Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 117(25): 14292-14298, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32522876

ABSTRACT

The mechanisms by which methylated mammalian promoters are transcriptionally silenced even in the presence of all of the factors required for their expression have long been a major unresolved issue in the field of epigenetics. Repression requires the assembly of a methylation-dependent silencing complex that contains the TRIM28 protein (also known as KAP1 and TIF1ß), a scaffolding protein without intrinsic repressive or DNA-binding properties. The identity of the key effector within this complex that represses transcription is unknown. We developed a methylation-sensitized interaction screen which revealed that TRIM28 was complexed with O-linked ß-N-acetylglucosamine transferase (OGT) only in cells that had normal genomic methylation patterns. OGT is the only glycosyltransferase that modifies cytoplasmic and nuclear protein by transfer of N-acetylglucosamine (O-GlcNAc) to serine and threonine hydroxyls. Whole-genome analysis showed that O-glycosylated proteins and TRIM28 were specifically bound to promoters of active retrotransposons and to imprinting control regions, the two major regulatory sequences controlled by DNA methylation. Furthermore, genome-wide loss of DNA methylation caused a loss of O-GlcNAc from multiple transcriptional repressor proteins associated with TRIM28. A newly developed Cas9-based editing method for targeted removal of O-GlcNAc was directed against retrotransposon promoters. Local chromatin de-GlcNAcylation specifically reactivated the expression of the targeted retrotransposon family without loss of DNA methylation. These data revealed that O-linked glycosylation of chromatin factors is essential for the transcriptional repression of methylated retrotransposons.


Subject(s)
Chromatin/metabolism , Promoter Regions, Genetic , Retroelements/physiology , Tripartite Motif-Containing Protein 28/metabolism , Acetylglucosamine/metabolism , Animals , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation , Epigenesis, Genetic , Gene Silencing , Glycosylation , Humans , Methylation , N-Acetylglucosaminyltransferases , Nuclear Proteins/metabolism , Protein Processing, Post-Translational , Proteomics , Repressor Proteins/metabolism , Transcription Factors/metabolism
2.
J Biol Chem ; 293(50): 19466-19475, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30341171

ABSTRACT

DNA methyltransferase 1 (DNMT1) is a multidomain protein believed to be involved only in the passive transmission of genomic methylation patterns via maintenance methylation. The mechanisms that regulate DNMT1 activity and targeting are complex and poorly understood. We used embryonic stem (ES) cells to investigate the function of the uncharacterized bromo-adjacent homology (BAH) domains and the glycine-lysine (GK) repeats that join the regulatory and catalytic domains of DNMT1. We removed the BAH domains by means of a CRISPR/Cas9-mediated deletion within the endogenous Dnmt1 locus. The internally deleted protein failed to associate with replication foci during S phase in vivo and lost the ability to mediate maintenance methylation. The data indicate that ablation of the BAH domains causes DNMT1 to be excluded from replication foci even in the presence of the replication focus-targeting sequence (RFTS). The GK repeats resemble the N-terminal tails of histones H2A and H4 and are normally acetylated. Substitution of lysines within the GK repeats with arginines to prevent acetylation did not alter the maintenance activity of DNMT1 but unexpectedly activated de novo methylation of paternal imprinting control regions (ICRs) in mouse ES cells; maternal ICRs remained unmethylated. We propose a model under which DNMT1 deposits paternal imprints in male germ cells in an acetylation-dependent manner. These data reveal that DNMT1 responds to multiple regulatory inputs that control its localization as well as its activity and is not purely a maintenance methyltransferase but can participate in the de novo methylation of a small but essential compartment of the genome.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/chemistry , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation , Histones/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Line , Humans , Mice , Models, Molecular , Protein Domains
3.
Photochem Photobiol Sci ; 17(8): 1049-1055, 2018 Aug 08.
Article in English | MEDLINE | ID: mdl-29926889

ABSTRACT

Epigenetic information is encoded in the mammalian genome in the form of cytosines methylated at the 5 position. Cytosine methylation has multiple biological effects, but our understanding of these effects has lagged because extant methods for mapping methylation sites genome-wide have severe shortcomings. For instance, the gold standard bisulfite sequencing approach suffers from the use of harsh reaction conditions resulting in DNA cleavage and incomplete conversion of unmethylated cytosine to uracil. We report here on a new photochemical method in which a DNA (cytosine-5)-methyltransferase can be used to covalently attach reactive functionalities which upon irradiation at ∼350 nm initiate photoinduced intramolecular reactions that convert modified C to T analogues. We synthesized a model compound, a cinnamyl ether-containing cytidine derivative, and demonstrated its conversion to a thymidine analogue using mild conditions and a DNA-compatible wavelength (∼350 nm), enabled by the use of a triplet sensitizer, thioxanthone. Transfer of a cinnamyl ether or comparable reactive functionality from an AdoMet analog to cytosine followed by the use of this photoconversion method would require only small amounts of DNA and allow complete methylation profiling on both long and short read sequencing platforms.


Subject(s)
Cytidine/chemistry , Thymidine/chemistry , CpG Islands , Cycloaddition Reaction , Cytidine/chemical synthesis , DNA/chemistry , DNA/metabolism , DNA Methylation , Lasers , Magnetic Resonance Spectroscopy , Photolysis , Spectrophotometry, Ultraviolet , Thioxanthenes/chemistry , Xanthones/chemistry
4.
Epigenetics Chromatin ; 11(1): 9, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29482658

ABSTRACT

BACKGROUND: It has been reported that USP7 (ubiquitin-specific protease 7) prevents ubiquitylation and degradation of DNA methyltransferase 1 (DNMT1) by direct binding of USP7 to the glycine-lysine (GK) repeats that join the N-terminal regulatory domain of DNMT1 to the C-terminal methyltransferase domain. The USP7-DNMT1 interaction was reported to be mediated by acetylation of lysine residues within the (GK) repeats. RESULTS: We found that DNMT1 is present at normal levels in mouse and human cells that contain undetectable levels of USP7. Substitution of the (GK) repeats by (GQ) repeats prevents lysine acetylation but does not affect the stability of DNMT1 or the ability of the mutant protein to restore genomic methylation levels when expressed in Dnmt1-null ES cells. Furthermore, both USP7 and PCNA are recruited to sites of DNA replication independently of the presence of DNMT1, and there is no evidence that DNMT1 is degraded in cycling cells after S phase. CONCLUSIONS: Multiple lines of evidence indicate that homeostasis of DNMT1 in somatic cells is controlled primarily at the level of transcription and that interaction of USP7 with the (GK) repeats of DNMT1 is unlikely to play a major role in the stabilization of DNMT1 protein.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Replication , DNA/genetics , Lysine/chemistry , Ubiquitin-Specific Peptidase 7/metabolism , Acetylation , Animals , Binding Sites , Cell Line , DNA (Cytosine-5-)-Methyltransferase 1/chemistry , Humans , Mice , Mouse Embryonic Stem Cells , Mutation , Protein Binding , Protein Stability , Ubiquitin-Specific Peptidase 7/chemistry
5.
Epigenetics Chromatin ; 10: 23, 2017.
Article in English | MEDLINE | ID: mdl-28503201

ABSTRACT

The prevailing views as to the form, function, and regulation of genomic methylation patterns have their origin many years in the past, at a time when the structure of the mammalian genome was only dimly perceived, when the number of protein-encoding mammalian genes was believed to be at least five times greater than the actual number, and when it was not understood that only ~10% of the genome is under selective pressure and likely to have biological function. We use more recent findings from genome biology and whole-genome methylation profiling to provide a reappraisal of the shape of genomic methylation patterns and the nature of the changes that they undergo during gametogenesis and early development. We observe that the sequences that undergo deep changes in methylation status during early development are largely sequences without regulatory function. We also discuss recent findings that begin to explain the remarkable fidelity of maintenance methylation. Rather than a general overview of DNA methylation in mammals (which has been the subject of many reviews), we present a new analysis of the distribution of methylated CpG dinucleotides across the multiple sequence compartments that make up the mammalian genome, and we offer an updated interpretation of the nature of the changes in methylation patterns that occur in germ cells and early embryos. We discuss the cues that might designate specific sequences for demethylation or de novo methylation during development, and we summarize recent findings on mechanisms that maintain methylation patterns in mammalian genomes. We also describe the several human disorders, each very different from the other, that are caused by mutations in DNA methyltransferase genes.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/genetics , Genomics , Genome, Human , Germ Cells/growth & development , Humans , Mutation
6.
Article in English | MEDLINE | ID: mdl-27252784

ABSTRACT

BACKGROUND: Almost all CpG-rich promoters in the mammalian genome are bound by the multidomain FBXL10 protein (also known as KDM2B, JHDM1B, CXXC2, and NDY1). FBXL10 is expressed as two isoforms: FBXL10-1, a longer form that contains an N-terminal histone demethylase domain with C-terminal F-box, CXXC, PHD, RING, and leucine-rich repeat domains, and FBXL10-2, a shorter form that initiates at an alternative internal exon and which lacks the histone demethylase domain but retains all other annotated domains. Selective deletion of Fbxl10-1 had been reported to produce a low penetrance and variable phenotype; most of the mutant animals were essentially normal. We constructed mutant mouse strains that were either null for Fbxl10-2 but wild type for Fbxl10-1 or null for both Fbxl10-1 and Fbxl10-2. RESULTS: Deletion of Fbxl10-2 (in a manner that does not perturb expression of Fbxl10-1) produced a phenotype very different from the Fbxl10-1 mutant, with craniofacial abnormalities, neural tube defects, and increased lethality, especially in females. Mutants that lacked both FBXL10-1 and FBXL10-2 showed embryonic lethality and even more extreme sexual dimorphism, with more severe gene dysregulation in mutant female embryos. X-linked genes were most severely dysregulated, and there was marked overexpression of Xist in mutant females although genes that encode factors that bind to Xist RNA were globally downregulated in mutant female as compared to male embryos. CONCLUSIONS: FBXL10 is the first factor shown to be required both for the normal expression and function of the Xist gene and for normal expression of proteins that associate with Xist RNA; it is proposed that FBXL10 coordinates the expression of Xist RNA with proteins that associate with this RNA. The function of FBXL10 is largely independent of the histone demethylase activity of the long form of the protein.

7.
PLoS Genet ; 11(10): e1005551, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26496195

ABSTRACT

Crumbs family proteins are apical transmembrane proteins with ancient roles in cell polarity. Mouse Crumbs2 mutants arrest at midgestation with abnormal neural plate morphology and a deficit of mesoderm caused by defects in gastrulation. We identified an ENU-induced mutation, wsnp, that phenocopies the Crumbs2 null phenotype. We show that wsnp is a null allele of Protein O-glucosyltransferase 1 (Poglut1), which encodes an enzyme previously shown to add O-glucose to EGF repeats in the extracellular domain of Drosophila and mammalian Notch, but the role of POGLUT1 in mammalian gastrulation has not been investigated. As predicted, we find that POGLUT1 is essential for Notch signaling in the early mouse embryo. However, the loss of mouse POGLUT1 causes an earlier and more dramatic phenotype than does the loss of activity of the Notch pathway, indicating that POGLUT1 has additional biologically relevant substrates. Using mass spectrometry, we show that POGLUT1 modifies EGF repeats in the extracellular domain of full-length mouse CRUMBS2. CRUMBS2 that lacks the O-glucose modification fails to be enriched on the apical plasma membrane and instead accumulates in the endoplasmic reticulum. The data demonstrate that CRUMBS2 is the target of POGLUT1 for the gastrulation epithelial-to-mesenchymal transitions (EMT) and that all activity of CRUMBS2 depends on modification by POGLUT1. Mutations in human POGLUT1 cause Dowling-Degos Disease, POGLUT1 is overexpressed in a variety of tumor cells, and mutations in the EGF repeats of human CRUMBS proteins are associated with human congenital nephrosis, retinitis pigmentosa and retinal degeneration, suggesting that O-glucosylation of CRUMBS proteins has broad roles in human health.


Subject(s)
Eye Proteins/genetics , Glucosyltransferases/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Receptor, Notch1/metabolism , Animals , Embryo, Mammalian , Embryonic Development , Eye Proteins/metabolism , Gastrulation/genetics , Glucosyltransferases/metabolism , Glycosylation , Humans , Membrane Proteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Phenotype , Protein Processing, Post-Translational/genetics , Receptor, Notch1/genetics , Signal Transduction
9.
Nat Genet ; 47(5): 479-85, 2015 May.
Article in English | MEDLINE | ID: mdl-25848754

ABSTRACT

Nearly all CpG-dense promoters are occupied by the multidomain chromosomal protein FBXL10. We show here that complete inactivation of the Fbxl10 gene leads to dense de novo methylation only of promoters that are co-occupied by both FBXL10 and Polycomb repressive complexes; this methylation results in pervasive defects in embryonic development and the death of homozygous Fbxl10-mutant embryos at midgestation. Deletion of key components of Polycomb repressive complexes 1 and 2 did not lead to ectopic genomic methylation. These results indicate that FBXL10 protects Polycomb-occupied promoters against ectopic de novo methylation. To our knowledge, FBXL10 is the first reported factor whose loss leads to a gain in genomic DNA methylation.


Subject(s)
DNA Methylation , F-Box Proteins/physiology , Jumonji Domain-Containing Histone Demethylases/physiology , Polycomb-Group Proteins/metabolism , Promoter Regions, Genetic , Animals , Cells, Cultured , CpG Islands , Embryonic Stem Cells/physiology , Epigenesis, Genetic , Gene Ontology , Male , Mice, Inbred C57BL , Mice, Transgenic , Polycomb Repressive Complex 1/metabolism , Transcription, Genetic , Ubiquitin-Protein Ligases/metabolism
10.
Proc Natl Acad Sci U S A ; 112(22): 6796-9, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-25368180

ABSTRACT

It has been nearly 40 y since it was suggested that genomic methylation patterns could be transmitted via maintenance methylation during S phase and might play a role in the dynamic regulation of gene expression during development [Holliday R, Pugh JE (1975) Science 187(4173):226-232; Riggs AD (1975) Cytogenet Cell Genet 14(1):9-25]. This revolutionary proposal was justified by "... our almost complete ignorance of the mechanism for the unfolding of the genetic program during development" that prevailed at the time. Many correlations between transcriptional activation and demethylation have since been reported, but causation has not been demonstrated and to date there is no reasonable proof of the existence of a complex biochemical system that activates and represses genes via reversible DNA methylation. Such a system would supplement or replace the conserved web of transcription factors that regulate cellular differentiation in organisms that have unmethylated genomes (such as Caenorhaditis elegans and the Dipteran insects) and those that methylate their genomes. DNA methylation does have essential roles in irreversible promoter silencing, as in the monoallelic expression of imprinted genes, in the silencing of transposons, and in X chromosome inactivation in female mammals. Rather than reinforcing or replacing regulatory pathways that are conserved between organisms that have either methylated or unmethylated genomes, DNA methylation endows genomes with the ability to subject specific sequences to irreversible transcriptional silencing even in the presence of all of the factors required for their expression, an ability that is generally unavailable to organisms that have unmethylated genomes.


Subject(s)
DNA Methylation/physiology , Epigenesis, Genetic/physiology , Gene Expression Regulation, Developmental/physiology , Mammals/growth & development , Animals , Promoter Regions, Genetic/genetics , X Chromosome Inactivation/genetics , X Chromosome Inactivation/physiology
11.
J Virol ; 88(18): 10680-95, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24991018

ABSTRACT

UNLABELLED: Mammalian genomes are replete with retrotransposable elements, including endogenous retroviruses. DNA methyltransferase 3-like (DNMT3L) is an epigenetic regulator expressed in prospermatogonia, growing oocytes, and embryonic stem (ES) cells. Here, we demonstrate that DNMT3L enhances the interaction of repressive epigenetic modifiers, including histone deacetylase 1 (HDAC1), SET domain, bifurcated 1 (SETDB1), DNA methyltransferase 3A (DNMT3A), and tripartite motif-containing protein 28 (TRIM28; also known as TIF1ß and KAP1) in ES cells and orchestrates retroviral silencing activity with TRIM28 through mechanisms including, but not limited to, de novo DNA methylation. Ectopic expression of DNMT3L in somatic cells causes methylation-independent retroviral silencing activity by recruitment of the TRIM28/HDAC1/SETDB1/DNMT3A/DNMT3L complex to newly integrated Moloney murine leukemia virus (Mo-MuLV) proviral DNA. Concurrent with this recruitment, we also observed the accumulation of histone H3 lysine 9 trimethylation (H3K9me3) and heterochromatin protein 1 gamma (HP1γ), as well as reduced H3K9 and H3K27 acetylation at Mo-MuLV proviral sequences. Ectopic expression of DNMT3L in late-passage mouse embryonic fibroblasts (MEFs) recruited cytoplasmically localized HDAC1 to the nucleus. The formation of this epigenetic modifying complex requires interaction of DNMT3L with DNMT3A as well as with histone H3. In fetal testes at embryonic day 17.5, endogenous DNMT3L also enhanced the binding among TRIM28, DNMT3A, SETDB1, and HDAC1. We propose that DNMT3L may be involved in initiating a cascade of repressive epigenetic modifications by assisting in the preparation of a chromatin context that further attracts DNMT3A-DNMT3L binding and installs longer-term DNA methylation marks at newly integrated retroviruses. IMPORTANCE: Almost half of the mammalian genome is composed of endogenous retroviruses and other retrotransposable elements that threaten genomic integrity. These elements are usually subject to epigenetic silencing. We discovered that two epigenetic regulators that lack enzymatic activity, DNA methyltransferase 3-like (DNMT3L) and tripartite motif-containing protein 28 (TRIM28), collaborate with each other to impose retroviral silencing. In addition to modulating de novo DNA methylation, we found that by interacting with TRIM28, DNMT3L can attract various enzymes to form a DNMT3L-induced repressive complex to remove active marks and add repressive marks to histone proteins. Collectively, these results reveal a novel and pivotal function of DNMT3L in shaping the chromatin modifications necessary for retroviral and retrotransposon silencing.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Gene Silencing , Leukemia, Experimental/enzymology , Leukemia, Experimental/genetics , Moloney murine leukemia virus/physiology , Repressor Proteins/metabolism , Animals , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Embryonic Stem Cells/enzymology , Embryonic Stem Cells/metabolism , Gene Expression , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histones/metabolism , Humans , Leukemia, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Moloney murine leukemia virus/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Repressor Proteins/genetics , Tripartite Motif-Containing Protein 28
12.
J Cancer Ther ; 5(14): 1311-1324, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25960928

ABSTRACT

Promoter silencing by ectopic de novo methylation of tumor suppressor genes has been proposed as comparable or equivalent to inactivating mutations as a factor in carcinogenesis. However, this hypotheses had not previously been tested by high resolution, high-coverage whole-genome methylation profiling in primary carcinomas. We have determined the genomic methylation status of a series of primary mammary carcinomas and matched control tissues by examination of more than 2.7 billion CpG dinucleotides. Most of the tumors showed variable losses of DNA methylation from all sequence compartments, but increases in promoter methylation were infrequent, very small in extent, and were observed largely at CpG-poor promoters. De novo methylation at the promoters of proto-oncogenes and tumor suppressor genes occurred at approximately the same frequency. The findings indicate that tumor suppressor silencing by de novo methylation is much less common than currently believed. We put forward a hypothesis under which the demethylation commonly observed in carcinomas is a manifestation of a defensive system that kills incipient cancer cells.

13.
Nat Genet ; 44(4): 398-405, S1-2, 2012 Mar 04.
Article in English | MEDLINE | ID: mdl-22387999

ABSTRACT

The generation of induced pluripotent stem cells (iPSCs) often results in aberrant epigenetic silencing of the imprinted Dlk1-Dio3 gene cluster, compromising the ability to generate entirely iPSC-derived adult mice ('all-iPSC mice'). Here, we show that reprogramming in the presence of ascorbic acid attenuates hypermethylation of Dlk1-Dio3 by enabling a chromatin configuration that interferes with binding of the de novo DNA methyltransferase Dnmt3a. This approach allowed us to generate all-iPSC mice from mature B cells, which have until now failed to support the development of exclusively iPSC-derived postnatal animals. Our data show that transcription factor-mediated reprogramming can endow a defined, terminally differentiated cell type with a developmental potential equivalent to that of embryonic stem cells. More generally, these findings indicate that culture conditions during cellular reprogramming can strongly influence the epigenetic and biological properties of the resultant iPSCs.


Subject(s)
Ascorbic Acid/pharmacology , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Genomic Imprinting , Induced Pluripotent Stem Cells , Intercellular Signaling Peptides and Proteins/genetics , Animals , Calcium-Binding Proteins , Cell Differentiation/genetics , Cellular Reprogramming/drug effects , Chromatin , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/drug effects , DNA Methyltransferase 3A , Mice , Octamer Transcription Factor-3/genetics , Promoter Regions, Genetic , Proteins/genetics , Proteins/metabolism , RNA, Long Noncoding
14.
Science ; 331(6020): 1036-40, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21163962

ABSTRACT

Maintenance of genomic methylation patterns is mediated primarily by DNA methyltransferase-1 (DNMT1). We have solved structures of mouse and human DNMT1 composed of CXXC, tandem bromo-adjacent homology (BAH1/2), and methyltransferase domains bound to DNA-containing unmethylated CpG sites. The CXXC specifically binds to unmethylated CpG dinucleotide and positions the CXXC-BAH1 linker between the DNA and the active site of DNMT1, preventing de novo methylation. In addition, a loop projecting from BAH2 interacts with the target recognition domain (TRD) of the methyltransferase, stabilizing the TRD in a retracted position and preventing it from inserting into the DNA major groove. Our studies identify an autoinhibitory mechanism, in which unmethylated CpG dinucleotides are occluded from the active site to ensure that only hemimethylated CpG dinucleotides undergo methylation.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA/chemistry , DNA/metabolism , Amino Acid Sequence , Animals , Catalytic Domain , Crystallography, X-Ray , Cysteine , DNA (Cytosine-5-)-Methyltransferase 1 , DNA-Cytosine Methylases/chemistry , DNA-Cytosine Methylases/metabolism , Dinucleoside Phosphates/chemistry , Dinucleoside Phosphates/metabolism , Humans , Mice , Models, Molecular , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Nucleic Acid Conformation , Protein Binding , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Substrate Specificity
15.
PLoS Genet ; 6(11): e1001214, 2010 Nov 18.
Article in English | MEDLINE | ID: mdl-21124941

ABSTRACT

In mammals, imprinted gene expression results from the sex-specific methylation of imprinted control regions (ICRs) in the parental germlines. Imprinting is linked to therian reproduction, that is, the placenta and imprinting emerged at roughly the same time and potentially co-evolved. We assessed the transcriptome-wide and ontology effect of maternally versus paternally methylated ICRs at the developmental stage of setting of the chorioallantoic placenta in the mouse (8.5dpc), using two models of imprinting deficiency including completely imprint-free embryos. Paternal and maternal imprints have a similar quantitative impact on the embryonic transcriptome. However, transcriptional effects of maternal ICRs are qualitatively focused on the fetal-maternal interface, while paternal ICRs weakly affect non-convergent biological processes, with little consequence for viability at 8.5dpc. Moreover, genes regulated by maternal ICRs indirectly influence genes regulated by paternal ICRs, while the reverse is not observed. The functional dominance of maternal imprints over early embryonic development is potentially linked to selection pressures favoring methylation-dependent control of maternal over paternal ICRs. We previously hypothesized that the different methylation histories of ICRs in the maternal versus the paternal germlines may have put paternal ICRs under higher mutational pressure to lose CpGs by deamination. Using comparative genomics of 17 extant mammalian species, we show here that, while ICRs in general have been constrained to maintain more CpGs than non-imprinted sequences, the rate of CpG loss at paternal ICRs has indeed been higher than at maternal ICRs during evolution. In fact, maternal ICRs, which have the characteristics of CpG-rich promoters, have gained CpGs compared to non-imprinted CpG-rich promoters. Thus, the numerical and, during early embryonic development, functional dominance of maternal ICRs can be explained as the consequence of two orthogonal evolutionary forces: pressure to tightly regulate genes affecting the fetal-maternal interface and pressure to avoid the mutagenic environment of the paternal germline.


Subject(s)
Biological Evolution , Embryonic Development/genetics , Genomic Imprinting/genetics , Mammals/embryology , Mammals/genetics , Animals , CpG Islands/genetics , DNA Methylation/genetics , Deamination/genetics , Embryo, Mammalian/metabolism , Female , Gene Expression Regulation, Developmental , Genome/genetics , Humans , Male , Mice , Phylogeny , Signal Transduction/genetics
16.
Epigenetics Chromatin ; 3(1): 17, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20868487

ABSTRACT

BACKGROUND: Genomic methylation patterns are established during gametogenesis, and perpetuated in somatic cells by faithful maintenance methylation. There have been previous indications that genomic methylation patterns may be less stable in embryonic stem (ES) cells than in differentiated somatic cells, but it is not known whether different mechanisms of de novo and maintenance methylation operate in pluripotent stem cells compared with differentiating somatic cells. RESULTS: In this paper, we show that ablation of the DNA methyltransferase regulator DNMT3L (DNA methyltransferase 3-like) in mouse ES cells renders them essentially incapable of de novo methylation of newly integrated retroviral DNA. We also show that ES cells lacking DNMT3L lose DNA methylation over time in culture, suggesting that DNA methylation in ES cells is the result of dynamic loss and gain of DNA methylation. We found that wild-type female ES cells lose DNA methylation at a much faster rate than do male ES cells; this defect could not be attributed to sex-specific differences in expression of DNMT3L or of any DNA methyltransferase. We also found that human ES and induced pluripotent stem cell lines showed marked but variable loss of methylation that could not be attributed to sex chromosome constitution or time in culture. CONCLUSIONS: These data indicate that DNA methylation in pluripotent stem cells is much more dynamic and error-prone than is maintenance methylation in differentiated cells. DNA methylation requires DNMT3L in stem cells, but DNMT3L is not expressed in differentiating somatic cells. Error-prone maintenance methylation will introduce unpredictable phenotypic variation into clonal populations of pluripotent stem cells, and this variation is likely to be much more pronounced in cultured female cells. This epigenetic variability has obvious negative implications for the clinical applications of stem cells.

17.
Genome Res ; 20(7): 972-80, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20488932

ABSTRACT

Abnormalities of genomic methylation patterns are lethal or cause disease, but the cues that normally designate CpG dinucleotides for methylation are poorly understood. We have developed a new method of methylation profiling that has single-CpG resolution and can address the methylation status of repeated sequences. We have used this method to determine the methylation status of >275 million CpG sites in human and mouse DNA from breast and brain tissues. Methylation density at most sequences was found to increase linearly with CpG density and to fall sharply at very high CpG densities, but transposons remained densely methylated even at higher CpG densities. The presence of histone H2A.Z and histone H3 di- or trimethylated at lysine 4 correlated strongly with unmethylated DNA and occurred primarily at promoter regions. We conclude that methylation is the default state of most CpG dinucleotides in the mammalian genome and that a combination of local dinucleotide frequencies, the interaction of repeated sequences, and the presence or absence of histone variants or modifications shields a population of CpG sites (most of which are in and around promoters) from DNA methyltransferases that lack intrinsic sequence specificity.


Subject(s)
Base Sequence/physiology , Chromatin/chemistry , Chromatin/physiology , DNA Methylation , Animals , Brain/metabolism , Breast/metabolism , Chromatin/genetics , Chromosome Mapping , CpG Islands/genetics , Female , Genome , Histones/metabolism , Humans , Mice , Sequence Analysis, DNA , Validation Studies as Topic
19.
Breast Cancer Res Treat ; 115(2): 397-404, 2009 May.
Article in English | MEDLINE | ID: mdl-18521744

ABSTRACT

Promoter-CpG island hypermethylation has been proposed as an alternative mechanism to inactivate BRCA1 in the breast where somatic mutations of BRCA1 are rare. To better understand breast cancer etiology and progression, we explored the association between BRCA1 promoter methylation status and prognostic factors as well as survival among women with breast cancer. Promoter methylation of BRCA1 was assessed in 851 archived tumor tissues collected from a population-based study of women diagnosed with invasive or in situ breast cancer in 1996-1997, and who were followed for vital status through the end of 2002. About 59% of the tumors were methylated at the promoter of BRCA1. The BRCA1 promoter methylation was more frequent in invasive cancers (P = 0.02) and among premenopausal cases (P = 0.05). BRCA1 promoter methylation was associated with increased risk of breast cancer-specific mortality (age-adjusted HR 1.71; 95% CI: 1.05-2.78) and all-cause mortality (age-adjusted HR 1.49; 95% CI: 1.02-2.18). Neither dietary methyl intakes in the year prior to the baseline interview nor the functional polymorphisms in one-carbon metabolism were associated with BRCA1 methylation status. Our study is the first epidemiological investigation on the prognostic value of BRCA1 promoter methylation in a large population-based cohort of breast cancer patients. Our results indicate that BRCA1 promoter methylation is an important factor to consider in predicting breast cancer survival.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/mortality , DNA Methylation/genetics , Genes, BRCA1 , Promoter Regions, Genetic/genetics , Breast Neoplasms/pathology , DNA Mutational Analysis , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Prognosis
20.
J Cell Biol ; 182(2): 263-76, 2008 Jul 28.
Article in English | MEDLINE | ID: mdl-18663141

ABSTRACT

Chromosome synapsis during zygotene is a prerequisite for the timely homologous recombinational repair of meiotic DNA double-strand breaks (DSBs). Unrepaired DSBs are thought to trigger apoptosis during midpachytene of male meiosis if synapsis fails. An early pachytene response to asynapsis is meiotic silencing of unsynapsed chromatin (MSUC), which, in normal males, silences the X and Y chromosomes (meiotic sex chromosome inactivation [MSCI]). In this study, we show that MSUC occurs in Spo11-null mouse spermatocytes with extensive asynapsis but lacking meiotic DSBs. In contrast, three mutants (Dnmt3l, Msh5, and Dmc1) with high levels of asynapsis and numerous persistent unrepaired DSBs have a severely impaired MSUC response. We suggest that MSUC-related proteins, including the MSUC initiator BRCA1, are sequestered at unrepaired DSBs. All four mutants fail to silence the X and Y chromosomes (MSCI failure), which is sufficient to explain the midpachytene apoptosis. Apoptosis does not occur in mice with a single additional asynapsed chromosome with unrepaired meiotic DSBs and no disturbance of MSCI.


Subject(s)
Chromosome Pairing/genetics , Gene Silencing/physiology , Meiosis/genetics , Sex Chromosomes/genetics , Spermatogenesis/genetics , X Chromosome Inactivation/genetics , Animals , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Chromosome Segregation/genetics , DNA Breaks, Double-Stranded , Endodeoxyribonucleases , Esterases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Multigene Family/genetics , Mutation/genetics , Spermatids/cytology , Spermatids/metabolism , Spermatocytes/cytology , Spermatocytes/metabolism , X Chromosome/genetics , Y Chromosome/genetics
SELECTION OF CITATIONS
SEARCH DETAIL