Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Cell ; 174(4): 1015-1030.e16, 2018 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-30096299

RESUMO

The mammalian brain is composed of diverse, specialized cell populations. To systematically ascertain and learn from these cellular specializations, we used Drop-seq to profile RNA expression in 690,000 individual cells sampled from 9 regions of the adult mouse brain. We identified 565 transcriptionally distinct groups of cells using computational approaches developed to distinguish biological from technical signals. Cross-region analysis of these 565 cell populations revealed features of brain organization, including a gene-expression module for synthesizing axonal and presynaptic components, patterns in the co-deployment of voltage-gated ion channels, functional distinctions among the cells of the vasculature and specialization of glutamatergic neurons across cortical regions. Systematic neuronal classifications for two complex basal ganglia nuclei and the striatum revealed a rare population of spiny projection neurons. This adult mouse brain cell atlas, accessible through interactive online software (DropViz), serves as a reference for development, disease, and evolution.


Assuntos
Encéfalo/metabolismo , Linhagem da Célula , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Análise de Célula Única/métodos , Transcriptoma , Animais , Encéfalo/crescimento & desenvolvimento , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Masculino , Camundongos , Camundongos Endogâmicos C57BL
2.
Immunity ; 50(1): 253-271.e6, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30471926

RESUMO

Microglia, the resident immune cells of the brain, rapidly change states in response to their environment, but we lack molecular and functional signatures of different microglial populations. Here, we analyzed the RNA expression patterns of more than 76,000 individual microglia in mice during development, in old age, and after brain injury. Our analysis uncovered at least nine transcriptionally distinct microglial states, which expressed unique sets of genes and were localized in the brain using specific markers. The greatest microglial heterogeneity was found at young ages; however, several states-including chemokine-enriched inflammatory microglia-persisted throughout the lifespan or increased in the aged brain. Multiple reactive microglial subtypes were also found following demyelinating injury in mice, at least one of which was also found in human multiple sclerosis lesions. These distinct microglia signatures can be used to better understand microglia function and to identify and manipulate specific subpopulations in health and disease.


Assuntos
Envelhecimento/imunologia , Lesões Encefálicas/imunologia , Encéfalo/fisiologia , Microglia/fisiologia , Esclerose Múltipla/imunologia , Adaptação Fisiológica , Envelhecimento/genética , Animais , Lesões Encefálicas/genética , Diferenciação Celular , Doenças Desmielinizantes , Humanos , Longevidade , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência de RNA , Análise de Célula Única
3.
Nature ; 586(7828): 262-269, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32999462

RESUMO

Primates and rodents, which descended from a common ancestor around 90 million years ago1, exhibit profound differences in behaviour and cognitive capacity; the cellular basis for these differences is unknown. Here we use single-nucleus RNA sequencing to profile RNA expression in 188,776 individual interneurons across homologous brain regions from three primates (human, macaque and marmoset), a rodent (mouse) and a weasel (ferret). Homologous interneuron types-which were readily identified by their RNA-expression patterns-varied in abundance and RNA expression among ferrets, mice and primates, but varied less among primates. Only a modest fraction of the genes identified as 'markers' of specific interneuron subtypes in any one species had this property in another species. In the primate neocortex, dozens of genes showed spatial expression gradients among interneurons of the same type, which suggests that regional variation in cortical contexts shapes the RNA expression patterns of adult neocortical interneurons. We found that an interneuron type that was previously associated with the mouse hippocampus-the 'ivy cell', which has neurogliaform characteristics-has become abundant across the neocortex of humans, macaques and marmosets but not mice or ferrets. We also found a notable subcortical innovation: an abundant striatal interneuron type in primates that had no molecularly homologous counterpart in mice or ferrets. These interneurons expressed a unique combination of genes that encode transcription factors, receptors and neuropeptides and constituted around 30% of striatal interneurons in marmosets and humans.


Assuntos
Interneurônios/citologia , Primatas , Animais , Callithrix , Córtex Cerebral/citologia , Feminino , Furões , Hipocampo/citologia , Humanos , Interneurônios/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Proteínas de Membrana Lisossomal/metabolismo , Macaca , Masculino , Camundongos , Neostriado/citologia , Proteínas do Tecido Nervoso/metabolismo , RNA/genética , Especificidade da Espécie , Fatores de Transcrição/metabolismo
5.
Nature ; 521(7550): 85-9, 2015 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-25739505

RESUMO

The basal ganglia are phylogenetically conserved subcortical nuclei necessary for coordinated motor action and reward learning. Current models postulate that the basal ganglia modulate cerebral cortex indirectly via an inhibitory output to thalamus, bidirectionally controlled by direct- and indirect-pathway striatal projection neurons (dSPNs and iSPNs, respectively). The basal ganglia thalamic output sculpts cortical activity by interacting with signals from sensory and motor systems. Here we describe a direct projection from the globus pallidus externus (GP), a central nucleus of the basal ganglia, to frontal regions of the cerebral cortex (FC). Two cell types make up the GP-FC projection, distinguished by their electrophysiological properties, cortical projections and expression of choline acetyltransferase (ChAT), a synthetic enzyme for the neurotransmitter acetylcholine (ACh). Despite these differences, ChAT(+) cells, which have been historically identified as an extension of the nucleus basalis, as well as ChAT(-) cells, release the inhibitory neurotransmitter GABA (γ-aminobutyric acid) and are inhibited by iSPNs and dSPNs of dorsal striatum. Thus, GP-FC cells comprise a direct GABAergic/cholinergic projection under the control of striatum that activates frontal cortex in vivo. Furthermore, iSPN inhibition of GP-FC cells is sensitive to dopamine 2 receptor signalling, revealing a pathway by which drugs that target dopamine receptors for the treatment of neuropsychiatric disorders can act in the basal ganglia to modulate frontal cortices.


Assuntos
Lobo Frontal/metabolismo , Globo Pálido/metabolismo , Ácido gama-Aminobutírico/metabolismo , Acetilcolina/metabolismo , Animais , Antipsicóticos/farmacologia , Núcleo Basal de Meynert/citologia , Núcleo Basal de Meynert/metabolismo , Colina O-Acetiltransferase/metabolismo , Fenômenos Eletrofisiológicos , Feminino , Lobo Frontal/citologia , Lobo Frontal/efeitos dos fármacos , Globo Pálido/citologia , Globo Pálido/efeitos dos fármacos , Globo Pálido/enzimologia , Macaca mulatta , Masculino , Camundongos , Vias Neurais , Receptores de Dopamina D2/metabolismo , Transdução de Sinais
6.
Nature ; 485(7400): 646-50, 2012 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-22660328

RESUMO

Neural activity during development critically shapes postnatal wiring of the mammalian brain. This is best illustrated by the sensory systems, in which the patterned feed-forward excitation provided by sensory organs and experience drives the formation of mature topographic circuits capable of extracting specific features of sensory stimuli. In contrast, little is known about the role of early activity in the development of the basal ganglia, a phylogenetically ancient group of nuclei fundamentally important for complex motor action and reward-based learning. These nuclei lack direct sensory input and are only loosely topographically organized, forming interlocking feed-forward and feed-back inhibitory circuits without laminar structure. Here we use transgenic mice and viral gene transfer methods to modulate neurotransmitter release and neuronal activity in vivo in the developing striatum. We find that the balance of activity between the two inhibitory and antagonist pathways in the striatum regulates excitatory innervation of the basal ganglia during development. These effects indicate that the propagation of activity through a multi-stage network regulates the wiring of the basal ganglia, revealing an important role of positive feedback in driving network maturation.


Assuntos
Gânglios da Base/embriologia , Gânglios da Base/fisiologia , Neostriado/embriologia , Neostriado/fisiologia , Vias Neurais/fisiologia , Sinapses/metabolismo , Animais , Gânglios da Base/citologia , Córtex Cerebral/citologia , Córtex Cerebral/fisiologia , Retroalimentação Fisiológica , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Modelos Neurológicos , Neostriado/citologia , Inibição Neural , Tálamo/citologia , Tálamo/fisiologia , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/deficiência , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo , Ácido gama-Aminobutírico/metabolismo
7.
Mol Biol Evol ; 32(10): 2694-706, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26104011

RESUMO

The fundamental asymmetry of female meiosis creates an arena for genetic elements to compete for inclusion in the egg, promoting the selfish evolution of centromere variants that maximize their transmission to the future egg. Such "female meiotic drive" has been hypothesized to explain the paradoxically complex and rapidly evolving nature of centromeric DNA and proteins. Although theoretically widespread, few cases of active drive have been observed, thereby limiting the opportunities to directly assess the impact of centromeric drive on molecular variation at centromeres and binding proteins. Here, we characterize the molecular evolutionary patterns of CENH3, the centromere-defining histone variant, in Mimulus monkeyflowers, a genus with one of the few known cases of active centromere-associated female meiotic drive. First, we identify a novel duplication of CENH3 in diploid Mimulus, including in lineages with actively driving centromeres. Second, we demonstrate long-term adaptive evolution at several sites in the N-terminus of CENH3, a region with some meiosis-specific functions that putatively interacts with centromeric DNA. Finally, we infer that the paralogs evolve under different selective regimes; some sites in the N-terminus evolve under positive selection in the pro-orthologs or only one paralog (CENH3_B) and the paralogs exhibit significantly different patterns of polymorphism within populations. Our finding of long-term, adaptive evolution at CENH3 in the context of centromere-associated meiotic drive supports an antagonistic, coevolutionary battle for evolutionary dominance between centromeric DNA and binding proteins.


Assuntos
Adaptação Fisiológica/genética , Evolução Biológica , Centrômero/metabolismo , Duplicação Gênica , Meiose , Mimulus/genética , Proteínas de Plantas/genética , Códon/genética , Regulação da Expressão Gênica de Plantas , Mimulus/citologia , Filogenia , Proteínas de Plantas/química , Polimorfismo Genético , Seleção Genética
8.
Proc Natl Acad Sci U S A ; 108(37): 15414-9, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21825165

RESUMO

To understand how the nervous system processes information, a map of the connections among neurons would be of great benefit. Here we describe the use of vesicular stomatitis virus (VSV) for tracing neuronal connections in vivo. We made VSV vectors that used glycoprotein (G) genes from several other viruses. The G protein from lymphocytic choriomeningitis virus endowed VSV with the ability to spread transsynaptically, specifically in an anterograde direction, whereas the rabies virus glycoprotein gave a specifically retrograde transsynaptic pattern. The use of an avian G protein fusion allowed specific targeting of cells expressing an avian receptor, which allowed a demonstration of monosynaptic anterograde tracing from defined cells. Synaptic connectivity of pairs of virally labeled cells was demonstrated by using slice cultures and electrophysiology. In vivo infections of several areas in the mouse brain led to the predicted patterns of spread for anterograde or retrograde tracers.


Assuntos
Sistema Nervoso Central/citologia , Vetores Genéticos/genética , Neurônios/metabolismo , Coloração e Rotulagem , Sinapses/metabolismo , Vesiculovirus/genética , Animais , Encéfalo/virologia , Olho/virologia , Vírus da Coriomeningite Linfocítica , Camundongos , Condutos Olfatórios/metabolismo , Condutos Olfatórios/virologia , Recombinação Genética/genética
9.
Nat Commun ; 13(1): 6993, 2022 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-36384944

RESUMO

Brain function depends on synaptic connections between specific neuron types, yet systematic descriptions of synaptic networks and their molecular properties are not readily available. Here, we introduce SBARRO (Synaptic Barcode Analysis by Retrograde Rabies ReadOut), a method that uses single-cell RNA sequencing to reveal directional, monosynaptic relationships based on the paths of a barcoded rabies virus from its "starter" postsynaptic cell to that cell's presynaptic partners. Thousands of these partner relationships can be ascertained in a single experiment, alongside genome-wide RNAs. We use SBARRO to describe synaptic networks formed by diverse mouse brain cell types in vitro, finding that different cell types have presynaptic networks with differences in average size and cell type composition. Patterns of RNA expression suggest that functioning synapses are critical for rabies virus uptake. By tracking individual rabies clones across cells, SBARRO offers new opportunities to map the synaptic organization of neural circuits.


Assuntos
Vírus da Raiva , Raiva , Camundongos , Animais , Vírus da Raiva/genética , Sinapses/fisiologia , Neurônios/fisiologia , RNA
10.
Neuron ; 109(16): 2590-2603.e13, 2021 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-34197733

RESUMO

Neuropsychiatric disorders are often accompanied by cognitive impairments/intellectual disability (ID). It is not clear whether there are converging mechanisms underlying these debilitating impairments. We found that many autism and schizophrenia risk genes are expressed in the anterodorsal subdivision (AD) of anterior thalamic nuclei, which has reciprocal connectivity with learning and memory structures. CRISPR-Cas9 knockdown of multiple risk genes selectively in AD thalamus led to memory deficits. While the AD is necessary for contextual memory encoding, the neighboring anteroventral subdivision (AV) regulates memory specificity. These distinct functions of AD and AV are mediated through their projections to retrosplenial cortex, using differential mechanisms. Furthermore, knockdown of autism and schizophrenia risk genes PTCHD1, YWHAG, or HERC1 from AD led to neuronal hyperexcitability, and normalization of hyperexcitability rescued memory deficits in these models. This study identifies converging cellular to circuit mechanisms underlying cognitive deficits in a subset of neuropsychiatric disease models.


Assuntos
Núcleos Anteriores do Tálamo/fisiopatologia , Disfunção Cognitiva/fisiopatologia , Vias Neurais/fisiopatologia , Núcleos Talâmicos/fisiopatologia , Animais , Núcleos Anteriores do Tálamo/fisiologia , Córtex Cerebral/fisiopatologia , Cognição/fisiologia , Camundongos , Vias Neurais/fisiologia , Núcleos Talâmicos/fisiologia
11.
Elife ; 92020 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-32613945

RESUMO

The mouse cerebral cortex contains neurons that express choline acetyltransferase (ChAT) and are a potential local source of acetylcholine. However, the neurotransmitters released by cortical ChAT+ neurons and their synaptic connectivity are unknown. We show that the nearly all cortical ChAT+ neurons in mice are specialized VIP+ interneurons that release GABA strongly onto other inhibitory interneurons and acetylcholine sparsely onto layer 1 interneurons and other VIP+/ChAT+ interneurons. This differential transmission of ACh and GABA based on the postsynaptic target neuron is reflected in VIP+/ChAT+ interneuron pre-synaptic terminals, as quantitative molecular analysis shows that only a subset of these are specialized to release acetylcholine. In addition, we identify a separate, sparse population of non-VIP ChAT+ neurons in the medial prefrontal cortex with a distinct developmental origin that robustly release acetylcholine in layer 1. These results demonstrate both cortex-region heterogeneity in cortical ChAT+ interneurons and target-specific co-release of acetylcholine and GABA.


Assuntos
Acetilcolina/metabolismo , Encéfalo/metabolismo , Colina O-Acetiltransferase/metabolismo , Neurônios/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Córtex Cerebral/metabolismo , Heterozigoto , Interneurônios/metabolismo , Camundongos , Córtex Pré-Frontal/metabolismo , Terminações Pré-Sinápticas/metabolismo
12.
Nat Genet ; 50(4): 621-629, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29632380

RESUMO

We introduce an approach to identify disease-relevant tissues and cell types by analyzing gene expression data together with genome-wide association study (GWAS) summary statistics. Our approach uses stratified linkage disequilibrium (LD) score regression to test whether disease heritability is enriched in regions surrounding genes with the highest specific expression in a given tissue. We applied our approach to gene expression data from several sources together with GWAS summary statistics for 48 diseases and traits (average N = 169,331) and found significant tissue-specific enrichments (false discovery rate (FDR) < 5%) for 34 traits. In our analysis of multiple tissues, we detected a broad range of enrichments that recapitulated known biology. In our brain-specific analysis, significant enrichments included an enrichment of inhibitory over excitatory neurons for bipolar disorder, and excitatory over inhibitory neurons for schizophrenia and body mass index. Our results demonstrate that our polygenic approach is a powerful way to leverage gene expression data for interpreting GWAS signals.


Assuntos
Expressão Gênica , Predisposição Genética para Doença , Transtorno Bipolar/genética , Índice de Massa Corporal , Encéfalo/metabolismo , Cromatina/genética , Epigênese Genética , Perfilação da Expressão Gênica/estatística & dados numéricos , Estudo de Associação Genômica Ampla/estatística & dados numéricos , Humanos , Doenças do Sistema Imunitário/genética , Desequilíbrio de Ligação , Modelos Genéticos , Herança Multifatorial , Neurônios/metabolismo , Esquizofrenia/genética , Distribuição Tecidual/genética
13.
Neuron ; 94(1): 138-152.e5, 2017 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-28384468

RESUMO

The basal ganglia (BG) integrate inputs from diverse sensorimotor, limbic, and associative regions to guide action-selection and goal-directed behaviors. The entopeduncular nucleus (EP) is a major BG output nucleus and has been suggested to channel signals from distinct BG nuclei to target regions involved in diverse functions. Here we use single-cell transcriptional and molecular analyses to demonstrate that the EP contains at least three classes of projection neurons-glutamate/GABA co-releasing somatostatin neurons, glutamatergic parvalbumin neurons, and GABAergic parvalbumin neurons. These classes comprise functionally and anatomically distinct output pathways that differentially affect EP target regions, such as the lateral habenula (LHb) and thalamus. Furthermore, LHb- and thalamic-projecting EP neurons are differentially innervated by subclasses of striatal and pallidal neurons. Therefore, we identify previously unknown subdivisions within the EP and reveal the existence of cascading, molecularly distinct projections through striatum and globus pallidus to EP targets within epithalamus and thalamus.


Assuntos
Gânglios da Base/metabolismo , Núcleo Entopeduncular/metabolismo , Neurônios GABAérgicos/metabolismo , Animais , Gânglios da Base/citologia , Núcleo Entopeduncular/citologia , Neurônios GABAérgicos/citologia , Perfilação da Expressão Gênica , Globo Pálido/citologia , Ácido Glutâmico/metabolismo , Habenula/citologia , Humanos , Hibridização in Situ Fluorescente , Sistema Límbico , Camundongos , Neostriado/citologia , Neurônios/citologia , Neurônios/metabolismo , Parvalbuminas/metabolismo , Córtex Sensório-Motor , Análise de Célula Única , Somatostatina/metabolismo , Tálamo/citologia
14.
PLoS One ; 11(2): e0149798, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26905595

RESUMO

The globus pallidus externus (GP) is a nucleus of the basal ganglia (BG), containing GABAergic projection neurons that arborize widely throughout the BG, thalamus and cortex. Ongoing work seeks to map axonal projection patterns from GP cell types, as defined by their electrophysiological and molecular properties. Here we use transgenic mice and recombinant viruses to characterize parvalbumin expressing (PV+) GP neurons within the BG circuit. We confirm that PV+ neurons 1) make up ~40% of the GP neurons 2) exhibit fast-firing spontaneous activity and 3) provide the major axonal arborization to the STN and substantia nigra reticulata/compacta (SNr/c). PV+ neurons also innervate the striatum. Retrograde labeling identifies ~17% of pallidostriatal neurons as PV+, at least a subset of which also innervate the STN and SNr. Optogenetic experiments in acute brain slices demonstrate that the PV+ pallidostriatal axons make potent inhibitory synapses on low threshold spiking (LTS) and fast-spiking interneurons (FS) in the striatum, but rarely on spiny projection neurons (SPNs). Thus PV+ GP neurons are synaptically positioned to directly coordinate activity between BG input nuclei, the striatum and STN, and thalamic-output from the SNr.


Assuntos
Axônios/metabolismo , Globo Pálido/metabolismo , Parvalbuminas/biossíntese , Núcleo Subtalâmico/metabolismo , Sinapses/metabolismo , Animais , Mapeamento Encefálico , Globo Pálido/citologia , Camundongos , Camundongos Transgênicos , Parvalbuminas/genética , Sinapses/genética
15.
Neuropharmacology ; 100: 40-6, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26220313

RESUMO

Neurons that produce acetylcholine (ACh) are positioned to broadly influence the brain, with axonal arborizations that extend throughout the cerebral cortex, striatum, and hippocampus. While the action of these neurons has typically been attributed entirely to ACh, neurons often release more than one primary neurotransmitter. Here, we review evidence for the cotransmission of the inhibitory neurotransmitter GABA from cholinergic neurons throughout the mammalian central nervous system. Functional cotransmission of ACh and GABA has been reported in the retina and cortex, and anatomical studies suggest that GABA cotransmission is a common feature of nearly all forebrain ACh-producing neurons. Further experiments are necessary to confirm the extent of GABA cotransmission from cholinergic neurons, and the contribution of GABA needs to be considered when studying the functional impact of activity in ACh-producing neurons. This article is part of the Special Issue entitled 'Synaptopathy--from Biology to Therapy'.


Assuntos
Acetilcolina/metabolismo , Encéfalo/metabolismo , Neurônios Colinérgicos/metabolismo , Transmissão Sináptica , Ácido gama-Aminobutírico/metabolismo , Animais , Humanos , Camundongos , Optogenética
16.
Curr Protoc Neurosci ; 72: 1.24.1-1.24.15, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26131660

RESUMO

Recombinant adeno-associated viruses (rAAVs) transcriptionally activated by Cre recombinase (Cre-On) are powerful tools for determining the anatomy and function of genetically defined neuronal types in transgenic Cre driver mice. Here we describe how rAAVs transcriptionally inactivated by Cre (Cre-Off) can be used in conjunction with Cre-On rAAVs or genomic Cre-reporter alleles to study brain circuits. Intracranial injection of Cre-On/Cre-Off rAAVs into spatially intermingled Cre(+) and Cre(-) neurons allows these populations to be differentially labeled or manipulated within individual animals. This comparison helps define the unique properties of Cre(+) neurons, highlighting the specialized role they play in their constituent brain circuits. This protocol touches on the conceptual and experimental background of Cre-Off rAAV systems, including caveats and methods of validation.


Assuntos
Dependovirus/genética , Vetores Genéticos/metabolismo , Integrases/metabolismo , Neurônios/metabolismo , Animais , Vetores Genéticos/genética , Integrases/genética , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Transgênicos , Transdução Genética
17.
Elife ; 42015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25723967

RESUMO

Neurotransmitter corelease is emerging as a common theme of central neuromodulatory systems. Though corelease of glutamate or GABA with acetylcholine has been reported within the cholinergic system, the full extent is unknown. To explore synaptic signaling of cholinergic forebrain neurons, we activated choline acetyltransferase expressing neurons using channelrhodopsin while recording post-synaptic currents (PSCs) in layer 1 interneurons. Surprisingly, we observed PSCs mediated by GABAA receptors in addition to nicotinic acetylcholine receptors. Based on PSC latency and pharmacological sensitivity, our results suggest monosynaptic release of both GABA and ACh. Anatomical analysis showed that forebrain cholinergic neurons express the GABA synthetic enzyme Gad2 and the vesicular GABA transporter (Slc32a1). We confirmed the direct release of GABA by knocking out Slc32a1 from cholinergic neurons. Our results identify GABA as an overlooked fast neurotransmitter utilized throughout the forebrain cholinergic system. GABA/ACh corelease may have major implications for modulation of cortical function by cholinergic neurons.


Assuntos
Acetilcolina/metabolismo , Neurônios Colinérgicos/metabolismo , Prosencéfalo/metabolismo , Ácido gama-Aminobutírico/metabolismo , 4-Aminopiridina/farmacologia , Animais , Colina O-Acetiltransferase/genética , Colina O-Acetiltransferase/metabolismo , Neurônios Colinérgicos/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Camundongos Transgênicos , Microscopia Confocal , Microscopia de Fluorescência por Excitação Multifotônica , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Prosencéfalo/citologia , Prosencéfalo/fisiologia , Receptores de GABA-A/metabolismo , Receptores Nicotínicos/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Tetrodotoxina/farmacologia , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
18.
Elife ; 42015 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-26551563

RESUMO

Dopamine is released in the striatum during development and impacts the activity of Protein Kinase A (PKA) in striatal spiny projection neurons (SPNs). We examined whether dopaminergic neuromodulation regulates activity-dependent glutamatergic synapse formation in the developing striatum. Systemic in vivo treatment with Gαs-coupled G-protein receptors (GPCRs) agonists enhanced excitatory synapses on direct pathway striatal spiny projection neurons (dSPNs), whereas rapid production of excitatory synapses on indirect pathway neurons (iSPNs) required the activation of Gαs GPCRs in SPNs of both pathways. Nevertheless, in vitro Gαs activation was sufficient to enhance spinogenesis induced by glutamate photolysis in both dSPNs and iSPNs, suggesting that iSPNs in intact neural circuits have additional requirements for rapid synaptic development. We evaluated the in vivo effects of enhanced glutamate release from corticostriatal axons and postsynaptic PKA and discovered a mechanism of developmental plasticity wherein rapid synaptogenesis is promoted by the coordinated actions of glutamate and postsynaptic Gαs-coupled receptors.


Assuntos
Dopaminérgicos/administração & dosagem , Plasticidade Neuronal/efeitos dos fármacos , Córtex Visual/fisiologia , Animais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Camundongos Endogâmicos C57BL , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Glutamato/metabolismo
19.
Artigo em Inglês | MEDLINE | ID: mdl-23403489

RESUMO

Defining the connections among neurons is critical to our understanding of the structure and function of the nervous system. Recombinant viruses engineered to transmit across synapses provide a powerful approach for the dissection of neuronal circuitry in vivo. We recently demonstrated that recombinant vesicular stomatitis virus (VSV) can be endowed with anterograde or retrograde transsynaptic tracing ability by providing the virus with different glycoproteins. Here we extend the characterization of the transmission and gene expression of recombinant VSV (rVSV) with the rabies virus glycoprotein (RABV-G), and provide examples of its activity relative to the anterograde transsynaptic tracer form of rVSV. rVSV with RABV-G was found to drive strong expression of transgenes and to spread rapidly from neuron to neuron in only a retrograde manner. Depending upon how the RABV-G was delivered, VSV served as a polysynaptic or monosynaptic tracer, or was able to define projections through axonal uptake and retrograde transport. In animals co-infected with rVSV in its anterograde form, rVSV with RABV-G could be used to begin to characterize the similarities and differences in connections to different areas. rVSV with RABV-G provides a flexible, rapid, and versatile tracing tool that complements the previously described VSV-based anterograde transsynaptic tracer.


Assuntos
Glicoproteínas de Membrana/fisiologia , Neurônios/fisiologia , Vírus da Raiva/fisiologia , Sinapses/fisiologia , Proteínas do Envelope Viral/fisiologia , Animais , Animais Recém-Nascidos , Células HEK293 , Humanos , Camundongos , Neurônios/virologia , Técnicas de Cultura de Órgãos , Transporte Proteico/fisiologia , Ratos , Ratos Sprague-Dawley , Sinapses/química , Sinapses/virologia
20.
Artigo em Inglês | MEDLINE | ID: mdl-22866029

RESUMO

Understanding the organization of the nervous system requires methods for dissecting the contributions of each component cell type to circuit function. One widely used approach combines genetic targeting of Cre recombinase to specific cell populations with infection of recombinant adeno-associated viruses (rAAVs) whose transgene expression is activated by Cre ("Cre-On"). Distinguishing how the Cre-expressing neurons differ functionally from neighboring Cre-negative neurons requires rAAVs that are inactivated by Cre ("Cre-Off") and can be used in tandem with Cre-On viruses. Here we introduce two rAAV vectors that are inactivated by Cre and carry different fluorophore and optogenetic constructs. We demonstrate single and dual rAAV systems to achieve Cre-On and Cre-Off expression in spatially-intermingled cell populations of the striatum. Using these systems, we uncovered cryptic genomic interactions that occur between multiple Cre-sensitive rAAVs or between Cre-sensitive rAAVs and somatic Cre-conditional alleles and devised methods to avoid these interactions. Our data highlight both important experimental caveats associated with Cre-dependent rAAV use as well as opportunities for the development of improved rAAVs for gene delivery.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA