Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
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
2.
Mol Neurobiol ; 56(7): 4838-4854, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30402709

RESUMEN

MeCP2 is a fundamental protein associated with several neurological disorders, including Rett syndrome. It is considered a multifunctional factor with a prominent role in regulating chromatin structure; however, a full comprehension of the consequences of its deficiency is still lacking. Here, we characterize a novel mouse model of Mecp2 bearing the human mutation Y120D, which is localized in the methyl-binding domain. As most models of Mecp2, the Mecp2Y120D mouse develops a severe Rett-like phenotype. This mutation alters the interaction of the protein with chromatin, but surprisingly, it also impairs its association with corepressors independently on the involved interacting domains. These features, which become overt mainly in the mature brain, cause a more accessible and transcriptionally active chromatin structure; conversely, in the Mecp2-null brain, we find a less accessible and transcriptionally inactive chromatin. By demonstrating that different MECP2 mutations can produce concordant neurological phenotypes but discordant molecular features, we highlight the importance of considering personalized approaches for the treatment of Rett syndrome.


Asunto(s)
Conducta Animal , Técnicas de Sustitución del Gen , Proteína 2 de Unión a Metil-CpG/metabolismo , Medicina de Precisión , Animales , Encéfalo/metabolismo , Encéfalo/patología , Cromatina/metabolismo , Femenino , Humanos , Longevidad , Masculino , Memoria a Corto Plazo , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Mutación/genética , Neuronas/metabolismo , Fenotipo , Síndrome de Rett
3.
Sci Rep ; 6: 28295, 2016 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-27323888

RESUMEN

MeCP2 is a transcriptional regulator whose functional alterations are responsible for several autism spectrum and mental disorders. Post-translational modifications (PTMs), and particularly differential phosphorylation, modulate MeCP2 function in response to diverse stimuli. Understanding the detailed role of MeCP2 phosphorylation is thus instrumental to ascertain how MeCP2 integrates the environmental signals and directs its adaptive transcriptional responses. The evolutionarily conserved serine 164 (S164) was found phosphorylated in rodent brain but its functional role has remained uncharacterized. We show here that phosphorylation of S164 in brain is dynamically regulated during neuronal maturation. S164 phosphorylation highly impairs MeCP2 binding to DNA in vitro and largely affects its nucleosome binding and chromatin affinity in vivo. Strikingly, the chromatin-binding properties of the global MeCP2 appear also extensively altered during the course of brain maturation. Functional assays reveal that proper temporal regulation of S164 phosphorylation controls the ability of MeCP2 to regulate neuronal morphology. Altogether, our results support the hypothesis of a complex PTM-mediated functional regulation of MeCP2 potentially involving a still poorly characterized epigenetic code. Furthermore, they demonstrate the relevance of the Intervening Domain of MeCP2 for binding to DNA.


Asunto(s)
Encéfalo/metabolismo , Cromatina/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , Animales , Metilación de ADN , Dendritas/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Células 3T3 NIH , Nucleosomas/metabolismo , Fosforilación , Unión Proteica , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Conejos , Serina/metabolismo
4.
PLoS One ; 11(4): e0153473, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27097329

RESUMEN

MECP2 mutations cause a number of neurological disorders of which Rett syndrome (RTT) represents the most thoroughly analysed condition. Many Mecp2 mouse models have been generated through the years; their validity is demonstrated by the presence of a broad spectrum of phenotypes largely mimicking those manifested by RTT patients. These mouse models, between which the C57BL/6 Mecp2tm1.1Bird strain probably represents the most used, enabled to disclose much of the roles of Mecp2. However, small litters with little viability and poor maternal care hamper the maintenance of the colony, thus limiting research on such animals. For this reason, past studies often used Mecp2 mouse models on mixed genetic backgrounds, thus opening questions on whether modifier genes could be responsible for at least part of the described effects. To verify this possibility, and facilitate the maintenance of the Mecp2 colony, we transferred the Mecp2tm1.1Bird allele on the stronger CD1 background. The CD1 strain is easier to maintain and largely recapitulates the phenotypes already described in Mecp2-null mice. We believe that this mouse model will foster the research on RTT.


Asunto(s)
Antecedentes Genéticos , Proteína 2 de Unión a Metil-CpG/deficiencia , Proteína 2 de Unión a Metil-CpG/genética , Modelos Animales , Alelos , Animales , Conducta Animal , Femenino , Eliminación de Gen , Masculino , Ratones , Neuronas/citología , Fenotipo , Especificidad de la Especie
6.
PLoS One ; 10(6): e0130183, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26098633

RESUMEN

Rett syndrome (RTT) is an autism spectrum disorder mainly caused by mutations in the X-linked MECP2 gene and affecting roughly 1 out of 10.000 born girls. Symptoms range in severity and include stereotypical movement, lack of spoken language, seizures, ataxia and severe intellectual disability. Notably, muscle tone is generally abnormal in RTT girls and women and the Mecp2-null mouse model constitutively reflects this disease feature. We hypothesized that MeCP2 in muscle might physiologically contribute to its development and/or homeostasis, and conversely its defects in RTT might alter the tissue integrity or function. We show here that a disorganized architecture, with hypotrophic fibres and tissue fibrosis, characterizes skeletal muscles retrieved from Mecp2-null mice. Alterations of the IGF-1/Akt/mTOR pathway accompany the muscle phenotype. A conditional mouse model selectively depleted of Mecp2 in skeletal muscles is characterized by healthy muscles that are morphologically and molecularly indistinguishable from those of wild-type mice raising the possibility that hypotonia in RTT is mainly, if not exclusively, mediated by non-cell autonomous effects. Our results suggest that defects in paracrine/endocrine signaling and, in particular, in the GH/IGF axis appear as the major cause of the observed muscular defects. Remarkably, this is the first study describing the selective deletion of Mecp2 outside the brain. Similar future studies will permit to unambiguously define the direct impact of MeCP2 on tissue dysfunctions.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Proteína 2 de Unión a Metil-CpG/genética , Hipotonía Muscular/patología , Atrofia Muscular/patología , Animales , Modelos Animales de Enfermedad , Femenino , Fibrosis/genética , Fibrosis/patología , Hormona del Crecimiento/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Hipotonía Muscular/genética , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/patología , Atrofia Muscular/genética , Comunicación Paracrina/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Síndrome de Rett/genética , Síndrome de Rett/patología , Serina-Treonina Quinasas TOR/metabolismo
7.
Neurosci Biobehav Rev ; 46 Pt 2: 187-201, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24594195

RESUMEN

Rett syndrome (RTT) is a devastating genetic disorder that worldwide represents the most common genetic cause of severe intellectual disability in females. Most cases are caused by mutations in the X-linked MECP2 gene. Several recent studies have demonstrated that RTT mimicking animal models do not develop an irreversible condition and phenotypic rescue is possible. However, no cure for RTT has been identified so far, and patients are only given symptomatic and supportive treatments. The development of clinical applications imposes a more comprehensive knowledge of MeCP2 functional role(s) and their relevance for RTT pathobiology. Herein, we thoroughly survey the knowledge about MeCP2 structure and functions, highlighting the necessity of identifying more functional domains and the value of molecular genetics. Given that, in our opinion, RTT ultimately is generated by perturbations in gene transcription and so far no genes/pathways have been consistently linked to a dysfunctional MeCP2, we have used higher-level bioinformatic analyses to identify commonly deregulated mechanisms in MeCP2-defective samples. In this review we present our results and discuss the possible value of the utilized approach.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Transducción de Señal/genética , Animales , Biología Computacional , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA