Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
J Med Chem ; 67(6): 4525-4540, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38294854

ABSTRACT

Ten-eleven translocation enzymes (TETs) are Fe(II)/2-oxoglutarate (2OG) oxygenases that catalyze the sequential oxidation of 5-methylcytosine to 5-hydroxymethylcytosine, 5-formylcytosine, and 5-carboxylcytosine in eukaryotic DNA. Despite their roles in epigenetic regulation, there is a lack of reported TET inhibitors. The extent to which 2OG oxygenase inhibitors, including clinically used inhibitors and oncometabolites, modulate DNA modifications via TETs has been unclear. Here, we report studies on human TET1-3 inhibition by a set of 2OG oxygenase-focused inhibitors, employing both enzyme-based and cellular assays. Most inhibitors manifested similar potencies for TET1-3 and caused increases in cellular 5hmC levels. (R)-2-Hydroxyglutarate, an oncometabolite elevated in isocitrate dehydrogenase mutant cancer cells, showed different degrees of inhibition, with TET1 being less potently inhibited than TET3 and TET2, potentially reflecting the proposed role of TET2 mutations in tumorigenesis. The results highlight the tractability of TETs as drug targets and provide starting points for selective inhibitor design.


Subject(s)
Dioxygenases , Glutarates , Oxygenases , Humans , Epigenesis, Genetic , Mixed Function Oxygenases , Dioxygenases/metabolism , DNA , DNA Methylation , Proto-Oncogene Proteins/metabolism
2.
J Am Chem Soc ; 145(13): 7095-7100, 2023 04 05.
Article in English | MEDLINE | ID: mdl-36961225

ABSTRACT

Selective, efficient, and controllable oxidation of cytosine modifications is valuable for epigenetic analyses, yet only limited progress has been made. Here, we present two modular chemical oxidation reactions: conversion of 5-hydroxymethylcytosine (5hmC) into 5-formylcytosine (5fC) using 4-acetamido-2,2,6,6-tetramethylpiperidine-1-oxoammonium tetrafluoroborate (ACT+BF4-) and further transformation of 5fC into 5-carboxycytosine (5caC) through Pinnick oxidation. Both reactions are mild and efficient on double-stranded DNA. We integrated these two oxidations with borane reduction to develop chemical-assisted pyridine borane sequencing plus (CAPS+), for direct and quantitative mapping of 5hmC. Compared with CAPS, CAPS+ improved the conversion rate and false-positive rate. We applied CAPS+ to mouse embryonic stem cells, human normal brain, and glioblastoma DNA samples and demonstrated its superior sensitivity in analyzing the hydroxymethylome.


Subject(s)
Cystine , Cystine/analysis , Humans , Animals , Mice , DNA Methylation , DNA/genetics , Oxidation-Reduction
3.
Life Sci Alliance ; 5(12)2022 12.
Article in English | MEDLINE | ID: mdl-36122935

ABSTRACT

The DNA-binding protein MeCP2 is reported to bind methylated cytosine in CG and CA motifs in genomic DNA, but it was recently proposed that arrays of tandemly repeated CA containing either methylated or hydroxymethylated cytosine are the primary targets for MeCP2 binding and function. Here we investigated the predictions of this hypothesis using a range of published datasets. We failed to detect enrichment of cytosine modification at genomic CA repeat arrays in mouse brain regions and found no evidence for preferential MeCP2 binding at CA repeats. Moreover, we did not observe a correlation between the CA repeat density near genes and their degree of transcriptional deregulation when MeCP2 was absent. Our results do not provide support for the hypothesis that CA repeats are key mediators of MeCP2 function. Instead, we found that CA repeats are subject to CAC methylation to a degree that is typical of the surrounding genome and contribute modestly to MeCP2-mediated modulation of gene expression in accordance with their content of this canonical target motif.


Subject(s)
Methyl-CpG-Binding Protein 2 , Animals , Cytosine/metabolism , DNA/metabolism , DNA Methylation , Methyl-CpG-Binding Protein 2/metabolism , Mice , Neurons/metabolism
4.
Neuro Oncol ; 24(12): 2093-2106, 2022 12 01.
Article in English | MEDLINE | ID: mdl-35468205

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most common and malignant primary brain tumor in adults. Despite maximal treatment, median survival remains dismal at 14-24 months. Immunotherapies, such as checkpoint inhibition, have revolutionized management of some cancers but have little benefit for GBM patients. This is, in part, due to the low mutational and neoantigen burden in this immunogenically "cold" tumor. METHODS: U87MG and patient-derived cell lines were treated with 5-aza-2'-deoxycytidine (DAC) and underwent whole-exome and transcriptome sequencing. Cell lines were then subjected to cellular assays with neoantigen and cancer testis antigen (CTA) specific T cells. RESULTS: We demonstrate that DAC increases neoantigen and CTA mRNA expression through DNA hypomethylation. This results in increased neoantigen presentation by MHC class I in tumor cells, leading to increased neoantigen- and CTA-specific T-cell activation and killing of DAC-treated cancer cells. In addition, we show that patients have endogenous cancer-specific T cells in both tumor and blood, which show increased tumor-specific activation in the presence of DAC-treated cells. CONCLUSIONS: Our work shows that DAC increases GBM immunogenicity and consequent susceptibility to T-cell responses in vitro. Our results support a potential use of DAC as a sensitizing agent for immunotherapy.


Subject(s)
Glioblastoma , Adult , Male , Humans , Glioblastoma/drug therapy , Glioblastoma/genetics , Decitabine/pharmacology , Antigens, Neoplasm/genetics , T-Lymphocytes , Testis , Cell Line, Tumor
7.
Nucleic Acids Res ; 49(13): e76, 2021 07 21.
Article in English | MEDLINE | ID: mdl-33905495

ABSTRACT

Whole genome base-resolution methylome sequencing allows for the most comprehensive analysis of DNA methylation, however, the considerable sequencing cost often limits its applications. While reduced representation sequencing can be an affordable alternative, over 80% of CpGs in the genome are not covered. Building on our recently developed TET-assisted pyridine borane sequencing (TAPS) method, we here described endonuclease enrichment TAPS (eeTAPS), which utilizes dihydrouracil (DHU)-cleaving endonuclease digestion of TAPS-converted DNA to enrich methylated CpG sites (mCpGs). eeTAPS can accurately detect 87% of mCpGs in the mouse genome with a sequencing depth equivalent to 4× whole genome sequencing. In comparison, reduced representation TAPS (rrTAPS) detected less than 4% of mCpGs with 2.5× sequencing depth. Our results demonstrate eeTAPS to be a new strategy for cost-effective genome-wide methylation analysis at single-CpG resolution that can fill the gap between whole-genome and reduced representation sequencing.


Subject(s)
DNA Methylation , Sequence Analysis, DNA/methods , Animals , Cells, Cultured , Cost-Benefit Analysis , CpG Islands , Deoxyribonuclease (Pyrimidine Dimer) , Embryonic Stem Cells/metabolism , Genomics/methods , Mice , Sequence Analysis, DNA/economics , Uracil-DNA Glycosidase
8.
Mol Cell ; 81(6): 1260-1275.e12, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33561390

ABSTRACT

DNA methylation is implicated in neuronal biology via the protein MeCP2, the mutation of which causes Rett syndrome. MeCP2 recruits the NCOR1/2 co-repressor complexes to methylated cytosine in the CG dinucleotide, but also to sites of non-CG methylation, which are abundant in neurons. To test the biological significance of the dual-binding specificity of MeCP2, we replaced its DNA binding domain with an orthologous domain from MBD2, which can only bind mCG motifs. Knockin mice expressing the domain-swap protein displayed severe Rett-syndrome-like phenotypes, indicating that normal brain function requires the interaction of MeCP2 with sites of non-CG methylation, specifically mCAC. The results support the notion that the delayed onset of Rett syndrome is due to the simultaneous post-natal accumulation of mCAC and its reader MeCP2. Intriguingly, genes dysregulated in both Mecp2 null and domain-swap mice are implicated in other neurological disorders, potentially highlighting targets of relevance to the Rett syndrome phenotype.


Subject(s)
DNA Methylation , Methyl-CpG-Binding Protein 2/metabolism , Neurons/metabolism , Animals , CpG Islands , Gene Knock-In Techniques , HeLa Cells , Humans , Male , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Transgenic , Mutation , NIH 3T3 Cells , Neurons/pathology , Protein Domains , Rett Syndrome/genetics , Rett Syndrome/metabolism , Rett Syndrome/pathology
9.
Genome Res ; 30(10): 1393-1406, 2020 10.
Article in English | MEDLINE | ID: mdl-32963030

ABSTRACT

Epigenetic modifications on chromatin play important roles in regulating gene expression. Although chromatin states are often governed by multilayered structure, how individual pathways contribute to gene expression remains poorly understood. For example, DNA methylation is known to regulate transcription factor binding but also to recruit methyl-CpG binding proteins that affect chromatin structure through the activity of histone deacetylase complexes (HDACs). Both of these mechanisms can potentially affect gene expression, but the importance of each, and whether these activities are integrated to achieve appropriate gene regulation, remains largely unknown. To address this important question, we measured gene expression, chromatin accessibility, and transcription factor occupancy in wild-type or DNA methylation-deficient mouse embryonic stem cells following HDAC inhibition. We observe widespread increases in chromatin accessibility at retrotransposons when HDACs are inhibited, and this is magnified when cells also lack DNA methylation. A subset of these elements has elevated binding of the YY1 and GABPA transcription factors and increased expression. The pronounced additive effect of HDAC inhibition in DNA methylation-deficient cells demonstrates that DNA methylation and histone deacetylation act largely independently to suppress transcription factor binding and gene expression.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Histone Deacetylases/metabolism , Histones/metabolism , Transcription Factors/metabolism , Acetylation , Chromatin/metabolism , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/enzymology , Embryonic Stem Cells/metabolism , Genome , Histone Deacetylase Inhibitors , Histone Deacetylases/pharmacology , Retroelements
10.
Nat Chem Biol ; 16(6): 604-605, 2020 06.
Article in English | MEDLINE | ID: mdl-32300239

Subject(s)
DNA , Genomics , Animals , Deoxyadenosines
11.
Mol Cell ; 77(6): 1159-1161, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32200796

ABSTRACT

Distal regulatory elements control gene expression during differentiation. In this issue of Molecular Cell, Barnett et al. (2020) develop a new technology, called ATAC-Me, and discover that removal of DNA methylation is not a pre-requisite for the creation of accessible chromatin at active gene regulatory elements during cellular differentiation.


Subject(s)
Chromatin , DNA Methylation , Cell Differentiation , Regulatory Sequences, Nucleic Acid
12.
Sci Rep ; 9(1): 13463, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31530880

ABSTRACT

Expression of the mismatch repair gene MutL homolog 1 (MLH1) is silenced in a clinically important subgroup of sporadic colorectal cancers. These cancers exhibit hypermutability with microsatellite instability (MSI) and differ from microsatellite-stable (MSS) colorectal cancers in both prognosis and response to therapies. Loss of MLH1 is usually due to epigenetic silencing with associated promoter methylation; coding somatic mutations rarely occur. Here we use the presence of a colorectal cancer (CRC) risk variant (rs1800734) within the MLH1 promoter to investigate the poorly understood mechanisms of MLH1 promoter methylation and loss of expression. We confirm the association of rs1800734 with MSI+ but not MSS cancer risk in our own data and by meta-analysis. Using sensitive allele-specific detection methods, we demonstrate that MLH1 is the target gene for rs1800734 mediated cancer risk. In normal colon tissue, small allele-specific differences exist only in MLH1 promoter methylation, but not gene expression. In contrast, allele-specific differences in both MLH1 methylation and expression are present in MSI+ cancers. We show that MLH1 transcriptional repression is dependent on DNA methylation and can be reversed by a methylation inhibitor. The rs1800734 allele influences the rate of methylation loss and amount of re-expression. The transcription factor TFAP4 binds to the rs1800734 region but with much weaker binding to the risk than the protective allele. TFAP4 binding is absent on both alleles when promoter methylation is present. Thus we propose that TFAP4 binding shields the protective rs1800734 allele of the MLH1 promoter from BRAF induced DNA methylation more effectively than the risk allele.


Subject(s)
Colorectal Neoplasms/genetics , DNA-Binding Proteins/metabolism , MutL Protein Homolog 1/genetics , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Transcription Factors/metabolism , Alleles , Case-Control Studies , CpG Islands , DNA Methylation , DNA-Binding Proteins/genetics , Databases, Factual , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Microsatellite Instability , MutL Protein Homolog 1/metabolism , RNA, Messenger/genetics , Transcription Factors/genetics
13.
Nat Methods ; 16(5): 429-436, 2019 05.
Article in English | MEDLINE | ID: mdl-31011185

ABSTRACT

Replication of eukaryotic genomes is highly stochastic, making it difficult to determine the replication dynamics of individual molecules with existing methods. We report a sequencing method for the measurement of replication fork movement on single molecules by detecting nucleotide analog signal currents on extremely long nanopore traces (D-NAscent). Using this method, we detect 5-bromodeoxyuridine (BrdU) incorporated by Saccharomyces cerevisiae to reveal, at a genomic scale and on single molecules, the DNA sequences replicated during a pulse-labeling period. Under conditions of limiting BrdU concentration, D-NAscent detects the differences in BrdU incorporation frequency across individual molecules to reveal the location of active replication origins, fork direction, termination sites, and fork pausing/stalling events. We used sequencing reads of 20-160 kilobases to generate a whole-genome single-molecule map of DNA replication dynamics and discover a class of low-frequency stochastic origins in budding yeast. The D-NAscent software is available at https://github.com/MBoemo/DNAscent.git .


Subject(s)
DNA Replication , Genome, Fungal , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Nanopores , Saccharomyces cerevisiae/genetics , Bromodeoxyuridine/metabolism , DNA, Fungal/genetics , Genome , Software
15.
Nat Commun ; 10(1): 30, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30604745

ABSTRACT

The inactive X chromosome (Xi) in female mammals adopts an atypical higher-order chromatin structure, manifested as a global loss of local topologically associated domains (TADs), A/B compartments and formation of two mega-domains. Here we demonstrate that the non-canonical SMC family protein, SmcHD1, which is important for gene silencing on Xi, contributes to this unique chromosome architecture. Specifically, allelic mapping of the transcriptome and epigenome in SmcHD1 mutant cells reveals the appearance of sub-megabase domains defined by gene activation, CpG hypermethylation and depletion of Polycomb-mediated H3K27me3. These domains, which correlate with sites of SmcHD1 enrichment on Xi in wild-type cells, additionally adopt features of active X chromosome higher-order chromosome architecture, including A/B compartments and partial restoration of TAD boundaries. Xi chromosome architecture changes also occurred following SmcHD1 knockout in a somatic cell model, but in this case, independent of Xi gene derepression. We conclude that SmcHD1 is a key factor in defining the unique chromosome architecture of Xi.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , DNA Methylation/genetics , Transcriptional Activation/genetics , X Chromosome Inactivation , Alleles , Animals , CRISPR-Cas Systems , Cell Line , Chromosomal Proteins, Non-Histone/metabolism , CpG Islands , Exons/genetics , Female , Fibroblasts , Gene Knockout Techniques , Histones/genetics , Histones/metabolism , Male , Mice , Point Mutation , Polycomb-Group Proteins/metabolism
16.
PLoS Genet ; 14(10): e1007643, 2018 10.
Article in English | MEDLINE | ID: mdl-30335751

ABSTRACT

The Fanconi Anemia (FA) pathway is important for repairing interstrand crosslinks (ICLs) between the Watson-Crick strands of the DNA double helix. An initial and essential stage in the repair process is the detection of the ICL. Here, we report the identification of UHRF2, a paralogue of UHRF1, as an ICL sensor protein. UHRF2 is recruited to ICLs in the genome within seconds of their appearance. We show that UHRF2 cooperates with UHRF1, to ensure recruitment of FANCD2 to ICLs. A direct protein-protein interaction is formed between UHRF1 and UHRF2, and between either UHRF1 and UHRF2, and FANCD2. Importantly, we demonstrate that the essential monoubiquitination of FANCD2 is stimulated by UHRF1/UHRF2. The stimulation is mediating by a retention of FANCD2 on chromatin, allowing for its monoubiquitination by the FA core complex. Taken together, we uncover a mechanism of ICL sensing by UHRF2, leading to FANCD2 recruitment and retention at ICLs, in turn facilitating activation of FANCD2 by monoubiquitination.


Subject(s)
DNA Repair/physiology , Fanconi Anemia Complementation Group D2 Protein/physiology , Ubiquitin-Protein Ligases/physiology , Amino Acid Sequence , CCAAT-Enhancer-Binding Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/physiology , Cell Line , Cell Nucleus/metabolism , Chromatin/metabolism , DNA/metabolism , DNA Damage/physiology , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Protein Interaction Domains and Motifs , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
17.
Genome Biol ; 19(1): 129, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30201020

ABSTRACT

BACKGROUND: DNA replication plays an important role in mutagenesis, yet little is known about how it interacts with other mutagenic processes. Here, we use somatic mutation signatures-each representing a mutagenic process-derived from 3056 patients spanning 19 cancer types to quantify the strand asymmetry of mutational signatures around replication origins and between early and late replicating regions. RESULTS: We observe that most of the detected mutational signatures are significantly correlated with the timing or direction of DNA replication. The properties of these associations are distinct for different signatures and shed new light on several mutagenic processes. For example, our results suggest that oxidative damage to the nucleotide pool substantially contributes to the mutational landscape of esophageal adenocarcinoma. CONCLUSIONS: Together, our results indicate an interaction between DNA replication, the associated damage repair, and most mutagenic processes.


Subject(s)
DNA Replication Timing , Mutagenesis , DNA Repair , Esophageal Neoplasms/genetics , Humans , Mutagens/toxicity , Mutation , Neoplasms/genetics
18.
Br J Cancer ; 118(12): 1683, 2018 06.
Article in English | MEDLINE | ID: mdl-29780161

ABSTRACT

Since the publication of this paper, the authors noticed that James E. East was assigned to the incorrect affiliation. The affiliation information is provided correctly, above.

19.
Cell Rep ; 23(8): 2225-2235, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29791834

ABSTRACT

Preclinical work has long focused on male animals, though biological sex clearly influences risk for certain diseases, including many psychiatric disorders. Such disorders are often treated by drugs targeting the CNS norepinephrine system. Despite roles for noradrenergic neurons in behavior and neuropsychiatric disease models, their molecular characterization has lagged. We profiled mouse noradrenergic neurons in vivo, defining over 3,000 high-confidence transcripts expressed therein, including druggable receptors. We uncovered remarkable sex differences in gene expression, including elevated expression of the EP3 receptor in females-which we leverage to illustrate the behavioral and pharmacologic relevance of these findings-and of Slc6a15 and Lin28b, both major depressive disorder (MDD)-associated genes. Broadly, we present a means of transcriptionally profiling locus coeruleus under baseline and experimental conditions. Our findings underscore the need for preclinical work to include both sexes and suggest that sex differences in noradrenergic neurons may underlie behavioral differences relevant to disease.


Subject(s)
Adrenergic Neurons/metabolism , Locus Coeruleus/metabolism , Sex Characteristics , Animals , Behavior, Animal , Female , Gene Expression Regulation , Lipopolysaccharides , Male , Mice , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Reproducibility of Results , Ribosomes/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...