Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters











Publication year range
1.
Clin Chem ; 67(6): 867-875, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33709101

ABSTRACT

BACKGROUND: Due to improved treatment, more patients with multiple myeloma (MM) reach a state of minimal residual disease (MRD). Different strategies for MM MRD monitoring include flow cytometry, allele-specific oligonucleotide-quantitative PCR, next-generation sequencing, and mass spectrometry (MS). The last 3 methods rely on the presence and the stability of a unique immunoglobulin fingerprint derived from the clonal plasma cell population. For MS-MRD monitoring it is imperative that MS-compatible clonotypic M-protein peptides are identified. To support implementation of molecular MRD techniques, we studied the presence and stability of these clonotypic features in the CoMMpass database. METHODS: An analysis pipeline based on MiXCR and HIGH-VQUEST was constructed to identify clonal molecular fingerprints and their clonotypic peptides based on transcriptomic datasets. To determine the stability of the clonal fingerprints, we compared the clonal fingerprints during disease progression for each patient. RESULTS: The analysis pipeline to establish the clonal fingerprint and MS-suitable clonotypic peptides was successfully validated in MM cell lines. In a cohort of 609 patients with MM, we demonstrated that the most abundant clone harbored a unique clonal molecular fingerprint and that multiple unique clonotypic peptides compatible with MS measurements could be identified for all patients. Furthermore, the clonal immunoglobulin gene fingerprints of both the light and heavy chain remained stable during MM disease progression. CONCLUSIONS: Our data support the use of the clonal immunoglobulin gene fingerprints in patients with MM as a suitable MRD target for MS-MRD analyses.


Subject(s)
Genes, Immunoglobulin/physiology , Multiple Myeloma , Peptides/chemistry , Biomarkers , Disease Progression , Humans , Multiple Myeloma/diagnosis , Multiple Myeloma/genetics , Neoplasm, Residual/genetics , Peptides/genetics
2.
Exp Dermatol ; 30(8): 1023-1032, 2021 08.
Article in English | MEDLINE | ID: mdl-32681572

ABSTRACT

The epidermal compartment of the skin is regenerated constantly by proliferation of epidermal keratinocytes. Differentiation of a subset of these keratinocytes allows the epidermis to retain its barrier properties. Regulation of keratinocyte fate-whether to remain proliferative or terminally differentiate-is complex and not fully understood. The objective of our study was to assess if DNA methylation changes contribute to the regulation of keratinocyte fate. We employed genome-wide MethylationEPIC beadchip array measuring approximately 850 000 probes combined with RNA sequencing of in vitro cultured non-differentiated and terminally differentiated adult human primary keratinocytes. We did not observe a correlation between methylation status and transcriptome changes. Moreover, only two differentially methylated probes were detected, of which one was located in the TRIM29 gene. Although TRIM29 knock-down resulted in lower expression levels of terminal differentiation genes, these changes were minor. From these results, we conclude that-in our in vitro experimental setup-it is unlikely that changes in DNA methylation have an important regulatory role in terminal keratinocyte differentiation.


Subject(s)
Cell Differentiation/genetics , DNA Methylation/genetics , Epigenome/genetics , Keratinocytes/metabolism , Adult , DNA-Binding Proteins/genetics , Humans , Transcription Factors/genetics
3.
Breast Cancer Res ; 21(1): 151, 2019 12 26.
Article in English | MEDLINE | ID: mdl-31878981

