Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 152(5): 984-96, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23452848

RESUMEN

Mutations in the X-linked MECP2 cause Rett syndrome, a devastating neurological disorder typified by a period of apparently normal development followed by loss of cognitive and psychomotor skills. Data from rare male patients suggest symptom onset and severity can be influenced by the location of the mutation, with amino acids 270 and 273 marking the difference between neonatal encephalopathy and death, on the one hand, and survival with deficits on the other. We therefore generated two mouse models expressing either MeCP2-R270X or MeCP2-G273X. The mice developed phenotypes at strikingly different rates and showed differential ATRX nuclear localization within the nervous system, over time, coinciding with phenotypic progression. We discovered that MeCP2 contains three AT-hook-like domains over a stretch of 250 amino acids, like HMGA DNA-bending proteins; one conserved AT-hook is disrupted in MeCP2-R270X, lending further support to the notion that one of MeCP2's key functions is to alter chromatin structure.


Asunto(s)
Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/metabolismo , Síndrome de Rett/metabolismo , Secuencia de Aminoácidos , Animales , ADN Helicasas/metabolismo , Modelos Animales de Enfermedad , Femenino , Heterocromatina/metabolismo , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Proteínas Nucleares/metabolismo , Estructura Terciaria de Proteína , Síndrome de Rett/genética , Síndrome de Rett/fisiopatología , Alineación de Secuencia , Transcripción Genética , Proteína Nuclear Ligada al Cromosoma X
2.
Annu Rev Cell Dev Biol ; 27: 631-52, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21721946

RESUMEN

Methyl-CpG binding protein 2 (MeCP2) was first identified in 1992 as a protein that binds specifically to methylated DNA. Mutations in the MECP2 gene were later found to be the cause of an autism spectrum disorder, Rett syndrome. Despite almost 20 years of research into the molecular mechanisms of MeCP2 function, many questions are yet to be answered conclusively. This review considers several key questions and attempts to evaluate the current state of evidence. For example, is MeCP2 just a methyl-CpG binding protein? Is it a multifunctional protein or primarily a transcriptional repressor? We also consider whether MeCP2, as a chromosome-binding protein, acts at specific sites within the genome or more globally, and in which cell types it is functionally important. Finally, we consider two alternative views of MeCP2 in the brain: as a regulator of brain development or as a factor that helps maintain neuronal/glial function.


Asunto(s)
Encéfalo/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , Animales , Encéfalo/anatomía & histología , Encéfalo/crecimiento & desarrollo , Islas de CpG , Metilación de ADN , Humanos , Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/genética , Neuroglía/metabolismo , Neuronas/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
3.
Nature ; 550(7676): 398-401, 2017 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-29019980

RESUMEN

Heterozygous mutations in the X-linked MECP2 gene cause the neurological disorder Rett syndrome. The methyl-CpG-binding protein 2 (MeCP2) protein is an epigenetic reader whose binding to chromatin primarily depends on 5-methylcytosine. Functionally, MeCP2 has been implicated in several cellular processes on the basis of its reported interaction with more than 40 binding partners, including transcriptional co-repressors (for example, the NCoR/SMRT complex), transcriptional activators, RNA, chromatin remodellers, microRNA-processing proteins and splicing factors. Accordingly, MeCP2 has been cast as a multi-functional hub that integrates diverse processes that are essential in mature neurons. At odds with the concept of broad functionality, missense mutations that cause Rett syndrome are concentrated in two discrete clusters coinciding with interaction sites for partner macromolecules: the methyl-CpG binding domain and the NCoR/SMRT interaction domain. Here we test the hypothesis that the single dominant function of MeCP2 is to physically connect DNA with the NCoR/SMRT complex, by removing almost all amino-acid sequences except the methyl-CpG binding and NCoR/SMRT interaction domains. We find that mice expressing truncated MeCP2 lacking both the N- and C-terminal regions (approximately half of the native protein) are phenotypically near-normal; and those expressing a minimal MeCP2 additionally lacking a central domain survive for over one year with only mild symptoms. This minimal protein is able to prevent or reverse neurological symptoms when introduced into MeCP2-deficient mice by genetic activation or virus-mediated delivery to the brain. Thus, despite evolutionary conservation of the entire MeCP2 protein sequence, the DNA and co-repressor binding domains alone are sufficient to avoid Rett syndrome-like defects and may therefore have therapeutic utility.


Asunto(s)
Prueba de Complementación Genética , Terapia Genética/métodos , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Síndrome de Rett/genética , Síndrome de Rett/terapia , Eliminación de Secuencia , Células 3T3 , Animales , Encéfalo/metabolismo , ADN/metabolismo , Células HeLa , Humanos , Masculino , Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/deficiencia , Ratones , Mutación Missense , Fenotipo , Dominios Proteicos/genética , Estabilidad Proteica , Síndrome de Rett/patología , Síndrome de Rett/fisiopatología , Transducción Genética
4.
J Am Chem Soc ; 144(7): 2987-2993, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35157801

RESUMEN

5-Methylcytosine (mC) and 5-hydroxymethylcytosine (hmC), the two main epigenetic modifications of mammalian DNA, exist in symmetric and asymmetric combinations in the two strands of CpG dyads. However, revealing such combinations in single DNA duplexes is a significant challenge. Here, we evolve methyl-CpG-binding domains (MBDs) derived from MeCP2 by bacterial cell surface display, resulting in the first affinity probes for hmC/mC CpGs. One mutant has low nanomolar affinity for a single hmC/mC CpG, discriminates against all 14 other modified CpG dyads, and rivals the selectivity of wild-type MeCP2. Structural studies indicate that this protein has a conserved scaffold and recognizes hmC and mC with two dedicated sets of residues. The mutant allows us to selectively address and enrich hmC/mC-containing DNA fragments from genomic DNA backgrounds. We anticipate that this novel probe will be a versatile tool to unravel the function of hmC/mC marks in diverse aspects of chromatin biology.


Asunto(s)
5-Metilcitosina/análogos & derivados , 5-Metilcitosina/química , ADN/aislamiento & purificación , Proteína 2 de Unión a Metil-CpG/química , Fragmentos de Péptidos/química , ADN/química , Metilación de ADN , Evolución Molecular Dirigida , Células HEK293 , Humanos , Proteína 2 de Unión a Metil-CpG/genética , Fragmentos de Péptidos/genética , Dominios Proteicos
5.
Int J Mol Sci ; 22(18)2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34576118

RESUMEN

Rett Syndrome (RTT) is an X linked neurodevelopmental disorder caused by mutations in the methyl-CpG-binding protein 2 (MECP2) gene, resulting in severe cognitive and physical disabilities. Despite an apparent normal prenatal and postnatal development period, symptoms usually present around 6 to 18 months of age. Little is known about the consequences of MeCP2 deficiency at a molecular and cellular level before the onset of symptoms in neural cells, and subtle changes at this highly sensitive developmental stage may begin earlier than symptomatic manifestation. Recent transcriptomic studies of patient induced pluripotent stem cells (iPSC)-differentiated neurons and brain organoids harbouring pathogenic mutations in MECP2, have unravelled new insights into the cellular and molecular changes caused by these mutations. Here we interrogated transcriptomic modifications in RTT patients using publicly available RNA-sequencing datasets of patient iPSCs harbouring pathogenic mutations and healthy control iPSCs by Weighted Gene Correlation Network Analysis (WGCNA). Preservation analysis identified core gene pathways involved in translation, ribosomal function, and ubiquitination perturbed in some MECP2 mutant iPSC lines. Furthermore, differential gene expression of the parental fibroblasts and iPSC-derived neurons revealed alterations in genes in the ubiquitination pathway and neurotransmission in fibroblasts and differentiated neurons respectively. These findings might suggest that global translational dysregulation and proteasome ubiquitin function in Rett syndrome begins in progenitor cells prior to lineage commitment and differentiation into neural cells.


Asunto(s)
Redes Reguladoras de Genes , Complejo de la Endopetidasa Proteasomal/metabolismo , Biosíntesis de Proteínas/genética , Síndrome de Rett/genética , Ubiquitina/metabolismo , Análisis por Conglomerados , Bases de Datos Genéticas , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Neuronas/metabolismo , Análisis de Componente Principal , Dominios Proteicos , Ubiquitina/genética
6.
Nucleic Acids Res ; 46(17): 9044-9056, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-30102379

RESUMEN

Despite their central importance in mammalian development, the mechanisms that regulate the DNA methylation machinery and thereby the generation of genomic methylation patterns are still poorly understood. Here, we identify the 5mC-binding protein MeCP2 as a direct and strong interactor of DNA methyltransferase 3 (DNMT3) proteins. We mapped the interaction interface to the transcriptional repression domain of MeCP2 and the ADD domain of DNMT3A and find that binding of MeCP2 strongly inhibits the activity of DNMT3A in vitro. This effect was reinforced by cellular studies where a global reduction of DNA methylation levels was observed after overexpression of MeCP2 in human cells. By engineering conformationally locked DNMT3A variants as novel tools to study the allosteric regulation of this enzyme, we show that MeCP2 stabilizes the closed, autoinhibitory conformation of DNMT3A. Interestingly, the interaction with MeCP2 and its resulting inhibition were relieved by the binding of K4 unmodified histone H3 N-terminal tail to the DNMT3A-ADD domain. Taken together, our data indicate that the localization and activity of DNMT3A are under the combined control of MeCP2 and H3 tail modifications where, depending on the modification status of the H3 tail at the binding sites, MeCP2 can act as either a repressor or activator of DNA methylation.


Asunto(s)
Cromatina/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , ADN/química , Epigénesis Genética , Histonas/genética , Proteína 2 de Unión a Metil-CpG/genética , Regulación Alostérica , Animales , Sitios de Unión , Química Encefálica , Cromatina/química , Clonación Molecular , ADN/metabolismo , ADN (Citosina-5-)-Metiltransferasas/química , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , ADN Metiltransferasa 3A , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Células HEK293 , Histonas/química , Histonas/metabolismo , Humanos , Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/metabolismo , Ratones , Mutagénesis Sitio-Dirigida/métodos , Unión Proteica , Ingeniería de Proteínas/métodos , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
7.
Proc Natl Acad Sci U S A ; 114(16): E3243-E3250, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28348241

RESUMEN

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.


Asunto(s)
Proteína 2 de Unión a Metil-CpG/química , Mutación Missense , Proteínas Nucleares/química , Receptores Citoplasmáticos y Nucleares/química , Proteínas Represoras/química , Síndrome de Rett/genética , Cristalografía por Rayos X , Células HeLa , Humanos , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Co-Represor 1 de Receptor Nuclear/química , Co-Represor 1 de Receptor Nuclear/genética , Co-Represor 1 de Receptor Nuclear/metabolismo , Conformación Proteica , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Síndrome de Rett/patología , Transducina/química , Transducina/genética , Transducina/metabolismo
8.
J Biol Chem ; 293(19): 7344-7354, 2018 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-29567833

RESUMEN

Cytosine methylation is a well-characterized epigenetic mark and occurs at both CG and non-CG sites in DNA. Both methylated CG (mCG)- and mCH (H = A, C, or T)-containing DNAs, especially mCAC-containing DNAs, are recognized by methyl-CpG-binding protein 2 (MeCP2) to regulate gene expression in neuron development. However, the molecular mechanism involved in the binding of methyl-CpG-binding domain (MBD) of MeCP2 to these different DNA motifs is unclear. Here, we systematically characterized the DNA-binding selectivities of the MBD domains in MeCP2 and MBD1-4 with isothermal titration calorimetry-based binding assays, mutagenesis studies, and X-ray crystallography. We found that the MBD domains of MeCP2 and MBD1-4 bind mCG-containing DNAs independently of the sequence identity outside the mCG dinucleotide. Moreover, some MBD domains bound to both methylated and unmethylated CA dinucleotide-containing DNAs, with a preference for the CAC sequence motif. We also found that the MBD domains bind to mCA or nonmethylated CA DNA by recognizing the complementary TG dinucleotide, which is consistent with an overlooked ligand of MeCP2, i.e. the matrix/scaffold attachment regions (MARs/SARs) with a consensus sequence of 5'-GGTGT-3' that was identified in early 1990s. Our results also explain why MeCP2 exhibits similar binding affinity to both mCA- and hmCA-containing dsDNAs. In summary, our results suggest that in addition to mCG sites, unmethylated CA or TG sites also serve as DNA-binding sites for MeCP2 and other MBD-containing proteins. This discovery expands the genome-wide activity of MBD-containing proteins in gene regulation.


Asunto(s)
Proteínas de Unión al ADN/química , ADN/metabolismo , Endodesoxirribonucleasas/química , Proteína 2 de Unión a Metil-CpG/química , Factores de Transcripción/química , Sitios de Unión , Calorimetría/métodos , Cristalografía por Rayos X , Citosina/química , Metilación de ADN , Proteínas de Unión al ADN/genética , Endodesoxirribonucleasas/genética , Regulación de la Expresión Génica , Guanina/química , Humanos , Proteína 2 de Unión a Metil-CpG/genética , Mutagénesis , Nucleótidos/metabolismo , Dominios Proteicos , Timina/química , Factores de Transcripción/genética
9.
Nature ; 499(7458): 341-5, 2013 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-23770587

RESUMEN

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.


Asunto(s)
Proteínas Co-Represoras/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , Treonina/metabolismo , Animales , Células Cultivadas , Humanos , Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Mutación , Neuronas/metabolismo , Fosforilación , Síndrome de Rett/genética , Transcripción Genética
10.
Nucleic Acids Res ; 45(13): 7751-7759, 2017 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-28486614

RESUMEN

Eukaryotic genomes contain numerous non-functional high-affinity sequences for transcription factors. These sequences potentially serve as natural decoys that sequester transcription factors. We have previously shown that the presence of sequences similar to the target sequence could substantially impede association of the transcription factor Egr-1 with its targets. In this study, using a stopped-flow fluorescence method, we examined the kinetic impact of DNA methylation of decoys on the search process of the Egr-1 zinc-finger protein. We analyzed its association with an unmethylated target site on fluorescence-labeled DNA in the presence of competitor DNA duplexes, including Egr-1 decoys. DNA methylation of decoys alone did not affect target search kinetics. In the presence of the MeCP2 methyl-CpG-binding domain (MBD), however, DNA methylation of decoys substantially (∼10-30-fold) accelerated the target search process of the Egr-1 zinc-finger protein. This acceleration did not occur when the target was also methylated. These results suggest that when decoys are methylated, MBD proteins can block them and thereby allow Egr-1 to avoid sequestration in non-functional locations. This effect may occur in vivo for DNA methylation outside CpG islands (CGIs) and could facilitate localization of some transcription factors within regulatory CGIs, where DNA methylation is rare.


Asunto(s)
Metilación de ADN , Proteína 2 de Unión a Metil-CpG/metabolismo , Sitios de Unión , Islas de CpG , ADN/química , ADN/genética , ADN/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Humanos , Cinética , Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/genética , Modelos Biológicos , Dominios Proteicos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Activación Transcripcional , Dedos de Zinc/genética
11.
Hum Mutat ; 39(5): 717-728, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29431277

RESUMEN

Mutations in the methyl-CpG-binding protein-2 gene (MECP2) are commonly associated with Rett syndrome. However, it has long been appreciated that there exists a spectrum of neuropsychiatric phenotypes associated with MECP2 variants. The most frequent Rett missense mutations are located in either the methyl-CpG-binding domain (MBD) or transcription repression domain (TRD). Clinical roles for mutations in other domains such as the intervening domain (ID) or AT-Hook domains have yet to be determined. Here, we report functional analysis of MECP2 missense mutations, located in AT-Hook1 within the ID, in a large Pakistani family with childhood onset cognitive decline and schizophrenia (SCZ), de novo in a girl with atypical Rett syndrome, and de novo in a woman with SCZ. We show that both p.Arg190His and p.Arg190Cys affect the ability of MeCP2 to bind to AT-rich DNA, also the brain-derived neurotrophic factor (BDNF) promoter, with the more drastic effects seen for p.Arg190Cys. Both mutations also affect nuclear chromatin clustering in vitro. These data support a possible molecular link between MECP2 AT-Hook1 mutations and psychosis. Given the ongoing large-scale whole exome and whole genome sequencing projects for psychiatric disorders, our findings suggest that rare missense variants in MECP2 be carefully evaluated for molecular consequences.


Asunto(s)
Secuencias AT-Hook , Cromatina/metabolismo , ADN/metabolismo , Discapacidad Intelectual/genética , Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/genética , Mutación/genética , Esquizofrenia/genética , Adulto , Animales , Secuencia de Bases , Línea Celular , Simulación por Computador , ADN/genética , Análisis Mutacional de ADN , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Linaje , Dominios Proteicos , Síndrome de Rett/genética
12.
Biochem Biophys Res Commun ; 495(1): 145-150, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29101034

RESUMEN

MeCP2 is a chromatin associated protein which is highly expressed in brain and relevant with Rett syndrome (RTT). There are AT-hook motifs in MeCP2 which can bind with AT-rich DNA, suggesting a role in chromatin binding. Here, we report the identification and characterization of another AT-rich DNA binding motif (residues 295 to 313) from the C-terminal transcription repression domain of MeCP2 by nuclear magnetic resonance (NMR) and isothermal calorimetry (ITC). This motif shows a micromolar affinity to AT-rich DNA, and it binds to the minor groove of DNA like AT-hook motifs. Together with the previous studies, our results provide an insight into a critical role of this motif in chromatin structure and function.


Asunto(s)
ADN/metabolismo , Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sitios de Unión , ADN/química , Humanos , Conformación de Ácido Nucleico , Unión Proteica , Dominios Proteicos , Síndrome de Rett/metabolismo
13.
Biochim Biophys Acta Mol Basis Dis ; 1864(2): 347-358, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29074463

RESUMEN

MECP2 is the causative gene for autism spectrum disorders, including Rett syndrome, a regressive neurodevelopmental rare disease mainly occurring in girls. Except for the distinct methyl-CpG binding domain and the transcriptional repression domain in MeCP2, three AT-hook-like domains have recently been identified. Several mutations in AT-hook 1 domain have been reported in autism cases or Rett database. However, the role of AT-hook 1 domain is still unclear. In this study, we generated a mouse line carrying deletion of eight conserved amino acids in AT-hook 1 domain by clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 technology. Mecp2ΔAT-hook1/y mutant male mice exhibited low locomotor activity, motor incoordination and cognitive deficit. In addition, these mutant mice exhibited increased anxiety. Moreover, pain insensitivity was noted in the mutant males. However, the social interactions were unaffected in AT-hook 1 mutant mice. Thinner CA1 region of the hippocampus was observed in the mutant mice. On the molecular basis, Western blot analysis showed increased expression of mutant MeCP2 protein in the cortex. Additionally, several genes expressed specifically in inhibitory neurons were markedly changed in the cerebrum. Taken together, these data demonstrate that disruption of AT-hook 1 domain in MeCP2 caused behavioral abnormality in mice, which suggests that AT-hook 1 is a critical region for the function of MeCP2 protein.


Asunto(s)
Conducta Animal , Cerebro/fisiología , Hipocampo/fisiología , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/fisiología , Animales , Peso Corporal , Mapeo Encefálico , Sistemas CRISPR-Cas , Eliminación de Gen , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Masculino , Aprendizaje por Laberinto , Proteína 2 de Unión a Metil-CpG/química , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Neuronas/fisiología , Percepción del Dolor , Unión Proteica , Dominios Proteicos
14.
Biochim Biophys Acta Gen Subj ; 1862(5): 1180-1189, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29428602

RESUMEN

Mutations in the X-linked MECP2 gene represent the main origin of Rett syndrome, causing a profound intellectual disability in females. MeCP2 is an epigenetic transcriptional regulator containing two main functional domains: a methyl-CpG binding domain (MBD) and a transcription repression domain (TRD). Over 600 pathogenic mutations were reported to affect the whole protein; almost half of missense mutations affect the MBD. Understanding the impact of these mutations on the MBD structure and interaction with DNA will foster the comprehension of their pathogenicity and possibly genotype/phenotype correlation studies. Herein, we use molecular dynamics simulations to obtain a detailed view of the dynamics of WT and mutated MBD in the presence and absence of DNA. The pathogenic mutation Y120D is used as paradigm for our studies. Further, since the Y120 residue was previously found to be a phosphorylation site, we characterize the dynamic profile of the MBD also in the presence of Y120 phosphorylation (pY120). We found that addition of a phosphate group to Y120 or mutation in aspartic acid affect domain mobility that samples an alternative conformational space with respect to the WT, leading to impaired ability to interact with DNA. Experimental assays showing a significant reduction in the binding affinity between the mutated MBD and the DNA confirmed our predictions.


Asunto(s)
ADN/química , Proteína 2 de Unión a Metil-CpG/química , Simulación de Dinámica Molecular , Mutación Missense , Síndrome de Rett , Sustitución de Aminoácidos , ADN/genética , ADN/metabolismo , Femenino , Humanos , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Dominios Proteicos
15.
J Biol Chem ; 291(10): 4873-81, 2016 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-26772194

RESUMEN

The epigenetic information encoded in the genomic DNA methylation pattern is translated by methylcytosine binding proteins like MeCP2 into chromatin topology and structure and gene activity states. We have shown previously that the MeCP2 level increases during differentiation and that it causes large-scale chromatin reorganization, which is disturbed by MeCP2 Rett syndrome mutations. Phosphorylation and other posttranslational modifications of MeCP2 have been described recently to modulate its function. Here we show poly(ADP-ribosyl)ation of endogenous MeCP2 in mouse brain tissue. Consequently, we found that MeCP2 induced aggregation of pericentric heterochromatin and that its chromatin accumulation was enhanced in poly(ADP-ribose) polymerase (PARP) 1(-/-) compared with wild-type cells. We mapped the poly(ADP-ribosyl)ation domains and engineered MeCP2 mutation constructs to further analyze potential effects on DNA binding affinity and large-scale chromatin remodeling. Single or double deletion of the poly(ADP-ribosyl)ated regions and PARP inhibition increased the heterochromatin clustering ability of MeCP2. Increased chromatin clustering may reflect increased binding affinity. In agreement with this hypothesis, we found that PARP-1 deficiency significantly increased the chromatin binding affinity of MeCP2 in vivo. These data provide novel mechanistic insights into the regulation of MeCP2-mediated, higher-order chromatin architecture and suggest therapeutic opportunities to manipulate MeCP2 function.


Asunto(s)
Cromatina/química , Proteína 2 de Unión a Metil-CpG/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Sitios de Unión , Encéfalo/metabolismo , Cromatina/metabolismo , Células HEK293 , Humanos , Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Mutación , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Unión Proteica , Ratas , Células Sf9 , Spodoptera
16.
Biochem Cell Biol ; 95(6): 593-608, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28796949

RESUMEN

This paper provides a brief introductory review of the most recent advances in our knowledge about the structural and functional aspects of two transcriptional regulators: MeCP2, a protein whose mutated forms are involved in Rett syndrome; and CTCF, a constitutive transcriptional insulator. This is followed by a description of the PTMs affecting these two proteins and an analysis of their known interacting partners. A special emphasis is placed on the recent studies connecting these two proteins, focusing on the still poorly understood potential structural and functional interactions between the two of them on the chromatin substrate. An overview is provided for some of the currently known genes that are dually regulated by these two proteins. Finally, a model is put forward to account for their possible involvement in their regulation of gene expression.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , Animales , Factor de Unión a CCCTC/química , Factor de Unión a CCCTC/genética , Silenciador del Gen , Humanos , Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/genética
17.
Biochemistry ; 55(31): 4275-85, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27420643

RESUMEN

Methyl-CpG binding protein 2 (MeCP2) is a multifunctional protein that guides neuronal development through its binding to DNA, recognition of sites of methyl-CpG (mCpG) DNA modification, and interaction with other regulatory proteins. Our study explores the relationship between mCpG and hydroxymethyl-CpG (hmCpG) recognition mediated by its mCpG binding domain (MBD) and binding cooperativity mediated by its C-terminal polypeptide. Previous study of the isolated MBD of MeCP2 documented an unusual mechanism by which ion uptake is required for discrimination of mCpG and hmCpG from CpG. MeCP2 binding cooperativity suppresses discrimination of modified DNA and is highly sensitive to both the total ion concentration and the type of counterions. Higher than physiological total ion concentrations completely suppress MeCP2 binding cooperativity, indicating a dominant electrostatic component to the interaction. Substitution of SO4(2-) for Cl(-) at physiological total ion concentrations also suppresses MeCP2 binding cooperativity, This effect is of particular note as the intracellular Cl(-) concentration changes during neuronal development. A related effect is that the protein-stabilizing solutes, TMAO and glutamate, reduce MeCP2 (but not isolated MBD) binding affinity by 2 orders of magnitude without affecting the apparent binding cooperativity. These observations suggest that polypeptide flexibility facilitates DNA binding by MeCP2. Consistent with this view, nuclear magnetic resonance (NMR) analyses show that ions have discrete effects on the structure of MeCP2, both MBD and the C-terminal domains. Notably, anion substitution results in changes in the NMR chemical shifts of residues, including some whose mutation causes the autism spectrum disorder Rett syndrome. Binding cooperativity makes MeCP2 an effective competitor with histone H1 for accessible DNA sites. The relationship between MeCP2 binding specificity and cooperativity is discussed in the context of chromatin binding, neuronal function, and neuronal development.


Asunto(s)
ADN/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , ADN/genética , Epigénesis Genética , Histonas/metabolismo , Humanos , Cinética , Dominio de Unión a CpG-Metil/genética , Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/genética , Neurogénesis , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Síndrome de Rett/genética , Síndrome de Rett/metabolismo
18.
Mol Cell ; 29(4): 525-31, 2008 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-18313390

RESUMEN

MeCP2 is an essential transcriptional repressor that mediates gene silencing through binding to methylated DNA. Binding specificity has been thought to depend on hydrophobic interactions between cytosine methyl groups and a hydrophobic patch within the methyl-CpG-binding domain (MBD). X-ray analysis of a methylated DNA-MBD cocrystal reveals, however, that the methyl groups make contact with a predominantly hydrophilic surface that includes tightly bound water molecules. This suggests that MeCP2 recognizes hydration of the major groove of methylated DNA rather than cytosine methylation per se. The MeCP2-DNA complex also identifies a unique structural role for T158, the residue most commonly mutated in Rett syndrome.


Asunto(s)
Islas de CpG , ADN/química , ADN/metabolismo , Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/metabolismo , Conformación de Ácido Nucleico , Conformación Proteica , Secuencia de Aminoácidos , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Cristalografía por Rayos X , ADN/genética , Metilación de ADN , Humanos , Sustancias Macromoleculares/química , Sustancias Macromoleculares/metabolismo , Proteína 2 de Unión a Metil-CpG/genética , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas , Unión Proteica , Síndrome de Rett/genética , Alineación de Secuencia , Timina/química , Timina/metabolismo
19.
Biochemistry ; 54(41): 6357-68, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26418480

RESUMEN

Rett syndrome causing missense mutations in the methyl-CpG-binding domain (MBD) of methyl CpG-binding protein 2 (MeCP2) were investigated both in silico and in vitro to reveal their effect on protein stability. It is demonstrated that the vast majority of frequently occurring mutations in the human population indeed alter the MBD folding free energy by a fraction of a kcal/mol up to more than 1 kcal/mol. While the absolute magnitude of the change of the free energy is small, the effect on the MBD functionality may be substantial since the folding free energy of MBD is about 2 kcal/mol only. Thus, it is emphasized that the effect of mutations on protein integrity should be evaluated with respect to the wild-type folding free energy but not with the absolute value of the folding free energy change. Furthermore, it was observed that the magnitude of the effect is correlated neither with the burial of the mutation sites nor with the basic amino acid physicochemical property change. Mutations that strongly perturb the immediate structural features were found to have little effect on folding free energy, while very conservative mutations resulted in large changes of the MBD stability. This observation was attributed to the protein's ability to structurally relax and reorganize to reduce the effect of mutation. Comparison between in silico and in vitro results indicated that some Web servers perform relatively well, while the free energy perturbation approach frequently overpredicts the magnitude of the free energy change especially when a charged amino acid is involved.


Asunto(s)
Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/genética , Mutación Puntual , Síndrome de Rett/genética , Islas de CpG , Metilación de ADN , Humanos , Proteína 2 de Unión a Metil-CpG/metabolismo , Modelos Moleculares , Unión Proteica , Pliegue de Proteína , Estabilidad Proteica , Estructura Terciaria de Proteína , Síndrome de Rett/metabolismo , Termodinámica
20.
Nucleic Acids Res ; 41(9): 4888-900, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23558747

RESUMEN

MeCP2 is a methyl-CpG-binding protein that is a main component of brain chromatin in vertebrates. In vitro studies have determined that in addition to its specific methyl-CpG-binding domain (MBD) MeCP2 also has several chromatin association domains. However, the specific interactions of MeCP2 with methylated or non-methylated chromatin regions and the structural characteristics of the resulting DNA associations in vivo remain poorly understood. We analysed the role of the MBD in MeCP2-chromatin associations in vivo using an MeCP2 mutant Rett syndrome mouse model (Mecp2(tm1.1Jae)) in which exon 3 deletion results in an N-terminal truncation of the protein, including most of the MBD. Our results show that in mutant mice, the truncated form of MeCP2 (ΔMeCP2) is expressed in different regions of the brain and liver, albeit at 50% of its wild-type (wt) counterpart. In contrast to the punctate nuclear distribution characteristic of wt MeCP2, ΔMeCP2 exhibits both diffuse nuclear localization and a substantial retention in the cytoplasm, suggesting a dysfunction of nuclear transport. In mutant brain tissue, neuronal nuclei are smaller, and ΔMeCP2 chromatin is digested faster by nucleases, producing a characteristic nuclease-resistant dinucleosome. Although a fraction of ΔMeCP2 is found associated with nucleosomes, its interaction with chromatin is transient and weak. Thus, our results unequivocally demonstrate that in vivo the MBD of MeCP2 together with its adjacent region in the N-terminal domain are critical for the proper interaction of the protein with chromatin, which cannot be replaced by any other of its protein domains.


Asunto(s)
Cromatina/metabolismo , Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/metabolismo , Animales , Encéfalo/metabolismo , Línea Celular Tumoral , Núcleo Celular/química , Núcleo Celular/ultraestructura , Exones , Hígado/metabolismo , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Transgénicos , Neuronas/química , Neuronas/ultraestructura , Nucleosomas/metabolismo , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA