Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 362
Filter
1.
Annu Rev Biochem ; 83: 585-614, 2014.
Article in English | MEDLINE | ID: mdl-24905787

ABSTRACT

The importance of eukaryotic DNA methylation [5-methylcytosine (5mC)] in transcriptional regulation and development was first suggested almost 40 years ago. However, the molecular mechanism underlying the dynamic nature of this epigenetic mark was not understood until recently, following the discovery that the TET proteins, a family of AlkB-like Fe(II)/α-ketoglutarate-dependent dioxygenases, can oxidize 5mC to generate 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and 5-carboxylcytosine (5caC). Since then, several mechanisms that are responsible for processing oxidized 5mC derivatives to achieve DNA demethylation have emerged. Our biochemical understanding of the DNA demethylation process has prompted new investigations into the biological functions of DNA demethylation. Characterization of two additional AlkB family proteins, FTO and ALKBH5, showed that they possess demethylase activity toward N(6)-methyladenosine (m(6)A) in RNA, indicating that members of this subfamily of dioxygenases have a general function in demethylating nucleic acids. In this review, we discuss recent advances in this emerging field, focusing on the mechanism and function of TET-mediated DNA demethylation.


Subject(s)
DNA Methylation , DNA/chemistry , Gene Expression Regulation , Oxygen/chemistry , RNA/chemistry , 5-Methylcytosine/chemistry , Animals , Cytosine/analogs & derivatives , Cytosine/chemistry , Escherichia coli/metabolism , Genome , Germ Cells/cytology , HEK293 Cells , Humans , Methylation , Mice , Neoplasms/genetics , Stem Cells/cytology , Transcriptome
2.
Methods ; 227: 37-47, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38729455

ABSTRACT

RNA modification serves as a pivotal component in numerous biological processes. Among the prevalent modifications, 5-methylcytosine (m5C) significantly influences mRNA export, translation efficiency and cell differentiation and are also associated with human diseases, including Alzheimer's disease, autoimmune disease, cancer, and cardiovascular diseases. Identification of m5C is critically responsible for understanding the RNA modification mechanisms and the epigenetic regulation of associated diseases. However, the large-scale experimental identification of m5C present significant challenges due to labor intensity and time requirements. Several computational tools, using machine learning, have been developed to supplement experimental methods, but identifying these sites lack accuracy and efficiency. In this study, we introduce a new predictor, MLm5C, for precise prediction of m5C sites using sequence data. Briefly, we evaluated eleven RNA sequence-derived features with four basic machine learning algorithms to generate baseline models. From these 44 models, we ranked them based on their performance and subsequently stacked the Top 20 baseline models as the best model, named MLm5C. The MLm5C outperformed the-state-of-the-art predictors. Notably, the optimization of the sequence length surrounding the modification sites significantly improved the prediction performance. MLm5C is an invaluable tool in accelerating the detection of m5C sites within the human genome, thereby facilitating in the characterization of their roles in post-transcriptional regulation.


Subject(s)
5-Methylcytosine , Machine Learning , RNA , Humans , 5-Methylcytosine/metabolism , 5-Methylcytosine/chemistry , RNA/genetics , RNA/chemistry , RNA/metabolism , Computational Biology/methods , RNA Processing, Post-Transcriptional , Algorithms
3.
Nature ; 569(7757): 581-585, 2019 05.
Article in English | MEDLINE | ID: mdl-31043749

ABSTRACT

Methylation of cytosine to 5-methylcytosine (5mC) is a prevalent DNA modification found in many organisms. Sequential oxidation of 5mC by ten-eleven translocation (TET) dioxygenases results in a cascade of additional epigenetic marks and promotes demethylation of DNA in mammals1,2. However, the enzymatic activity and function of TET homologues in other eukaryotes remains largely unexplored. Here we show that the green alga Chlamydomonas reinhardtii contains a 5mC-modifying enzyme (CMD1) that is a TET homologue and catalyses the conjugation of a glyceryl moiety to the methyl group of 5mC through a carbon-carbon bond, resulting in two stereoisomeric nucleobase products. The catalytic activity of CMD1 requires Fe(II) and the integrity of its binding motif His-X-Asp, which is conserved in Fe-dependent dioxygenases3. However, unlike previously described TET enzymes, which use 2-oxoglutarate as a co-substrate4, CMD1 uses L-ascorbic acid (vitamin C) as an essential co-substrate. Vitamin C donates the glyceryl moiety to 5mC with concurrent formation of glyoxylic acid and CO2. The vitamin-C-derived DNA modification is present in the genome of wild-type C. reinhardtii but at a substantially lower level in a CMD1 mutant strain. The fitness of CMD1 mutant cells during exposure to high light levels is reduced. LHCSR3, a gene that is critical for the protection of C. reinhardtii from photo-oxidative damage under high light conditions, is hypermethylated and downregulated in CMD1 mutant cells compared to wild-type cells, causing a reduced capacity for photoprotective non-photochemical quenching. Our study thus identifies a eukaryotic DNA base modification that is catalysed by a divergent TET homologue and unexpectedly derived from vitamin C, and describes its role as a potential epigenetic mark that may counteract DNA methylation in the regulation of photosynthesis.


