Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Mol Cell ; 81(6): 1260-1275.e12, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33561390

RESUMEN

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.


Asunto(s)
Metilación de ADN , Proteína 2 de Unión a Metil-CpG/metabolismo , Neuronas/metabolismo , Animales , Islas de CpG , Técnicas de Sustitución del Gen , Células HeLa , Humanos , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Transgénicos , Mutación , Células 3T3 NIH , Neuronas/patología , Dominios Proteicos , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Síndrome de Rett/patología
2.
Genes Dev ; 32(23-24): 1514-1524, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30463906

RESUMEN

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.


Asunto(s)
Expresión Génica , Histona Desacetilasas/metabolismo , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/toxicidad , Animales , Proteínas Co-Represoras/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática/genética , Técnicas de Inactivación de Genes , Histona Desacetilasas/genética , Masculino , Discapacidad Intelectual Ligada al Cromosoma X/genética , Discapacidad Intelectual Ligada al Cromosoma X/fisiopatología , Ratones , Mutación , Enfermedades del Sistema Nervioso/genética , Neuroglía/metabolismo , Neuronas/metabolismo , Co-Represor 1 de Receptor Nuclear/metabolismo , Co-Represor 2 de Receptor Nuclear/metabolismo , Dominios Proteicos , Proteínas tau/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.
Hum Mol Genet ; 25(3): 558-70, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26647311

RESUMEN

Rett syndrome is caused by mutations in the X-linked MECP2 gene, which encodes a chromosomal protein that binds to methylated DNA. Mouse models mirror the human disorder and therefore allow investigation of phenotypes at a molecular level. We describe an Mecp2 allelic series representing the three most common missense Rett syndrome (RTT) mutations, including first reports of Mecp2[R133C] and Mecp2[T158M] knock-in mice, in addition to Mecp2[R306C] mutant mice. Together these three alleles comprise ∼25% of all RTT mutations in humans, but they vary significantly in average severity. This spectrum is mimicked in the mouse models; R133C being least severe, T158M most severe and R306C of intermediate severity. Both R133C and T158M mutations cause compound phenotypes at the molecular level, combining compromised DNA binding with reduced stability, the destabilizing effect of T158M being more severe. Our findings contradict the hypothesis that the R133C mutation exclusively abolishes binding to hydroxymethylated DNA, as interactions with DNA containing methyl-CG, methyl-CA and hydroxymethyl-CA are all reduced in vivo. We find that MeCP2[T158M] is significantly less stable than MeCP2[R133C], which may account for the divergent clinical impact of the mutations. Overall, this allelic series recapitulates human RTT severity, reveals compound molecular aetiologies and provides a valuable resource in the search for personalized therapeutic interventions.


Asunto(s)
Alelos , Proteína 2 de Unión a Metil-CpG/genética , Mutación Missense , Síndrome de Rett/genética , Síndrome de Rett/patología , Sustitución de Aminoácidos , Animales , ADN/genética , ADN/metabolismo , Metilación de ADN , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Humanos , Masculino , Proteína 2 de Unión a Metil-CpG/metabolismo , Ratones , Ratones Transgénicos , Modelos Moleculares , Fenotipo , Unión Proteica , Síndrome de Rett/metabolismo , Síndrome de Rett/mortalidad , Índice de Severidad de la Enfermedad , Transducción de Señal , Análisis de Supervivencia
5.
Nucleic Acids Res ; 43(2): 732-44, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25572316

RESUMEN

The possibility that alterations in DNA methylation are mechanistic drivers of development, aging and susceptibility to disease is widely acknowledged, but evidence remains patchy or inconclusive. Of particular interest in this regard is the brain, where it has been reported that DNA methylation impacts on neuronal activity, learning and memory, drug addiction and neurodegeneration. Until recently, however, little was known about the 'landscape' of the human brain methylome. Here we assay 1.9 million CpGs in each of 43 brain samples representing different individuals and brain regions. The cerebellum was a consistent outlier compared to all other regions, and showed over 16 000 differentially methylated regions (DMRs). Unexpectedly, the sequence characteristics of hypo- and hypermethylated domains in cerebellum were distinct. In contrast, very few DMRs distinguished regions of the cortex, limbic system and brain stem. Inter-individual DMRs were readily detectable in these regions. These results lead to the surprising conclusion that, with the exception of cerebellum, DNA methylation patterns are more homogeneous between different brain regions from the same individual, than they are for a single brain region between different individuals. This finding suggests that DNA sequence composition, not developmental status, is the principal determinant of the human brain DNA methylome.


Asunto(s)
Encéfalo/metabolismo , Metilación de ADN , Secuencia de Bases , Cerebelo/metabolismo , Islas de CpG , ADN/química , Humanos
6.
Eur J Immunol ; 44(6): 1835-41, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24578067

RESUMEN

Th1 and Th2 cell fates are traditionally viewed as mutually exclusive, but recent work suggests that these lineages may be more plastic than previously thought. When isolating splenic CD4(+) T cells from mice infected with the parasitic helminth Schistosoma mansoni, we observed a defined population of IFN-γ/IL-4 double-positive cells. These IFN-γ(+) IL-4(+) cells showed differences in DNA methylation at the Ifng and Il4 loci when compared with IFN-γ(+) IL-4(-) (Th1) and IFN-γ(-) IL-4(+) (Th2) cells, demonstrating that they represent a distinct effector cell population. IFN-γ(+) IL-4(+) cells also displayed a discrete DNA methylation pattern at a CpG island within the body of the Gata3 gene, which encodes the master regulator of Th2 identity. DNA methylation at this region correlated with decreased Gata3 levels, suggesting a possible role in controlling Gata3 expression. These data provide important insight into the molecular mechanisms behind the co-existence of Th1 and Th2 characteristics.


Asunto(s)
Metilación de ADN/inmunología , Interferón gamma/inmunología , Interleucina-4/inmunología , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/inmunología , Células TH1/inmunología , Células Th2/inmunología , Animales , Islas de CpG/inmunología , Femenino , Factor de Transcripción GATA3/inmunología , Regulación de la Expresión Génica/inmunología , Ratones , Esquistosomiasis mansoni/patología , Células TH1/patología , Células Th2/patología
7.
Hum Mol Genet ; 21(17): 3806-14, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22653753

RESUMEN

Rett Syndrome is a neurological disorder caused by mutations in the X-linked MECP2 gene. Mouse models where Mecp2 is inactivated or mutated recapitulate several features of the disorder and have demonstrated a requirement for the protein to ensure brain function in adult mice. We deleted the Mecp2 gene in ~80% of brain cells at three postnatal ages to determine whether the need for MeCP2 varies with age. Inactivation at all three time points induced Rett-like phenotypes and caused premature death of the animals. We find two threshold ages beyond which the requirement for MeCP2 markedly increases in stringency. The earlier threshold (8-14 weeks), when inactivated mice develop symptoms, represents early adulthood in the mouse and coincides with the period when Mecp2-null mice exhibit terminal symptoms. Unexpectedly, we identified a later age threshold (30-45 weeks) beyond which an 80% reduction in MeCP2 is incompatible with life. This finding suggests an enhanced role for MeCP2 in the aging brain.


Asunto(s)
Envejecimiento/genética , Silenciador del Gen , Proteína 2 de Unión a Metil-CpG/genética , Envejecimiento/efectos de los fármacos , Animales , Animales Recién Nacidos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Sitios Genéticos/genética , Aprendizaje/efectos de los fármacos , Masculino , Proteína 2 de Unión a Metil-CpG/metabolismo , Ratones , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Fenotipo , Recombinación Genética/genética , Síndrome de Rett/genética , Síndrome de Rett/fisiopatología , Análisis de Supervivencia , Tamoxifeno/farmacología
8.
Digit Biomark ; 8(1): 30-39, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38510264

RESUMEN

Background: Fatigue is a prominent symptom in many diseases and is strongly associated with impaired daily function. The measurement of daily function is currently almost always done with questionnaires, which are subjective and imprecise. With the recent advances of digital wearable technologies, novel approaches to evaluate daily function quantitatively and objectively in real-life conditions are increasingly possible. This also creates new possibilities to measure fatigue-related changes of daily function using such technologies. Summary: This review examines which digitally assessable parameters in immune-mediated inflammatory and neurodegenerative diseases may have the greatest potential to reflect fatigue-related changes of daily function. Key Messages: Results of a standardized analysis of the literature reporting about perception-, capacity-, and performance-evaluating assessment tools indicate that changes of the following parameters: physical activity, independence of daily living, social participation, working life, mental status, cognitive and aerobic capacity, and supervised and unsupervised mobility performance have the highest potential to reflect fatigue-related changes of daily function. These parameters thus hold the greatest potential for quantitatively measuring fatigue in representative diseases in real-life conditions, e.g., with digital wearable technologies. Furthermore, to the best of our knowledge, this is a new approach to analysing evidence for the design of performance-based digital assessment protocols in human research, which may stimulate further systematic research in this area.

9.
Brain ; 135(Pt 9): 2699-710, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22525157

RESUMEN

Rett syndrome is a neurological disorder caused by mutation of the X-linked MECP2 gene. Mice lacking functional Mecp2 display a spectrum of Rett syndrome-like signs, including disturbances in motor function and abnormal patterns of breathing, accompanied by structural defects in central motor areas and the brainstem. Although routinely classified as a neurodevelopmental disorder, many aspects of the mouse phenotype can be effectively reversed by activation of a quiescent Mecp2 gene in adults. This suggests that absence of Mecp2 during brain development does not irreversibly compromise brain function. It is conceivable, however, that deep-seated neurological defects persist in mice rescued by late activation of Mecp2. To test this possibility, we have quantitatively analysed structural and functional plasticity of the rescued adult male mouse brain. Activation of Mecp2 in ∼70% of neurons reversed many morphological defects in the motor cortex, including neuronal size and dendritic complexity. Restoration of Mecp2 expression was also accompanied by a significant improvement in respiratory and sensory-motor functions, including breathing pattern, grip strength, balance beam and rotarod performance. Our findings sustain the view that MeCP2 does not play a pivotal role in brain development, but may instead be required to maintain full neurological function once development is complete.


Asunto(s)
Conducta Animal/fisiología , Corteza Cerebral/patología , Proteína 2 de Unión a Metil-CpG/genética , Neuronas/patología , Fenotipo , Síndrome de Rett/genética , Animales , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Silenciador del Gen , Fuerza de la Mano/fisiología , Humanos , Proteína 2 de Unión a Metil-CpG/metabolismo , Ratones , Neuronas/metabolismo , Síndrome de Rett/metabolismo , Síndrome de Rett/patología , Síndrome de Rett/fisiopatología , Prueba de Desempeño de Rotación con Aceleración Constante
10.
J Pers Med ; 11(8)2021 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-34442459

RESUMEN

There has been great progress in Huntington's disease (HD) research. Yet, effective treatments to halt disease before the onset of disabling symptoms are still unavailable. Scientific breakthroughs require an active and lasting commitment from families. However, they are traditionally less involved and heard in studies. Accordingly, the European Huntington Association (EHA) surveyed individuals at risk (HDRisk) and with premanifest HD (PreHD) to determine which factors affect their willingness to participate in research. Questions assessed research experience and knowledge, information sources, reasons for involvement and noninvolvement, and factors preventing and facilitating participation. The survey included 525 individuals, of which 68.8% never participated in studies and 38.6% reported limited research knowledge. Furthermore, 52% trusted patient organizations to get research information. Reasons for involvement were altruistic and more important than reasons for noninvolvement, which were related to negative emotions. Obstacles included time/financial constraints and invasive procedures, while professional support was seen as a facilitator. PreHD individuals reported less obstacles to research participation than HDRisk individuals. Overall, a high motivation to participate in research was noted, despite limited experience and literacy. This motivation is influenced by subjective and objective factors and, importantly, by HD status. Patient organizations have a key role in fostering motivation through education and support.

11.
Epigenomes ; 3(1): 7, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31354981

RESUMEN

Most human genes are associated with promoters embedded in non-methylated, G + C-rich CpG islands (CGIs). Not all CGIs are found at annotated promoters, however, raising the possibility that many serve as promoters for transcripts that do not code for proteins. To test this hypothesis, we searched for novel transcripts in embryonic stem cells (ESCs) that originate within orphan CGIs. Among several candidates, we detected a transcript that included three members of the let-7 micro-RNA family: Let-7a-1, let-7f-1, and let-7d. Deletion of the CGI prevented expression of the precursor RNA and depleted the included miRNAs. Mice homozygous for this mutation were sub-viable and showed growth and other defects. The results suggest that despite the identity of their seed sequences, members of the let-7 miRNA family exert distinct functions that cannot be complemented by other members.

12.
Cell Rep ; 24(9): 2213-2220, 2018 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-30157418

RESUMEN

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.


Asunto(s)
ADN/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , Señales de Localización Nuclear/metabolismo , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , Islas de CpG , ADN/genética , Modelos Animales de Enfermedad , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Síndrome de Rett/metabolismo , alfa Carioferinas/metabolismo
13.
PLoS One ; 5(4): e9937, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20376339

RESUMEN

Differentiation of embryonic stem (ES) cells is accompanied by silencing of the Oct-4 gene and de novo DNA methylation of its regulatory region. Previous studies have focused on the requirements for promoter region methylation. We therefore undertook to analyse the progression of DNA methylation of the approximately 2000 base pair regulatory region of Oct-4 in ES cells that are wildtype or deficient for key proteins. We find that de novo methylation is initially seeded at two discrete sites, the proximal enhancer and distal promoter, spreading later to neighboring regions, including the remainder of the promoter. De novo methyltransferases Dnmt3a and Dnmt3b cooperate in the initial targeted stage of de novo methylation. Efficient completion of the pattern requires Dnmt3a and Dnmt1, but not Dnmt3b. Methylation of the Oct-4 promoter depends on the histone H3 lysine 9 methyltransferase G9a, as shown previously, but CpG methylation throughout most of the regulatory region accumulates even in the absence of G9a. Analysis of the Oct-4 regulatory domain as a whole has allowed us to detect targeted de novo methylation and to refine our understanding the roles of key protein components in this process.


Asunto(s)
Diferenciación Celular/genética , Metilación de ADN , Células Madre Embrionarias/citología , Factor 3 de Transcripción de Unión a Octámeros/fisiología , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Línea Celular , Islas de CpG , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Ratones , Regiones Promotoras Genéticas
14.
Genome Res ; 17(5): 625-31, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17420183

RESUMEN

DNA is methylated at the dinucleotide CpG in genomes of a wide range of plants and animals. Among animals, variable patterns of genomic CpG methylation have been described, ranging from undetectable levels (e.g., in Caenorhabditis elegans) to high levels of global methylation in the vertebrates. The most frequent pattern in invertebrate animals, however, is mosaic methylation, comprising domains of methylated DNA interspersed with unmethylated domains. To understand the origin of mosaic DNA methylation patterns, we examined the distribution of DNA methylation in the Ciona intestinalis genome. Bisulfite sequencing and computational analysis revealed methylated domains with sharp boundaries that strongly colocalize with approximately 60% of transcription units. By contrast, promoters, intergenic DNA, and transposons are not preferentially targeted by DNA methylation. Methylated transcription units include evolutionarily conserved genes, whereas the most highly expressed genes preferentially belong to the unmethylated fraction. The results lend support to the hypothesis that CpG methylation functions to suppress spurious transcriptional initiation within infrequently transcribed genes.


Asunto(s)
Ciona intestinalis/genética , Islas de CpG/genética , Metilación de ADN , ADN/metabolismo , Transcripción Genética , Animales , ADN/genética , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA