Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Genome Biol ; 22(1): 186, 2021 06 22.
Article in English | MEDLINE | ID: mdl-34158086

ABSTRACT

BACKGROUND: TET enzymes mediate DNA demethylation by oxidizing 5-methylcytosine (5mC) in DNA to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and 5-carboxylcytosine (5caC). Since these oxidized methylcytosines (oxi-mCs) are not recognized by the maintenance methyltransferase DNMT1, DNA demethylation can occur through "passive," replication-dependent dilution when cells divide. A distinct, replication-independent ("active") mechanism of DNA demethylation involves excision of 5fC and 5caC by the DNA repair enzyme thymine DNA glycosylase (TDG), followed by base excision repair. RESULTS: Here by analyzing inducible gene-disrupted mice, we show that DNA demethylation during primary T cell differentiation occurs mainly through passive replication-dependent dilution of all three oxi-mCs, with only a negligible contribution from TDG. In addition, by pyridine borane sequencing (PB-seq), a simple recently developed method that directly maps 5fC/5caC at single-base resolution, we detect the accumulation of 5fC/5caC in TDG-deleted T cells. We also quantify the occurrence of concordant demethylation within and near enhancer regions in the Il4 locus. In an independent system that does not involve cell division, macrophages treated with liposaccharide accumulate 5hmC at enhancers and show altered gene expression without DNA demethylation; loss of TET enzymes disrupts gene expression, but loss of TDG has no effect. We also observe that mice with long-term (1 year) deletion of Tdg are healthy and show normal survival and hematopoiesis. CONCLUSIONS: We have quantified the relative contributions of TET and TDG to cell differentiation and DNA demethylation at representative loci in proliferating T cells. We find that TET enzymes regulate T cell differentiation and DNA demethylation primarily through passive dilution of oxi-mCs. In contrast, while we observe a low level of active, replication-independent DNA demethylation mediated by TDG, this process does not appear to be essential for immune cell activation or differentiation.


Subject(s)
DNA Methylation , DNA-Binding Proteins/genetics , Dioxygenases/genetics , Macrophages/enzymology , T-Lymphocytes/enzymology , Thymine DNA Glycosylase/genetics , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cytosine/analogs & derivatives , Cytosine/metabolism , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/deficiency , Dioxygenases/deficiency , Enhancer Elements, Genetic , Gene Expression , Genetic Loci , Hematopoiesis/genetics , Interleukin-4/genetics , Interleukin-4/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Lipopolysaccharides/pharmacology , Longevity/genetics , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Knockout , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymine DNA Glycosylase/deficiency
2.
Cell Physiol Biochem ; 39(3): 975-84, 2016.
Article in English | MEDLINE | ID: mdl-27513857

ABSTRACT

AIMS: To study the effect of thymine DNA glycosylase (TDG) gene knockdown on the differentiation of pig preadipocytes. METHODS: Preadipocytes were obtained from subcutaneous adipose tissue from the neck of 1- to 7-day-old pigs. The TDG gene was knocked down using siRNA, and cell differentiation was induced. The mRNA expression level was measured using fluorescence quantitative PCR, and the protein expression level was determined using Western blot analysis. The DNA methylation levels in promoter regions of differentiation-related genes were also evaluated. RESULTS: TDG gene knockdown decreased the mRNA expression levels of the peroxisome proliferator-activated receptorγ (PPARγ) and Fatty acid binding proteins 4(FABP4 Also known as aP2) genes (P<0.01), while the mRNA expression level of the CCAAT/enhancer binding protein alpha(C/EBPα) gene did not change significantly (P>0.05). In addition, after induced differentiation, the lipid droplet production significantly decreased, and the percentages of methylation in the promoter regions of C/EBPα, PPARγ, and aP2 genes were 0.9%, 80%, and 76%, respectively. In contrast, the percentages of methylation in the negative control groups were 0.5%, 67.5%, and 58%, respectively. CONCLUSION: TDG gene knockdown could inhibit the differentiation of pig preadipocytes and affect the DNA methylation levels of some transcription factors.


Subject(s)
Adipocytes, White/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , Epigenesis, Genetic , Fatty Acid-Binding Proteins/genetics , PPAR gamma/genetics , Thymine DNA Glycosylase/genetics , Adipocytes, White/cytology , Animals , Animals, Newborn , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation , DNA Methylation , Fatty Acid-Binding Proteins/metabolism , Gene Expression , Gene Knockdown Techniques , Lipid Droplets/metabolism , Male , PPAR gamma/metabolism , Primary Cell Culture , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Subcutaneous Fat/cytology , Subcutaneous Fat/metabolism , Swine , Thymine DNA Glycosylase/deficiency
3.
DNA Repair (Amst) ; 43: 78-88, 2016 07.
Article in English | MEDLINE | ID: mdl-27289557

ABSTRACT

The family of Ten-Eleven Translocation (TET) proteins is implicated in the process of active DNA demethylation and thus in epigenetic regulation. TET 1, 2 and 3 proteins are oxygenases that can hydroxylate 5-methylcytosine (5-mC) into 5-hydroxymethylcytosine (5-hmC) and further oxidize 5-hmC into 5-formylcytosine (5-fC) and 5-carboxylcytosine (5-caC). The base excision repair (BER) pathway removes the resulting 5-fC and 5-caC bases paired with a guanine and replaces them with regular cytosine. The question arises whether active modification of 5-mC residues and their subsequent elimination could affect the genomic DNA stability. Here, we generated two inducible cell lines (Ba/F3-EPOR, and UT7) overexpressing wild-type or catalytically inactive human TET2 proteins. Wild-type TET2 induction resulted in an increased level of 5-hmC and a cell cycle defect in S phase associated with higher level of phosphorylated P53, chromosomal and centrosomal abnormalities. Furthermore, in a thymine-DNA glycosylase (Tdg) deficient context, the TET2-mediated increase of 5-hmC induces mutagenesis characterized by GC>AT transitions in CpG context suggesting a mutagenic potential of 5-hmC metabolites. Altogether, these data suggest that TET2 activity and the levels of 5-hmC and its derivatives should be tightly controlled to avoid genetic and chromosomal instabilities. Moreover, TET2-mediated active demethylation might be a very dangerous process if used to entirely demethylate the genome and might rather be used only at specific loci.


Subject(s)
5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , DNA Repair , DNA-Binding Proteins/genetics , Genomic Instability , Mutagenesis , Proto-Oncogene Proteins/genetics , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Base Sequence , Cell Line , Cytosine/analogs & derivatives , Cytosine/metabolism , DNA-Binding Proteins/metabolism , Dioxygenases , Epigenesis, Genetic , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Hydroxylation , Megakaryocyte Progenitor Cells/cytology , Megakaryocyte Progenitor Cells/metabolism , Mice , Proto-Oncogene Proteins/metabolism , S Phase , Thymine DNA Glycosylase/deficiency , Thymine DNA Glycosylase/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
Epigenetics ; 10(11): 1006-13, 2015.
Article in English | MEDLINE | ID: mdl-26440216

ABSTRACT

Oxidation of 5-methylcytosine by TET family proteins can induce DNA replication-dependent (passive) DNA demethylation and base excision repair (BER)-based (active) DNA demethylation. The balance of active vs. passive TET-induced demethylation remains incompletely determined. In the context of large scale DNA demethylation, active demethylation may require massive induction of the DNA repair machinery and thus compromise genome stability. To study this issue, we constructed a tetracycline-controlled TET-induced global DNA demethylation system in HEK293T cells. Upon TET overexpression, we observed induction of DNA damage and activation of a DNA damage response; however, BER genes are not upregulated to promote DNA repair. Depletion of TDG (thymine DNA glycosylase) or APEX1 (apurinic/apyrimidinic endonuclease 1), two key BER enzymes, enhances rather than impairs global DNA demethylation, which can be explained by stimulated proliferation. By contrast, growth arrest dramatically blocks TET-induced global DNA demethylation. Thus, in the context of TET-induction in HEK293T cells, the DNA replication-dependent passive mechanism functions as the predominant pathway for global DNA demethylation. In the same context, BER-based active demethylation is markedly restricted by limited BER upregulation, thus potentially preventing a disastrous DNA damage response to extensive active DNA demethylation.


Subject(s)
DNA Methylation , DNA Repair , Mixed Function Oxygenases/metabolism , Proto-Oncogene Proteins/metabolism , Cell Proliferation , DNA Damage , DNA-(Apurinic or Apyrimidinic Site) Lyase/deficiency , HEK293 Cells , Humans , Thymine DNA Glycosylase/deficiency
5.
Tohoku J Exp Med ; 226(1): 75-83, 2012 01.
Article in English | MEDLINE | ID: mdl-22200605

ABSTRACT

Thymine DNA glycosylase (TDG) is involved in the repair of G:T and G:U mismatches caused by hydrolytic deamination of 5-methylcytosine and cytosine, respectively. Recent studies have shown that TDG not only has G-T/U glycosylase activities but also acts in the maintaining proper epigenetic status. In order to investigate the function of TDG in vivo, mice lacking Tdg, Tdg (-/-), were generated. Tdg mutant mice died in utero by 11.5 days post coitum (dpc), although there were no significant differences in the spontaneous mutant frequencies between wild type and Tdg (-/-) embryos. On the other hand, the levels of noradrenaline in 10.5 dpc whole embryos, which is necessary for normal embryogenesis, were dramatically reduced in Tdg (-/-) embryos. Consequently, we tested the effect of D, L-threo-3, 4-dihydroxyphenylserine (DOPS), a synthetic precursor of noradrenaline, on the survival of the Tdg (-/-) embryos. DOPS was given to pregnant Tdg (+/-) mice from 6.5 dpc through drinking water. Most of the Tdg (-/-) embryos were alive at 11.5 dpc, and they were partially rescued up to 14.5 dpc by the administration of DOPS. In contrast, the administration of L-3, 4-dihydroxyphenylalanine (L-DOPA) had marginal effects on Tdg (-/-) embryonic lethality. No embryo was alive without DOPS beyond 11.5 dpc, suggesting that the lethality in (-/-) embryos is partially due to the reduction of noradrenaline. These results suggest that embryonic lethality in Tdg (-/-) embryos is due, in part, to the reduction of noradrenaline levels.


Subject(s)
Droxidopa/metabolism , Embryo, Mammalian/enzymology , Epigenesis, Genetic/physiology , Norepinephrine/metabolism , Thymine DNA Glycosylase/metabolism , Animals , Chromatography, High Pressure Liquid , DNA Primers/genetics , Dopamine/metabolism , Droxidopa/pharmacology , Embryo, Mammalian/drug effects , Epigenesis, Genetic/genetics , Female , Levodopa/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Polymerase Chain Reaction , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Thymine DNA Glycosylase/deficiency , Thymine DNA Glycosylase/genetics
6.
Nature ; 470(7334): 419-23, 2011 Feb 17.
Article in English | MEDLINE | ID: mdl-21278727

ABSTRACT

Thymine DNA glycosylase (TDG) is a member of the uracil DNA glycosylase (UDG) superfamily of DNA repair enzymes. Owing to its ability to excise thymine when mispaired with guanine, it was proposed to act against the mutability of 5-methylcytosine (5-mC) deamination in mammalian DNA. However, TDG was also found to interact with transcription factors, histone acetyltransferases and de novo DNA methyltransferases, and it has been associated with DNA demethylation in gene promoters following activation of transcription, altogether implicating an engagement in gene regulation rather than DNA repair. Here we use a mouse genetic approach to determine the biological function of this multifaceted DNA repair enzyme. We find that, unlike other DNA glycosylases, TDG is essential for embryonic development, and that this phenotype is associated with epigenetic aberrations affecting the expression of developmental genes. Fibroblasts derived from Tdg null embryos (mouse embryonic fibroblasts, MEFs) show impaired gene regulation, coincident with imbalanced histone modification and CpG methylation at promoters of affected genes. TDG associates with the promoters of such genes both in fibroblasts and in embryonic stem cells (ESCs), but epigenetic aberrations only appear upon cell lineage commitment. We show that TDG contributes to the maintenance of active and bivalent chromatin throughout cell differentiation, facilitating a proper assembly of chromatin-modifying complexes and initiating base excision repair to counter aberrant de novo methylation. We thus conclude that TDG-dependent DNA repair has evolved to provide epigenetic stability in lineage committed cells.


Subject(s)
Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Epigenesis, Genetic/genetics , Genes, Lethal/genetics , Phenotype , Thymine DNA Glycosylase/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Chromatin/genetics , Chromatin/metabolism , CpG Islands/genetics , DNA Methylation , DNA Repair , Embryo, Mammalian/enzymology , Fibroblasts/metabolism , Gene Deletion , Gene Expression Regulation, Developmental , Genes, Essential/genetics , Histones/metabolism , Mice , Mice, Knockout , Promoter Regions, Genetic/genetics , Thymine DNA Glycosylase/deficiency , Thymine DNA Glycosylase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...