomically identified PVALB neurons analyzed by Patch-seq, we find that PVALB neurons typically show depressing synaptic characteristics, whereas other interneuron types including SST-positive neurons show facilitating characteristics. Together, these data support the existence of target cell-specific synaptic properties in human cortex that are similar to rodent, thereby indicating evolutionary conservation of local circuit connectivity motifs from excitatory to inhibitory neurons and their synaptic dynamics.


Assuntos
Neocórtex , Humanos , Neocórtex/fisiologia , Transmissão Sináptica/fisiologia , Hibridização in Situ Fluorescente , Estudos Prospectivos , Neurônios/fisiologia , Células Piramidais/fisiologia , Sinapses/fisiologia , Interneurônios/fisiologia
11.
Nat Commun ; 14(1): 3345, 2023 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-37291094

RESUMO

Delivering genes to and across the brain vasculature efficiently and specifically across species remains a critical challenge for addressing neurological diseases. We have evolved adeno-associated virus (AAV9) capsids into vectors that transduce brain endothelial cells specifically and efficiently following systemic administration in wild-type mice with diverse genetic backgrounds, and in rats. These AAVs also exhibit superior transduction of the CNS across non-human primates (marmosets and rhesus macaques), and in ex vivo human brain slices, although the endothelial tropism is not conserved across species. The capsid modifications translate from AAV9 to other serotypes such as AAV1 and AAV-DJ, enabling serotype switching for sequential AAV administration in mice. We demonstrate that the endothelial-specific mouse capsids can be used to genetically engineer the blood-brain barrier by transforming the mouse brain vasculature into a functional biofactory. We apply this approach to Hevin knockout mice, where AAV-X1-mediated ectopic expression of the synaptogenic protein Sparcl1/Hevin in brain endothelial cells rescued synaptic deficits.


Assuntos
Células Endoteliais , Roedores , Camundongos , Ratos , Animais , Células Endoteliais/metabolismo , Roedores/genética , Macaca mulatta/genética , Encéfalo/metabolismo , Tropismo/genética , Camundongos Knockout , Dependovirus/metabolismo , Vetores Genéticos/genética , Transdução Genética , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas da Matriz Extracelular/genética
12.
Science ; 382(6667): eadf2359, 2023 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-37824649

RESUMO

Single-cell transcriptomic studies have identified a conserved set of neocortical cell types from small postmortem cohorts. We extended these efforts by assessing cell type variation across 75 adult individuals undergoing epilepsy and tumor surgeries. Nearly all nuclei map to one of 125 robust cell types identified in the middle temporal gyrus. However, we found interindividual variance in abundances and gene expression signatures, particularly in deep-layer glutamatergic neurons and microglia. A minority of donor variance is explainable by age, sex, ancestry, disease state, and cell state. Genomic variation was associated with expression of 150 to 250 genes for most cell types. This characterization of cellular variation provides a baseline for cell typing in health and disease.


Assuntos
Lobo Temporal , Transcriptoma , Adulto , Humanos , Epilepsia/metabolismo , Perfilação da Expressão Gênica , Neurônios/metabolismo , Lobo Temporal/citologia , Lobo Temporal/metabolismo , Doenças do Sistema Nervoso/genética , Transtornos Mentais/genética
13.
Science ; 382(6667): eadf5357, 2023 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-37824674

RESUMO

Delineating the gene-regulatory programs underlying complex cell types is fundamental for understanding brain function in health and disease. Here, we comprehensively examined human brain cell epigenomes by probing DNA methylation and chromatin conformation at single-cell resolution in 517 thousand cells (399 thousand neurons and 118 thousand non-neurons) from 46 regions of three adult male brains. We identified 188 cell types and characterized their molecular signatures. Integrative analyses revealed concordant changes in DNA methylation, chromatin accessibility, chromatin organization, and gene expression across cell types, cortical areas, and basal ganglia structures. We further developed single-cell methylation barcodes that reliably predict brain cell types using the methylation status of select genomic sites. This multimodal epigenomic brain cell atlas provides new insights into the complexity of cell-type-specific gene regulation in adult human brains.


Assuntos
Encéfalo , Metilação de DNA , Epigênese Genética , Adulto , Humanos , Masculino , Encéfalo/citologia , Encéfalo/metabolismo , Cromatina/metabolismo , Genoma Humano , Análise de Célula Única , Imageamento Tridimensional , Atlas como Assunto
14.
Science ; 382(6667): eadf6812, 2023 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-37824655

RESUMO

Variation in cytoarchitecture is the basis for the histological definition of cortical areas. We used single cell transcriptomics and performed cellular characterization of the human cortex to better understand cortical areal specialization. Single-nucleus RNA-sequencing of 8 areas spanning cortical structural variation showed a highly consistent cellular makeup for 24 cell subclasses. However, proportions of excitatory neuron subclasses varied substantially, likely reflecting differences in connectivity across primary sensorimotor and association cortices. Laminar organization of astrocytes and oligodendrocytes also differed across areas. Primary visual cortex showed characteristic organization with major changes in the excitatory to inhibitory neuron ratio, expansion of layer 4 excitatory neurons, and specialized inhibitory neurons. These results lay the groundwork for a refined cellular and molecular characterization of human cortical cytoarchitecture and areal specialization.


Assuntos
Neocórtex , Humanos , Neocórtex/metabolismo , Neocórtex/ultraestrutura , Neurônios/classificação , Neurônios/metabolismo , Transcriptoma , Análise da Expressão Gênica de Célula Única , Filogenia
15.
Nat Commun ; 13(1): 5628, 2022 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-36163250

RESUMO

After spinal cord injury, tissue distal to the lesion contains undamaged cells that could support or augment recovery. Targeting these cells requires a clearer understanding of their injury responses and capacity for repair. Here, we use single nucleus RNA sequencing to profile how each cell type in the lumbar spinal cord changes after a thoracic injury in mice. We present an atlas of these dynamic responses across dozens of cell types in the acute, subacute, and chronically injured spinal cord. Using this resource, we find rare spinal neurons that express a signature of regeneration in response to injury, including a major population that represent spinocerebellar projection neurons. We characterize these cells anatomically and observed axonal sparing, outgrowth, and remodeling in the spinal cord and cerebellum. Together, this work provides a key resource for studying cellular responses to injury and uncovers the spontaneous plasticity of spinocerebellar neurons, uncovering a potential candidate for targeted therapy.


Assuntos
Traumatismos da Medula Espinal , Animais , Axônios/metabolismo , Cerebelo/metabolismo , Camundongos , Regeneração Nervosa/fisiologia , Neurônios/metabolismo , Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia
16.
Blood ; 113(8): 1670-80, 2009 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-18971422

RESUMO

High levels of aldehyde dehydrogenase (ALDH) activity have been proposed to be a common feature of stem cells. Adult hematopoietic, neural, and cancer stem cells have all been reported to have high ALDH activity, detected using Aldefluor, a fluorogenic substrate for ALDH. This activity has been attributed to Aldh1a1, an enzyme that is expressed at high levels in stem cells and that has been suggested to regulate stem cell function. Nonetheless, Aldh1a1 function in stem cells has never been tested genetically. We observed that Aldh1a1 was preferentially expressed in mouse hematopoietic stem cells (HSCs) and expression increased with age. Hematopoietic cells from Aldh1a1-deficient mice exhibited increased sensitivity to cyclophosphamide in a non-cell-autonomous manner, consistent with its role in cyclophosphamide metabolism in the liver. However, Aldh1a1 deficiency did not affect hematopoiesis, HSC function, or the capacity to reconstitute irradiated recipients in young or old adult mice. Aldh1a1 deficiency also did not affect Aldefluor staining of hematopoietic cells. Finally, Aldh1a1 deficiency did not affect the function of stem cells from the adult central or peripheral nervous systems. Aldh1a1 is not a critical regulator of adult stem cell function or Aldefluor staining in mice.


Assuntos
Aldeído Desidrogenase/metabolismo , Sistema Nervoso Central/citologia , Células-Tronco Hematopoéticas/enzimologia , Sistema Nervoso Periférico/citologia , Células-Tronco/enzimologia , Envelhecimento/fisiologia , Aldeído Desidrogenase/genética , Família Aldeído Desidrogenase 1 , Animais , Células Cultivadas , Ciclofosfamida/farmacologia , Corantes Fluorescentes , Regulação Enzimológica da Expressão Gênica , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Agonistas Mieloablativos/farmacologia , Retinal Desidrogenase , Células-Tronco/citologia
17.
Elife ; 102021 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-34473054

RESUMO

Abundant evidence supports the presence of at least three distinct types of thalamocortical (TC) neurons in the primate dorsal lateral geniculate nucleus (dLGN) of the thalamus, the brain region that conveys visual information from the retina to the primary visual cortex (V1). Different types of TC neurons in mice, humans, and macaques have distinct morphologies, distinct connectivity patterns, and convey different aspects of visual information to the cortex. To investigate the molecular underpinnings of these cell types, and how these relate to differences in dLGN between human, macaque, and mice, we profiled gene expression in single nuclei and cells using RNA-sequencing. These efforts identified four distinct types of TC neurons in the primate dLGN: magnocellular (M) neurons, parvocellular (P) neurons, and two types of koniocellular (K) neurons. Despite extensively documented morphological and physiological differences between M and P neurons, we identified few genes with significant differential expression between transcriptomic cell types corresponding to these two neuronal populations. Likewise, the dominant feature of TC neurons of the adult mouse dLGN is high transcriptomic similarity, with an axis of heterogeneity that aligns with core vs. shell portions of mouse dLGN. Together, these data show that transcriptomic differences between principal cell types in the mature mammalian dLGN are subtle relative to the observed differences in morphology and cortical projection targets. Finally, alignment of transcriptome profiles across species highlights expanded diversity of GABAergic neurons in primate versus mouse dLGN and homologous types of TC neurons in primates that are distinct from TC neurons in mouse.


Assuntos
Núcleo Celular/genética , Corpos Geniculados/metabolismo , Neurônios/metabolismo , Córtex Visual/metabolismo , Animais , Perfilação da Expressão Gênica , Humanos , Macaca , Camundongos , RNA-Seq , Análise de Célula Única , Tálamo/metabolismo , Vias Visuais/metabolismo
18.
Neuron ; 109(9): 1449-1464.e13, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33789083

RESUMO

Rapid cell type identification by new genomic single-cell analysis methods has not been met with efficient experimental access to these cell types. To facilitate access to specific neural populations in mouse cortex, we collected chromatin accessibility data from individual cells and identified enhancers specific for cell subclasses and types. When cloned into recombinant adeno-associated viruses (AAVs) and delivered to the brain, these enhancers drive transgene expression in specific cortical cell subclasses. We extensively characterized several enhancer AAVs to show that they label different projection neuron subclasses as well as a homologous neuron subclass in human cortical slices. We also show how coupling enhancer viruses expressing recombinases to a newly generated transgenic mouse, Ai213, enables strong labeling of three different neuronal classes/subclasses in the brain of a single transgenic animal. This approach combines unprecedented flexibility with specificity for investigation of cell types in the mouse brain and beyond.


Assuntos
Encéfalo/citologia , Neurônios/classificação , Neurônios/citologia , Análise de Célula Única/métodos , Animais , Conjuntos de Dados como Assunto , Dependovirus , Humanos , Camundongos , Camundongos Transgênicos
19.
Cell Rep ; 34(13): 108754, 2021 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-33789096

RESUMO

Viral genetic tools that target specific brain cell types could transform basic neuroscience and targeted gene therapy. Here, we use comparative open chromatin analysis to identify thousands of human-neocortical-subclass-specific putative enhancers from across the genome to control gene expression in adeno-associated virus (AAV) vectors. The cellular specificity of reporter expression from enhancer-AAVs is established by molecular profiling after systemic AAV delivery in mouse. Over 30% of enhancer-AAVs produce specific expression in the targeted subclass, including both excitatory and inhibitory subclasses. We present a collection of Parvalbumin (PVALB) enhancer-AAVs that show highly enriched expression not only in cortical PVALB cells but also in some subcortical PVALB populations. Five vectors maintain PVALB-enriched expression in primate neocortex. These results demonstrate how genome-wide open chromatin data mining and cross-species AAV validation can be used to create the next generation of non-species-restricted viral genetic tools.


Assuntos
Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Neocórtex/metabolismo , Animais , Cromatina/genética , Cromatina/metabolismo , Bases de Dados Genéticas , Dependovirus/genética , Doença/genética , Epigênese Genética , Vetores Genéticos/metabolismo , Genoma , Humanos , Camundongos , Neurônios/metabolismo , Parvalbuminas/metabolismo , Primatas , Especificidade da Espécie
20.
Cell Stem Cell ; 21(3): 289-290, 2017 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-28886360

RESUMO

3D organoids enable in vitro human brain development models, but they have not yet recapitulated some essential features of brain circuit formation. Recently, several studies appearing in Nature, Nature Methods, and Cell Stem Cell generated fused organoid models of inhibitory and excitatory neuron development, which can now achieve functional circuit integration.


Assuntos
Neurogênese , Organoides , Encéfalo , Movimento Celular , Humanos , Interneurônios
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa