Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 152
Filter
1.
Cell ; 162(5): 961-73, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26317465

ABSTRACT

DNA-demethylating agents have shown clinical anti-tumor efficacy via an unknown mechanism of action. Using a combination of experimental and bioinformatics analyses in colorectal cancer cells, we demonstrate that low-dose 5-AZA-CdR targets colorectal cancer-initiating cells (CICs) by inducing viral mimicry. This is associated with induction of dsRNAs derived at least in part from endogenous retroviral elements, activation of the MDA5/MAVS RNA recognition pathway, and downstream activation of IRF7. Indeed, disruption of virus recognition pathways, by individually knocking down MDA5, MAVS, or IRF7, inhibits the ability of 5-AZA-CdR to target colorectal CICs and significantly decreases 5-AZA-CdR long-term growth effects. Moreover, transfection of dsRNA into CICs can mimic the effects of 5-AZA-CdR. Together, our results represent a major shift in understanding the anti-tumor mechanisms of DNA-demethylating agents and highlight the MDA5/MAVS/IRF7 pathway as a potentially druggable target against CICs.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Azacitidine/pharmacology , Cells, Cultured , DEAD-box RNA Helicases/metabolism , DNA Methylation/drug effects , Decitabine , Endogenous Retroviruses/metabolism , Humans , Interferon Regulatory Factor-7/metabolism , Interferon-Induced Helicase, IFIH1 , Mice , RNA, Double-Stranded/metabolism , Receptors, Retinoic Acid/metabolism , Signal Transduction
2.
Cell ; 147(6): 1283-94, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22153073

ABSTRACT

Key regulatory genes, suppressed by Polycomb and H3K27me3, become active during normal differentiation and induced reprogramming. Using the well-characterized enhancer/promoter pair of MYOD1 as a model, we have identified a critical role for enhancers in reprogramming. We observed an unexpected nucleosome-depleted region (NDR) at the H3K4me1-enriched enhancer at which transcriptional regulators initially bind, leading to subsequent changes in the chromatin at the cognate promoter. Exogenous Myod1 activates its own transcription by binding first at the enhancer, leading to an NDR and transcription-permissive chromatin at the associated MYOD1 promoter. Exogenous OCT4 also binds first to the permissive MYOD1 enhancer but has a different effect on the cognate promoter, where the monovalent H3K27me3 marks are converted to the bivalent state characteristic of stem cells. Genome-wide, a high percentage of Polycomb targets are associated with putative enhancers in permissive states, suggesting that they may provide a widespread avenue for the initiation of cell-fate reprogramming.


Subject(s)
Enhancer Elements, Genetic , Repressor Proteins/metabolism , Animals , Cell Line , Epigenomics , Fibroblasts/metabolism , Humans , Mice , MyoD Protein/genetics , Nucleosomes/metabolism , Octamer Transcription Factor-3/metabolism , Polycomb-Group Proteins , Promoter Regions, Genetic
3.
Nature ; 586(7827): 151-155, 2020 10.
Article in English | MEDLINE | ID: mdl-32968275

ABSTRACT

CpG methylation by de novo DNA methyltransferases (DNMTs) 3A and 3B is essential for mammalian development and differentiation and is frequently dysregulated in cancer1. These two DNMTs preferentially bind to nucleosomes, yet cannot methylate the DNA wrapped around the nucleosome core2, and they favour the methylation of linker DNA at positioned nucleosomes3,4. Here we present the cryo-electron microscopy structure of a ternary complex of catalytically competent DNMT3A2, the catalytically inactive accessory subunit DNMT3B3 and a nucleosome core particle flanked by linker DNA. The catalytic-like domain of the accessory DNMT3B3 binds to the acidic patch of the nucleosome core, which orients the binding of DNMT3A2 to the linker DNA. The steric constraints of this arrangement suggest that nucleosomal DNA must be moved relative to the nucleosome core for de novo methylation to occur.


Subject(s)
Cryoelectron Microscopy , DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA (Cytosine-5-)-Methyltransferases/metabolism , Nucleosomes/metabolism , Animals , Biocatalysis , Chromatin Assembly and Disassembly , DNA/chemistry , DNA/metabolism , DNA Methylation , DNA Methyltransferase 3A , Histones/chemistry , Histones/genetics , Histones/metabolism , Humans , Models, Molecular , Nucleosomes/chemistry , Protein Binding , Protein Domains , Xenopus/genetics , DNA Methyltransferase 3B
4.
Nucleic Acids Res ; 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38874474

ABSTRACT

Long terminal repeats (LTRs), which often contain promoter and enhancer sequences of intact endogenous retroviruses (ERVs), are known to be co-opted as cis-regulatory elements for fine-tuning host-coding gene expression. Since LTRs are mainly silenced by the deposition of repressive epigenetic marks, substantial activation of LTRs has been found in human cells after treatment with epigenetic inhibitors. Although the LTR12C family makes up the majority of ERVs activated by epigenetic inhibitors, how these epigenetically and transcriptionally activated LTR12C elements can regulate the host-coding gene expression remains unclear due to genome-wide alteration of transcriptional changes after epigenetic inhibitor treatments. Here, we specifically transactivated >600 LTR12C elements by using single guide RNA-based dCas9-SunTag-VP64, a site-specific targeting CRISPR activation (CRISPRa) system, with minimal off-target events. Interestingly, most of the transactivated LTR12C elements acquired the H3K27ac-marked enhancer feature, while only 20% were co-marked with promoter-associated H3K4me3 modifications. The enrichment of the H3K4me3 signal was intricately associated with downstream regions of LTR12C, such as internal regions of intact ERV9 or other types of retrotransposons. Here, we leverage an optimized CRISPRa system to identify two distinct epigenetic signatures that define LTR12C transcriptional activation, which modulate the expression of proximal protein-coding genes.

5.
Nucleic Acids Res ; 51(12): 5997-6005, 2023 07 07.
Article in English | MEDLINE | ID: mdl-37094063

ABSTRACT

CpG methylation generally occurs on both DNA strands and is essential for mammalian development and differentiation. Until recently, hemimethylation, in which only one strand is methylated, was considered to be simply a transitory state generated during DNA synthesis. The discovery that a subset of CCCTC-binding factor (CTCF) binding sites is heritably hemimethylated suggests that hemimethylation might have an unknown biological function. Here we show that the binding of CTCF is profoundly altered by which DNA strand is methylated and by the specific CTCF binding motif. CpG methylation on the motif strand can inhibit CTCF binding by up to 7-fold, whereas methylation on the opposite strand can stimulate binding by up to 4-fold. Thus, hemimethylation can alter binding by up to 28-fold in a strand-specific manner. The mechanism for sensing methylation on the opposite strand requires two critical residues, V454 and S364, within CTCF zinc fingers 7 and 4. Similar to methylation, CpG hydroxymethylation on the motif strand can inhibit CTCF binding by up to 4-fold. However, hydroxymethylation on the opposite strand removes the stimulatory effect. Strand-specific methylation states may therefore provide a mechanism to explain the transient and dynamic nature of CTCF-mediated chromatin interactions.


Subject(s)
CCCTC-Binding Factor , DNA Methylation , Repressor Proteins , Animals , Binding Sites , CCCTC-Binding Factor/metabolism , Chromatin , CpG Islands , DNA/metabolism , Mammals/genetics , Repressor Proteins/metabolism
6.
Mol Cell ; 62(3): 422-431, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27153539

ABSTRACT

The role of cytosine methylation in the structure and function of enhancers is not well understood. In this study, we investigate the role of DNA methylation at enhancers by comparing the epigenomes of the HCT116 cell line and its highly demethylated derivative, DKO1. Unlike promoters, a portion of regular and super- or stretch enhancers show active H3K27ac marks co-existing with extensive DNA methylation, demonstrating the unexpected presence of bivalent chromatin in both cultured and uncultured cells. Furthermore, our findings also show that bivalent regions have fewer nucleosome-depleted regions and transcription factor-binding sites than monovalent regions. Reduction of DNA methylation genetically or pharmacologically leads to a decrease of the H3K27ac mark. Thus, DNA methylation plays an unexpected dual role at enhancer regions, being anti-correlated focally at transcription factor-binding sites but positively correlated globally with the active H3K27ac mark to ensure structural enhancer integrity.


Subject(s)
DNA Methylation , Enhancer Elements, Genetic , Epigenesis, Genetic , Histones/metabolism , Protein Processing, Post-Translational , Acetylation , Binding Sites , Chromatin Assembly and Disassembly , Cytosine , HCT116 Cells , Histones/genetics , Humans , Time Factors
7.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Article in English | MEDLINE | ID: mdl-33723081

ABSTRACT

Genomic imprinting occurs before fertilization, impacts every cell of the developing child, and may be sensitive to environmental perturbations. The noncoding RNA, nc886 (also called VTRNA2-1) is the only known example of the ∼100 human genes imprinted by DNA methylation, that shows polymorphic imprinting in the population. The nc886 gene is part of an ∼1.6-kb differentially methylated region (DMR) that is methylated in the oocyte and silenced on the maternal allele in about 75% of humans worldwide. Here, we show that the presence or absence of imprinting at the nc886 DMR in an individual is consistent across different tissues, confirming that the imprint is established before cellular differentiation and is maintained into adulthood. We investigated the relationships between the frequency of imprinting in newborns and maternal age, alcohol consumption and cigarette smoking before conception in more than 1,100 mother/child pairs from South Africa. The probability of imprinting in newborns was increased in older mothers and decreased in mothers who drank alcohol before conception. On the other hand, cigarette smoking had no apparent relationship with the frequency of imprinting. These data show an epigenetic change during oocyte maturation which is potentially subject to environmental influence. Much focus has been placed on avoiding alcohol consumption during pregnancy, but our data suggest that drinking before conception may affect the epigenome of the newborn.


Subject(s)
Alcohol Drinking/adverse effects , DNA Methylation , Epigenesis, Genetic , Genomic Imprinting , Maternal Exposure/adverse effects , Oocytes/metabolism , RNA, Untranslated/genetics , Alleles , CpG Islands , Female , High-Throughput Nucleotide Sequencing , Humans , Maternal Age , Pregnancy
8.
Proc Natl Acad Sci U S A ; 117(32): 19359-19366, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32719115

ABSTRACT

Multicellular eukaryotic genomes show enormous differences in size. A substantial part of this variation is due to the presence of transposable elements (TEs). They contribute significantly to a cell's mass of DNA and have the potential to become involved in host gene control. We argue that the suppression of their activities by methylation of the C-phosphate-G (CpG) dinucleotide in DNA is essential for their long-term accommodation in the host genome and, therefore, to its expansion. An inevitable consequence of cytosine methylation is an increase in C-to-T transition mutations via deamination, which causes CpG loss. Cytosine deamination is often needed for TEs to take on regulatory functions in the host genome. Our study of the whole-genome sequences of 53 organisms showed a positive correlation between the size of a genome and the percentage of TEs it contains, as well as a negative correlation between size and the CpG observed/expected (O/E) ratio in both TEs and the host DNA. TEs are seldom found at promoters and transcription start sites, but they are found more at enhancers, particularly after they have accumulated C-to-T and other mutations. Therefore, the methylation of TE DNA allows for genome expansion and also leads to new opportunities for gene control by TE-based regulatory sites.


Subject(s)
DNA Methylation , Eukaryota/genetics , Genome , CpG Islands , Cytosine/metabolism , DNA Transposable Elements , Eukaryota/metabolism , Gene Expression Regulation , Genome Size , Mutation , Promoter Regions, Genetic
9.
Nat Rev Genet ; 17(10): 630-41, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27629931

ABSTRACT

Next-generation sequencing has revealed that more than 50% of human cancers harbour mutations in enzymes that are involved in chromatin organization. Tumour cells not only are activated by genetic and epigenetic alterations, but also routinely use epigenetic processes to ensure their escape from chemotherapy and host immune surveillance. Hence, a growing emphasis of recent drug discovery efforts has been on targeting the epigenome, including DNA methylation and histone modifications, with several new drugs being tested and some already approved by the US Food and Drug Administration (FDA). The future will see the increasing success of combining epigenetic drugs with other therapies. As epigenetic drugs target the epigenome as a whole, these true 'genomic medicines' lessen the need for precision approaches to individualized therapies.


Subject(s)
DNA Methylation/drug effects , Epigenomics/methods , Histone Deacetylase Inhibitors/pharmacology , Neoplasms/drug therapy , Neoplasms/genetics , Epigenesis, Genetic , High-Throughput Nucleotide Sequencing/methods , Histones/metabolism , Humans
10.
Genome Res ; 28(8): 1147-1157, 2018 08.
Article in English | MEDLINE | ID: mdl-29970451

ABSTRACT

We provide a comprehensive genomic and epigenomic map of the more than 500,000 endogenous retroviruses (ERVs) and fragments that populate the intergenic regions of the human genome. The repressive epigenetic marks associated with the ERVs, particularly long terminal repeats (LTRs), show a remarkable switch in silencing mechanisms, depending on the evolutionary age of the LTRs. Young LTRs tend to be CpG rich and are mainly suppressed by DNA methylation, whereas intermediate age LTRs are associated predominantly with histone modifications, particularly histone H3 lysine 9 (H3K9) methylation. Young LTRs can be reactivated by treatment with the DNA methylation inhibitor 5-aza-2'-deoxycytidine (5-aza-CdR) alone, but their level of expression is much increased by 5-aza-CdR treatment plus knockdown of one of several H3K9 methyltransferases or of the H3K27 methyltransferase EZH2. The removal of cytosine methylation led to rapid, widespread increases in H3K9me3 in the LTRs. Intermediate age LTRs had lower CpG densities and were not up-regulated by 5-aza-CdR treatment, but they were sensitive to knockdown of H3K9 methyltransferases. Unlike the situation in embryonic stem cells, the polycomb repressive complex (PRC2) has a minor role in LTR suppression by itself and is only a player after removal of cytosine methylation in the analyzed cancer cell line. Up-regulation of LTRs and induction of "viral mimicry" is rapidly becoming of interest for predicting cancer patient response to epigenetic therapies. Understanding the mechanism for LTR suppression is of major importance in order to improve patient treatment strategies.


Subject(s)
CpG Islands/genetics , DNA Methylation/genetics , Endogenous Retroviruses/genetics , Terminal Repeat Sequences/genetics , Embryonic Stem Cells/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Gene Silencing , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Humans , Polycomb Repressive Complex 2/genetics , Protein Processing, Post-Translational
11.
Proc Natl Acad Sci U S A ; 115(51): E11970-E11977, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30509985

ABSTRACT

Genomic imprinting mediated by DNA methylation restricts gene expression to a single allele determined by parental origin and is not generally considered to be under genetic or environmental influence. Here, we focused on a differentially methylated region (DMR) of approximately 1.9 kb that includes a 101-bp noncoding RNA gene (nc886/VTRNA2-1), which is maternally imprinted in ∼75% of humans. This is unlike other imprinted genes, which demonstrate monoallelic methylation in 100% of individuals. The DMR includes a CTCF binding site on the centromeric side defining the DMR boundary and is flanked by a CTCF binding site on the telomeric side. The centromeric CTCF binding site contains an A/C polymorphism (rs2346018); the C allele is associated with less imprinting. The frequency of imprinting of the nc886 DMR in infants was linked to at least two nongenetic factors, maternal age at delivery and season of conception. In a separate cohort, nc886 imprinting was associated with lower body mass index in children at 5 y of age. Thus, we propose that the imprinting status of the nc886 DMR is "tunable" in that it is associated with maternal haplotype and prenatal environment. This provides a potential mechanism for transmitting information, with phenotypic consequences, from mother to child.


Subject(s)
DNA Methylation , Epigenomics , Genomic Imprinting , Polymorphism, Genetic , Alleles , Binding Sites , CCCTC-Binding Factor , Child , Child, Preschool , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation , Haplotypes , Humans , Maternal Age , MicroRNAs/genetics , Mothers , Pregnancy , RNA, Untranslated/genetics
12.
Semin Cancer Biol ; 51: 59-67, 2018 08.
Article in English | MEDLINE | ID: mdl-29102482

ABSTRACT

Over the past few years it has become clear that vitamin C, as a provider of reduced iron, is an essential factor for the function of epigenetic regulators that initiate the demethylation of DNA and histones. Vitamin C deficiency is rare in the general population, but is frequently observed in patients with cancer. Genes encoding epigenetic regulators are often mutated in cancer, underscoring their central roles in carcinogenesis. In hematological cancers, such as acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS), drugs that reverse epigenetic aberrations are now the standard of care. Recent in vitro studies suggest that vitamin C at physiological concentrations, combined with hypomethylating agents may act synergistically to cause DNA demethylation through active and passive mechanisms, respectively. Additionally, several recent studies have renewed interest in the use of pharmacological doses of vitamin C injected intravenously to selectively kill tumor cells. This review will focus on the potential of vitamin C to optimize the outcome of epigenetic therapy in cancer patients and alternatively to act as a therapeutic at high doses.


Subject(s)
Antioxidants/therapeutic use , Ascorbic Acid/therapeutic use , DNA Methylation , Epigenesis, Genetic , Epigenomics/methods , Gene Expression Regulation, Neoplastic/drug effects , Neoplasms/genetics , Animals , Humans , Neoplasms/drug therapy , Neoplasms/pathology
13.
Proc Natl Acad Sci U S A ; 113(37): 10238-44, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27573823

ABSTRACT

Vitamin C deficiency is found in patients with cancer and might complicate various therapy paradigms. Here we show how this deficiency may influence the use of DNA methyltransferase inhibitors (DNMTis) for treatment of hematological neoplasias. In vitro, when vitamin C is added at physiological levels to low doses of the DNMTi 5-aza-2'-deoxycytidine (5-aza-CdR), there is a synergistic inhibition of cancer-cell proliferation and increased apoptosis. These effects are associated with enhanced immune signals including increased expression of bidirectionally transcribed endogenous retrovirus (ERV) transcripts, increased cytosolic dsRNA, and activation of an IFN-inducing cellular response. This synergistic effect is likely the result of both passive DNA demethylation by DNMTi and active conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) by ten-eleven translocation (TET) enzymes at LTR regions of ERVs, because vitamin C acts as a cofactor for TET proteins. In addition, TET2 knockout reduces the synergy between the two compounds. Furthermore, we show that many patients with hematological neoplasia are markedly vitamin C deficient. Thus, our data suggest that correction of vitamin C deficiency in patients with hematological and other cancers may improve responses to epigenetic therapy with DNMTis.


Subject(s)
Ascorbic Acid/administration & dosage , Azacitidine/analogs & derivatives , Enzyme Inhibitors/administration & dosage , Hematologic Neoplasms/drug therapy , Apoptosis/drug effects , Ascorbic Acid Deficiency/complications , Ascorbic Acid Deficiency/drug therapy , Ascorbic Acid Deficiency/metabolism , Ascorbic Acid Deficiency/pathology , Azacitidine/administration & dosage , Cell Proliferation/drug effects , DNA Methylation/drug effects , DNA-Binding Proteins/genetics , Decitabine , Dioxygenases , Drug Synergism , Endogenous Retroviruses/genetics , Female , Hematologic Neoplasms/complications , Hematologic Neoplasms/pathology , Humans , Interferons/genetics , Male , Methyltransferases/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , RNA, Double-Stranded/drug effects
14.
Genome Res ; 25(4): 467-77, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25747664

ABSTRACT

The holistic role of DNA methylation in the organization of the cancer epigenome is not well understood. Here we perform a comprehensive, high-resolution analysis of chromatin structure to compare the landscapes of HCT116 colon cancer cells and a DNA methylation-deficient derivative. The NOMe-seq accessibility assay unexpectedly revealed symmetrical and transcription-independent nucleosomal phasing across active, poised, and inactive genomic elements. DNA methylation abolished this phasing primarily at enhancers and CpG island (CGI) promoters, with little effect on insulators and non-CGI promoters. Abolishment of DNA methylation led to the context-specific reestablishment of the poised and active states of normal colon cells, which were marked in methylation-deficient cells by distinct H3K27 modifications and the presence of either well-phased nucleosomes or nucleosome-depleted regions, respectively. At higher-order genomic scales, we found that long, H3K9me3-marked domains had lower accessibility, consistent with a more compact chromatin structure. Taken together, our results demonstrate the nuanced and context-dependent role of DNA methylation in the functional, multiscale organization of cancer epigenomes.


Subject(s)
Chromatin/genetics , Colonic Neoplasms/genetics , DNA Methylation/genetics , Cell Line, Tumor , CpG Islands/genetics , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/genetics , Epigenesis, Genetic , HCT116 Cells , Histones/genetics , Humans , Nucleosomes/genetics , Promoter Regions, Genetic/genetics , DNA Methyltransferase 3B
15.
Nat Rev Genet ; 13(7): 484-92, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22641018

ABSTRACT

DNA methylation is frequently described as a 'silencing' epigenetic mark, and indeed this function of 5-methylcytosine was originally proposed in the 1970s. Now, thanks to improved genome-scale mapping of methylation, we can evaluate DNA methylation in different genomic contexts: transcriptional start sites with or without CpG islands, in gene bodies, at regulatory elements and at repeat sequences. The emerging picture is that the function of DNA methylation seems to vary with context, and the relationship between DNA methylation and transcription is more nuanced than we realized at first. Improving our understanding of the functions of DNA methylation is necessary for interpreting changes in this mark that are observed in diseases such as cancer.


Subject(s)
CpG Islands/genetics , DNA Methylation/genetics , DNA Methylation/physiology , Gene Expression Regulation/physiology , Genes/genetics , Regulatory Elements, Transcriptional/genetics , Repetitive Sequences, Nucleic Acid/genetics , Transcription Initiation Site/physiology , CpG Islands/physiology , Gene Expression Regulation/genetics , Genes/physiology , Models, Molecular , Regulatory Elements, Transcriptional/physiology , Repetitive Sequences, Nucleic Acid/physiology
16.
Mol Cell ; 39(6): 901-11, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20864037

ABSTRACT

Profound chromatin changes occur during mitosis to allow for gene silencing and chromosome segregation followed by reactivation of memorized transcription states in daughter cells. Using genome-wide sequencing, we found H2A.Z-containing +1 nucleosomes of active genes shift upstream to occupy TSSs during mitosis, significantly reducing nucleosome-depleted regions. Single-molecule analysis confirmed nucleosome shifting and demonstrated that mitotic shifting is specific to active genes that are silenced during mitosis and, thus, is not seen on promoters, which are silenced by methylation or mitotically expressed genes. Using the GRP78 promoter as a model, we found H3K4 trimethylation is also maintained while other indicators of active chromatin are lost and expression is decreased. These key changes provide a potential mechanism for rapid silencing and reactivation of genes during the cell cycle.


Subject(s)
Gene Silencing , Histones/metabolism , Mitosis/genetics , Nucleosomes/metabolism , Acetylation , CCAAT-Binding Factor/metabolism , Cell Cycle Proteins/genetics , Cell Division/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Methylation/physiology , DNA Polymerase II/metabolism , Endoplasmic Reticulum Chaperone BiP , G1 Phase/genetics , Gene Expression/genetics , Genes, p16/physiology , Heat-Shock Proteins/genetics , Humans , Membrane Proteins/genetics , Methylation , Models, Genetic , Phosphorylation/physiology , Promoter Regions, Genetic/physiology , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Resting Phase, Cell Cycle/genetics , Sequence Analysis, DNA , TATA-Box Binding Protein/metabolism , Transcription Initiation Site/physiology , Polo-Like Kinase 1
17.
Genome Res ; 24(9): 1421-32, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24916973

ABSTRACT

It is well established that cancer-associated epigenetic repression occurs concomitant with CpG island hypermethylation and loss of nucleosomes at promoters, but the role of nucleosome occupancy and epigenetic reprogramming at distal regulatory elements in cancer is still poorly understood. Here, we evaluate the scope of global epigenetic alterations at enhancers and insulator elements in prostate and breast cancer cells using simultaneous genome-wide mapping of DNA methylation and nucleosome occupancy (NOMe-seq). We find that the genomic location of nucleosome-depleted regions (NDRs) is mostly cell type specific and preferentially found at enhancers in normal cells. In cancer cells, however, we observe a global reconfiguration of NDRs at distal regulatory elements coupled with a substantial reorganization of the cancer methylome. Aberrant acquisition of nucleosomes at enhancer-associated NDRs is associated with hypermethylation and epigenetic silencing marks, and conversely, loss of nucleosomes with demethylation and epigenetic activation. Remarkably, we show that nucleosomes remain strongly organized and phased at many facultative distal regulatory elements, even in the absence of a NDR as an anchor. Finally, we find that key transcription factor (TF) binding sites also show extensive peripheral nucleosome phasing, suggesting the potential for TFs to organize NDRs genome-wide and contribute to deregulation of cancer epigenomes. Together, our findings suggest that "decommissioning" of NDRs and TFs at distal regulatory elements in cancer cells is accompanied by DNA hypermethylation susceptibility of enhancers and insulator elements, which in turn may contribute to an altered genome-wide architecture and epigenetic deregulation in malignancy.


Subject(s)
DNA Methylation , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Insulator Elements , Nucleosomes/genetics , Epigenesis, Genetic , Humans , MCF-7 Cells , Nucleosomes/metabolism
18.
Genome Res ; 24(4): 545-53, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24515120

ABSTRACT

Extracellular cues play critical roles in the establishment of the epigenome during development and may also contribute to epigenetic perturbations found in disease states. The direct role of the local tissue environment on the post-development human epigenome, however, remains unclear due to limitations in studies of human subjects. Here, we use an isogenic human ileal neobladder surgical model and compare global DNA methylation levels of intestinal epithelial cells pre- and post-neobladder construction using the Infinium HumanMethylation450 BeadChip. Our study is the first to quantify the effect of environmental cues on the human epigenome and show that the local tissue environment directly modulates DNA methylation patterns in normal differentiated cells in vivo. In the neobladder, the intestinal epithelial cells lose their tissue-specific epigenetic landscape in a time-dependent manner following the tissue's exposure to a bladder environment. We find that de novo methylation of many intestine-specific enhancers occurs at the rate of 0.41% per month (P < 0.01, Pearson = 0.71), while demethylation of primarily non-intestine-specific transcribed regions occurs at the rate of -0.37% per month (P < 0.01, Pearson = -0.57). The dynamic resetting of the DNA methylome in the neobladder not only implicates local environmental cues in the shaping and maintenance of the epigenome but also illustrates an unexpected cross-talk between the epigenome and the cellular environment.


Subject(s)
Cell Differentiation/genetics , DNA Methylation/genetics , Epigenesis, Genetic , Intestines/growth & development , Aged , CpG Islands , Genome, Human , Humans , Intestines/surgery , Intestines/transplantation , Middle Aged , Tissue Transplantation
19.
Genome Res ; 24(3): 454-66, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24299735

ABSTRACT

Epigenetic information is available from contemporary organisms, but is difficult to track back in evolutionary time. Here, we show that genome-wide epigenetic information can be gathered directly from next-generation sequence reads of DNA isolated from ancient remains. Using the genome sequence data generated from hair shafts of a 4000-yr-old Paleo-Eskimo belonging to the Saqqaq culture, we generate the first ancient nucleosome map coupled with a genome-wide survey of cytosine methylation levels. The validity of both nucleosome map and methylation levels were confirmed by the recovery of the expected signals at promoter regions, exon/intron boundaries, and CTCF sites. The top-scoring nucleosome calls revealed distinct DNA positioning biases, attesting to nucleotide-level accuracy. The ancient methylation levels exhibited high conservation over time, clustering closely with modern hair tissues. Using ancient methylation information, we estimated the age at death of the Saqqaq individual and illustrate how epigenetic information can be used to infer ancient gene expression. Similar epigenetic signatures were found in other fossil material, such as 110,000- to 130,000-yr-old bones, supporting the contention that ancient epigenomic information can be reconstructed from a deep past. Our findings lay the foundation for extracting epigenomic information from ancient samples, allowing shifts in epialleles to be tracked through evolutionary time, as well as providing an original window into modern epigenomics.


Subject(s)
Cytosine/metabolism , DNA Methylation , Genome, Human , Inuit/genetics , Nucleosomes/genetics , Animals , Chromosome Mapping , Epigenesis, Genetic , Epigenomics , Evolution, Molecular , Gene Expression , Gene Expression Regulation , Humans , Phylogeny , Promoter Regions, Genetic , Sequence Analysis, DNA
20.
Nucleic Acids Res ; 42(4): 2245-56, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24288367

ABSTRACT

A number of genome-wide analyses have revealed that estrogen receptor α binding to and regulation of its target genes correlate with binding of FOXA1, a pioneer factor, to nearby DNA sites in MCF-7 breast cancer cells. The enhancer element-specific histone H3K4me1/2 mark is enriched at the specific FOXA1/ERα recruitment sites in chromatin, but the mechanism by which these enhancer marks are established in chromatin before hormone treatment is unclear. Here, we show that mixed-lineage leukemia 1 (MLL1) protein is a key determinant that maintains permissive chromatin structure of the TFF1 enhancer region. MLL1 occupies the TFF1 enhancer region and methylates H3K4 before hormone stimulation. In vitro, MLL1 binds directly to the CpG-rich region of the TFF1 enhancer, and its binding is dependent on hypomethylation of DNA. Furthermore, the depletion of MLL1 in MCF-7 cells results in a dramatic decrease of chromatin accessibility and recruitment of FOXA1 and ERα to the enhancer element. Our study defines the mechanism by which MLL1 nucleates histone H3K4 methylation marks in CpG-enriched regions to maintain permissive chromatin architecture and allow FOXA1 and estrogen receptor α binding to transcriptional regulatory sites in breast cancer cells.


Subject(s)
Chromatin/chemistry , Enhancer Elements, Genetic , Estrogen Receptor alpha/metabolism , Gene Expression Regulation , Myeloid-Lymphoid Leukemia Protein/metabolism , Transcription, Genetic , Cell Line , Chromatin/metabolism , CpG Islands , Hepatocyte Nuclear Factor 3-alpha/metabolism , Histone-Lysine N-Methyltransferase , Humans , MCF-7 Cells , Trefoil Factor-1 , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL