Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 43(6): 114329, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38850535

RESUMO

Many autism spectrum disorder (ASD)-associated genes act as transcriptional regulators (TRs). Chromatin immunoprecipitation sequencing (ChIP-seq) was used to identify the regulatory targets of ARID1B, BCL11A, FOXP1, TBR1, and TCF7L2, ASD-associated TRs in the developing human and mouse cortex. These TRs shared substantial overlap in the binding sites, especially within open chromatin. The overlap within a promoter region, 1-2,000 bp upstream of the transcription start site, was highly predictive of brain-expressed genes. This signature was observed in 96 out of 102 ASD-associated genes. In vitro CRISPRi against ARID1B and TBR1 delineated downstream convergent biology in mouse cortical cultures. After 8 days, NeuN+ and CALB+ cells were decreased, GFAP+ cells were increased, and transcriptomic signatures correlated with the postmortem brain samples from individuals with ASD. We suggest that functional convergence across five ASD-associated TRs leads to shared neurodevelopmental outcomes of haploinsufficient disruption.

2.
bioRxiv ; 2023 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-37425940

RESUMO

Transcription factors (TFs) bind combinatorially to genomic cis-regulatory elements (cREs), orchestrating transcription programs. While studies of chromatin state and chromosomal interactions have revealed dynamic neurodevelopmental cRE landscapes, parallel understanding of the underlying TF binding lags. To elucidate the combinatorial TF-cRE interactions driving mouse basal ganglia development, we integrated ChIP-seq for twelve TFs, H3K4me3-associated enhancer-promoter interactions, chromatin and transcriptional state, and transgenic enhancer assays. We identified TF-cREs modules with distinct chromatin features and enhancer activity that have complementary roles driving GABAergic neurogenesis and suppressing other developmental fates. While the majority of distal cREs were bound by one or two TFs, a small proportion were extensively bound, and these enhancers also exhibited exceptional evolutionary conservation, motif density, and complex chromosomal interactions. Our results provide new insights into how modules of combinatorial TF-cRE interactions activate and repress developmental expression programs and demonstrate the value of TF binding data in modeling gene regulatory wiring.

3.
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
4.
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
5.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34921112

RESUMO

We uncovered a transcription factor (TF) network that regulates cortical regional patterning in radial glial stem cells. Screening the expression of hundreds of TFs in the developing mouse cortex identified 38 TFs that are expressed in gradients in the ventricular zone (VZ). We tested whether their cortical expression was altered in mutant mice with known patterning defects (Emx2, Nr2f1, and Pax6), which enabled us to define a cortical regionalization TF network (CRTFN). To identify genomic programming underlying this network, we performed TF ChIP-seq and chromatin-looping conformation to identify enhancer-gene interactions. To map enhancers involved in regional patterning of cortical progenitors, we performed assays for epigenomic marks and DNA accessibility in VZ cells purified from wild-type and patterning mutant mice. This integrated approach has identified a CRTFN and VZ enhancers involved in cortical regional patterning in the mouse.


Assuntos
Córtex Cerebral/embriologia , Redes Reguladoras de Genes , Elementos Reguladores de Transcrição , Fatores de Transcrição/metabolismo , Animais , Fator I de Transcrição COUP/metabolismo , Córtex Cerebral/metabolismo , Epigenoma , Proteínas de Homeodomínio/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Fator de Transcrição PAX6/metabolismo , Fator de Transcrição 1 de Leucemia de Células Pré-B/metabolismo , Fatores de Transcrição/genética
6.
Cell Rep ; 37(10): 110089, 2021 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-34879283

RESUMO

Deleterious genetic variants in POGZ, which encodes the chromatin regulator Pogo Transposable Element with ZNF Domain protein, are strongly associated with autism spectrum disorder (ASD). Although it is a high-confidence ASD risk gene, the neurodevelopmental functions of POGZ remain unclear. Here we reveal the genomic binding of POGZ in the developing forebrain at euchromatic loci and gene regulatory elements (REs). We profile chromatin accessibility and gene expression in Pogz-/- mice and show that POGZ promotes the active chromatin state and transcription of clustered synaptic genes. We further demonstrate that POGZ forms a nuclear complex and co-occupies loci with ADNP, another high-confidence ASD risk gene, and provide evidence that POGZ regulates other neurodevelopmental disorder risk genes as well. Our results reveal a neurodevelopmental function of an ASD risk gene and identify molecular targets that may elucidate its function in ASD.


Assuntos
Transtorno Autístico/enzimologia , Encéfalo/enzimologia , Proteínas de Ciclo Celular/fisiologia , Montagem e Desmontagem da Cromatina , Proteínas de Ligação a DNA/fisiologia , Eucromatina/metabolismo , Sinapses/enzimologia , Transposases/metabolismo , Animais , Transtorno Autístico/genética , Transtorno Autístico/fisiopatologia , Sítios de Ligação , Encéfalo/crescimento & desenvolvimento , Proteínas de Ciclo Celular/genética , Elementos de DNA Transponíveis , Proteínas de Ligação a DNA/genética , Elementos Facilitadores Genéticos , Eucromatina/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Predisposição Genética para Doença , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurogênese , Regiões Promotoras Genéticas , Sinapses/genética , Transposases/genética
7.
Nat Genet ; 53(4): 521-528, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33782603

RESUMO

Ultraconserved enhancer sequences show perfect conservation between human and rodent genomes, suggesting that their functions are highly sensitive to mutation. However, current models of enhancer function do not sufficiently explain this extreme evolutionary constraint. We subjected 23 ultraconserved enhancers to different levels of mutagenesis, collectively introducing 1,547 mutations, and examined their activities in transgenic mouse reporter assays. Overall, we find that the regulatory properties of ultraconserved enhancers are robust to mutation. Upon mutagenesis, nearly all (19/23, 83%) still functioned as enhancers at one developmental stage, as did most of those tested again later in development (5/9, 56%). Replacement of endogenous enhancers with mutated alleles in mice corroborated results of transgenic assays, including the functional resilience of ultraconserved enhancers to mutation. Our findings show that the currently known activities of ultraconserved enhancers do not necessarily require the perfect conservation observed in evolution and suggest that additional regulatory or other functions contribute to their sequence constraint.


Assuntos
Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Mutação , Fatores de Transcrição/genética , Alelos , Animais , Sequência de Bases , Sequência Conservada , Embrião de Mamíferos , Humanos , Camundongos , Mutagênese Sítio-Dirigida , Ratos , Fatores de Transcrição/metabolismo
8.
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
9.
J Comp Neurol ; 524(3): 609-29, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26304102

RESUMO

The development of the cortex is an elaborate process that integrates a plethora of finely tuned molecular processes ranging from carefully regulated gradients of transcription factors, dynamic changes in the chromatin landscape, or formation of protein complexes to elicit and regulate transcription. Combined with cellular processes such as cell type specification, proliferation, differentiation, and migration, all of these developmental processes result in the establishment of an adult mammalian cortex with its typical lamination and regional patterning. By examining in-depth the role of one transcription factor, Pax6, on the regulation of cortical development, its integration in the regulation of chromatin state, and its regulation by cis-regulatory elements, we aim to demonstrate the importance of integrating each level of regulation in our understanding of cortical development.


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Epigênese Genética , Proteínas do Olho/metabolismo , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Repressoras/metabolismo , Animais , Evolução Biológica , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina/fisiologia , Humanos , Fator de Transcrição PAX6
10.
Neuron ; 88(6): 1192-1207, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26671461

RESUMO

We demonstrate using conditional mutagenesis that Pbx1, with and without Pbx2(+/-) sensitization, regulates regional identity and laminar patterning of the developing mouse neocortex in cortical progenitors (Emx1-Cre) and in newly generated neurons (Nex1-Cre). Pbx1/2 mutants have three salient molecular phenotypes of cortical regional and laminar organization: hypoplasia of the frontal cortex, ventral expansion of the dorsomedial cortex, and ventral expansion of Reelin expression in the cortical plate of the frontal cortex, concomitant with an inversion of cortical layering in the rostral cortex. Molecular analyses, including PBX ChIP-seq, provide evidence that PBX promotes frontal cortex identity by repressing genes that promote dorsocaudal fate.


Assuntos
Córtex Cerebral/embriologia , Córtex Cerebral/crescimento & desenvolvimento , Proteínas de Homeodomínio/fisiologia , Células-Tronco Neurais/fisiologia , Neurônios/fisiologia , Fatores de Transcrição/fisiologia , Animais , Animais Recém-Nascidos , Camundongos , Camundongos Transgênicos , Mitose/fisiologia , Fator de Transcrição 1 de Leucemia de Células Pré-B , Proteína Reelina , Células-Tronco/fisiologia
11.
Nat Commun ; 6: 6887, 2015 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-25904499

RESUMO

Guidepost cells present at and surrounding the midline provide guidance cues that orient the growing axons through commissures. Here we show that the transcription factor Nkx2.1 known to control the specification of GABAergic interneurons also regulates the differentiation of astroglia and polydendrocytes within the mouse anterior commissure (AC). Nkx2.1-positive glia were found to originate from three germinal regions of the ventral telencephalon. Nkx2.1-derived glia were observed in and around the AC region by E14.5. Thereafter, a selective cell ablation strategy showed a synergistic role of Nkx2.1-derived cells, both GABAergic interneurons and astroglia, towards the proper formation of the AC. Finally, our results reveal that the Nkx2.1-regulated cells mediate AC axon guidance through the expression of the repellent cue, Slit2. These results bring forth interesting insights about the spatial and temporal origin of midline telencephalic glia, and highlight the importance of neurons and astroglia towards the formation of midline commissures.


Assuntos
Comissura Anterior/embriologia , Astrócitos/metabolismo , Neurônios GABAérgicos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Interneurônios/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Comissura Anterior/citologia , Comissura Anterior/metabolismo , Astrócitos/citologia , Axônios , Movimento Celular , Eletroporação , Embrião de Mamíferos , Neurônios GABAérgicos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Técnicas In Vitro , Interneurônios/citologia , Camundongos , Neuroglia/citologia , Neuroglia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Telencéfalo/citologia , Telencéfalo/embriologia , Telencéfalo/metabolismo , Fator Nuclear 1 de Tireoide
12.
Adv Neurobiol ; 8: 133-64, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25300136

RESUMO

Roundabout receptors (Robo) and their Slit ligands were discovered in the 1990s and found to be key players in axon guidance. Slit was initially described s an extracellular matrix protein that was expressed by midline glia in Drosophila. A few years later, it was shown that, in vertebrates and invertebrates, Slits acted as chemorepellents for axons crossing the midline. Robo proteins were originally discovered in Drosophila in a mutant screen for genes involved in the regulation of midline crossing. This ligand-receptor pair has since been implicated in a variety of other neuronal and non-neuronal processes ranging from cell migration to angiogenesis, tumourigenesis and even organogenesis of tissues such as kidneys, lungs and breasts.


Assuntos
Movimento Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Receptores Imunológicos/metabolismo , Animais , Axônios/fisiologia , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Modelos Biológicos , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Receptores Imunológicos/química , Receptores Imunológicos/genética , Transdução de Sinais/fisiologia , Proteínas Roundabout
13.
J Neurosci ; 34(16): 5717-31, 2014 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-24741061

RESUMO

The elaborate cytoarchitecture of the mammalian neocortex requires the timely production of its constituent pyramidal neurons and interneurons and their disposition in appropriate layers. Numerous chemotropic factors present in the forebrain throughout cortical development play important roles in the orchestration of these events. The Roundabout (Robo) family of receptors and their ligands, the Slit proteins, are expressed in the developing forebrain, and are known to play important roles in the generation and migration of cortical interneurons. However, few studies have investigated their function(s) in the development of pyramidal cells. Here, we observed expression of Robo1 and Slit genes (Slit1, Slit2) in cells lining the telencephalic ventricles, and found significant increases in progenitor cells (basal and apical) at embryonic day (E)12.5 and E14.5 in the developing cortex of Robo1(-/-), Slit1(-/-), and Slit1(-/-)/Slit2(-/-), but not in mice lacking the other Robo or Slit genes. Using layer-specific markers, we found that both early- and late-born pyramidal neuron populations were significantly increased in the cortices of Robo1(-/-) mice at the end of corticogenesis (E18.5). The excess number of cortical pyramidal neurons generated prenatally appears to die in early postnatal life. The observed increase in pyramidal neurons was due to prolonged proliferative activity of their progenitors and not due to changes in cell cycle events. This finding, confirmed by in utero electroporation with Robo1 short hairpin RNA (shRNA) or control constructs into progenitors along the ventricular zone as well as in dissociated cortical cell cultures, points to a novel role for Robo1 in regulating the proliferation and generation of pyramidal neurons.


Assuntos
Proliferação de Células , Regulação da Expressão Gênica no Desenvolvimento/genética , Neocórtex , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/genética , Neurônios/fisiologia , Receptores Imunológicos/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Ventrículos Cerebrais/citologia , Ventrículos Cerebrais/embriologia , Ventrículos Cerebrais/crescimento & desenvolvimento , Embrião de Mamíferos , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neocórtex/citologia , Neocórtex/embriologia , Neocórtex/crescimento & desenvolvimento , Proteínas do Tecido Nervoso/genética , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores Imunológicos/genética , Proteínas Roundabout
14.
Schizophr Res ; 150(1): 252-7, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23911257

RESUMO

Astrocyte dysregulation has been implicated in the pathophysiology of schizophrenia (SCZ) and bipolar disorder (BPD), however the exact nature of astrocytic alterations remains to be identified. In this study we investigated whether levels of four astrocyte-specific proteins; glial fibrillary acidic protein (GFAP), aldehyde dehydrogenase 1L1 (ALDH1L1), vimentin and excitatory amino acid transporter 1 (EAAT1) are altered in SCZ and BPD. Relative concentrations of GFAP, ALDH1L1, vimentin and EAAT1 were assessed post-mortem in dorsolateral prefrontal cortex in SCZ (n=35), BPD (n=34) and non-psychiatric controls (n=35) by western blotting. The same proteins were also quantified in cingulate cortex of rats administered the antipsychotics haloperidol and clozapine. Elevated levels of GFAP were observed in SCZ and BPD, when compared to controls. GFAP was also significantly increased when comparing individuals with psychotic symptoms against those without. Vimentin, ALDH1L1 and EAAT1 levels did not differ between groups. Rats exposed to antipsychotics did not exhibit significant differences in any astrocytic protein, suggesting that increased GFAP in SCZ is not attributable to antipsychotic treatment. Our findings indicate that astrocyte pathology may be associated with psychotic symptoms. Lack of ALDH1L1 and vimentin variability and increased GFAP levels may imply that astrocyte numbers are unchanged but astrocytes are partially activated, or may indicate a specific dysregulation of GFAP.


Assuntos
Regulação da Expressão Gênica/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Córtex Pré-Frontal/metabolismo , Transtornos Psicóticos/patologia , Adulto , Aldeído Desidrogenase/metabolismo , Análise de Variância , Animais , Antipsicóticos/farmacologia , Transtorno Bipolar/patologia , Distribuição de Qui-Quadrado , Transportador 1 de Aminoácido Excitatório/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Peso Molecular , Oxirredutases atuantes sobre Doadores de Grupo CH-NH , Ratos , Ratos Sprague-Dawley , Vimentina/metabolismo
15.
Circ Res ; 112(3): 465-75, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23255421

RESUMO

RATIONALE: The Slit-Roundabout (Robo) signaling pathway has pleiotropic functions during Drosophila heart development. However, its role in mammalian heart development is largely unknown. OBJECTIVE: To analyze the role of Slit-Robo signaling in the formation of the pericardium and the systemic venous return in the murine heart. METHODS AND RESULTS: Expression of genes encoding Robo1 and Robo2 receptors and their ligands Slit2 and Slit3 was found in or around the systemic venous return and pericardium during development. Analysis of embryos lacking Robo1 revealed partial absence of the pericardium, whereas Robo1/2 double mutants additionally showed severely reduced sinus horn myocardium, hypoplastic caval veins, and a persistent left inferior caval vein. Mice lacking Slit3 recapitulated the defects in the myocardialization, alignment, and morphology of the caval veins. Ligand binding assays confirmed Slit3 as the preferred ligand for the Robo1 receptor, whereas Slit2 showed preference for Robo2. Sinus node development was mostly unaffected in all mutants. In addition, we show absence of cross-regulation with previously identified regulators Tbx18 and Wt1. We provide evidence that pericardial defects are created by abnormal localization of the caval veins combined with ectopic pericardial cavity formation. Local increase in neural crest cell death and impaired neural crest adhesive and migratory properties underlie the ectopic pericardium formation. CONCLUSIONS: A novel Slit-Robo signaling pathway is involved in the development of the pericardium, the sinus horn myocardium, and the alignment of the caval veins. Reduced Slit3 binding in the absence of Robo1, causing impaired cardiac neural crest survival, adhesion, and migration, underlies the pericardial defects.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Pericárdio/metabolismo , Receptores Imunológicos/metabolismo , Transdução de Sinais , Veias Cavas/metabolismo , Animais , Apoptose , Adesão Celular , Movimento Celular , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfogênese , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Crista Neural/anormalidades , Crista Neural/metabolismo , Pericárdio/anormalidades , Receptores Imunológicos/deficiência , Receptores Imunológicos/genética , Nó Sinoatrial/anormalidades , Nó Sinoatrial/metabolismo , Proteínas com Domínio T/metabolismo , Técnicas de Cultura de Tecidos , Veias Cavas/anormalidades , Proteínas WT1/metabolismo , Proteínas Roundabout
16.
J Neurosci ; 32(47): 16892-905, 2012 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-23175841

RESUMO

In the postnatal forebrain, the subventricular zone (SVZ) contains a pool of undifferentiated cells, which proliferate and migrate along the rostral migratory stream (RMS) to the olfactory bulb and differentiate into granule cells and periglomerular cells. Plexin-B2 is a semaphorin receptor previously known to act on neuronal proliferation in the embryonic brain and neuronal migration in the cerebellum. We show here that, in the postnatal and adult CNS, Plexin-B2 is expressed in the subventricular zone lining the telencephalic ventricles and in the rostral migratory stream. We analyzed Plxnb2(-/-) mice and found that there is a marked reduction in the proliferation of SVZ cells in the mutant. Plexin-B2 expression is downregulated in the olfactory bulb as interneurons initiate radial migration. BrdU labeling and GFP electroporation into postnatal SVZ, in addition to time-lapse videomicroscopy, revealed that neuroblasts deficient for Plexin-B2 migrate faster than control ones and leave the RMS more rapidly. Overall, these results show that Plexin-B2 plays a role in postnatal neurogenesis and in the migration of SVZ-derived neuroblasts.


Assuntos
Movimento Celular/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Prosencéfalo/fisiologia , Animais , Antimetabólitos , Transplante de Tecido Encefálico , Bromodesoxiuridina , Movimento Celular/genética , Eletroporação , Feminino , Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Fator de Crescimento de Hepatócito/fisiologia , Imuno-Histoquímica , Hibridização In Situ , Masculino , Camundongos , Camundongos Knockout , Mutação/fisiologia , Proteínas do Tecido Nervoso/genética , Neurogênese/genética , Bulbo Olfatório/fisiologia , Prosencéfalo/citologia
17.
Development ; 139(18): 3326-31, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22912413

RESUMO

Gonadotropin-releasing hormone (GnRH) neurons are born in the nasal placode and migrate along olfactory and vomeronasal axons to reach the forebrain and settle in the hypothalamus, where they control reproduction. The molecular cues that guide their migration have not been fully identified, but are thought to control either cell movement directly or the patterning of their axonal substrates. Using genetically altered mouse models we show that the migration of GnRH neurons is directly modulated by Slit2 and Robo3, members of the axon guidance Slit ligand and Robo receptor families. Mice lacking Slit2 or Robo3 have a reduced number of GnRH neurons in the forebrain, but a normal complement of their supporting axons, pointing to a direct role for these molecules in GnRH neuron migration.


Assuntos
Movimento Celular/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Animais , Células COS , Movimento Celular/genética , Chlorocebus aethiops , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Mutantes , Proteínas do Tecido Nervoso/genética , Receptores de Superfície Celular
18.
J Alzheimers Dis ; 32(2): 267-72, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22776965

RESUMO

Accumulation of amyloid-ß peptides (Aß) and cholinergic degeneration are hallmarks of Alzheimer's disease (AD). In a triple transgenic mouse model of AD (3xTg-AD), soluble Aß42 levels were detected in the septum by 2 months of age, reaching their highest levels at 3-6 months and decreasing at 12 months. Deficits in the number of septal cholinergic neurons and the length of hippocampal cholinergic axons were observed starting at 4 months in 3xTg-AD mice. Our results show that septal Aß and septohippocampal cholinergic pathology in 3xTg-AD mice occur at an early stage of disease.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Neurônios Colinérgicos/patologia , Hipocampo/metabolismo , Septo do Cérebro/metabolismo , Doença de Alzheimer/patologia , Animais , Axônios/metabolismo , Axônios/patologia , Neurônios Colinérgicos/metabolismo , Hipocampo/patologia , Camundongos , Camundongos Transgênicos , Septo do Cérebro/patologia
19.
Development ; 137(12): 1939-52, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20501589

RESUMO

In most tissues, the precise control of cell migration and cell-cell interaction is of paramount importance to the development of a functional structure. Several families of secreted molecules have been implicated in regulating these aspects of development, including the Slits and their Robo receptors. These proteins have well described roles in axon guidance but by influencing cell polarity and adhesion, they participate in many developmental processes in diverse cell types. We review recent progress in understanding both the molecular mechanisms that modulate Slit/Robo expression and their functions in neural and non-neural tissue.


Assuntos
Movimento Celular/genética , Polaridade Celular/genética , Citoesqueleto/metabolismo , Sistema Nervoso/metabolismo , Neurogênese/fisiologia , Animais , Citoesqueleto/genética , Drosophila/genética , Drosophila/metabolismo , Camundongos , Camundongos Knockout
20.
Prog Neuropsychopharmacol Biol Psychiatry ; 34(4): 669-73, 2010 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-20227455

RESUMO

Abnormal lipid profiles have been reported in the central nervous system (CNS) in individuals with schizophrenia, although the etiology of these changes remains to be elucidated. While treatment with second-generation antipsychotics has been associated with alterations in peripheral lipid levels and changes in erythrocyte membrane composition, the relationship between peripheral and CNS lipid levels is complex and the effect of antipsychotics on CNS lipid regulation is not yet understood. In this study we investigated whether sub-chronic administration of the second-generation antipsychotic clozapine and the first-generation antipsychotic haloperidol alters brain membrane lipid composition in male Sprague-Dawley rats. The relationship between brain membrane lipid composition and plasma cholesterol concentrations was also assessed. Our results indicate that brain lipid composition and plasma cholesterol concentrations are not altered following administration of antipsychotics. No correlation was observed between plasma and brain membrane cholesterol levels. Our findings suggest that observed alterations in brain lipid profiles in individuals with schizophrenia are not a consequence of treatment with antipsychotic medications.


Assuntos
Química Encefálica/efeitos dos fármacos , Colesterol/análise , Clozapina/farmacologia , Haloperidol/farmacologia , Fosfolipídeos/análise , Análise de Variância , Animais , Antipsicóticos/farmacologia , Peso Corporal/efeitos dos fármacos , Colesterol/sangue , Masculino , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA