Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nucleic Acids Res ; 44(16): 7592-604, 2016 09 19.
Article in English | MEDLINE | ID: mdl-27179028

ABSTRACT

Methylation of DNA at carbon 5 of cytosine is essential for mammalian development and implicated in transcriptional repression of genes and transposons. New patterns of DNA methylation characteristic of lineage-committed cells are established at the exit from pluripotency by de novo DNA methyltransferases enzymes, DNMT3A and DNMT3B, which are regulated by developmental signaling and require access to chromatin-organized DNA. Whether or not the capacity for de novo DNA methylation of developmentally regulated loci is preserved in differentiated somatic cells and can occur in the absence of exogenous signals is currently unknown. Here, we demonstrate that fibroblasts derived from chromatin remodeling ATPase LSH (HELLS)-null mouse embryos, which lack DNA methylation from centromeric repeats, transposons and a number of gene promoters, are capable of reestablishing DNA methylation and silencing of misregulated genes upon re-expression of LSH. We also show that the ability of LSH to bind ATP and the cellular concentration of DNMT3B are critical for cell-autonomous de novo DNA methylation in somatic cells. These data suggest the existence of cellular memory that persists in differentiated cells through many cell generations and changes in transcriptional state.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA Helicases/metabolism , DNA Methylation , Fibroblasts/metabolism , 5-Methylcytosine/metabolism , Animals , Cell Differentiation/genetics , Cell Line , DNA (Cytosine-5-)-Methyltransferases/metabolism , Embryo, Mammalian/cytology , Fibroblasts/cytology , Gene Expression Regulation, Developmental , Gene Silencing , Mice , Mutation/genetics , NIH 3T3 Cells , Promoter Regions, Genetic/genetics , Repetitive Sequences, Nucleic Acid/genetics , Retroelements/genetics , DNA Methyltransferase 3B
2.
Cell Rep ; 15(1): 77-85, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27052169

ABSTRACT

DNA methylation at imprinting control regions (ICRs) is established in gametes in a sex-specific manner and has to be stably maintained during development and in somatic cells to ensure the correct monoallelic expression of imprinted genes. In addition to DNA methylation, the ICRs are marked by allele-specific histone modifications. Whether these marks are essential for maintenance of genomic imprinting is largely unclear. Here, we show that the histone H3 lysine 9 methylases G9a and GLP are required for stable maintenance of imprinted DNA methylation in embryonic stem cells; however, their catalytic activity and the G9a/GLP-dependent H3K9me2 mark are completely dispensable for imprinting maintenance despite the genome-wide loss of non-imprinted DNA methylation in H3K9me2-depleted cells. We provide additional evidence that the G9a/GLP complex protects imprinted DNA methylation by recruitment of de novo DNA methyltransferases, which antagonize TET dioxygenass-dependent erosion of DNA methylation at ICRs.


Subject(s)
DNA Methylation , Embryonic Stem Cells/metabolism , Genomic Imprinting , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Cell Line , Histocompatibility Antigens/genetics , Histone-Lysine N-Methyltransferase/genetics , Humans
3.
J Cell Sci ; 125(Pt 22): 5524-34, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22946062

ABSTRACT

LSH, a protein related to the SNF2 family of chromatin-remodelling ATPases, is essential for the correct establishment of DNA methylation levels and patterns in plants and mammalian cells. However, some of the phenotypes resulting from LSH deficiency cannot be explained easily by defects in DNA methylation. Here we show that LSH-deficient mouse and human fibroblasts show reduced viability after exposure to ionizing radiation and repair DNA double-strand breaks less efficiently than wild-type cells. A more detailed characterisation of this phenotype revealed that, in the absence of LSH, the histone variant H2AX is not efficiently phosphorylated in response to DNA damage. This results in impaired recruitment of MDC1 and 53BP1 proteins to DNA double-strand breaks and compromises phosphorylation of checkpoint kinase CHK2. Furthermore, we demonstrate that the ability of LSH to hydrolyse ATP is necessary for efficient phosphorylation of H2AX at DNA double-strand breaks and successful repair of DNA damage. Taken together, our data reveal a previously unsuspected role of LSH ATPase in the maintenance of genome stability in mammalian somatic cells, which is independent of its function in de novo DNA methylation during development.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA Breaks, Double-Stranded , DNA Helicases/metabolism , DNA Repair , Histones/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cell Cycle Proteins , Checkpoint Kinase 2 , DNA Breaks, Double-Stranded/radiation effects , DNA Helicases/deficiency , DNA Methylation/radiation effects , DNA Repair/radiation effects , Enzyme Activation/radiation effects , Fibroblasts/metabolism , Fibroblasts/radiation effects , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kinetics , Mammals/metabolism , Mice , Mutant Proteins/metabolism , Phosphorylation/radiation effects , Protein Serine-Threonine Kinases/metabolism , Radiation, Ionizing , Signal Transduction/radiation effects
4.
Dev Cell ; 23(2): 265-79, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22841499

ABSTRACT

X chromosome inactivation involves multiple levels of chromatin modification, established progressively and in a stepwise manner during early development. The chromosomal protein Smchd1 was recently shown to play an important role in DNA methylation of CpG islands (CGIs), a late step in the X inactivation pathway that is required for long-term maintenance of gene silencing. Here we show that inactive X chromosome (Xi) CGI methylation can occur via either Smchd1-dependent or -independent pathways. Smchd1-dependent CGI methylation, the primary pathway, is acquired gradually over an extended period, whereas Smchd1-independent CGI methylation occurs rapidly after the onset of X inactivation. The de novo methyltransferase Dnmt3b is required for methylation of both classes of CGI, whereas Dnmt3a and Dnmt3L are dispensable. Xi CGIs methylated by these distinct pathways differ with respect to their sequence characteristics and immediate chromosomal environment. We discuss the implications of these results for understanding CGI methylation during development.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , CpG Islands , DNA Methylation , X Chromosome Inactivation , Alleles , Animals , Cell Line , Chromosomal Proteins, Non-Histone/genetics , Mice , Protein Isoforms/genetics , Protein Isoforms/metabolism
5.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...