Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 52(6): 2995-3010, 2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38224953

RESUMO

Meiosis is a key step during germ cell differentiation, accompanied by the activation of thousands of genes through germline-specific chromatin reorganization. The chromatin remodeling mechanisms underpinning early meiotic stages remain poorly understood. Here we focus on the function of one of the major autism genes, CHD8, in spermatogenesis, based on the epidemiological association between autism and low fertility rates. Specific ablation of Chd8 in germ cells results in gradual depletion of undifferentiated spermatogonia and the failure of meiotic double-strand break (DSB) formation, leading to meiotic prophase I arrest and cell death. Transcriptional analyses demonstrate that CHD8 is required for extensive activation of spermatogenic genes in spermatogonia, necessary for spermatogonial proliferation and meiosis. CHD8 directly binds and regulates genes crucial for meiosis, including H3K4me3 histone methyltransferase genes, meiotic cohesin genes, HORMA domain-containing genes, synaptonemal complex genes, and DNA damage response genes. We infer that CHD8 contributes to meiotic DSB formation and subsequent meiotic progression through combined regulation of these meiosis-related genes. Our study uncovers an essential role of CHD8 in the proliferation of undifferentiated spermatogonia and the successful progression of meiotic prophase I.


Assuntos
Meiose , Espermatogônias , Masculino , Proliferação de Células/genética , Cromatina/genética , Cromatina/metabolismo , Meiose/genética , Espermatogênese/genética , Animais , Camundongos
2.
Hum Mol Genet ; 29(8): 1274-1291, 2020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32142125

RESUMO

Mutations in the gene encoding the chromatin remodeler CHD8 are strongly associated with autism spectrum disorder (ASD). CHD8 haploinsufficiency also results in autistic phenotypes in humans and mice. Although myelination defects have been observed in individuals with ASD, whether oligodendrocyte dysfunction is responsible for autistic phenotypes has remained unknown. Here we show that reduced expression of CHD8 in oligodendrocytes gives rise to abnormal behavioral phenotypes in mice. CHD8 was found to regulate the expression of many myelination-related genes and to be required for oligodendrocyte maturation and myelination. Ablation of Chd8 specifically in oligodendrocytes of mice impaired myelination, slowed action potential propagation and resulted in behavioral deficits including increased social interaction and anxiety-like behavior, with similar effects being apparent in Chd8 heterozygous mutant mice. Our results thus indicate that CHD8 is essential for myelination and that dysfunction of oligodendrocytes as a result of CHD8 haploinsufficiency gives rise to several neuropsychiatric phenotypes.


Assuntos
Transtorno do Espectro Autista/genética , Proteínas de Ligação a DNA/genética , Neurogênese/genética , Fatores de Transcrição/genética , Animais , Transtorno do Espectro Autista/patologia , Montagem e Desmontagem da Cromatina/genética , Modelos Animais de Doenças , Haploinsuficiência/genética , Heterozigoto , Humanos , Camundongos , Mutação/genética , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Fenótipo
3.
Nature ; 537(7622): 675-679, 2016 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-27602517

RESUMO

Autism spectrum disorder (ASD) comprises a range of neurodevelopmental disorders characterized by deficits in social interaction and communication as well as by restricted and repetitive behaviours. ASD has a strong genetic component with high heritability. Exome sequencing analysis has recently identified many de novo mutations in a variety of genes in individuals with ASD, with CHD8, a gene encoding a chromatin remodeller, being most frequently affected. Whether CHD8 mutations are causative for ASD and how they might establish ASD traits have remained unknown. Here we show that mice heterozygous for Chd8 mutations manifest ASD-like behavioural characteristics including increased anxiety, repetitive behaviour, and altered social behaviour. CHD8 haploinsufficiency did not result in prominent changes in the expression of a few specific genes but instead gave rise to small but global changes in gene expression in the mouse brain, reminiscent of those in the brains of patients with ASD. Gene set enrichment analysis revealed that neurodevelopment was delayed in the mutant mouse embryos. Furthermore, reduced expression of CHD8 was associated with abnormal activation of RE-1 silencing transcription factor (REST), which suppresses the transcription of many neuronal genes. REST activation was also observed in the brains of humans with ASD, and CHD8 was found to interact physically with REST in the mouse brain. Our results are thus consistent with the notion that CHD8 haploinsufficiency is a highly penetrant risk factor for ASD, with disease pathogenesis probably resulting from a delay in neurodevelopment.


Assuntos
Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/psicologia , Proteínas de Ligação a DNA/genética , Haploinsuficiência/genética , Animais , Ansiedade/complicações , Ansiedade/genética , Transtorno do Espectro Autista/complicações , Encéfalo/metabolismo , Proteínas de Ligação a DNA/deficiência , Deficiências do Desenvolvimento/genética , Modelos Animais de Doenças , Regulação para Baixo , Predisposição Genética para Doença , Heterozigoto , Masculino , Megalencefalia/complicações , Megalencefalia/genética , Camundongos , Camundongos Knockout , Mutação , Penetrância , Fenótipo , Proteínas Repressoras/metabolismo , Comportamento Social , Transcriptoma
4.
J Biol Chem ; 293(15): 5705-5714, 2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29472293

RESUMO

The ubiquitin-proteasome system regulates the abundance of many cellular proteins by mediating their targeted degradation. We previously developed a method-differential proteomics-based identification of ubiquitylation substrates (DiPIUS)-for the comprehensive identification of substrates for a given F-box protein subunit of SCF-type ubiquitin ligases. We have now applied DiPIUS to the F-box protein Fbxw7 in three cell lines (mHepa, Neuro2A, and C2C12) and thereby identified myelin regulatory factor (MyRF), an endoplasmic reticulum-anchored transcription factor that is essential for myelination of nerves in the central nervous system, as a candidate substrate of Fbxw7 specifically in mHepa cells. Co-immunoprecipitation analysis confirmed that the NH2-terminal cytoplasmic domain of MyRF interacted with Fbxw7 in these cells. Furthermore, an in vitro ubiquitylation assay revealed that MyRF undergoes polyubiquitylation in the presence of purified recombinant SCFFbxw7 In addition, the stability of MyRF in mHepa cells was increased by mutation of a putative phosphodegron sequence or by exposure of the cells to an inhibitor of glycogen synthase kinase-3 (GSK-3). We found that MyRF mRNA is not restricted to the central nervous system but is instead distributed widely among mouse tissues. Furthermore, with the use of RNA sequencing in mHepa cells overexpressing or depleted of MyRF, we identified many novel potential target genes of MyRF. Our results thus suggest that Fbxw7 controls the transcription of MyRF target genes in various tissues through regulation of MyRF protein stability in a manner dependent on MyRF phosphorylation by GSK-3.


Assuntos
Retículo Endoplasmático/metabolismo , Proteína 7 com Repetições F-Box-WD/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Proteólise , Fatores de Transcrição/biossíntese , Transcrição Gênica/fisiologia , Linhagem Celular , Clotrimazol/análogos & derivados , Retículo Endoplasmático/genética , Proteína 7 com Repetições F-Box-WD/genética , Quinase 3 da Glicogênio Sintase/genética , Especificidade de Órgãos , Fosforilação/fisiologia , Domínios Proteicos , Fatores de Transcrição/genética
5.
Commun Biol ; 6(1): 593, 2023 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-37268684

RESUMO

CHD8 encodes chromodomain helicase DNA-binding protein 8 and its mutation is a highly penetrant risk factor for autism spectrum disorder (ASD). CHD8 serves as a key transcriptional regulator on the basis of its chromatin-remodeling activity and thereby controls the proliferation and differentiation of neural progenitor cells. However, the function of CHD8 in postmitotic neurons and the adult brain has remained unclear. Here we show that Chd8 homozygous deletion in mouse postmitotic neurons results in downregulation of the expression of neuronal genes as well as alters the expression of activity-dependent genes induced by KCl-mediated neuronal depolarization. Furthermore, homozygous ablation of CHD8 in adult mice was associated with attenuation of activity-dependent transcriptional responses in the hippocampus to kainic acid-induced seizures. Our findings implicate CHD8 in transcriptional regulation in postmitotic neurons and the adult brain, and they suggest that disruption of this function might contribute to ASD pathogenesis associated with CHD8 haploinsufficiency.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Camundongos , Animais , Transtorno Autístico/genética , Transtorno Autístico/metabolismo , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/metabolismo , Homozigoto , Deleção de Sequência , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Neurônios/metabolismo
6.
Cell Rep ; 35(1): 108932, 2021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-33826902

RESUMO

Mutations in the gene encoding the chromatin remodeler chromodomain helicase DNA-binding protein 8 (CHD8) are a highly penetrant risk factor for autism spectrum disorder (ASD). Although cerebellar abnormalities have long been thought to be related to ASD pathogenesis, it has remained largely unknown whether dysfunction of CHD8 in the cerebellum contributes to ASD phenotypes. We here show that cerebellar granule neuron progenitor (GNP)-specific deletion of Chd8 in mice impairs the proliferation and differentiation of these cells as well as gives rise to cerebellar hypoplasia and a motor coordination defect, but not to ASD-like behavioral abnormalities. CHD8 is found to regulate the expression of neuronal genes in GNPs. It also binds preferentially to promoter regions and modulates local chromatin accessibility of transcriptionally active genes in these cells. Our results have thus uncovered a key role for CHD8 in cerebellar development, with important implications for understanding the contribution of this brain region to ASD pathogenesis.


Assuntos
Transtorno Autístico/patologia , Cerebelo/embriologia , Cerebelo/fisiopatologia , Proteínas de Ligação a DNA/metabolismo , Atividade Motora , Animais , Comportamento Animal , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Cerebelo/anormalidades , Cromatina/metabolismo , Proteínas de Ligação a DNA/deficiência , Deficiências do Desenvolvimento , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos Endogâmicos C57BL , Malformações do Sistema Nervoso , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Sinapses/metabolismo
7.
Mol Brain ; 13(1): 160, 2020 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-33228730

RESUMO

CHD8 encodes a chromatin-remodeling factor and is one of the most recurrently mutated genes in individuals with autism spectrum disorder (ASD). Although we have recently shown that mice heterozygous for Chd8 mutation manifest myelination defects and ASD-like behaviors, the detailed mechanisms underlying ASD pathogenesis have remained unclear. Here we performed diffusion tensor imaging (DTI) and resting-state functional magnetic resonance imaging (rsfMRI) in oligodendrocyte lineage-specific Chd8 heterozygous mutant mice. DTI revealed that ablation of Chd8 specifically in oligodendrocytes of mice was associated with microstructural changes of specific brain regions including the cortex and striatum. The extent of these changes in white matter including the corpus callosum and fornix was correlated with total contact time in the reciprocal social interaction test. Analysis with rsfMRI revealed changes in functional brain connectivity in the mutant mice, and the extent of such changes in the cortex, hippocampus, and amygdala was also correlated with the change in social interaction. Our results thus suggest that changes in brain microstructure and functional connectivity induced by oligodendrocyte dysfunction might underlie altered social interaction in mice with oligodendrocyte-specific CHD8 haploinsufficiency.


Assuntos
Encéfalo/patologia , Encéfalo/fisiopatologia , Proteínas de Ligação a DNA/genética , Mutação/genética , Rede Nervosa/fisiopatologia , Oligodendroglia/metabolismo , Animais , Comportamento Animal , Encéfalo/diagnóstico por imagem , Linhagem da Célula , Imagem de Tensor de Difusão , Heterozigoto , Camundongos , Camundongos Mutantes , Comportamento Social
8.
Mol Cell Biol ; 37(8)2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28069738

RESUMO

FBXL5 is the substrate recognition subunit of an SCF-type ubiquitin ligase that serves as a master regulator of iron metabolism in mammalian cells. We previously showed that mice with systemic deficiency of FBXL5 fail to sense intracellular iron levels and die in utero at embryonic day 8.5 (E8.5) as a result of iron overload and subsequent oxidative stress. This early embryonic mortality has thus impeded study of the role of FBXL5 in brain development. We have now generated mice lacking FBXL5 specifically in nestin-expressing neural stem progenitor cells (NSPCs) in the brain. Unexpectedly, the mutant embryos manifested a progressive increase in the number of NSPCs and astroglia in the cerebral cortex. Stabilization of iron regulatory protein 2 (IRP2) as a result of FBXL5 deficiency led to accumulation of ferrous and ferric iron as well as to generation of reactive oxygen species. Pharmacological manipulation suggested that the phenotypes of FBXL5 deficiency are attributable to aberrant activation of mammalian target of rapamycin (mTOR) signaling. Our results thus show that FBXL5 contributes to regulation of NSPC proliferation during mammalian brain development.


Assuntos
Encéfalo/metabolismo , Proteínas F-Box/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Animais , Animais Recém-Nascidos , Proliferação de Células , Embrião de Mamíferos/citologia , Deleção de Genes , Ferro/metabolismo , Camundongos Knockout , Estresse Oxidativo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA