Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Genome Res ; 32(2): 367-377, 2022 02.
Article in English | MEDLINE | ID: mdl-34916251

ABSTRACT

Sequence-specific DNA-binding transcription factors are central to gene regulation. They are often associated with consensus binding sites that predict far more genomic sites than are bound in vivo. One explanation is that most sites are blocked by nucleosomes, such that only sites in nucleosome-depleted regulatory regions are bound. We compared the binding of the yeast transcription factor Gcn4 in vivo using published ChIP-seq data (546 sites) and in vitro, using a modified SELEX method ("G-SELEX"), which utilizes short genomic DNA fragments to quantify binding at all sites. We confirm that Gcn4 binds strongly to an AP-1-like sequence (TGACTCA) and weakly to half-sites. However, Gcn4 binds only some of the 1078 exact matches to this sequence, even in vitro. We show that there are only 166 copies of the high-affinity RTGACTCAY site (exact match) in the yeast genome, all occupied in vivo, largely independently of whether they are located in nucleosome-depleted or nucleosomal regions. Generally, RTGACTCAR/YTGACTCAY sites are bound much more weakly and YTGACTCAR sites are unbound, with biological implications for determining induction levels. We conclude that, to a first approximation, Gcn4 binding can be predicted using the high-affinity site, without reference to chromatin structure. We propose that transcription factor binding sites should be defined more precisely using quantitative data, allowing more accurate genome-wide prediction of binding sites and greater insight into gene regulation.


Subject(s)
Saccharomyces cerevisiae Proteins , Transcription Factors , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Binding Sites , Nucleosomes/genetics , Nucleosomes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Nucleic Acids Res ; 46(10): 5159-5170, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29660017

ABSTRACT

Thymine DNA glycosylase (TDG) excises thymine from mutagenic G·T mispairs generated by deamination of 5-methylcytosine (mC) and it removes two mC derivatives, 5-formylcytosine (fC) and 5-carboxylcytosine (caC), in a multistep pathway for DNA demethylation. TDG is modified by small ubiquitin-like modifier (SUMO) proteins, but the impact of sumoylation on TDG activity is poorly defined and the functions of TDG sumoylation remain unclear. We determined the effect of TDG sumoylation, by SUMO-1 or SUMO-2, on substrate binding and catalytic parameters. Single turnover experiments reveal that sumoylation dramatically impairs TDG base-excision activity, such that G·T activity is reduced by ≥45-fold and fC and caC are excised slowly, with a reaction half-life of ≥9 min (37°C). Fluorescence anisotropy studies reveal that unmodified TDG binds tightly to G·fC and G·caC substrates, with dissociation constants in the low nanomolar range. While sumoylation of TDG weakens substrate binding, the residual affinity is substantial and is comparable to that of biochemically-characterized readers of fC and caC. Our findings raise the possibility that sumoylation enables TDG to function, at least transiently, as reader of fC and caC. Notably, sumoylation could potentially facilitate TDG recruitment of other proteins, including transcription factors or epigenetic regulators, to these sites in DNA.


Subject(s)
Thymine DNA Glycosylase/metabolism , Catalysis , Cytosine/analogs & derivatives , Cytosine/metabolism , Fluorescence Polarization , Humans , SUMO-1 Protein/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Thymine DNA Glycosylase/chemistry , Thymine DNA Glycosylase/genetics
3.
Methods Enzymol ; 592: 357-376, 2017.
Article in English | MEDLINE | ID: mdl-28668127

ABSTRACT

Base excision repair (BER) is a conserved and ubiquitous pathway that is initiated by DNA glycosylases, which recognize and remove damaged or mismatched nucleobases, setting the stage for restoration of the correct DNA sequence by follow-on BER enzymes. DNA glycosylases employ a nucleotide-flipping step prior to cleavage of the N-glycosyl bond, and most exhibit slow release of the abasic DNA product and/or strong product inhibition. As such, studying the catalytic mechanism of these enzymes requires care in the design, execution, and interpretation of single- and multiple-turnover kinetics experiments, which is the topic of this chapter.


Subject(s)
DNA Glycosylases/metabolism , DNA Repair , Enzyme Assays/methods , Animals , DNA Damage , Humans , Kinetics
4.
Biochemistry ; 55(45): 6205-6208, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27805810

ABSTRACT

Thymine DNA glycosylase (TDG) is a base excision repair enzyme with key functions in epigenetic regulation. Performing a critical step in a pathway for active DNA demethylation, TDG removes 5-formylcytosine and 5-carboxylcytosine, oxidized derivatives of 5-methylcytosine that are generated by TET (ten-eleven translocation) enzymes. We determined a crystal structure of TDG bound to DNA with a noncleavable (2'-fluoroarabino) analogue of 5-formyldeoxycytidine flipped into its active site, revealing how it recognizes and hydrolytically excises fC. Together with previous structural and biochemical findings, the results illustrate how TDG employs an adaptable active site to excise a broad variety of nucleobases from DNA.


Subject(s)
Cytosine/analogs & derivatives , DNA/metabolism , Thymine DNA Glycosylase/metabolism , 5-Methylcytosine/chemistry , 5-Methylcytosine/metabolism , Catalytic Domain , Crystallography, X-Ray , Cytosine/chemistry , Cytosine/metabolism , DNA/chemistry , DNA/genetics , DNA Methylation , DNA Repair , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Structure , Nucleic Acid Conformation , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/metabolism , Oxidation-Reduction , Protein Binding , Protein Domains , Substrate Specificity , Thymine DNA Glycosylase/chemistry
5.
Nucleic Acids Res ; 44(21): 10248-10258, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27580719

ABSTRACT

Thymine DNA Glycosylase (TDG) is a base excision repair enzyme functioning in DNA repair and epigenetic regulation. TDG removes thymine from mutagenic G·T mispairs arising from deamination of 5-methylcytosine (mC), and it processes other deamination-derived lesions including uracil (U). Essential for DNA demethylation, TDG excises 5-formylcytosine and 5-carboxylcytosine, derivatives of mC generated by Tet (ten-eleven translocation) enzymes. Here, we report structural and functional studies of TDG82-308, a new construct containing 29 more N-terminal residues than TDG111-308, the construct used for previous structures of DNA-bound TDG. Crystal structures and NMR experiments demonstrate that most of these N-terminal residues are disordered, for substrate- or product-bound TDG82-308 Nevertheless, G·T substrate affinity and glycosylase activity of TDG82-308 greatly exceeds that of TDG111-308 and is equivalent to full-length TDG. We report the first high-resolution structures of TDG in an enzyme-substrate complex, for G·U bound to TDG82-308 (1.54 Å) and TDG111-308 (1.71 Å), revealing new enzyme-substrate contacts, direct and water-mediated. We also report a structure of the TDG82-308 product complex (1.70 Å). TDG82-308 forms unique enzyme-DNA interactions, supporting its value for structure-function studies. The results advance understanding of how TDG recognizes and removes modified bases from DNA, particularly those resulting from deamination.


Subject(s)
DNA Damage , DNA/chemistry , DNA/metabolism , Thymine DNA Glycosylase/chemistry , Thymine DNA Glycosylase/metabolism , Amino Acid Sequence , Binding Sites , DNA/genetics , Enzyme Activation , Humans , Hydrophobic and Hydrophilic Interactions , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Conformation , Protein Binding , Protein Interaction Domains and Motifs , Recombinant Proteins , Structure-Activity Relationship , Substrate Specificity
6.
Chem Rev ; 116(20): 12711-12729, 2016 10 26.
Article in English | MEDLINE | ID: mdl-27501078

ABSTRACT

Base excision repair (BER) is one of several DNA repair pathways found in all three domains of life. BER counters the mutagenic and cytotoxic effects of damage that occurs continuously to the nitrogenous bases in DNA, and its critical role in maintaining genomic integrity is well established. However, BER also performs essential functions in processes other than DNA repair, where it acts on naturally modified bases in DNA. A prominent example is the central role of BER in mediating active DNA demethylation, a multistep process that erases the epigenetic mark 5-methylcytosine (5mC), and derivatives thereof, converting them back to cytosine. Herein, we review recent advances in the understanding of how BER mediates this critical component of epigenetic regulation in plants and animals.


Subject(s)
DNA Repair , DNA/genetics , Epigenesis, Genetic , Animals , DNA Methylation , DNA, Plant/genetics , Plants/genetics , Vertebrates/genetics
7.
J Biol Chem ; 291(17): 9014-24, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-26917720

ABSTRACT

Thymine-DNA glycosylase (TDG) plays critical roles in DNA base excision repair and DNA demethylation. It has been proposed, based on structural studies and in vitro biochemistry, that sumoylation is required for efficient TDG enzymatic turnover following base excision. However, whether sumoylation is required for TDG activity in vivo has not previously been tested. We have developed an in vivo assay for TDG activity that takes advantage of its recently discovered role in DNA demethylation and selective recognition and repair of 5-carboxylcytosine. Using this assay, we investigated the role of sumoylation in regulating TDG activity through the use of TDG mutants defective for sumoylation and Small Ubiquitin-like Modifier (SUMO) binding and by altering TDG sumoylation through SUMO and SUMO protease overexpression experiments. Our findings indicate that sumoylation and SUMO binding are not essential for TDG-mediated excision and repair of 5-carboxylcytosine bases. Moreover, in vitro assays revealed that apurinic/apyrimidinic nuclease 1 provides nearly maximum stimulation of TDG processing of G·caC substrates. Thus, under our assay conditions, apurinic/apyrimidinic nuclease 1-mediated stimulation or other mechanisms sufficiently alleviate TDG product inhibition and promote its enzymatic turnover in vivo.


Subject(s)
Cytosine/analogs & derivatives , DNA Methylation/physiology , Mutation , SUMO-1 Protein/metabolism , Sumoylation/physiology , Thymine DNA Glycosylase/metabolism , Cytosine/metabolism , HEK293 Cells , Humans , SUMO-1 Protein/genetics , Thymine DNA Glycosylase/genetics
8.
Nucleic Acids Res ; 43(19): 9541-52, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26358812

ABSTRACT

Thymine DNA Glycosylase (TDG) performs essential functions in maintaining genetic integrity and epigenetic regulation. Initiating base excision repair, TDG removes thymine from mutagenic G ·: T mispairs caused by 5-methylcytosine (mC) deamination and other lesions including uracil (U) and 5-hydroxymethyluracil (hmU). In DNA demethylation, TDG excises 5-formylcytosine (fC) and 5-carboxylcytosine (caC), which are generated from mC by Tet (ten-eleven translocation) enzymes. Using improved crystallization conditions, we solved high-resolution (up to 1.45 Å) structures of TDG enzyme-product complexes generated from substrates including G·U, G·T, G·hmU, G·fC and G·caC. The structures reveal many new features, including key water-mediated enzyme-substrate interactions. Together with nuclear magnetic resonance experiments, the structures demonstrate that TDG releases the excised base from its tight product complex with abasic DNA, contrary to previous reports. Moreover, DNA-free TDG exhibits no significant binding to free nucleobases (U, T, hmU), indicating a Kd >> 10 mM. The structures reveal a solvent-filled channel to the active site, which might facilitate dissociation of the excised base and enable caC excision, which involves solvent-mediated acid catalysis. Dissociation of the excised base allows TDG to bind the beta rather than the alpha anomer of the abasic sugar, which might stabilize the enzyme-product complex.


Subject(s)
Base Pair Mismatch , DNA/chemistry , Thymine DNA Glycosylase/chemistry , Catalytic Domain , Crystallography, X-Ray , DNA/metabolism , Humans , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Pentoxyl/analogs & derivatives , Pentoxyl/chemistry , Pentoxyl/metabolism , Protein Binding , Thymine/metabolism , Thymine DNA Glycosylase/metabolism , Uracil/metabolism
9.
J Biol Chem ; 289(22): 15810-9, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24753249

ABSTRACT

Thymine DNA glycosylase (TDG) initiates the repair of G·T mismatches that arise by deamination of 5-methylcytosine (mC), and it excises 5-formylcytosine and 5-carboxylcytosine, oxidized forms of mC. TDG functions in active DNA demethylation and is essential for embryonic development. TDG forms a tight enzyme-product complex with abasic DNA, which severely impedes enzymatic turnover. Modification of TDG by small ubiquitin-like modifier (SUMO) proteins weakens its binding to abasic DNA. It was proposed that sumoylation of product-bound TDG regulates product release, with SUMO conjugation and deconjugation needed for each catalytic cycle, but this model remains unsubstantiated. We examined the efficiency and specificity of TDG sumoylation using in vitro assays with purified E1 and E2 enzymes, finding that TDG is modified efficiently by SUMO-1 and SUMO-2. Remarkably, we observed similar modification rates for free TDG and TDG bound to abasic or undamaged DNA. To examine the conjugation step directly, we determined modification rates (kobs) using preformed E2∼SUMO-1 thioester. The hyperbolic dependence of kobs on TDG concentration gives kmax = 1.6 min(-1) and K1/2 = 0.55 µM, suggesting that E2∼SUMO-1 has higher affinity for TDG than for the SUMO targets RanGAP1 and p53 (peptide). Whereas sumoylation substantially weakens TDG binding to DNA, TDG∼SUMO-1 still binds relatively tightly to AP-DNA (Kd ∼50 nM). Although E2∼SUMO-1 exhibits no specificity for product-bound TDG, the relatively high conjugation efficiency raises the possibility that E2-mediated sumoylation could stimulate product release in vivo. This and other implications for the biological role and mechanism of TDG sumoylation are discussed.


Subject(s)
DNA Repair/physiology , SUMO-1 Protein/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Thymine DNA Glycosylase/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , DNA Methylation/physiology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Glycosylation , HeLa Cells , Humans , Protein Processing, Post-Translational/physiology , Protein Structure, Secondary , Protein Structure, Tertiary , SUMO-1 Protein/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , Substrate Specificity , Sumoylation/physiology , Thymine DNA Glycosylase/chemistry , Thymine DNA Glycosylase/genetics , Ubiquitin-Conjugating Enzymes/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...