Subject(s)
5-Methylcytosine/metabolism , Algal Proteins/metabolism , Ascorbic Acid/metabolism , Biocatalysis , Chlamydomonas reinhardtii/enzymology , DNA/chemistry , DNA/metabolism , 5-Methylcytosine/chemistry , Carbon Dioxide/metabolism , DNA Methylation , Glyoxylates/metabolism , Nucleosides/chemistry , Nucleosides/metabolism , Photosynthesis
4.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article in English | MEDLINE | ID: mdl-35210361

ABSTRACT

5-methylcytosine (m5C) is an important epitranscriptomic modification involved in messenger RNA (mRNA) stability and translation efficiency in various biological processes. However, it remains unclear if m5C modification contributes to the dynamic regulation of the transcriptome during the developmental cycles of Plasmodium parasites. Here, we characterize the landscape of m5C mRNA modifications at single nucleotide resolution in the asexual replication stages and gametocyte sexual stages of rodent (Plasmodium yoelii) and human (Plasmodium falciparum) malaria parasites. While different representations of m5C-modified mRNAs are associated with the different stages, the abundance of the m5C marker is strikingly enhanced in the transcriptomes of gametocytes. Our results show that m5C modifications confer stability to the Plasmodium transcripts and that a Plasmodium ortholog of NSUN2 is a major mRNA m5C methyltransferase in malaria parasites. Upon knockout of P. yoelii nsun2 (pynsun2), marked reductions of m5C modification were observed in a panel of gametocytogenesis-associated transcripts. These reductions correlated with impaired gametocyte production in the knockout rodent malaria parasites. Restoration of the nsun2 gene in the knockout parasites rescued the gametocyte production phenotype as well as m5C modification of the gametocytogenesis-associated transcripts. Together with the mRNA m5C profiles for two species of Plasmodium, our findings demonstrate a major role for NSUN2-mediated m5C modifications in mRNA transcript stability and sexual differentiation in malaria parasites.


Subject(s)
5-Methylcytosine/chemistry , Plasmodium falciparum/metabolism , Plasmodium yoelii/growth & development , Plasmodium yoelii/metabolism , Protozoan Proteins/metabolism , RNA, Messenger/metabolism , Germ Cells , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Plasmodium yoelii/genetics , Transcriptome
5.
Anal Chem ; 96(24): 9984-9993, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38833588

ABSTRACT

Metal-organic frameworks (MOFs) show unique advantages in simulating the dynamics and fidelity of natural coordination. Inspired by zinc finger protein, a second linker was introduced to affect the homogeneous MOF system and thus facilitate the emergence of diverse functionalities. Under the systematic identification of 12 MOF species (i.e., metal ions, linkers) and 6 second linkers (trigger), a dissipative system consisting of Co-BDC-NO2 and o-phenylenediamine (oPD) was screened out, which can rapidly and in situ generate a high photothermal complex (η = 36.9%). Meanwhile, both the carboxylation of epigenetic modifications and metal ion (Fe3+, Ni2+, Cu2+, Zn2+, Co2+ and Mn2+) screening were utilized to improve the local coordination environment so that the adaptable Co-MOF growth on the DNA strand was realized. Thus, epigenetic modification information on DNA was converted to an amplified metal ion signal, and then oPD was further introduced to generate bimodal dissipative signals by which a simple, high-sensitivity detection strategy of 5-hydroxymethylcytosine (LOD = 0.02%) and 5-formylcytosine (LOD = 0.025‰) was developed. The strategy provides one low-cost method (< 0.01 $/sample) for quantifying global epigenetic modifications, which greatly promotes epigenetic modification-based early disease diagnosis. This work also proposes a general heterocoordination design concept for molecular recognition and signal transduction, opening a new MOF-based sensing paradigm.


Subject(s)
Cobalt , DNA , Epigenesis, Genetic , Metal-Organic Frameworks , Phenylenediamines , Metal-Organic Frameworks/chemistry , Cobalt/chemistry , DNA/chemistry , Phenylenediamines/chemistry , 5-Methylcytosine/chemistry , 5-Methylcytosine/analysis , 5-Methylcytosine/analogs & derivatives , Cytosine/chemistry , Cytosine/analogs & derivatives , Limit of Detection
6.
Nucleic Acids Res ; 50(D1): D196-D203, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34986603

ABSTRACT

5-Methylcytosine (m5C) is one of the most prevalent covalent modifications on RNA. It is known to regulate a broad variety of RNA functions, including nuclear export, RNA stability and translation. Here, we present m5C-Atlas, a database for comprehensive collection and annotation of RNA 5-methylcytosine. The database contains 166 540 m5C sites in 13 species identified from 5 base-resolution epitranscriptome profiling technologies. Moreover, condition-specific methylation levels are quantified from 351 RNA bisulfite sequencing samples gathered from 22 different studies via an integrative pipeline. The database also presents several novel features, such as the evolutionary conservation of a m5C locus, its association with SNPs, and any relevance to RNA secondary structure. All m5C-atlas data are accessible through a user-friendly interface, in which the m5C epitranscriptomes can be freely explored, shared, and annotated with putative post-transcriptional mechanisms (e.g. RBP intermolecular interaction with RNA, microRNA interaction and splicing sites). Together, these resources offer unprecedented opportunities for exploring m5C epitranscriptomes. The m5C-Atlas database is freely accessible at https://www.xjtlu.edu.cn/biologicalsciences/m5c-atlas.


Subject(s)
Databases, Genetic , Epigenome/genetics , Software , Transcriptome/genetics , 5-Methylcytosine/chemistry , 5-Methylcytosine/metabolism , Humans , MicroRNAs/genetics , Polymorphism, Single Nucleotide/genetics , RNA Processing, Post-Transcriptional/genetics , Sequence Analysis, RNA
7.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Article in English | MEDLINE | ID: mdl-34551979

ABSTRACT

Reduced succinate dehydrogenase (SDH) activity resulting in adverse succinate accumulation was previously considered relevant only in 0.05 to 0.5% of kidney cancers associated with germline SDH mutations. Here, we sought to examine a broader role for SDH loss in kidney cancer pathogenesis/progression. We report that underexpression of SDH subunits resulting in accumulation of oncogenic succinate is a common feature in clear cell renal cell carcinoma (ccRCC) (∼80% of all kidney cancers), with a marked adverse impact on survival in ccRCC patients (n = 516). We show that SDH down-regulation is a critical brake in the TCA cycle during ccRCC pathogenesis and progression. In exploring mechanisms of SDH down-regulation in ccRCC, we report that Von Hippel-Lindau loss-induced hypoxia-inducible factor-dependent up-regulation of miR-210 causes direct inhibition of the SDHD transcript. Moreover, shallow deletion of SDHB occurs in ∼20% of ccRCC. We then demonstrate that SDH loss-induced succinate accumulation contributes to adverse loss of 5-hydroxymethylcytosine, gain of 5-methylcytosine, and enhanced invasiveness in ccRCC via inhibition of ten-eleven translocation (TET)-2 activity. Intriguingly, binding affinity between the catalytic domain of recombinant TET-2 and succinate was found to be very low, suggesting that the mechanism of succinate-induced attenuation of TET-2 activity is likely via product inhibition rather than competitive inhibition. Finally, exogenous ascorbic acid, a TET-activating demethylating agent, led to reversal of the above oncogenic effects of succinate in ccRCC cells. Collectively, our study demonstrates that functional SDH deficiency is a common adverse feature of ccRCC and not just limited to the kidney cancers associated with germline SDH mutations.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/pathology , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/pathology , Succinate Dehydrogenase/metabolism , 5-Methylcytosine/chemistry , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Cycle , Cell Movement , Cell Proliferation , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Mutation , Neoplasm Invasiveness , Prognosis , Succinate Dehydrogenase/genetics , Survival Rate , Tumor Cells, Cultured
8.
Bioinformatics ; 38(18): 4271-4277, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35866985

ABSTRACT

MOTIVATION: 5-Methylcytosine (m5C) is a crucial post-transcriptional modification. With the development of technology, it is widely found in various RNAs. Numerous studies have indicated that m5C plays an essential role in various activities of organisms, such as tRNA recognition, stabilization of RNA structure, RNA metabolism and so on. Traditional identification is costly and time-consuming by wet biological experiments. Therefore, computational models are commonly used to identify the m5C sites. Due to the vast computing advantages of deep learning, it is feasible to construct the predictive model through deep learning algorithms. RESULTS: In this study, we construct a model to identify m5C based on a deep fusion approach with an improved residual network. First, sequence features are extracted from the RNA sequences using Kmer, K-tuple nucleotide frequency component (KNFC), Pseudo dinucleotide composition (PseDNC) and Physical and chemical property (PCP). Kmer and KNFC extract information from a statistical point of view. PseDNC and PCP extract information from the physicochemical properties of RNA sequences. Then, two parts of information are fused with new features using bidirectional long- and short-term memory and attention mechanisms, respectively. Immediately after, the fused features are fed into the improved residual network for classification. Finally, 10-fold cross-validation and independent set testing are used to verify the credibility of the model. The results show that the accuracy reaches 91.87%, 95.55%, 92.27% and 95.60% on the training sets and independent test sets of Arabidopsis thaliana and M.musculus, respectively. This is a considerable improvement compared to previous studies and demonstrates the robust performance of our model. AVAILABILITY AND IMPLEMENTATION: The data and code related to the study are available at https://github.com/alivelxj/m5c-DFRESG.


Subject(s)
5-Methylcytosine , RNA , RNA/chemistry , 5-Methylcytosine/chemistry , Nucleotides/chemistry , Algorithms , Base Sequence
9.
PLoS Biol ; 18(4): e3000684, 2020 04.
Article in English | MEDLINE | ID: mdl-32275660

ABSTRACT

5-hydroxymethylcytosine (5hmC) is the most prevalent intermediate on the oxidative DNA demethylation pathway and is implicated in regulation of embryogenesis, neurological processes, and cancerogenesis. Profiling of this relatively scarce genomic modification in clinical samples requires cost-effective high-resolution techniques that avoid harsh chemical treatment. Here, we present a bisulfite-free approach for 5hmC profiling at single-nucleotide resolution, named hmTOP-seq (5hmC-specific tethered oligonucleotide-primed sequencing), which is based on direct sequence readout primed at covalently labeled 5hmC sites from an in situ tethered DNA oligonucleotide. Examination of distinct conjugation chemistries suggested a structural model for the tether-directed nonhomologous polymerase priming enabling theoretical evaluation of suitable tethers at the design stage. The hmTOP-seq procedure was optimized and validated on a small model genome and mouse embryonic stem cells, which allowed construction of single-nucleotide 5hmC maps reflecting subtle differences in strand-specific CG hydroxymethylation. Collectively, hmTOP-seq provides a new valuable tool for cost-effective and precise identification of 5hmC in characterizing its biological role and epigenetic changes associated with human disease.


Subject(s)
5-Methylcytosine/analogs & derivatives , Sequence Analysis, DNA/methods , 5-Methylcytosine/chemistry , Acetylation , Animals , Bacteriophage lambda/genetics , Cell Line , DNA Methylation , Embryonic Stem Cells/physiology , Genome , Histones/metabolism , Lysine/metabolism , Mice , Oligonucleotides , Reproducibility of Results , Sulfites
10.
Cell Biochem Funct ; 41(6): 704-712, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37349892

ABSTRACT

The ten-eleven translocation (TET) isoforms (TET1-3) play critical roles in epigenetic transcription regulation. In addition, mutations in the TET2 gene are frequently detected in patients with glioma and myeloid malignancies. TET isoforms can oxidize 5-methylcytosine to 5-hydroxymethylcytosine, 5-formylcytosine, and 5-carboxylcytosine, by iterative oxidation. The in vivo DNA demethylation activity of TET isoforms may depend on many factors including enzyme's structural features, its interaction with DNA-binding proteins, chromatin context, DNA sequence, DNA length, and configuration. The rationale for this study is to identify the preferred DNA length and configuration in the substrates of TET isoforms. We have used a highly sensitive LC-MS/MS-based method to compare the substrate preference of TET isoforms. To this end, four DNA substrate sets (S1, S2, S3, S4) of different sequences were chosen. In addition, in each set, four different lengths of DNA substrates comprising 7-, 13-, 19-, and 25-mer nucleotides were synthesized. Each DNA substrate was further used in three different configurations, that is, double stranded symmetrically-methylated, double stranded hemi-methylated, and single stranded single-methylated to evaluate their effect on TET-mediated 5mC oxidation. We demonstrate that mouse TET1 (mTET1) and human TET2 (hTET2) have highest preference for 13-mer dsDNA substrates. Increasing or decreasing the length of dsDNA substrate reduces product formation. In contrast to their dsDNA counterparts, the length of ssDNA substrates did not have a predictable effect on 5mC oxidation. Finally, we show that substrate specificity of TET isoforms correlates with their DNA binding efficiency. Our results demonstrate that mTET1 and hTET2 prefer 13-mer dsDNA as a substrate over ssDNA. These results may help elucidate novel properties of TET-mediated 5mC oxidation and help develop novel diagnostic tools to detect TET2 function in patients.


Subject(s)
5-Methylcytosine , Dioxygenases , Humans , Animals , Mice , 5-Methylcytosine/chemistry , 5-Methylcytosine/metabolism , Dioxygenases/genetics , Dioxygenases/metabolism , Chromatography, Liquid , Tandem Mass Spectrometry , DNA/metabolism , DNA Methylation , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism
11.
Nucleic Acids Res ; 49(2): 1152-1162, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33337470

ABSTRACT

Modification of cytosine plays an important role in epigenetic regulation of gene expression and genome stability. Cytosine is converted to 5-methylcytosine (5mC) by DNA methyltransferase; in turn, 5mC may be oxidized to 5-hydroxymethylcytosine (5hmC) by ten-eleven translocation enzyme. The structural flexibility of DNA is known to affect the binding of proteins to methylated DNA. Here, we have carried out a semi-quantitative analysis of the dynamics of double-stranded DNA (dsDNA) containing various epigenetic modifications by combining data from imino 1H exchange and imino 1H R1ρ relaxation dispersion NMR experiments in a complementary way. Using this approach, we characterized the base-opening (kopen) and base-closing (kclose) rates, facilitating a comparison of the base-opening and -closing process of dsDNA containing cytosine in different states of epigenetic modification. A particularly striking result is the increase in the kopen rate of hemi-methylated dsDNA 5mC/C relative to unmodified or fully methylated dsDNA, indicating that the Watson-Crick base pairs undergo selective destabilization in 5mC/C. Collectively, our findings imply that the epigenetic modulation of cytosine dynamics in dsDNA mediates destabilization of the GC Watson-Crick base pair to allow base-flipping in living cells.


Subject(s)
5-Methylcytosine/chemistry , DNA Methylation , DNA/chemistry , Epigenome , Base Pairing , DNA/chemical synthesis , DNA/genetics , Genomic Instability , Guanine/chemistry , Humans , Kinetics , Molecular Dynamics Simulation , Nuclear Magnetic Resonance, Biomolecular , Nucleic Acid Denaturation , Protons
12.
Nucleic Acids Res ; 49(3): 1708-1723, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33450012

ABSTRACT

Many modification-dependent restriction endonucleases (MDREs) are fusions of a PUA superfamily modification sensor domain and a nuclease catalytic domain. EVE domains belong to the PUA superfamily, and are present in MDREs in combination with HNH nuclease domains. Here, we present a biochemical characterization of the EVE-HNH endonuclease VcaM4I and crystal structures of the protein alone, with EVE domain bound to either 5mC modified dsDNA or to 5mC/5hmC containing ssDNA. The EVE domain is moderately specific for 5mC/5hmC containing DNA according to EMSA experiments. It flips the modified nucleotide, to accommodate it in a hydrophobic pocket of the enzyme, primarily formed by P24, W82 and Y130 residues. In the crystallized conformation, the EVE domain and linker helix between the two domains block DNA binding to the catalytic domain. Removal of the EVE domain and inter-domain linker, but not of the EVE domain alone converts VcaM4I into a non-specific toxic nuclease. The role of the key residues in the EVE and HNH domains of VcaM4I is confirmed by digestion and restriction assays with the enzyme variants that differ from the wild-type by changes to the base binding pocket or to the catalytic residues.


Subject(s)
DNA Restriction Enzymes/chemistry , DNA/chemistry , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/chemistry , Catalytic Domain , Crystallography, X-Ray , DNA, Single-Stranded/chemistry , Models, Molecular , Nucleotide Motifs , Protein Domains , Scattering, Small Angle , Vibrio/enzymology , X-Ray Diffraction
13.
Angew Chem Int Ed Engl ; 62(26): e202304756, 2023 06 26.
Article in English | MEDLINE | ID: mdl-37118885

ABSTRACT

The epigenetic modification 5-methylcytosine plays a vital role in development, cell specific gene expression and disease states. The selective chemical modification of the 5-methylcytosine methyl group is challenging. Currently, no such chemistry exists. Direct functionalisation of 5-methylcytosine would improve the detection and study of this epigenetic feature. We report a xanthone-photosensitised process that introduces a 4-pyridine modification at a C(sp3 )-H bond in the methyl group of 5-methylcytosine. We propose a reaction mechanism for this type of reaction based on density functional calculations and apply transition state analysis to rationalise differences in observed reaction efficiencies between cyanopyridine derivatives. The reaction is initiated by single electron oxidation of 5-methylcytosine followed by deprotonation to generate the methyl group radical. Cross coupling of the methyl radical with 4-cyanopyridine installs a 4-pyridine label at 5-methylcytosine. We demonstrate use of the pyridination reaction to enrich 5-methylcytosine-containing ribonucleic acid.


Subject(s)
5-Methylcytosine , Electrons , 5-Methylcytosine/chemistry , Oxidation-Reduction , Catalysis , Epigenesis, Genetic
14.
J Biol Chem ; 296: 100087, 2021.
Article in English | MEDLINE | ID: mdl-33199375

ABSTRACT

Oxidation of 5-methylcytosine (5mC) in DNA by the ten-eleven translocation (TET) family of enzymes is indispensable for gene regulation in mammals. More recently, evidence has emerged to support a biological function for TET-mediated m5C oxidation in messenger RNA. Here, we describe a previously uncharacterized role of TET-mediated m5C oxidation in transfer RNA (tRNA). We found that the TET-mediated oxidation product 5-hydroxylmethylcytosine (hm5C) is specifically enriched in tRNA inside cells and that the oxidation activity of TET2 on m5C in tRNAs can be readily observed in vitro. We further observed that hm5C levels in tRNA were significantly decreased in Tet2 KO mouse embryonic stem cells (mESCs) in comparison with wild-type mESCs. Reciprocally, induced expression of the catalytic domain of TET2 led to an obvious increase in hm5C and a decrease in m5C in tRNAs relative to uninduced cells. Strikingly, we also show that TET2-mediated m5C oxidation in tRNA promotes translation in vitro. These results suggest TET2 may influence translation through impacting tRNA methylation and reveal an unexpected role for TET enzymes in regulating multiple nodes of the central dogma.


Subject(s)
5-Methylcytosine/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Transfer/metabolism , 5-Methylcytosine/chemistry , Animals , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Dioxygenases , Embryonic Stem Cells/metabolism , Mice , Mice, Knockout , Protein Biosynthesis , Proto-Oncogene Proteins/chemistry , RNA, Transfer/chemistry
15.
J Am Chem Soc ; 144(7): 2987-2993, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35157801

ABSTRACT

5-Methylcytosine (mC) and 5-hydroxymethylcytosine (hmC), the two main epigenetic modifications of mammalian DNA, exist in symmetric and asymmetric combinations in the two strands of CpG dyads. However, revealing such combinations in single DNA duplexes is a significant challenge. Here, we evolve methyl-CpG-binding domains (MBDs) derived from MeCP2 by bacterial cell surface display, resulting in the first affinity probes for hmC/mC CpGs. One mutant has low nanomolar affinity for a single hmC/mC CpG, discriminates against all 14 other modified CpG dyads, and rivals the selectivity of wild-type MeCP2. Structural studies indicate that this protein has a conserved scaffold and recognizes hmC and mC with two dedicated sets of residues. The mutant allows us to selectively address and enrich hmC/mC-containing DNA fragments from genomic DNA backgrounds. We anticipate that this novel probe will be a versatile tool to unravel the function of hmC/mC marks in diverse aspects of chromatin biology.


Subject(s)
5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/chemistry , DNA/isolation & purification , Methyl-CpG-Binding Protein 2/chemistry , Peptide Fragments/chemistry , DNA/chemistry , DNA Methylation , Directed Molecular Evolution , HEK293 Cells , Humans , Methyl-CpG-Binding Protein 2/genetics , Peptide Fragments/genetics , Protein Domains
16.
Genome Res ; 29(5): 750-761, 2019 05.
Article in English | MEDLINE | ID: mdl-30948436

ABSTRACT

Coordinated changes of DNA (de)methylation, nucleosome positioning, and chromatin binding of the architectural protein CTCF play an important role for establishing cell-type-specific chromatin states during differentiation. To elucidate molecular mechanisms that link these processes, we studied the perturbed DNA modification landscape in mouse embryonic stem cells (ESCs) carrying a double knockout (DKO) of the Tet1 and Tet2 dioxygenases. These enzymes are responsible for the conversion of 5-methylcytosine (5mC) into its hydroxymethylated (5hmC), formylated (5fC), or carboxylated (5caC) forms. We determined changes in nucleosome positioning, CTCF binding, DNA methylation, and gene expression in DKO ESCs and developed biophysical models to predict differential CTCF binding. Methylation-sensitive nucleosome repositioning accounted for a significant portion of CTCF binding loss in DKO ESCs, whereas unmethylated and nucleosome-depleted CpG islands were enriched for CTCF sites that remained occupied. A number of CTCF sites also displayed direct correlations with the CpG modification state: CTCF was preferentially lost from sites that were marked with 5hmC in wild-type (WT) cells but not from 5fC-enriched sites. In addition, we found that some CTCF sites can act as bifurcation points defining the differential methylation landscape. CTCF loss from such sites, for example, at promoters, boundaries of chromatin loops, and topologically associated domains (TADs), was correlated with DNA methylation/demethylation spreading and can be linked to down-regulation of neighboring genes. Our results reveal a hierarchical interplay between cytosine modifications, nucleosome positions, and DNA sequence that determines differential CTCF binding and regulates gene expression.


Subject(s)
DNA Methylation , DNA-Binding Proteins/genetics , Epigenesis, Genetic , Mouse Embryonic Stem Cells/enzymology , Proto-Oncogene Proteins/genetics , 5-Methylcytosine/chemistry , Animals , CCCTC-Binding Factor/metabolism , Cell Line , DNA-Binding Proteins/metabolism , Dioxygenases , Insulator Elements/genetics , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/metabolism , Nucleosomes/enzymology , Proto-Oncogene Proteins/metabolism
17.
PLoS Comput Biol ; 17(11): e1009547, 2021 11.
Article in English | MEDLINE | ID: mdl-34748533

ABSTRACT

We present a comprehensive, experimental and theoretical study of the impact of 5-hydroxymethylation of DNA cytosine. Using molecular dynamics, biophysical experiments and NMR spectroscopy, we found that Ten-Eleven translocation (TET) dioxygenases generate an epigenetic variant with structural and physical properties similar to those of 5-methylcytosine. Experiments and simulations demonstrate that 5-methylcytosine (mC) and 5-hydroxymethylcytosine (hmC) generally lead to stiffer DNA than normal cytosine, with poorer circularization efficiencies and lower ability to form nucleosomes. In particular, we can rule out the hypothesis that hydroxymethylation reverts to unmodified cytosine physical properties, as hmC is even more rigid than mC. Thus, we do not expect dramatic changes in the chromatin structure induced by differences in physical properties between d(mCpG) and d(hmCpG). Conversely, our simulations suggest that methylated-DNA binding domains (MBDs), associated with repression activities, are sensitive to the substitution d(mCpG) ➔ d(hmCpG), while MBD3 which has a dual activation/repression activity is not sensitive to the d(mCpG) d(hmCpG) change. Overall, while gene activity changes due to cytosine methylation are the result of the combination of stiffness-related chromatin reorganization and MBD binding, those associated to 5-hydroxylation of methylcytosine could be explained by a change in the balance of repression/activation pathways related to differential MBD binding.


Subject(s)
5-Methylcytosine/analogs & derivatives , DNA Methylation , DNA/chemistry , DNA/metabolism , Epigenesis, Genetic , 5-Methylcytosine/chemistry , 5-Methylcytosine/metabolism , Binding Sites , Biophysical Phenomena , Computational Biology , DNA/genetics , Humans , Magnetic Resonance Spectroscopy , Models, Biological , Molecular Dynamics Simulation , Nucleic Acid Conformation
18.
Methods ; 187: 13-27, 2021 03.
Article in English | MEDLINE | ID: mdl-32755621

ABSTRACT

Cytosine methylation is one of the best studied epigenetic modifications. In mammals, DNA methylation patterns vary among cells and is mainly found in the CpG context. DNA methylation is involved in important processes during development and differentiation and its dysregulation can lead to or is associated with diseases, such as cancer, loss-of-imprinting syndromes and neurological disorders. It has been also shown that DNA methylation at the cellular, tissue and organism level varies with age. To overcome the costs of Whole-Genome Bisulfite Sequencing, the gold standard method to detect 5-methylcytosines at a single base resolution, DNA methylation arrays have been developed and extensively used. This method allows one to assess the status of a fraction of the CpG sites present in the genome of an organism. In order to combine the relatively low cost of Methylation Arrays and digital signals of bisulfite sequencing, we developed a Targeted Bisulfite Sequencing method that can be applied to biomarker discovery for virtually any phenotype. Here we describe a comprehensive step-by-step protocol to build a DNA methylation-based epigenetic clock.


Subject(s)
DNA Methylation , Epigenomics/methods , High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, DNA/methods , 5-Methylcytosine/analysis , 5-Methylcytosine/chemistry , 5-Methylcytosine/metabolism , Age Factors , Aging/genetics , Biomarkers/analysis , Epigenesis, Genetic , Humans , Models, Genetic , Sulfites/chemistry
19.
Nucleic Acids Res ; 48(7): 3962-3974, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32095813

ABSTRACT

Modified DNA bases functionally distinguish the taxonomic forms of life-5-methylcytosine separates prokaryotes from eukaryotes and 5-hydroxymethylcytosine (5hmC) invertebrates from vertebrates. We demonstrate here that mouse endonuclease G (mEndoG) shows specificity for both 5hmC and Holliday junctions. The enzyme has higher affinity (>50-fold) for junctions over duplex DNAs. A 5hmC-modification shifts the position of the cut site and increases the rate of DNA cleavage in modified versus unmodified junctions. The crystal structure of mEndoG shows that a cysteine (Cys69) is positioned to recognize 5hmC through a thiol-hydroxyl hydrogen bond. Although this Cys is conserved from worms to mammals, a two amino acid deletion in the vertebrate relative to the invertebrate sequence unwinds an α-helix, placing the thiol of Cys69 into the mEndoG active site. Mutations of Cys69 with alanine or serine show 5hmC-specificity that mirrors the hydrogen bonding potential of the side chain (C-H < S-H < O-H). A second orthogonal DNA binding site identified in the mEndoG structure accommodates a second arm of a junction. Thus, the specificity of mEndoG for 5hmC and junctions derives from structural adaptations that distinguish the vertebrate from the invertebrate enzyme, thereby thereby supporting a role for 5hmC in recombination processes.


Subject(s)
5-Methylcytosine/analogs & derivatives , DNA/chemistry , Endodeoxyribonucleases/chemistry , 5-Methylcytosine/chemistry , 5-Methylcytosine/metabolism , Animals , Binding Sites , DNA/metabolism , DNA Cleavage , DNA, Cruciform/metabolism , Endodeoxyribonucleases/metabolism , Mice , Models, Molecular , Substrate Specificity
20.
Nucleic Acids Res ; 48(1): 55-62, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31777919

ABSTRACT

i-Motifs are widely used in nanotechnology, play a part in gene regulation and have been detected in human nuclei. As these structures are composed of cytosine, they are potential sites for epigenetic modification. In addition to 5-methyl- and 5-hydroxymethylcytosine modifications, recent evidence has suggested biological roles for 5-formylcytosine and 5-carboxylcytosine. Herein the human telomeric i-motif sequence was used to examine how these four epigenetic modifications alter the thermal and pH stability of i-motifs. Changes in melting temperature and transitional pH depended on both the type of modification and its position within the i-motif forming sequence. The cytosines most sensitive to modification were next to the first and third loops within the structure. Using previously described i-motif forming sequences, we screened the MCF-7 and MCF-10A methylomes to map 5-methylcytosine and found the majority of sequences were differentially methylated in MCF7 (cancerous) and MCF10A (non-cancerous) cell lines. Furthermore, i-motif forming sequences stable at neutral pH were significantly more likely to be epigenetically modified than traditional acidic i-motif forming sequences. This work has implications not only in the epigenetic regulation of DNA, but also allows discreet tunability of i-motif stability for nanotechnological applications.


Subject(s)
5-Methylcytosine/analogs & derivatives , Cytosine/analogs & derivatives , Cytosine/metabolism , DNA/metabolism , Epigenesis, Genetic , 5-Methylcytosine/chemistry , 5-Methylcytosine/metabolism , Cell Line , Cytosine/chemistry , DNA/chemistry , DNA/genetics , DNA Methylation , Humans , Hydrogen-Ion Concentration , MCF-7 Cells , Nucleotide Motifs
SELECTION OF CITATIONS
SEARCH DETAIL