Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 13(1): 3868, 2023 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-36890145

RESUMO

Understanding the molecular pathology of neurodevelopmental disorders should aid the development of therapies for these conditions. In MeCP2 duplication syndrome (MDS)-a severe autism spectrum disorder-neuronal dysfunction is caused by increased levels of MeCP2. MeCP2 is a nuclear protein that binds to methylated DNA and recruits the nuclear co-repressor (NCoR) complex to chromatin via an interaction with the WD repeat-containing proteins TBL1 and TBLR1. The peptide motif in MeCP2 that binds to TBL1/TBLR1 is essential for the toxicity of excess MeCP2 in animal models of MDS, suggesting that small molecules capable of disrupting this interaction might be useful therapeutically. To facilitate the search for such compounds, we devised a simple and scalable NanoLuc luciferase complementation assay for measuring the interaction of MeCP2 with TBL1/TBLR1. The assay allowed excellent separation between positive and negative controls, and had low signal variance (Z-factor = 0.85). We interrogated compound libraries using this assay in combination with a counter-screen based on luciferase complementation by the two subunits of protein kinase A (PKA). Using this dual screening approach, we identified candidate inhibitors of the interaction between MeCP2 and TBL1/TBLR1. This work demonstrates the feasibility of future screens of large compound collections, which we anticipate will enable the development of small molecule therapeutics to ameliorate MDS.


Assuntos
Transtorno do Espectro Autista , Receptores Citoplasmáticos e Nucleares , Animais , Proteínas Repressoras/genética , Luminescência , Proteína 2 de Ligação a Metil-CpG/metabolismo , Proteínas Nucleares/metabolismo
2.
Mol Cell ; 81(6): 1260-1275.e12, 2021 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-33561390

RESUMO

DNA methylation is implicated in neuronal biology via the protein MeCP2, the mutation of which causes Rett syndrome. MeCP2 recruits the NCOR1/2 co-repressor complexes to methylated cytosine in the CG dinucleotide, but also to sites of non-CG methylation, which are abundant in neurons. To test the biological significance of the dual-binding specificity of MeCP2, we replaced its DNA binding domain with an orthologous domain from MBD2, which can only bind mCG motifs. Knockin mice expressing the domain-swap protein displayed severe Rett-syndrome-like phenotypes, indicating that normal brain function requires the interaction of MeCP2 with sites of non-CG methylation, specifically mCAC. The results support the notion that the delayed onset of Rett syndrome is due to the simultaneous post-natal accumulation of mCAC and its reader MeCP2. Intriguingly, genes dysregulated in both Mecp2 null and domain-swap mice are implicated in other neurological disorders, potentially highlighting targets of relevance to the Rett syndrome phenotype.


Assuntos
Metilação de DNA , Proteína 2 de Ligação a Metil-CpG/metabolismo , Neurônios/metabolismo , Animais , Ilhas de CpG , Técnicas de Introdução de Genes , Células HeLa , Humanos , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Camundongos Transgênicos , Mutação , Células NIH 3T3 , Neurônios/patologia , Domínios Proteicos , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Síndrome de Rett/patologia
4.
Genes Dev ; 32(23-24): 1514-1524, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30463906

RESUMO

Duplication of the X-linked MECP2 gene causes a severe neurological syndrome whose molecular basis is poorly understood. To determine the contribution of known functional domains to overexpression toxicity, we engineered a mouse model that expresses wild-type or mutated MeCP2 from the Mapt (Tau) locus in addition to the endogenous protein. Animals that expressed approximately four times the wild-type level of MeCP2 failed to survive to weaning. Strikingly, a single amino acid substitution that prevents MeCP2 from binding to the TBL1X(R1) subunit of nuclear receptor corepressor 1/2 (NCoR1/2) complexes, when expressed at equivalent high levels, was phenotypically indistinguishable from wild type, suggesting that excessive corepressor recruitment underlies toxicity. In contrast, mutations affecting the DNA-binding domain were toxic when overexpressed. As the NCoR1/2 corepressors are thought to act through histone deacetylation by histone deacetylase 3 (HDAC3), we asked whether mutations in NCoR1 and NCoR2 that drastically reduced their ability to activate this enzyme would relieve the MeCP2 overexpression phenotype. Surprisingly, severity was unaffected, indicating that the catalytic activity of HDAC3 is not the mediator of toxicity. Our findings shed light on the molecular mechanisms underlying MECP2 duplication syndrome and call for a re-evaluation of the precise biological role played by corepressor recruitment.


Assuntos
Expressão Gênica , Histona Desacetilases/metabolismo , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/toxicidade , Animais , Proteínas Correpressoras/metabolismo , Modelos Animais de Doenças , Ativação Enzimática/genética , Técnicas de Inativação de Genes , Histona Desacetilases/genética , Masculino , Deficiência Intelectual Ligada ao Cromossomo X/genética , Deficiência Intelectual Ligada ao Cromossomo X/fisiopatologia , Camundongos , Mutação , Doenças do Sistema Nervoso/genética , Neuroglia/metabolismo , Neurônios/metabolismo , Correpressor 1 de Receptor Nuclear/metabolismo , Correpressor 2 de Receptor Nuclear/metabolismo , Domínios Proteicos , Proteínas tau/metabolismo
5.
Cell Rep ; 24(9): 2213-2220, 2018 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-30157418

RESUMO

MeCP2 is a nuclear protein that is mutated in the severe neurological disorder Rett syndrome (RTT). The ability to target ß-galactosidase to the nucleus was previously used to identify a conserved nuclear localization signal (NLS) in MeCP2 that interacts with the nuclear import factors KPNA3 and KPNA4. Here, we report that nuclear localization of MeCP2 does not depend on its NLS. Instead, our data reveal that an intact methyl-CpG binding domain (MBD) is sufficient for nuclear localization, suggesting that MeCP2 can be retained in the nucleus by its affinity for DNA. Consistent with these findings, we demonstrate that disease progression in a mouse model of RTT is unaffected by an inactivating mutation in the NLS of MeCP2. Taken together, our work reveals an unexpected redundancy between functional domains of MeCP2 in targeting this protein to the nucleus, potentially explaining why NLS-inactivating mutations are rarely associated with disease.


Assuntos
DNA/metabolismo , Proteína 2 de Ligação a Metil-CpG/metabolismo , Sinais de Localização Nuclear/metabolismo , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Ilhas de CpG , DNA/genética , Modelos Animais de Doenças , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Síndrome de Rett/metabolismo , alfa Carioferinas/metabolismo
6.
Proc Natl Acad Sci U S A ; 114(16): E3243-E3250, 2017 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-28348241

RESUMO

Rett syndrome (RTT) is an X-linked neurological disorder caused by mutations in the methyl-CpG-binding protein 2 (MeCP2) gene. The majority of RTT missense mutations disrupt the interaction of the MeCP2 with DNA or the nuclear receptor corepressor (NCoR)/silencing mediator of retinoic acid and thyroid receptors (SMRT) corepressor complex. Here, we show that the "NCoR/SMRT interaction domain" (NID) of MeCP2 directly contacts transducin beta-like 1 (TBL1) and TBL1 related (TBLR1), two paralogs that are core components of NCoR/SMRT. We determine the cocrystal structure of the MeCP2 NID in complex with the WD40 domain of TBLR1 and confirm by in vitro and ex vivo assays that mutation of interacting residues of TBLR1 and TBL1 disrupts binding to MeCP2. Strikingly, the four MeCP2-NID residues mutated in RTT are those residues that make the most extensive contacts with TBLR1. Moreover, missense mutations in the gene for TBLR1 that are associated with intellectual disability also prevent MeCP2 binding. Our study therefore reveals the molecular basis of an interaction that is crucial for optimal brain function.


Assuntos
Proteína 2 de Ligação a Metil-CpG/química , Mutação de Sentido Incorreto , Proteínas Nucleares/química , Receptores Citoplasmáticos e Nucleares/química , Proteínas Repressoras/química , Síndrome de Rett/genética , Cristalografia por Raios X , Células HeLa , Humanos , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Correpressor 1 de Receptor Nuclear/química , Correpressor 1 de Receptor Nuclear/genética , Correpressor 1 de Receptor Nuclear/metabolismo , Conformação Proteica , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Síndrome de Rett/patologia , Transducina/química , Transducina/genética , Transducina/metabolismo
7.
FEBS Lett ; 590(17): 2927-33, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27461740

RESUMO

MeCP2 is a chromatin-associated protein that is mutated in Rett syndrome. Its methyl-CpG-binding domain interacts with DNA containing methylated cytosine, but other modes of recruitment to the genome have also been proposed. Here, we use in vitro and in vivo assays to investigate the DNA binding specificity of two AT-hook motifs in MeCP2. One exhibits robust sequence-specific DNA binding, whereas the other is a much weaker AT-hook. Our data indicate that these motifs are secondary contributors to DNA binding by MeCP2, and this view is supported by the absence of disease-causing missense mutations at these sites.


Assuntos
Motivos AT-Hook/genética , Metilação de DNA/genética , Proteína 2 de Ligação a Metil-CpG/genética , Síndrome de Rett/genética , Sequência de Bases/genética , Cromatina/genética , Citosina/metabolismo , Proteínas de Ligação a DNA/genética , Humanos , Mutação de Sentido Incorreto/genética , Síndrome de Rett/patologia
8.
Nat Rev Genet ; 16(5): 261-75, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25732612

RESUMO

Rett syndrome (RTT) is a severe neurological disorder caused by mutations in the X-linked gene MECP2 (methyl-CpG-binding protein 2). Two decades of research have fostered the view that MeCP2 is a multifunctional chromatin protein that integrates diverse aspects of neuronal biology. More recently, studies have focused on specific RTT-associated mutations within the protein. This work has yielded molecular insights into the critical functions of MeCP2 that promise to simplify our understanding of RTT pathology.


Assuntos
Síndrome de Rett/genética , Animais , Encéfalo/patologia , Encéfalo/fisiopatologia , DNA/química , DNA/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/fisiologia , Camundongos , Camundongos Knockout , Modelos Neurológicos , Mutação , Neurônios/patologia , Neurônios/fisiologia , Ligação Proteica , Síndrome de Rett/patologia , Síndrome de Rett/fisiopatologia , Transativadores/genética , Transativadores/fisiologia
9.
Nat Neurosci ; 16(7): 898-902, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23770565

RESUMO

Rett syndrome (RTT) is a severe neurological disorder that is caused by mutations in the MECP2 gene. Many missense mutations causing RTT are clustered in the DNA-binding domain of MeCP2, suggesting that association with chromatin is critical for its function. We identified a second mutational cluster in a previously uncharacterized region of MeCP2. We found that RTT mutations in this region abolished the interaction between MeCP2 and the NCoR/SMRT co-repressor complexes. Mice bearing a common missense RTT mutation in this domain exhibited severe RTT-like phenotypes. Our data are compatible with the hypothesis that brain dysfunction in RTT is caused by a loss of the MeCP2 'bridge' between the NCoR/SMRT co-repressors and chromatin.


Assuntos
Proteína 2 de Ligação a Metil-CpG/genética , Mutação/genética , Correpressor 1 de Receptor Nuclear/metabolismo , Correpressor 2 de Receptor Nuclear/metabolismo , Síndrome de Rett/genética , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Células Cultivadas , Modelos Animais de Doenças , Comportamento Exploratório/fisiologia , Proteínas de Fluorescência Verde/genética , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Moleculares , Correpressor 1 de Receptor Nuclear/genética , Correpressor 2 de Receptor Nuclear/genética , Síndrome de Rett/patologia , Síndrome de Rett/fisiopatologia
10.
Nature ; 499(7458): 341-5, 2013 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-23770587

RESUMO

Rett syndrome (RTT) is an X-linked human neurodevelopmental disorder with features of autism and severe neurological dysfunction in females. RTT is caused by mutations in methyl-CpG-binding protein 2 (MeCP2), a nuclear protein that, in neurons, regulates transcription, is expressed at high levels similar to that of histones, and binds to methylated cytosines broadly across the genome. By phosphotryptic mapping, we identify three sites (S86, S274 and T308) of activity-dependent MeCP2 phosphorylation. Phosphorylation of these sites is differentially induced by neuronal activity, brain-derived neurotrophic factor, or agents that elevate the intracellular level of 3',5'-cyclic AMP (cAMP), indicating that MeCP2 may function as an epigenetic regulator of gene expression that integrates diverse signals from the environment. Here we show that the phosphorylation of T308 blocks the interaction of the repressor domain of MeCP2 with the nuclear receptor co-repressor (NCoR) complex and suppresses the ability of MeCP2 to repress transcription. In knock-in mice bearing the common human RTT missense mutation R306C, neuronal activity fails to induce MeCP2 T308 phosphorylation, suggesting that the loss of T308 phosphorylation might contribute to RTT. Consistent with this possibility, the mutation of MeCP2 T308A in mice leads to a decrease in the induction of a subset of activity-regulated genes and to RTT-like symptoms. These findings indicate that the activity-dependent phosphorylation of MeCP2 at T308 regulates the interaction of MeCP2 with the NCoR complex, and that RTT in humans may be due, in part, to the loss of activity-dependent MeCP2 T308 phosphorylation and a disruption of the phosphorylation-regulated interaction of MeCP2 with the NCoR complex.


Assuntos
Proteínas Correpressoras/metabolismo , Proteína 2 de Ligação a Metil-CpG/metabolismo , Treonina/metabolismo , Animais , Células Cultivadas , Humanos , Proteína 2 de Ligação a Metil-CpG/química , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Mutação , Neurônios/metabolismo , Fosforilação , Síndrome de Rett/genética , Transcrição Gênica
11.
EMBO J ; 25(22): 5317-28, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17066076

RESUMO

In mammalian cells, DNA methylation is associated with heritable and stable gene repression, mediated in part by methyl-CpG-binding domain (MBD) proteins that recruit corepressors to modify chromatin. MBD1 protein, a member of the MBD family, forms a complex with SETDB1 histone methylase to silence transcription at target promoters by methylation of lysine 9 of histone H3. How MBD1-mediated transcriptional repression is regulated is currently unknown. Here we show that MBD1 is a target for sumoylation by PIAS1 (Protein Inhibitors of Activated STAT 1) and PIAS3 E3 SUMO (small ubiquitin-like modifier)-ligases, at two conserved lysine residues within the C-terminus of MBD1. Although sumoylated MBD1 binds to methylated DNA, it does not incorporate into a complex with SETDB1 and does not efficiently repress transcription of a target gene, p53BP2, in HeLa cells. Our data suggest that transcriptional silencing by MBD1 is regulated by a PIAS-mediated conjugation of SUMO1, which antagonizes the formation of a repressive complex with SETDB1.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Chaperonas Moleculares/fisiologia , Proteínas Inibidoras de STAT Ativados/fisiologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/fisiologia , Fatores de Transcrição/fisiologia , Transcrição Gênica , Sequência de Aminoácidos , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Transporte/fisiologia , Cromatina/genética , Cromatina/fisiologia , Sequência Conservada , Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Inativação Gênica , Células HeLa , Histona-Lisina N-Metiltransferase , Histonas/metabolismo , Humanos , Chaperonas Moleculares/genética , Dados de Sequência Molecular , Ligação Proteica , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Metiltransferases/metabolismo , Proteína SUMO-1 , Homologia de Sequência de Aminoácidos , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...