Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 184, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38167803

ABSTRACT

The intracellular ATP-ribosyltransferases PARP1 and PARP2, contribute to DNA base excision repair (BER) and DNA demethylation and have been implicated in epigenetic programming in early mammalian development. Recently, proteomic analyses identified BER proteins to be covalently poly-ADP-ribosylated by PARPs. The role of this posttranslational modification in the BER process is unknown. Here, we show that PARP1 senses AP-sites and SSBs generated during TET-TDG mediated active DNA demethylation and covalently attaches PAR to each BER protein engaged. Covalent PARylation dissociates BER proteins from DNA, which accelerates the completion of the repair process. Consistently, inhibition of PARylation in mESC resulted both in reduced locus-specific TET-TDG-targeted DNA demethylation, and in reduced general repair of random DNA damage. Our findings establish a critical function of covalent protein PARylation in coordinating molecular processes associated with dynamic DNA methylation.


Subject(s)
DNA Repair , Excision Repair , Animals , Poly ADP Ribosylation , DNA Demethylation , Proteomics , Poly (ADP-Ribose) Polymerase-1/metabolism , DNA Damage , DNA/genetics , DNA/metabolism , Mammals/genetics
2.
J Natl Cancer Inst ; 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38273663

ABSTRACT

BACKGROUND: The phenomenon of field cancerization reflects the transition of normal cells into those predisposed to cancer. Assessing the scope and intensity of this process in the colon may support risk prediction and colorectal cancer prevention. METHODS: The SWEPIC study, encompassing 1,111 participants for DNA methylation analysis and a subset of 84 for RNA-seq, was employed to detect field cancerization in individuals with adenomatous polyps (AP). Methylation variations were evaluated for their discriminative capability, including in external cohorts, genomic localization, clinical correlations, and associated RNA expression patterns. RESULTS: Normal cecal tissue of individuals harboring an AP in the proximal colon manifested dysregulated DNA methylation compared to tissue from healthy individuals at 558 unique loci. Leveraging these adenoma-related differentially variable and methylated CpGs (aDVMCs), our classifier discerned between healthy and AP-adjacent tissues across SWEPIC datasets (cross-validated ROC AUC [0.63-0.81]), including within age-stratified cohorts. This discriminative capacity was validated in three external sets, differentiating healthy from cancer-adjacent tissue (ROC AUC: [0.82-0.88]). Notably, aDVMC dysregulation correlated with polyp multiplicity. More than 50% of aDVMCs were significantly associated with age. These aDVMCs were enriched in active regions of the genome (p < .001), and associated genes exhibited altered expression in AP-adjacent tissues. CONCLUSIONS: Our findings underscore the early onset of field cancerization in the right colon during the neoplastic transformation process. A more extensive validation of aDVMC dysregulation as a stratification tool could pave the way for enhanced surveillance approaches, especially given its linkage to adenoma emergence.

3.
EMBO J ; 38(1)2019 01 03.
Article in English | MEDLINE | ID: mdl-30523148

ABSTRACT

During active DNA demethylation, 5-methylcytosine (5mC) is oxidized by TET proteins to 5-formyl-/5-carboxylcytosine (5fC/5caC) for replacement by unmethylated C by TDG-initiated DNA base excision repair (BER). Base excision generates fragile abasic sites (AP-sites) in DNA and has to be coordinated with subsequent repair steps to limit accumulation of genome destabilizing secondary DNA lesions. Here, we show that 5fC/5caC is generated at a high rate in genomes of differentiating mouse embryonic stem cells and that SUMOylation and the BER protein XRCC1 play critical roles in orchestrating TDG-initiated BER of these lesions. SUMOylation of XRCC1 facilitates physical interaction with TDG and promotes the assembly of a TDG-BER core complex. Within this TDG-BERosome, SUMO is transferred from XRCC1 and coupled to the SUMO acceptor lysine in TDG, promoting its dissociation while assuring the engagement of the BER machinery to complete demethylation. Although well-studied, the biological importance of TDG SUMOylation has remained obscure. Here, we demonstrate that SUMOylation of TDG suppresses DNA strand-break accumulation and toxicity to PARP inhibition in differentiating mESCs and is essential for neural lineage commitment.


Subject(s)
Cell Differentiation/genetics , DNA Demethylation , DNA Repair/physiology , Embryonic Stem Cells/physiology , Sumoylation/physiology , X-ray Repair Cross Complementing Protein 1/metabolism , 5-Methylcytosine/metabolism , Animals , Cells, Cultured , Cytosine/analogs & derivatives , Cytosine/metabolism , Humans , Mice , Multiprotein Complexes/metabolism , Protein Multimerization/physiology
4.
PLoS Genet ; 11(1): e1004899, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25569253

ABSTRACT

Protein modifications regulate both DNA repair levels and pathway choice. How each modification achieves regulatory effects and how different modifications collaborate with each other are important questions to be answered. Here, we show that sumoylation regulates double-strand break repair partly by modifying the end resection factor Sae2. This modification is conserved from yeast to humans, and is induced by DNA damage. We mapped the sumoylation site of Sae2 to a single lysine in its self-association domain. Abolishing Sae2 sumoylation by mutating this lysine to arginine impaired Sae2 function in the processing and repair of multiple types of DNA breaks. We found that Sae2 sumoylation occurs independently of its phosphorylation, and the two modifications act in synergy to increase soluble forms of Sae2. We also provide evidence that sumoylation of the Sae2-binding nuclease, the Mre11-Rad50-Xrs2 complex, further increases end resection. These findings reveal a novel role for sumoylation in DNA repair by regulating the solubility of an end resection factor. They also show that collaboration between different modifications and among multiple substrates leads to a stronger biological effect.


Subject(s)
DNA End-Joining Repair/genetics , DNA Repair/genetics , Endonucleases/genetics , Saccharomyces cerevisiae Proteins/genetics , Sumoylation/genetics , DNA Breaks, Double-Stranded , DNA Damage/genetics , DNA-Binding Proteins/genetics , Endodeoxyribonucleases/genetics , Exodeoxyribonucleases/genetics , Humans , Phosphorylation , Saccharomyces cerevisiae , Solubility
5.
PLoS One ; 8(8): e71960, 2013.
Article in English | MEDLINE | ID: mdl-23936535

ABSTRACT

The SUMO-dependent ubiquitin ligase Slx8 plays key roles in promoting genome stability, including the processing of trapped Topoisomerase I (Top1) cleavage complexes and removal of toxic SUMO conjugates. We show that it is the latter function that constitutes Slx8's primary role in fission yeast. The SUMO conjugates in question are formed by the SUMO ligase Pli1, which is necessary for limiting spontaneous homologous recombination when Top1 is present. Surprisingly there is no requirement for Pli1 to limit recombination in the vicinity of a replication fork blocked at the programmed barrier RTS1. Notably, once committed to Pli1-mediated SUMOylation Slx8 becomes essential for genotoxin resistance, limiting both spontaneous and RTS1 induced recombination, and promoting normal chromosome segregation. We show that Slx8 removes Pli1-dependent Top1-SUMO conjugates and in doing so helps to constrain recombination at RTS1. Overall our data highlight how SUMOylation and SUMO-dependent ubiquitylation by the Pli1-Slx8 axis contribute in different ways to maintain genome stability.


Subject(s)
DNA Topoisomerases, Type I/metabolism , Genomic Instability , SUMO-1 Protein/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Sumoylation , Ubiquitin-Protein Ligases/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Chromosome Segregation/drug effects , Chromosome Segregation/genetics , DNA Replication/drug effects , DNA Replication/genetics , DNA Topoisomerases, Type I/deficiency , DNA Topoisomerases, Type I/genetics , DNA, Fungal/biosynthesis , DNA, Fungal/genetics , Gene Deletion , Genomic Instability/drug effects , Ligases , Mutagens/toxicity , Recombination, Genetic/drug effects , Recombination, Genetic/genetics , Schizosaccharomyces/cytology , Schizosaccharomyces/enzymology , Sumoylation/drug effects
6.
Science ; 336(6088): 1585-8, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22723423

ABSTRACT

The formation of healthy gametes depends on programmed DNA double-strand breaks (DSBs), which are each repaired as a crossover (CO) or non-crossover (NCO) from a homologous template. Although most of these DSBs are repaired without giving COs, little is known about the genetic requirements of NCO-specific recombination. We show that Fml1, the Fanconi anemia complementation group M (FANCM)-ortholog of Schizosaccharomyces pombe, directs the formation of NCOs during meiosis in competition with the Mus81-dependent pro-CO pathway. We also define the Rad51/Dmc1-mediator Swi5-Sfr1 as a major determinant in biasing the recombination process in favor of Mus81, to ensure the appropriate amount of COs to guide meiotic chromosome segregation. The conservation of these proteins from yeast to humans suggests that this interplay may be a general feature of meiotic recombination.


Subject(s)
Crossing Over, Genetic , DNA Helicases/metabolism , Homologous Recombination , Meiosis , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/genetics , Chromosome Segregation , Chromosomes, Fungal/physiology , DNA Breaks, Double-Stranded , DNA Helicases/genetics , DNA Repair , DNA, Fungal/chemistry , DNA, Fungal/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endonucleases/genetics , Endonucleases/metabolism , Mutation , Recombinases/genetics , Recombinases/metabolism , Schizosaccharomyces/physiology , Schizosaccharomyces pombe Proteins/genetics
7.
Genes Dev ; 26(6): 594-602, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22426535

ABSTRACT

Bidirectionally moving DNA replication forks merge at termination sites composed of accidental or programmed DNA-protein barriers. If merging fails, then regions of unreplicated DNA can result in the breakage of DNA during mitosis, which in turn can give rise to genome instability. Despite its importance, little is known about the mechanisms that promote the final stages of fork merging in eukaryotes. Here we show that the Pif1 family DNA helicase Pfh1 plays a dual role in promoting replication fork termination. First, it facilitates replication past DNA-protein barriers, and second, it promotes the merging of replication forks. A failure of these processes in Pfh1-deficient cells results in aberrant chromosome segregation and heightened genome instability.


Subject(s)
DNA Helicases/metabolism , DNA Replication , Genomic Instability , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/genetics , Chromosome Segregation , DNA Helicases/genetics , DNA, Fungal/genetics , DNA, Ribosomal/genetics , Schizosaccharomyces/enzymology , Schizosaccharomyces pombe Proteins/genetics
8.
Nucleic Acids Res ; 39(15): 6568-84, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21576223

ABSTRACT

Most DNA double-strand breaks (DSBs) in S- and G2-phase cells are repaired accurately by Rad51-dependent sister chromatid recombination. However, a minority give rise to gross chromosome rearrangements (GCRs), which can result in disease/death. What determines whether a DSB is repaired accurately or inaccurately is currently unclear. We provide evidence that suggests that perturbing replication by a non-programmed protein-DNA replication fork barrier results in the persistence of replication intermediates (most likely regions of unreplicated DNA) into mitosis, which results in anaphase bridge formation and ultimately to DNA breakage. However, unlike previously characterised replication-associated DSBs, these breaks are repaired mainly by Rad51-independent processes such as single-strand annealing, and are therefore prone to generate GCRs. These data highlight how a replication-associated DSB can be predisposed to give rise to genome rearrangements in eukaryotes.


Subject(s)
DNA Breaks, Double-Stranded , DNA Replication , Recombination, Genetic , Anaphase/genetics , Chromosome Deletion , DNA/ultrastructure , DNA Helicases/metabolism , Epstein-Barr Virus Nuclear Antigens/metabolism , Lac Repressors/metabolism , Mitosis , Mutation , Operator Regions, Genetic , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/metabolism
9.
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
10.
DNA Repair (Amst) ; 6(4): 489-504, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17116428

ABSTRACT

When it was first isolated from extracts of HeLa cells in Josef Jiricny's laboratory, the thymine DNA glycosylase (TDG) attracted attention because of its ability to remove thymine, i.e. a normal DNA base, from G.T mispairs. This implicated a function of DNA base excision repair in the restoration of G.C base pairs following the deamination of a 5-methylcytosine. TDG turned out to be the founding member of a newly emerging family of mismatch-directed uracil-DNA glycosylases, the MUG proteins, that act on a comparably broad spectrum of base lesion including G.U as the common, most efficiently processed substrate. However, because of its apparent catalytic inefficiency, some have considered TDG a poor DNA repair enzyme without an important biological function. Others have reported 5-meC DNA glycosylase activity to be associated with TDG, thrusting the enzyme into limelight as a possible DNA demethylase. Yet others have found the glycosylase to interact with transcription factors, implicating a function in gene regulation, which appears to be critically important in developmental processes. This article reviews all these developments in view of possible biological functions of this multifaceted DNA glycosylase.


Subject(s)
DNA Mismatch Repair , DNA Repair , Thymine DNA Glycosylase/metabolism , Amino Acid Sequence , Evolution, Molecular , Humans , Molecular Sequence Data , Protein Conformation , Sequence Alignment , Thymine DNA Glycosylase/chemistry , Thymine DNA Glycosylase/genetics
11.
Mol Cell ; 22(1): 51-62, 2006 Apr 07.
Article in English | MEDLINE | ID: mdl-16600869

ABSTRACT

Alterations in DNA repair lead to genomic instability and higher risk of cancer. DNA base excision repair (BER) corrects damaged bases, apurinic sites, and single-strand DNA breaks. Here, a regulatory mechanism for DNA polymerase beta (Pol beta) is described. Pol beta was found to form a complex with the protein arginine methyltransferase 6 (PRMT6) and was specifically methylated in vitro and in vivo. Methylation of Pol beta by PRMT6 strongly stimulated DNA polymerase activity by enhancing DNA binding and processivity, while single nucleotide insertion and dRP-lyase activity were not affected. Two residues, R83 and R152, were identified in Pol beta as the sites of methylation by PRMT6. Genetic complementation of Pol beta knockout cells with R83/152K mutant revealed the importance of these residues for the cellular resistance to DNA alkylating agent. Based on our findings, we propose that PRMT6 plays a role as a regulator of BER.


Subject(s)
Arginine/metabolism , DNA Methylation , DNA Polymerase beta/metabolism , Gene Expression Regulation , Nuclear Proteins/physiology , Protein-Arginine N-Methyltransferases/physiology , Animals , Arginine/chemistry , DNA Damage , DNA Ligases/physiology , DNA Polymerase beta/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/enzymology , Fibroblasts/cytology , Fibroblasts/enzymology , Genetic Complementation Test , Humans , Mice , Mice, Knockout
12.
Curr Biol ; 15(7): 616-23, 2005 Apr 12.
Article in English | MEDLINE | ID: mdl-15823533

ABSTRACT

BACKGROUND: Base excision repair initiated by human thymine-DNA glycosylase (TDG) results in the generation of abasic sites (AP sites) in DNA. TDG remains bound to this unstable repair intermediate, indicating that its transmission to the downstream-acting AP endonuclease is a coordinated process. Previously, we established that posttranslational modification of TDG with Small Ubiquitin-like MOdifiers (SUMOs) facilitates the dissociation of the DNA glycosylase from the product AP site, but the underlying molecular mechanism remained unclear. RESULTS: We now show that upon DNA interaction, TDG undergoes a dramatic conformational change, which involves its flexible N-terminal domain and accounts for the nonspecific DNA binding ability of the enzyme. This function is required for efficient processing of the G.T mismatch but then cooperates with the specific DNA contacts established in the active site pocket of TDG to prevent its dissociation from the product AP site after base release. SUMO1 conjugation to the C-terminal K330 of TDG modulates the DNA binding function of the N terminus to induce dissociation of the glycosylase from the AP site while it leaves the catalytic properties of base release in the active site pocket of the enzyme unaffected. CONCLUSION: Our data provide insight into the molecular mechanism of SUMO modification mediated modulation of enzymatic properties of TDG. A conformational change, involving the N-terminal domain of TDG, provides unspecific DNA interactions that facilitate processing of a wider spectrum of substrates at the expense of enzymatic turnover. SUMOylation then reverses this structural change in the product bound TDG.


Subject(s)
DNA Repair/physiology , Small Ubiquitin-Related Modifier Proteins/metabolism , Thymine DNA Glycosylase/metabolism , Blotting, Western , DNA Primers , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Electrophoretic Mobility Shift Assay , Genetic Vectors , Humans , Models, Biological , Protein Conformation , Protein Structure, Tertiary , SUMO-1 Protein , Thymine DNA Glycosylase/genetics
13.
EMBO J ; 21(6): 1456-64, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11889051

ABSTRACT

DNA glycosylases initiate base excision repair (BER) through the generation of potentially harmful abasic sites (AP sites) in DNA. Human thymine-DNA glycosylase (TDG) is a mismatch-specific uracil/thymine-DNA glycosylase with an implicated function in the restoration of G*C base pairs at sites of cytosine or 5-methylcytosine deamination. The rate-limiting step in the action of TDG in vitro is its dissociation from the product AP site, suggesting the existence of a specific enzyme release mechanism in vivo. We show here that TDG interacts with and is covalently modified by the ubiquitin-like proteins SUMO-1 and SUMO-2/3. SUMO conjugation dramatically reduces the DNA substrate and AP site binding affinity of TDG, and this is associated with a significant increase in enzymatic turnover in reactions with a G*U substrate and the loss of G*T processing activity. Sumoylation also potentiates the stimulatory effect of APE1 on TDG. These observations implicate a function of sumoylation in the controlled dissociation of TDG from the AP site and open up novel perspectives for the understanding of the molecular mechanisms coordinating the early steps of BER.


Subject(s)
DNA Repair , Endodeoxyribonucleases/metabolism , SUMO-1 Protein/metabolism , Small Ubiquitin-Related Modifier Proteins , Ubiquitins/metabolism , Binding Sites , Deoxyribonuclease (Pyrimidine Dimer) , Endodeoxyribonucleases/genetics , HeLa Cells , Humans , Lysine/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , SUMO-1 Protein/genetics , Ubiquitins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...