ABSTRACT

BACKGROUND: Breast cancer (BC) immune infiltrates play a critical role in tumor progression and response to treatment. Besides stromal tumor infiltrating lymphocytes (sTILs) which have recently reached level 1B evidence as a prognostic marker in triple negative BC, a plethora of methods to assess immune infiltration exists, and it is unclear how these compare to each other and if they can be used interchangeably. METHODS: Two experienced pathologists scored sTIL, intra-tumoral TIL (itTIL), and 6 immune cell types (CD3+, CD4+, CD8+, CD20+, CD68+, FOXP3+) in the International Cancer Genomics Consortium breast cancer cohort using hematoxylin and eosin-stained (n = 243) and immunohistochemistry-stained tissue microarrays (n = 254) and whole slides (n = 82). The same traits were evaluated using transcriptomic- and methylomic-based deconvolution methods or signatures. RESULTS: The concordance correlation coefficient (CCC) between pathologists for sTIL was very good (0.84) and for cell-specific immune infiltrates slightly lower (0.63-0.66). Comparison between tissue microarray and whole slide pathology scores revealed systematically higher values in whole slides (ratio 2.60-5.98). The Spearman correlations between microscopic sTIL and transcriptomic- or methylomic-based assessment of immune infiltrates were highly variable (r = 0.01-0.56). Similar observations were made for cell type-specific quantifications (r = 0.001-0.54). We observed a strong inter-method variability between the omics-derived estimations, which is further cell type dependent. Finally, we demonstrated that most methods more accurately identify highly infiltrated (sTIL ≥ 60%; area under the curve, AUC, 0.64-0.99) as compared to lowly infiltrated tumors (sTIL ≤ 10%; AUC 0.52-0.82). CONCLUSIONS: There is a lower inter-pathologist concordance for cell-specific quantification as compared to overall infiltration quantification. Microscopic assessments are underestimated when considering small cores (tissue microarray) instead of whole slides. Results further highlight considerable differences between the microscopic-, transcriptomic-, and methylomic-based methods in the assessment of overall and cell-specific immune infiltration in BC. We therefore call for extreme caution when assessing immune infiltrates using current methods and emphasize the need for standardized immune characterization beyond TIL.


Subject(s)
Breast Neoplasms/etiology , Disease Susceptibility , Lymphocytes, Tumor-Infiltrating/immunology , Biomarkers, Tumor , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Epigenome , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/pathology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Tissue Array Analysis , Transcriptome , Tumor Microenvironment/immunology
4.
Nat Commun ; 10(1): 1749, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30988298

ABSTRACT

Global loss of DNA methylation and CpG island (CGI) hypermethylation are key epigenomic aberrations in cancer. Global loss manifests itself in partially methylated domains (PMDs) which extend up to megabases. However, the distribution of PMDs within and between tumor types, and their effects on key functional genomic elements including CGIs are poorly defined. We comprehensively show that loss of methylation in PMDs occurs in a large fraction of the genome and represents the prime source of DNA methylation variation. PMDs are hypervariable in methylation level, size and distribution, and display elevated mutation rates. They impose intermediate DNA methylation levels incognizant of functional genomic elements including CGIs, underpinning a CGI methylator phenotype (CIMP). Repression effects on tumor suppressor genes are negligible as they are generally excluded from PMDs. The genomic distribution of PMDs reports tissue-of-origin and may represent tissue-specific silent regions which tolerate instability at the epigenetic, transcriptomic and genetic level.


Subject(s)
Breast Neoplasms/genetics , CpG Islands , DNA Methylation , Epigenesis, Genetic , Female , Humans , Logistic Models
6.
Cell Stem Cell ; 24(1): 123-137.e8, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30472157

ABSTRACT

The pluripotent ground state is defined as a basal state free of epigenetic restrictions, which influence lineage specification. While naive embryonic stem cells (ESCs) can be maintained in a hypomethylated state with open chromatin when grown using two small-molecule inhibitors (2i)/leukemia inhibitory factor (LIF), in contrast to serum/LIF-grown ESCs that resemble early post-implantation embryos, broader features of the ground-state pluripotent epigenome are not well understood. We identified epigenetic features of mouse ESCs cultured using 2i/LIF or serum/LIF by proteomic profiling of chromatin-associated complexes and histone modifications. Polycomb-repressive complex 2 (PRC2) and its product H3K27me3 are highly abundant in 2i/LIF ESCs, and H3K27me3 is distributed genome-wide in a CpG-dependent fashion. Consistently, PRC2-deficient ESCs showed increased DNA methylation at sites normally occupied by H3K27me3 and increased H4 acetylation. Inhibiting DNA methylation in PRC2-deficient ESCs did not affect their viability or transcriptome. Our findings suggest a unique H3K27me3 configuration protects naive ESCs from lineage priming, and they reveal widespread epigenetic crosstalk in ground-state pluripotency.


Subject(s)
Chromatin/metabolism , DNA Methylation , Epigenesis, Genetic , Mouse Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Polycomb Repressive Complex 2/metabolism , Proteome/analysis , Animals , Cell Differentiation , Chromatin/genetics , Histones/genetics , Histones/metabolism , Mice , Mouse Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Polycomb Repressive Complex 2/genetics , Protein Processing, Post-Translational
8.
Nat Genet ; 49(3): 341-348, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28112740

ABSTRACT

Somatic rearrangements contribute to the mutagenized landscape of cancer genomes. Here, we systematically interrogated rearrangements in 560 breast cancers by using a piecewise constant fitting approach. We identified 33 hotspots of large (>100 kb) tandem duplications, a mutational signature associated with homologous-recombination-repair deficiency. Notably, these tandem-duplication hotspots were enriched in breast cancer germline susceptibility loci (odds ratio (OR) = 4.28) and breast-specific 'super-enhancer' regulatory elements (OR = 3.54). These hotspots may be sites of selective susceptibility to double-strand-break damage due to high transcriptional activity or, through incrementally increasing copy number, may be sites of secondary selective pressure. The transcriptomic consequences ranged from strong individual oncogene effects to weak but quantifiable multigene expression effects. We thus present a somatic-rearrangement mutational process affecting coding sequences and noncoding regulatory elements and contributing a continuum of driver consequences, from modest to strong effects, thereby supporting a polygenic model of cancer development.


Subject(s)
Breast Neoplasms/genetics , Genetic Loci/genetics , Mutation/genetics , Regulatory Sequences, Nucleic Acid/genetics , DNA Breaks, Double-Stranded , DNA Repair/genetics , Female , Gene Expression/genetics , Genome/genetics , Humans , Transcriptome/genetics
9.
Nat Commun ; 7: 12910, 2016 Sep 26.
Article in English | MEDLINE | ID: mdl-27666519

ABSTRACT

A recent comprehensive whole genome analysis of a large breast cancer cohort was used to link known and novel drivers and substitution signatures to the transcriptome of 266 cases. Here, we validate that subtype-specific aberrations show concordant expression changes for, for example, TP53, PIK3CA, PTEN, CCND1 and CDH1. We find that CCND3 expression levels do not correlate with amplification, while increased GATA3 expression in mutant GATA3 cancers suggests GATA3 is an oncogene. In luminal cases the total number of substitutions, irrespective of type, associates with cell cycle gene expression and adverse outcome, whereas the number of mutations of signatures 3 and 13 associates with immune-response specific gene expression, increased numbers of tumour-infiltrating lymphocytes and better outcome. Thus, while earlier reports imply that the sheer number of somatic aberrations could trigger an immune-response, our data suggests that substitutions of a particular type are more effective in doing so than others.

10.
Nature ; 534(7605): 47-54, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27135926

ABSTRACT

We analysed whole-genome sequences of 560 breast cancers to advance understanding of the driver mutations conferring clonal advantage and the mutational processes generating somatic mutations. We found that 93 protein-coding cancer genes carried probable driver mutations. Some non-coding regions exhibited high mutation frequencies, but most have distinctive structural features probably causing elevated mutation rates and do not contain driver mutations. Mutational signature analysis was extended to genome rearrangements and revealed twelve base substitution and six rearrangement signatures. Three rearrangement signatures, characterized by tandem duplications or deletions, appear associated with defective homologous-recombination-based DNA repair: one with deficient BRCA1 function, another with deficient BRCA1 or BRCA2 function, the cause of the third is unknown. This analysis of all classes of somatic mutation across exons, introns and intergenic regions highlights the repertoire of cancer genes and mutational processes operating, and progresses towards a comprehensive account of the somatic genetic basis of breast cancer.


Subject(s)
Breast Neoplasms/genetics , Genome, Human/genetics , Mutation/genetics , Cohort Studies , DNA Mutational Analysis , DNA Replication/genetics , DNA, Neoplasm/genetics , Female , Genes, BRCA1 , Genes, BRCA2 , Genomics , Humans , Male , Mutagenesis , Mutation Rate , Oncogenes/genetics , Recombinational DNA Repair/genetics
11.
Nat Commun ; 7: 11383, 2016 May 02.
Article in English | MEDLINE | ID: mdl-27136393

ABSTRACT

Somatic mutations in human cancers show unevenness in genomic distribution that correlate with aspects of genome structure and function. These mutations are, however, generated by multiple mutational processes operating through the cellular lineage between the fertilized egg and the cancer cell, each composed of specific DNA damage and repair components and leaving its own characteristic mutational signature on the genome. Using somatic mutation catalogues from 560 breast cancer whole-genome sequences, here we show that each of 12 base substitution, 2 insertion/deletion (indel) and 6 rearrangement mutational signatures present in breast tissue, exhibit distinct relationships with genomic features relating to transcription, DNA replication and chromatin organization. This signature-based approach permits visualization of the genomic distribution of mutational processes associated with APOBEC enzymes, mismatch repair deficiency and homologous recombinational repair deficiency, as well as mutational processes of unknown aetiology. Furthermore, it highlights mechanistic insights including a putative replication-dependent mechanism of APOBEC-related mutagenesis.


Subject(s)
Apolipoproteins B/genetics , Breast Neoplasms/genetics , DNA Repair , Genome, Human , Mutation , Apolipoproteins B/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chromatin/chemistry , Chromatin/metabolism , DNA Damage , DNA Replication , Female , Humans , MCF-7 Cells , Mutagenesis , Sequence Analysis, DNA , Transcription, Genetic
12.
Cell Rep ; 8(3): 743-53, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25066127

ABSTRACT

DNA methylation is a central epigenetic modification that is established by de novo DNA methyltransferases. The mechanisms underlying the generation of genomic methylation patterns are still poorly understood. Using mass spectrometry and a phosphospecific Dnmt3a antibody, we demonstrate that CK2 phosphorylates endogenous Dnmt3a at two key residues located near its PWWP domain, thereby downregulating the ability of Dnmt3a to methylate DNA. Genome-wide DNA methylation analysis shows that CK2 primarily modulates CpG methylation of several repeats, most notably of Alu SINEs. This modulation can be directly attributed to CK2-mediated phosphorylation of Dnmt3a. We also find that CK2-mediated phosphorylation is required for localization of Dnmt3a to heterochromatin. By revealing phosphorylation as a mode of regulation of de novo DNA methyltransferase function and by uncovering a mechanism for the regulation of methylation at repetitive elements, our results shed light on the origin of DNA methylation patterns.


Subject(s)
Casein Kinase II/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Protein Processing, Post-Translational , 3T3 Cells , Animals , Cell Line, Tumor , CpG Islands , DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA Methyltransferase 3A , Down-Regulation , Heterochromatin/metabolism , Humans , Mice , Phosphorylation , Protein Structure, Tertiary , Short Interspersed Nucleotide Elements
13.
PLoS One ; 9(6): e99603, 2014.
Article in English | MEDLINE | ID: mdl-24927503

ABSTRACT

MBD2 is a subunit of the NuRD complex that is postulated to mediate gene repression via recruitment of the complex to methylated DNA. In this study we adopted an MBD2 tagging-approach to study its genome wide binding characteristics. We show that in vivo MBD2 is mainly recruited to CpG island promoters that are highly methylated. Interestingly, MBD2 binds around 1 kb downstream of the transcription start site of a subset of ∼ 400 CpG island promoters that are characterized by the presence of active histone marks, RNA polymerase II (Pol2) and low to medium gene expression levels and H3K36me3 deposition. These tagged-MBD2 binding sites in MCF-7 show increased methylation in a cohort of primary breast cancers but not in normal breast samples, suggesting a putative role for MBD2 in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , DNA Methylation , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , RNA Polymerase II/metabolism , Binding Sites , CpG Islands , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Molecular Sequence Data , Promoter Regions, Genetic , Sequence Analysis, RNA
14.
Cell Stem Cell ; 13(3): 360-9, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23850244

ABSTRACT

The use of two kinase inhibitors (2i) enables derivation of mouse embryonic stem cells (ESCs) in the pluripotent ground state. Using whole-genome bisulfite sequencing (WGBS), we show that male 2i ESCs are globally hypomethylated compared to conventional ESCs maintained in serum. In serum, female ESCs are hypomethyated similarly to male ESCs in 2i, and DNA methylation is further reduced in 2i. Regions with elevated DNA methylation in 2i strongly correlate with the presence of H3K9me3 on endogenous retroviruses (ERVs) and imprinted loci. The methylome of male ESCs in serum parallels postimplantation blastocyst cells, while 2i stalls ESCs in a hypomethylated, ICM-like state. WGBS analysis during adaptation of 2i ESCs to serum suggests that deposition of DNA methylation is largely random, while loss of DNA methylation during reversion to 2i occurs passively, initiating at TET1 binding sites. Together, our analysis provides insight into DNA methylation dynamics in cultured ESCs paralleling early developmental processes.


Subject(s)
Blastocyst/physiology , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/physiology , Histone Demethylases/metabolism , Pluripotent Stem Cells/physiology , Proto-Oncogene Proteins/metabolism , Animals , Cells, Cultured , DNA Methylation/drug effects , DNA-Binding Proteins/genetics , Embryonic Stem Cells/drug effects , Female , Fetal Development , Genome/genetics , Histones/metabolism , Leukemia Inhibitory Factor/metabolism , Male , Methylation , Mice , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/genetics , Sequence Analysis, DNA , Sulfites/chemistry
15.
Genes Dev ; 27(9): 1059-71, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23651858

ABSTRACT

Notch signaling plays crucial roles in mediating cell fate choices in all metazoans largely by specifying the transcriptional output of one cell in response to a neighboring cell. The DNA-binding protein RBPJ is the principle effector of this pathway in mammals and, together with the transcription factor moiety of Notch (NICD), regulates the expression of target genes. The prevalent view presumes that RBPJ statically occupies consensus binding sites while exchanging repressors for activators in response to NICD. We present the first specific RBPJ chromatin immunoprecipitation and high-throughput sequencing study in mammalian cells. To dissect the mode of transcriptional regulation by RBPJ and identify its direct targets, whole-genome binding profiles were generated for RBPJ; its coactivator, p300; NICD; and the histone H3 modifications H3 Lys 4 trimethylation (H3K4me3), H3 Lys 4 monomethylation (H3K4me1), and histone H3 Lys 27 acetylation (H3K27ac) in myogenic cells under active or inhibitory Notch signaling conditions. Our results demonstrate dynamic binding of RBPJ in response to Notch activation at essentially all sites co-occupied by NICD. Additionally, we identify a distinct set of sites where RBPJ recruits neither NICD nor p300 and binds DNA statically, irrespective of Notch activity. These findings significantly modify our views on how RBPJ and Notch signaling mediate their activities and consequently impact on cell fate decisions.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Binding Sites , Cell Line , Chromatin/genetics , Genome-Wide Association Study , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Protein Binding , Regulatory Elements, Transcriptional/genetics
16.
Cell ; 152(5): 1146-59, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23434322

ABSTRACT

Tet proteins oxidize 5-methylcytosine (mC) to generate 5-hydroxymethyl (hmC), 5-formyl (fC), and 5-carboxylcytosine (caC). The exact function of these oxidative cytosine bases remains elusive. We applied quantitative mass-spectrometry-based proteomics to identify readers for mC and hmC in mouse embryonic stem cells (mESC), neuronal progenitor cells (NPC), and adult mouse brain tissue. Readers for these modifications are only partially overlapping, and some readers, such as Rfx proteins, display strong specificity. Interactions are dynamic during differentiation, as for example evidenced by the mESC-specific binding of Klf4 to mC and the NPC-specific binding of Uhrf2 to hmC, suggesting specific biological roles for mC and hmC. Oxidized derivatives of mC recruit distinct transcription regulators as well as a large number of DNA repair proteins in mouse ES cells, implicating the DNA damage response as a major player in active DNA demethylation.


Subject(s)
5-Methylcytosine/analysis , Cytosine/analogs & derivatives , DNA Methylation , 5-Methylcytosine/metabolism , Animals , Brain/cytology , Brain/metabolism , Cytosine/analysis , Cytosine/metabolism , DNA Glycosylases/metabolism , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Kruppel-Like Factor 4 , Mass Spectrometry , Mice , Oxidation-Reduction , Proto-Oncogene Proteins/metabolism , Regulatory Factor X Transcription Factors , Stem Cells/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism
17.
Epigenetics ; 7(12): 1355-67, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23079744

ABSTRACT

Aberrant DNA methylation often occurs in colorectal cancer (CRC). In our study we applied a genome-wide DNA methylation analysis approach, MethylCap-seq, to map the differentially methylated regions (DMRs) in 24 tumors and matched normal colon samples. In total, 2687 frequently hypermethylated and 468 frequently hypomethylated regions were identified, which include potential biomarkers for CRC diagnosis. Hypermethylation in the tumor samples was enriched at CpG islands and gene promoters, while hypomethylation was distributed throughout the genome. Using epigenetic data from human embryonic stem cells, we show that frequently hypermethylated regions coincide with bivalent loci in human embryonic stem cells. DNA methylation is commonly thought to lead to gene silencing; however, integration of publically available gene expression data indicates that 75% of the frequently hypermethylated genes were most likely already lowly or not expressed in normal tissue. Collectively, our study provides genome-wide DNA methylation maps of CRC, comprehensive lists of DMRs, and gives insights into the role of aberrant DNA methylation in CRC formation.


Subject(s)
Colorectal Neoplasms/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic , Biomarkers, Tumor/genetics , Case-Control Studies , Cell Line, Tumor , Colon/physiology , Embryonic Stem Cells/physiology , Epigenesis, Genetic , Genome-Wide Association Study , Histones/genetics , Histones/metabolism , Humans , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Reference Values
18.
Genome Res ; 22(6): 1128-38, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22466170

ABSTRACT

Cross-talk between DNA methylation and histone modifications drives the establishment of composite epigenetic signatures and is traditionally studied using correlative rather than direct approaches. Here, we present sequential ChIP-bisulfite-sequencing (ChIP-BS-seq) as an approach to quantitatively assess DNA methylation patterns associated with chromatin modifications or chromatin-associated factors directly. A chromatin-immunoprecipitation (ChIP)-capturing step is used to obtain a restricted representation of the genome occupied by the epigenetic feature of interest, for which a single-base resolution DNA methylation map is then generated. When applied to H3 lysine 27 trimethylation (H3K27me3), we found that H3K27me3 and DNA methylation are compatible throughout most of the genome, except for CpG islands, where these two marks are mutually exclusive. Further ChIP-BS-seq-based analysis in Dnmt triple-knockout (TKO) embryonic stem cells revealed that total loss of CpG methylation is associated with alteration of H3K27me3 levels throughout the genome: H3K27me3 in localized peaks is decreased while broad local enrichments (BLOCs) of H3K27me3 are formed. At an even broader scale, these BLOCs correspond to regions of high DNA methylation in wild-type ES cells, suggesting that DNA methylation prevents H3K27me3 deposition locally and at a megabase scale. Our strategy provides a unique way of investigating global interdependencies between DNA methylation and other chromatin features.


Subject(s)
Chromatin Immunoprecipitation/methods , Chromatin/genetics , CpG Islands , DNA Methylation , Histones/metabolism , Sequence Analysis, DNA/methods , Animals , Cell Line , Cell Line, Tumor , Chromatin/drug effects , Colonic Neoplasms/genetics , Embryonic Stem Cells/physiology , Epigenesis, Genetic , Gene Knockout Techniques , Genomics/methods , Humans , Lysine/metabolism , Mice , Sulfites/pharmacology
19.
PLoS One ; 6(10): e25884, 2011.
Article in English | MEDLINE | ID: mdl-21991380

ABSTRACT

BACKGROUND: DNA methylation is an epigenetic modification that plays a crucial role in a variety of biological processes. Methylated DNA is specifically bound by Methyl-CpG Binding Proteins (MBPs). Three different types of MBPs have been identified so far: the Methyl-CpG Binding Domain (MBD) family proteins, three BTB/POZ-Zn-finger proteins, and UHRF1. Most of the known MBPs have been identified via homology with the MBD and Zn-finger domains as present in MeCP2 and Kaiso, respectively. It is conceivable that other proteins are capable of recognizing methylated DNA. METHODOLOGY/PRINCIPAL FINDINGS: For the purpose of identifying novel 'readers' we set up a methyl-CpG pull-down assay combined with stable-isotope labeling by amino acids in cell culture (SILAC). In a methyl-CpG pull-down with U937 nuclear extracts, we recovered several known MBPs and almost all subunits of the MBD2/NuRD complex as methylation specific binders, providing proof-of-principle. Interestingly, RBP-J, the transcription factor downstream of Notch receptors, also bound the DNA in a methylation dependent manner. Follow-up pull-downs and electrophoretic mobility shift assays (EMSAs) showed that RBP-J binds methylated DNA in the context of a mutated RBP-J consensus motif. CONCLUSIONS/SIGNIFICANCE: The here described SILAC/methyl-CpG pull-down constitutes a new approach to identify potential novel DNAme readers and will advance unraveling of the complete methyl-DNA interactome.


Subject(s)
CpG Islands/genetics , DNA Methylation/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Isotope Labeling/methods , Base Sequence , Consensus Sequence/genetics , Humans , Molecular Sequence Data , Mutation/genetics , Nucleotide Motifs , Protein Binding , U937 Cells
20.
Genome Res ; 21(8): 1313-27, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21636662

ABSTRACT

DNA methylation is a tightly regulated epigenetic mark associated with transcriptional repression. Next-generation sequencing of purified methylated DNA obtained from early Xenopus tropicalis embryos demonstrates that this genome is heavily methylated during blastula and gastrula stages. Although DNA methylation is largely absent from transcriptional start sites marked with histone H3 lysine 4 trimethylation (H3K4me3), we find both promoters and gene bodies of active genes robustly methylated. In contrast, DNA methylation is absent in large H3K27me3 domains, indicating that these two repression pathways have different roles. Comparison with chromatin state maps of human ES cells reveals strong conservation of epigenetic makeup and gene regulation between the two systems. Strikingly, genes that are highly expressed in pluripotent cells and in Xenopus embryos but not in differentiated cells exhibit relatively high DNA methylation. Therefore, we tested the repressive potential of DNA methylation using transient and transgenic approaches and show that methylated promoters are robustly transcribed in blastula- and gastrula-stage embryos, but not in oocytes or late embryos. These findings have implications for reprogramming and the epigenetic regulation of pluripotency and differentiation and suggest a relatively open, pliable chromatin state in early embryos followed by reestablished methylation-dependent transcriptional repression during organogenesis and differentiation.


Subject(s)
DNA/metabolism , Transcription, Genetic , Xenopus/embryology , Animals , Cell Differentiation , Chromatin/metabolism , DNA Methylation , Embryo, Nonmammalian/metabolism , Embryonic Development , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Histones/metabolism , Humans , Promoter Regions, Genetic , Xenopus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL