Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cell Rep ; 24(9): 2213-2220, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30157418

ABSTRACT

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.


Subject(s)
DNA/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Nuclear Localization Signals/metabolism , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , CpG Islands , DNA/genetics , Disease Models, Animal , Male , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Rett Syndrome/metabolism , alpha Karyopherins/metabolism
2.
FEBS Lett ; 590(17): 2927-33, 2016 09.
Article in English | MEDLINE | ID: mdl-27461740

ABSTRACT

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.


Subject(s)
AT-Hook Motifs/genetics , DNA Methylation/genetics , Methyl-CpG-Binding Protein 2/genetics , Rett Syndrome/genetics , Base Sequence/genetics , Chromatin/genetics , Cytosine/metabolism , DNA-Binding Proteins/genetics , Humans , Mutation, Missense/genetics , Rett Syndrome/pathology
3.
Hum Mol Genet ; 25(3): 558-70, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26647311

ABSTRACT

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.


Subject(s)
Alleles , Methyl-CpG-Binding Protein 2/genetics , Mutation, Missense , Rett Syndrome/genetics , Rett Syndrome/pathology , Amino Acid Substitution , Animals , DNA/genetics , DNA/metabolism , DNA Methylation , Disease Models, Animal , Gene Expression Regulation , Gene Knock-In Techniques , Humans , Male , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mice, Transgenic , Models, Molecular , Phenotype , Protein Binding , Rett Syndrome/metabolism , Rett Syndrome/mortality , Severity of Illness Index , Signal Transduction , Survival Analysis
4.
Nucleic Acids Res ; 43(2): 732-44, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25572316

ABSTRACT

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.


Subject(s)
Brain/metabolism , DNA Methylation , Base Sequence , Cerebellum/metabolism , CpG Islands , DNA/chemistry , Humans
5.
Elife ; 3: e03397, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25259796

ABSTRACT

The mammalian genome is punctuated by CpG islands (CGIs), which differ sharply from the bulk genome by being rich in G + C and the dinucleotide CpG. CGIs often include transcription initiation sites and display 'active' histone marks, notably histone H3 lysine 4 methylation. In embryonic stem cells (ESCs) some CGIs adopt a 'bivalent' chromatin state bearing simultaneous 'active' and 'inactive' chromatin marks. To determine whether CGI chromatin is developmentally programmed at specific genes or is imposed by shared features of CGI DNA, we integrated artificial CGI-like DNA sequences into the ESC genome. We found that bivalency is the default chromatin structure for CpG-rich, G + C-rich DNA. A high CpG density alone is not sufficient for this effect, as A + T-rich sequence settings invariably provoke de novo DNA methylation leading to loss of CGI signature chromatin. We conclude that both CpG-richness and G + C-richness are required for induction of signature chromatin structures at CGIs.


Subject(s)
Chromatin/chemistry , CpG Islands/genetics , AT Rich Sequence/genetics , Animals , Base Composition/genetics , Base Sequence , Cell Line , DNA Methylation , Embryonic Stem Cells/metabolism , Histones/metabolism , Lysine/metabolism , Mice , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Trans-Activators/metabolism
6.
Nat Neurosci ; 16(7): 898-902, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23770565

ABSTRACT

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.


Subject(s)
Methyl-CpG-Binding Protein 2/genetics , Mutation/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Co-Repressor 2/metabolism , Rett Syndrome/genetics , Animals , Brain/metabolism , Brain/pathology , Cells, Cultured , Disease Models, Animal , Exploratory Behavior/physiology , Green Fluorescent Proteins/genetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Molecular , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 2/genetics , Rett Syndrome/pathology , Rett Syndrome/physiopathology
7.
Hum Mol Genet ; 21(17): 3806-14, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22653753

ABSTRACT

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.


Subject(s)
Aging/genetics , Gene Silencing , Methyl-CpG-Binding Protein 2/genetics , Aging/drug effects , Animals , Animals, Newborn , Gene Expression Regulation, Developmental/drug effects , Gene Silencing/drug effects , Genetic Loci/genetics , Learning/drug effects , Male , Methyl-CpG-Binding Protein 2/metabolism , Mice , Motor Activity/drug effects , Motor Activity/genetics , Phenotype , Recombination, Genetic/genetics , Rett Syndrome/genetics , Rett Syndrome/physiopathology , Survival Analysis , Tamoxifen/pharmacology
8.
Brain ; 135(Pt 9): 2699-710, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22525157

ABSTRACT

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.


Subject(s)
Behavior, Animal/physiology , Cerebral Cortex/pathology , Methyl-CpG-Binding Protein 2/genetics , Neurons/pathology , Phenotype , Rett Syndrome/genetics , Animals , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Disease Models, Animal , Gene Silencing , Hand Strength/physiology , Humans , Methyl-CpG-Binding Protein 2/metabolism , Mice , Neurons/metabolism , Rett Syndrome/metabolism , Rett Syndrome/pathology , Rett Syndrome/physiopathology , Rotarod Performance Test
9.
Genome Res ; 21(1): 83-94, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21149390

ABSTRACT

LSH, a member of the SNF2 family of chromatin remodeling ATPases encoded by the Hells gene, is essential for normal levels of DNA methylation in the mammalian genome. While the role of LSH in the methylation of repetitive DNA sequences is well characterized, its contribution to the regulation of DNA methylation and the expression of protein-coding genes has not been studied in detail. In this report we investigate genome-wide patterns of DNA methylation at gene promoters in Hells(-/-) mouse embryonic fibroblasts (MEFs). We find that in the absence of LSH, DNA methylation is lost or significantly reduced at ∼20% of all normally methylated promoter sequences. As a consequence, a large number of genes are misexpressed in Hells(-/-) MEFs. Comparison of Hells(-/-) MEFs with wild-type MEFs and embryonic stem (ES) cells suggests that LSH is important for de novo DNA methylation events that accompany the establishment and differentiation of embryonic lineage cells. We further show that the generation of normal DNA methylation patterns and stable gene silencing at specific promoters require cooperation between LSH and the G9a/GLP complex of histone methylases. At such loci, G9a recruitment is compromised when LSH is absent or greatly reduced. Taken together, our data suggest a mechanism whereby LSH promotes binding of DNA methyltransferases and the G9a/GLP complex to specific loci and facilitates developmentally programmed DNA methylation and stable gene silencing during lineage commitment and differentiation.


Subject(s)
DNA Helicases/metabolism , DNA Methylation , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/metabolism , Animals , Cell Differentiation , Cells, Cultured , DNA Helicases/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblasts/metabolism , Gene Silencing , Histone-Lysine N-Methyltransferase/genetics , Methyltransferases/genetics , Methyltransferases/metabolism , Mice
10.
Nucleic Acids Res ; 38(14): 4620-34, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20378711

ABSTRACT

MBD1, a member of the methyl-CpG-binding domain family of proteins, has been reported to repress transcription of methylated and unmethylated promoters. As some MBD1 isoforms contain two DNA-binding domains-an MBD, which recognizes methylated DNA; and a CXXC3 zinc finger, which binds unmethylated CpG-it is unclear whether these two domains function independently of each other or if they cooperate in facilitating recruitment of MBD1 to particular genomic loci. In this report we investigate DNA-binding specificity of MBD and CXXC3 domains in vitro and in vivo. We find that the methyl-CpG-binding domain of MBD1 binds more efficiently to methylated DNA within a specific sequence context. We identify genes that are targeted by MBD1 in human cells and demonstrate that a functional MBD domain is necessary and sufficient for recruitment of MBD1 to specific sites at these loci, while DNA binding by the CXXC3 motif is largely dispensable. In summary, the binding preferences of MBD1, although dependent upon the presence of methylated DNA, are clearly distinct from those of other methyl-CpG-binding proteins, MBD2 and MeCP2.


Subject(s)
DNA Methylation , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Cell Line , CpG Islands , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Silencing , HeLa Cells , Herpes Simplex Virus Protein Vmw65/metabolism , Humans , Point Mutation , Promoter Regions, Genetic , Protein Binding , Protein Interaction Domains and Motifs , Receptors, Nerve Growth Factor/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , rho GTP-Binding Proteins/genetics
11.
PLoS One ; 5(4): e9937, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20376339

ABSTRACT

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.


Subject(s)
Cell Differentiation/genetics , DNA Methylation , Embryonic Stem Cells/cytology , Octamer Transcription Factor-3/physiology , Regulatory Sequences, Nucleic Acid/genetics , Animals , Cell Line , CpG Islands , DNA (Cytosine-5-)-Methyltransferases/metabolism , Mice , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...