Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 180
Filtrar
1.
Cell ; 172(3): 491-499.e15, 2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-29358049

RESUMO

Non-coding "ultraconserved" regions containing hundreds of consecutive bases of perfect sequence conservation across mammalian genomes can function as distant-acting enhancers. However, initial deletion studies in mice revealed that loss of such extraordinarily constrained sequences had no immediate impact on viability. Here, we show that ultraconserved enhancers are required for normal development. Focusing on some of the longest ultraconserved sites genome wide, located near the essential neuronal transcription factor Arx, we used genome editing to create an expanded series of knockout mice lacking individual or combinations of ultraconserved enhancers. Mice with single or pairwise deletions of ultraconserved enhancers were viable and fertile but in nearly all cases showed neurological or growth abnormalities, including substantial alterations of neuron populations and structural brain defects. Our results demonstrate the functional importance of ultraconserved enhancers and indicate that remarkably strong sequence conservation likely results from fitness deficits that appear subtle in a laboratory setting.


Assuntos
Sequência Conservada , Desenvolvimento Embrionário/genética , Elementos Facilitadores Genéticos , Animais , Encéfalo/anormalidades , Encéfalo/embriologia , Encéfalo/metabolismo , Feminino , Deleção de Genes , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Masculino , Camundongos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
Cell ; 155(7): 1521-31, 2013 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-24360275

RESUMO

Enhancers are distal regulatory elements that can activate tissue-specific gene expression and are abundant throughout mammalian genomes. Although substantial progress has been made toward genome-wide annotation of mammalian enhancers, their temporal activity patterns and global contributions in the context of developmental in vivo processes remain poorly explored. Here we used epigenomic profiling for H3K27ac, a mark of active enhancers, coupled to transgenic mouse assays to examine the genome-wide utilization of enhancers in three different mouse tissues across seven developmental stages. The majority of the ∼90,000 enhancers identified exhibited tightly temporally restricted predicted activity windows and were associated with stage-specific biological functions and regulatory pathways in individual tissues. Comparative genomic analysis revealed that evolutionary conservation of enhancers decreases following midgestation across all tissues examined. The dynamic enhancer activities uncovered in this study illuminate rapid and pervasive temporal in vivo changes in enhancer usage that underlie processes central to development and disease.


Assuntos
Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Estudo de Associação Genômica Ampla , Acetilação , Animais , Epigênese Genética , Evolução Molecular , Histonas/metabolismo , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos
3.
Cell ; 152(4): 895-908, 2013 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-23375746

RESUMO

The mammalian telencephalon plays critical roles in cognition, motor function, and emotion. Though many of the genes required for its development have been identified, the distant-acting regulatory sequences orchestrating their in vivo expression are mostly unknown. Here, we describe a digital atlas of in vivo enhancers active in subregions of the developing telencephalon. We identified more than 4,600 candidate embryonic forebrain enhancers and studied the in vivo activity of 329 of these sequences in transgenic mouse embryos. We generated serial sets of histological brain sections for 145 reproducible forebrain enhancers, resulting in a publicly accessible web-based data collection comprising more than 32,000 sections. We also used epigenomic analysis of human and mouse cortex tissue to directly compare the genome-wide enhancer architecture in these species. These data provide a primary resource for investigating gene regulatory mechanisms of telencephalon development and enable studies of the role of distant-acting enhancers in neurodevelopmental disorders.


Assuntos
Elementos Facilitadores Genéticos , Telencéfalo/metabolismo , Animais , Embrião de Mamíferos/metabolismo , Feto/metabolismo , Estudo de Associação Genômica Ampla , Humanos , Camundongos , Telencéfalo/embriologia , Transcriptoma , Fatores de Transcrição de p300-CBP/metabolismo
4.
Development ; 149(11)2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35695185

RESUMO

In the developing subpallium, the fate decision between neurons and glia is driven by expression of Dlx1/2 or Olig1/2, respectively, two sets of transcription factors with a mutually repressive relationship. The mechanism by which Dlx1/2 repress progenitor and oligodendrocyte fate, while promoting transcription of genes needed for differentiation, is not fully understood. We identified a motif within DLX1 that binds RBBP4, a NuRD complex subunit. ChIP-seq studies of genomic occupancy of DLX1 and six different members of the NuRD complex show that DLX1 and NuRD colocalize to putative regulatory elements enriched near other transcription factor genes. Loss of Dlx1/2 leads to dysregulation of genome accessibility at putative regulatory elements near genes repressed by Dlx1/2, including Olig2. Consequently, heterozygosity of Dlx1/2 and Rbbp4 leads to an increase in the production of OLIG2+ cells. These findings highlight the importance of the interplay between transcription factors and chromatin remodelers in regulating cell-fate decisions.


Assuntos
Proteínas de Homeodomínio , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase , Diferenciação Celular/genética , Genes Homeobox , Proteínas de Homeodomínio/metabolismo , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
Proc Natl Acad Sci U S A ; 119(15): e2108760119, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35377797

RESUMO

Enhancers integrate transcription factor signaling pathways that drive cell fate specification in the developing brain. We paired enhancer labeling and single-cell RNA-sequencing (scRNA-seq) to delineate and distinguish specification of neuronal lineages in mouse medial, lateral, and caudal ganglionic eminences (MGE, LGE, and CGE) at embryonic day (E)11.5. We show that scRNA-seq clustering using transcription factors improves resolution of regional and developmental populations, and that enhancer activities identify specific and overlapping GE-derived neuronal populations. First, we mapped the activities of seven evolutionarily conserved brain enhancers at single-cell resolution in vivo, finding that the selected enhancers had diverse activities in specific progenitor and neuronal populations across the GEs. We then applied enhancer-based labeling, scRNA-seq, and analysis of in situ hybridization data to distinguish transcriptionally distinct and spatially defined subtypes of MGE-derived GABAergic and cholinergic projection neurons and interneurons. Our results map developmental origins and specification paths underlying neurogenesis in the embryonic basal ganglia and showcase the power of scRNA-seq combined with enhancer-based labeling to resolve the complex paths of neuronal specification underlying mouse brain development.


Assuntos
Gânglios da Base , Neurônios Colinérgicos , Elementos Facilitadores Genéticos , Neurônios GABAérgicos , Neurogênese , Animais , Gânglios da Base/citologia , Gânglios da Base/embriologia , Linhagem da Célula/genética , Neurônios Colinérgicos/metabolismo , Neurônios GABAérgicos/metabolismo , Camundongos , Neurogênese/genética , RNA-Seq , Análise de Célula Única , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
6.
Dev Biol ; 503: 10-24, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37532091

RESUMO

The external globus pallidus (GPe) is an essential component of the basal ganglia, a group of subcortical nuclei that are involved in control of action. Changes in the firing of GPe neurons are associated with both passive and active body movements. Aberrant activity of GPe neurons has been linked to motor symptoms of a variety of movement disorders, such as Parkinson's Disease, Huntington's disease and dystonia. Recent studies have helped delineate functionally distinct subtypes of GABAergic GPe projection neurons. However, not much is known about specific molecular mechanisms underlying the development of GPe neuronal subtypes. We show that the transcriptional regulator Lmo3 is required for the development of medial ganglionic eminence derived Nkx2.1+ and PV+ GPe neurons, but not lateral ganglionic eminence derived FoxP2+ neurons. As a consequence of the reduction in PV+ neurons, Lmo3-null mice have a reduced GPe input to the subthalamic nucleus.


Assuntos
Neurônios GABAérgicos , Globo Pálido , Proteínas com Domínio LIM , Movimento , Animais , Camundongos , Neurônios GABAérgicos/metabolismo , Globo Pálido/metabolismo , Camundongos Knockout , Movimento/fisiologia , Transtornos dos Movimentos/genética , Transtornos dos Movimentos/metabolismo , Transtornos dos Movimentos/fisiopatologia , Proteínas com Domínio LIM/genética , Proteínas com Domínio LIM/metabolismo
7.
Cell ; 135(3): 396-400, 2008 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-18984148

RESUMO

Genetic studies are refining our understanding of neurodevelopmental mechanisms in autism. Some autism-related mutations appear to disrupt genes regulated by neuronal activity, which are especially important in development of the postnatal nervous system. Gene replacement studies in mice indicate that the developmental window to ameliorate symptoms may be wider than previously anticipated.


Assuntos
Transtorno Autístico/fisiopatologia , Encéfalo/crescimento & desenvolvimento , Animais , Transtorno Autístico/genética , Encéfalo/fisiologia , Feminino , Humanos , Masculino , Sinapses/fisiologia
8.
Development ; 144(15): 2837-2851, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28694260

RESUMO

Distinct cortical interneuron (CIN) subtypes have unique circuit functions; dysfunction in specific subtypes is implicated in neuropsychiatric disorders. Somatostatin- and parvalbumin-expressing (SST+ and PV+) interneurons are the two major subtypes generated by medial ganglionic eminence (MGE) progenitors. Spatial and temporal mechanisms governing their cell-fate specification and differential integration into cortical layers are largely unknown. We provide evidence that Coup-TF1 and Coup-TF2 (Nr2f1 and Nr2f2) transcription factor expression in an arc-shaped progenitor domain within the MGE promotes time-dependent survival of this neuroepithelium and the time-dependent specification of layer V SST+ CINs. Coup-TF1 and Coup-TF2 autonomously repress PV+ fate in MGE progenitors, in part through directly driving Sox6 expression. These results have identified, in mouse, a transcriptional pathway that controls SST-PV fate.


Assuntos
Fator II de Transcrição COUP/metabolismo , Fator I de Transcrição COUP/metabolismo , Interneurônios/metabolismo , Neocórtex/citologia , Animais , Fator I de Transcrição COUP/genética , Fator II de Transcrição COUP/genética , Células Cultivadas , Imunoprecipitação da Cromatina , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Imuno-Histoquímica , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Parvalbuminas/genética , Parvalbuminas/metabolismo , Fatores de Transcrição SOXD/genética , Fatores de Transcrição SOXD/metabolismo , Somatostatina/genética , Somatostatina/metabolismo
9.
Mol Psychiatry ; 24(9): 1248-1257, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31089192

RESUMO

In 2003 Rubenstein and Merzenich hypothesized that some forms of Autism (ASD) might be caused by a reduction in signal-to-noise in key neural circuits, which could be the result of changes in excitatory-inhibitory (E-I) balance. Here, we have clarified the concept of E-I balance, and updated the original hypothesis in light of the field's increasingly sophisticated understanding of neuronal circuits. We discuss how specific developmental mechanisms, which reduce inhibition, affect cortical and hippocampal functions. After describing how mutations of some ASD genes disrupt inhibition in mice, we close by suggesting that E-I balance represents an organizing framework for understanding findings related to pathophysiology and for identifying appropriate treatments.


Assuntos
Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/fisiopatologia , Rede Nervosa/fisiologia , Animais , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Hipocampo/fisiopatologia , Humanos , Inibição Psicológica , Transtornos Mentais/fisiopatologia , Camundongos , Neurônios/fisiologia
10.
Genome Res ; 26(8): 1013-22, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27325115

RESUMO

Exome sequencing studies have identified multiple genes harboring de novo loss-of-function (LoF) variants in individuals with autism spectrum disorders (ASD), including TBR1, a master regulator of cortical development. We performed ChIP-seq for TBR1 during mouse cortical neurogenesis and show that TBR1-bound regions are enriched adjacent to ASD genes. ASD genes were also enriched among genes that are differentially expressed in Tbr1 knockouts, which together with the ChIP-seq data, suggests direct transcriptional regulation. Of the nine ASD genes examined, seven were misexpressed in the cortices of Tbr1 knockout mice, including six with increased expression in the deep cortical layers. ASD genes with adjacent cortical TBR1 ChIP-seq peaks also showed unusually low levels of LoF mutations in a reference human population and among Icelanders. We then leveraged TBR1 binding to identify an appealing subset of candidate ASD genes. Our findings highlight a TBR1-regulated network of ASD genes in the developing neocortex that are relatively intolerant to LoF mutations, indicating that these genes may play critical roles in normal cortical development.


Assuntos
Transtorno do Espectro Autista/genética , Proteínas de Ligação a DNA/genética , Neocórtex/fisiopatologia , Neurogênese/genética , Animais , Transtorno do Espectro Autista/fisiopatologia , Modelos Animais de Doenças , Exoma/genética , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Camundongos , Mutação , Neocórtex/crescimento & desenvolvimento , Neurônios/metabolismo , Neurônios/patologia , Fatores de Risco , Proteínas com Domínio T
11.
Cereb Cortex ; 28(11): 3868-3879, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-29028946

RESUMO

Human mutations in CNTNAP2 are associated with an array of neuropsychiatric and neurological syndromes, including speech and language disorders, epilepsy, and autism spectrum disorder (ASD). We examined Cntnap2's expression and function in GABAergic cortical interneurons (CINs), where its RNA is present at highest levels in chandelier neurons, PV+ neurons and VIP+ neurons. In vivo functions were studied using both constitutive Cntnap2 null mice and a transplantation assay, the latter to assess cell autonomous phenotypes of medial ganglionic eminence (MGE)-derived CINs. We found that Cntnap2 constitutive null mutants had normal numbers of MGE-derived CINs, but had reduced PV+ CINs. Transplantation assays showed that Cntnap2 cell autonomously regulated the physiology of parvalbumin (PV)+, fast-spiking CINs; no phenotypes were observed in somatostatin+, regular spiking, CINs. We also tested the effects of 4 human CNTNAP2 ASD missense mutations in vivo, and found that they impaired PV+ CIN development. Together, these data reveal that reduced CNTNAP2 function impairs PV+ CINs, a cell type with important roles in regulating cortical circuits.


Assuntos
Neurônios GABAérgicos/fisiologia , Interneurônios/fisiologia , Proteínas de Membrana/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Alelos , Animais , Transtorno do Espectro Autista , Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Humanos , Masculino , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação de Sentido Incorreto , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Parvalbuminas/metabolismo , Proteína Reelina , Serina Endopeptidases/metabolismo , Córtex Somatossensorial/fisiologia , Telencéfalo/crescimento & desenvolvimento
12.
Cereb Cortex ; 28(11): 3797-3815, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-29028947

RESUMO

The postnatal functions of the Dlx1&2 transcription factors in cortical interneurons (CINs) are unknown. Here, using conditional Dlx1, Dlx2, and Dlx1&2 knockouts (CKOs), we defined their roles in specific CINs. The CKOs had dendritic, synaptic, and survival defects, affecting even PV+ CINs. We provide evidence that DLX2 directly drives Gad1, Gad2, and Vgat expression, and show that mutants had reduced mIPSC amplitude. In addition, the mutants formed fewer GABAergic synapses on excitatory neurons and had reduced mIPSC frequency. Furthermore, Dlx1/2 CKO had hypoplastic dendrites, fewer excitatory synapses, and reduced excitatory input. We provide evidence that some of these phenotypes were due to reduced expression of GRIN2B (a subunit of the NMDA receptor), a high confidence Autism gene. Thus, Dlx1&2 coordinate key components of CIN postnatal development by promoting their excitability, inhibitory output, and survival.


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Neurônios GABAérgicos/fisiologia , Proteínas de Homeodomínio/fisiologia , Interneurônios/fisiologia , Sinapses/fisiologia , Fatores de Transcrição/fisiologia , Ácido gama-Aminobutírico/biossíntese , Animais , Córtex Cerebral/citologia , Feminino , Neurônios GABAérgicos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Glutamato Descarboxilase/metabolismo , Proteínas de Homeodomínio/genética , Interneurônios/citologia , Masculino , Camundongos Knockout , Potenciais Pós-Sinápticos em Miniatura , Fatores de Transcrição/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
13.
Development ; 142(21): 3746-57, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26534986

RESUMO

Transcription factors act during cortical development as master regulatory genes that specify cortical arealization and cellular identities. Although numerous transcription factors have been identified as being crucial for cortical development, little is known about their downstream targets and how they mediate the emergence of specific neuronal connections via selective axon guidance. The EMX transcription factors are essential for early patterning of the cerebral cortex, but whether EMX1 mediates interhemispheric connectivity by controlling corpus callosum formation remains unclear. Here, we demonstrate that in mice on the C57Bl/6 background EMX1 plays an essential role in the midline crossing of an axonal subpopulation of the corpus callosum derived from the anterior cingulate cortex. In the absence of EMX1, cingulate axons display reduced expression of the axon guidance receptor NRP1 and form aberrant axonal bundles within the rostral corpus callosum. EMX1 also functions as a transcriptional activator of Nrp1 expression in vitro, and overexpression of this protein in Emx1 knockout mice rescues the midline-crossing phenotype. These findings reveal a novel role for the EMX1 transcription factor in establishing cortical connectivity by regulating the interhemispheric wiring of a subpopulation of neurons within the mouse anterior cingulate cortex.


Assuntos
Giro do Cíngulo/metabolismo , Proteínas de Homeodomínio/metabolismo , Neuropilina-1/metabolismo , Fatores de Transcrição/metabolismo , Agenesia do Corpo Caloso/embriologia , Agenesia do Corpo Caloso/genética , Animais , Axônios/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Semaforinas/metabolismo
14.
Nat Rev Neurosci ; 14(3): 202-16, 2013 03.
Artigo em Inglês | MEDLINE | ID: mdl-23385869

RESUMO

A systematic classification and accepted nomenclature of neuron types is much needed but is currently lacking. This article describes a possible taxonomical solution for classifying GABAergic interneurons of the cerebral cortex based on a novel, web-based interactive system that allows experts to classify neurons with pre-determined criteria. Using Bayesian analysis and clustering algorithms on the resulting data, we investigated the suitability of several anatomical terms and neuron names for cortical GABAergic interneurons. Moreover, we show that supervised classification models could automatically categorize interneurons in agreement with experts' assignments. These results demonstrate a practical and objective approach to the naming, characterization and classification of neurons based on community consensus.


Assuntos
Algoritmos , Córtex Cerebral/citologia , Interneurônios/classificação , Interneurônios/citologia , Terminologia como Assunto , Ácido gama-Aminobutírico/metabolismo , Animais , Teorema de Bayes , Córtex Cerebral/metabolismo , Análise por Conglomerados , Humanos , Interneurônios/metabolismo
15.
Cereb Cortex ; 27(9): 4303-4313, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27497284

RESUMO

Prenatally, the cytokine CXCL12 regulates cortical interneuron migration, whereas its postnatal functions are poorly understood. Here, we report that CXCL12 is expressed postnatally in layer V pyramidal neurons and localizes on their cell bodies in the medial prefrontal cortex (mPFC), while its receptors CXCR4/CXCR7 localize to the axon terminals of parvalbumin (PV) interneurons. Conditionally eliminating CXCL12 in neonatal layer V pyramidal neurons led to decreased axon targeting and reduced inhibitory perisomatic synapses from PV+ basket interneurons onto layer V pyramidal neurons. Consequently, the mPFC of Cxcl12 conditional mutants displayed attenuated inhibitory postsynaptic currents onto layer V pyramidal neurons. Thus, postnatal CXCL12 signaling promotes a specific interneuron circuit that inhibits mPFC activity.


Assuntos
Quimiocina CXCL12/metabolismo , Interneurônios/metabolismo , Córtex Pré-Frontal/metabolismo , Sinapses/fisiologia , Animais , Axônios/metabolismo , Quimiocina CXCL12/genética , Potenciais Pós-Sinápticos Inibidores/fisiologia , Camundongos Transgênicos , Parvalbuminas/metabolismo , Células Piramidais/fisiologia , Receptores CXCR4/metabolismo , Ácido gama-Aminobutírico/metabolismo
16.
Proc Natl Acad Sci U S A ; 111(1): 492-7, 2014 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-24344303

RESUMO

Chronic changes in excitability and activity can induce homeostatic plasticity. These perturbations may be associated with neurological disorders, particularly those involving loss or dysfunction of GABA interneurons. In distal-less homeobox 1 (Dlx1(-/-)) mice with late-onset interneuron loss and reduced inhibition, we observed both excitatory synaptic silencing and decreased intrinsic neuronal excitability. These homeostatic changes do not fully restore normal circuit function, because synaptic silencing results in enhanced potential for long-term potentiation and abnormal gamma oscillations. Transplanting medial ganglionic eminence interneuron progenitors to introduce new GABAergic interneurons, we demonstrate restoration of hippocampal function. Specifically, miniature excitatory postsynaptic currents, input resistance, hippocampal long-term potentiation, and gamma oscillations are all normalized. Thus, in vivo homeostatic plasticity is a highly dynamic and bidirectional process that responds to changes in inhibition.


Assuntos
Proteínas de Homeodomínio/genética , Interneurônios/patologia , Células-Tronco Neurais/transplante , Plasticidade Neuronal , Fatores de Transcrição/genética , Animais , Morte Celular , Transplante de Células , Eletrofisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Neurônios GABAérgicos/metabolismo , Inativação Gênica , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/metabolismo , Homeostase , Imuno-Histoquímica , Interneurônios/metabolismo , Potenciação de Longa Duração , Masculino , Camundongos , Neurônios/metabolismo , Oscilometria , Sinapses/fisiologia , Transmissão Sináptica/fisiologia , Ácido gama-Aminobutírico/metabolismo
17.
Nature ; 468(7321): 263-9, 2010 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-21068835

RESUMO

Mutations in the X-linked MECP2 gene, which encodes the transcriptional regulator methyl-CpG-binding protein 2 (MeCP2), cause Rett syndrome and several neurodevelopmental disorders including cognitive disorders, autism, juvenile-onset schizophrenia and encephalopathy with early lethality. Rett syndrome is characterized by apparently normal early development followed by regression, motor abnormalities, seizures and features of autism, especially stereotyped behaviours. The mechanisms mediating these features are poorly understood. Here we show that mice lacking Mecp2 from GABA (γ-aminobutyric acid)-releasing neurons recapitulate numerous Rett syndrome and autistic features, including repetitive behaviours. Loss of MeCP2 from a subset of forebrain GABAergic neurons also recapitulates many features of Rett syndrome. MeCP2-deficient GABAergic neurons show reduced inhibitory quantal size, consistent with a presynaptic reduction in glutamic acid decarboxylase 1 (Gad1) and glutamic acid decarboxylase 2 (Gad2) levels, and GABA immunoreactivity. These data demonstrate that MeCP2 is critical for normal function of GABA-releasing neurons and that subtle dysfunction of GABAergic neurons contributes to numerous neuropsychiatric phenotypes.


Assuntos
Transtorno Autístico/fisiopatologia , Proteína 2 de Ligação a Metil-CpG/deficiência , Proteína 2 de Ligação a Metil-CpG/metabolismo , Síndrome de Rett/fisiopatologia , Transdução de Sinais , Transtorno de Movimento Estereotipado/fisiopatologia , Ácido gama-Aminobutírico/metabolismo , Animais , Transtorno Autístico/complicações , Transtorno Autístico/genética , Transtorno Autístico/patologia , Encéfalo/citologia , Comportamento Compulsivo/complicações , Comportamento Compulsivo/genética , Comportamento Compulsivo/fisiopatologia , Modelos Animais de Doenças , Eletroencefalografia , Genótipo , Glutamato Descarboxilase/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Proteínas de Homeodomínio/genética , Potenciais Pós-Sinápticos Inibidores , Potenciação de Longa Duração , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Camundongos Transgênicos , Inibição Neural , Plasticidade Neuronal , Neurônios/metabolismo , Fenótipo , Terminações Pré-Sinápticas/metabolismo , Transtornos Psicomotores/complicações , Transtornos Psicomotores/genética , Transtornos Psicomotores/fisiopatologia , Reflexo de Sobressalto/genética , Respiração , Síndrome de Rett/complicações , Síndrome de Rett/genética , Síndrome de Rett/patologia , Comportamento Autodestrutivo/complicações , Comportamento Autodestrutivo/genética , Comportamento Autodestrutivo/fisiopatologia , Transtorno de Movimento Estereotipado/complicações , Transtorno de Movimento Estereotipado/genética , Transtorno de Movimento Estereotipado/patologia , Taxa de Sobrevida , Transmissão Sináptica , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética
18.
J Neurosci ; 34(29): 9506-15, 2014 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-25031394

RESUMO

Excitatory and inhibitory balance of neuronal network activity is essential for normal brain function and may be of particular importance to memory. Apolipoprotein (apo) E4 and amyloid-ß (Aß) peptides, two major players in Alzheimer's disease (AD), cause inhibitory interneuron impairments and aberrant neuronal activity in the hippocampal dentate gyrus in AD-related mouse models and humans, leading to learning and memory deficits. To determine whether replacing the lost or impaired interneurons rescues neuronal signaling and behavioral deficits, we transplanted embryonic interneuron progenitors into the hippocampal hilus of aged apoE4 knock-in mice without or with Aß accumulation. In both conditions, the transplanted cells developed into mature interneurons, functionally integrated into the hippocampal circuitry, and restored normal learning and memory. Thus, restricted hilar transplantation of inhibitory interneurons restores normal cognitive function in two widely used AD-related mouse models, highlighting the importance of interneuron impairments in AD pathogenesis and the potential of cell replacement therapy for AD. More broadly, it demonstrates that excitatory and inhibitory balance are crucial for learning and memory, and suggests an avenue for investigating the processes of learning and memory and their alterations in healthy aging and diseases.


Assuntos
Doença de Alzheimer , Apolipoproteína E4/genética , Hipocampo/patologia , Interneurônios/fisiologia , Aprendizagem/fisiologia , Memória/fisiologia , Células-Tronco Neurais/transplante , Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Doença de Alzheimer/cirurgia , Precursor de Proteína beta-Amiloide/genética , Animais , Modelos Animais de Doenças , Feminino , Humanos , Técnicas In Vitro , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Proteínas do Tecido Nervoso/metabolismo
19.
Dev Biol ; 385(1): 94-106, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24157949

RESUMO

The progenitor zones of the embryonic mouse ventral telencephalon give rise to GABAergic and cholinergic neurons. We have shown previously that two LIM-homeodomain (LIM-HD) transcription factors, Lhx6 and Lhx8, that are downstream of Nkx2.1, are critical for the development of telencephalic GABAergic and cholinergic neurons. Here we investigate the role of Ldb1, a nuclear protein that binds directly to all LIM-HD factors, in the development of these ventral telencephalon derived neurons. We show that Ldb1 is expressed in the Nkx2.1 cell lineage during embryonic development and in mature neurons. Conditional deletion of Ldb1 causes defects in the expression of a series of genes in the ventral telencephalon and severe impairment in the tangential migration of cortical interneurons from the ventral telencephalon. Similar to the phenotypes observed in Lhx6 or Lhx8 mutant mice, the Ldb1 conditional mutants show a reduction in the number of both GABAergic and cholinergic neurons in the telencephalon. Furthermore, our analysis reveals defects in the development of the parvalbumin-positive neurons in the globus pallidus and striatum of the Ldb1 mutants. These results provide evidence that Ldb1 plays an essential role as a transcription co-regulator of Lhx6 and Lhx8 in the control of mammalian telencephalon development.


Assuntos
Neurônios Colinérgicos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Neurônios GABAérgicos/metabolismo , Proteínas com Domínio LIM/metabolismo , Proteínas Nucleares/metabolismo , Telencéfalo/embriologia , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Proteínas de Ligação a DNA/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Globo Pálido/embriologia , Proteínas Hedgehog/biossíntese , Proteínas Hedgehog/metabolismo , Proteínas com Domínio LIM/genética , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Telencéfalo/crescimento & desenvolvimento , Fator Nuclear 1 de Tireoide
20.
Development ; 139(9): 1630-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22492355

RESUMO

The development of the progenitor zones in the pallium, lateral ganglionic eminence (LGE) and medial ganglionic eminence (MGE) in the subpallium has been well studied; however, so far the role of the caudal ganglionic eminence (CGE), a posterior subpallial domain, in telencephalon patterning remains poorly understood. COUP-TFII, an orphan nuclear receptor, is preferentially expressed in the CGE. We generated COUP-TFII mouse mutants, using Rx-Cre (RxCre;COUP-TFII(F/F)), to study its function in telencephalon development. In these mutants, we found severe defects in the formation of the amygdala complex, including the lateral (LA), basolateral (BLA) and basomedial (BMA) amygdala nuclei. Molecular analysis provided evidence that the migration of CGE-derived Pax6(+) cells failed to settle into the BMA nucleus, owing to reduced expression of neuropilin 1 (Nrp1) and Nrp2, two semaphorin receptors that regulate neuronal cell migration and axon guidance. Our ChIP assays revealed that Nrp1 and Nrp2 genes are the direct targets of COUP-TFII in the telencephalon in vivo. Furthermore, our results showed that the coordinated development between the CGE originated subpallial population (Pax6(+) cells) and pallial populations (Tbr1(+) and Lhx2(+) cells) was essential for patterning the amygdala assembly. Our study presented novel genetic evidence that the caudal ganglionic eminence, a distinct subpallial progenitor zone, contributes cells to the basal telencephalon, such as the BMA nucleus.


Assuntos
Tonsila do Cerebelo/embriologia , Fator II de Transcrição COUP/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neuropilina-1/metabolismo , Neuropilina-2/metabolismo , Animais , Fator II de Transcrição COUP/genética , Imunoprecipitação da Cromatina , Primers do DNA/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Imuno-Histoquímica , Camundongos , Camundongos Mutantes , Reação em Cadeia da Polimerase em Tempo Real
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA