Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Biochemistry ; 48(50): 11994-2004, 2009 Dec 22.
Article in English | MEDLINE | ID: mdl-19899814

ABSTRACT

The selection of nucleoside triphosphates by a polymerase is controlled by several energetic and structural features, including base pairing geometry as well as sugar structure and conformation. Whereas base pairing has been considered exhaustively, substantially less is known about the role of sugar modifications for both nucleotide incorporation and primer extension. In this study, we synthesized oligonucleotides containing 2'-fluoro-modified nucleosides with constrained sugar pucker in an internucleotide position and, for the first time, at a primer 3'-end. The thermodynamic stability of these duplexes was examined. The nucleoside 2'-deoxy-2'-fluoroarabinofuranosyluracil [U(2'F(ara))] favors the 2'-endo conformation (DNA-like), while 2'-deoxy-2'-fluororibofuranosyluracil [U(2'F(ribo))] favors the 3'-endo conformation (RNA-like). Oligonucleotides containing U(2'F(ara)) have slightly higher melting temperatures (T(m)) than those containing U(2'F(ribo)) when located in internucleotide positions or at the 3'-end and when correctly paired with adenine or mispaired with guanine. However, both modifications decrease the magnitude of DeltaH degrees and DeltaS degrees for duplex formation in all sequence contexts. In examining the thermodynamic properties for this set of oligonucleotides, we find entropy-enthalpy compensation is apparent. Our thermodynamic findings led to a series of experiments with DNA ligase that reveal, contrary to expectation based upon observed T(m) values, that the duplex containing the U(2'F(ribo)) analogue is more easily ligated. The 2'-fluoro-2'-deoxynucleosides examined here are valuable probes of the impact of sugar constraint and are also members of an important class of antitumor and antiviral agents. The data reported here may facilitate an understanding of the biological properties of these agents, as well as the contribution of sugar conformation to replication fidelity.


Subject(s)
Base Pair Mismatch , Base Pairing , Carbohydrates/chemistry , Thermodynamics , Carbohydrate Conformation , Carbohydrate Sequence , DNA Ligase ATP , DNA Ligases/chemistry , Deoxyuridine/analogs & derivatives , Deoxyuridine/chemistry , Escherichia coli Proteins/chemistry , Humans , Molecular Sequence Data , Nucleic Acid Heteroduplexes/chemistry , Oligonucleotides/chemical synthesis , Poly-ADP-Ribose Binding Proteins , Xenopus Proteins
2.
Chem Res Toxicol ; 22(6): 1194-204, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19480391

ABSTRACT

The nucleoside analogue 5-aza-2'-deoxycytidine (Decitabine, DAC) is one of several drugs in clinical use that inhibit DNA methyltransferases, leading to a decrease of 5-methylcytosine in newly replicated DNA and subsequent transcriptional activation of genes silenced by cytosine methylation. In addition to methyltransferase inhibition, DAC has demonstrated toxicity and potential mutagenicity, and can induce a DNA-repair response. The mechanisms accounting for these events are not well understood. DAC is chemically unstable in aqueous solutions, but there is little consensus between previous reports as to its half-life and corresponding products of decomposition at physiological temperature and pH, potentially confounding studies on its mechanism of action and long-term use in humans. Here, we have employed a battery of analytical methods to estimate kinetic rates and to characterize DAC decomposition products under conditions of physiological temperature and pH. Our results indicate that DAC decomposes into a plethora of products, formed by hydrolytic opening and deformylation of the triazine ring, in addition to anomerization and possibly other changes in the sugar ring structure. We also discuss the advantages and problems associated with each analytical method used. The results reported here will facilitate ongoing studies and clinical trials aimed at understanding the mechanisms of action, toxicity, and possible mutagenicity of DAC and related analogues.


Subject(s)
Azacitidine/analogs & derivatives , Enzyme Inhibitors/chemistry , Azacitidine/chemistry , Chromatography, High Pressure Liquid , DNA Modification Methylases/antagonists & inhibitors , DNA Modification Methylases/metabolism , Decitabine , Gas Chromatography-Mass Spectrometry , Half-Life , Hydrogen-Ion Concentration , Kinetics , Magnetic Resonance Spectroscopy , Spectrophotometry, Ultraviolet , Temperature , Time Factors
3.
Anal Biochem ; 379(2): 196-207, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18485883

ABSTRACT

Oligonucleotides containing modified bases are commonly used for biochemical and biophysical studies to assess the impact of specific types of chemical damage on DNA structure and function. In contrast to the synthesis of oligonucleotides with normal DNA bases, oligonucleotide synthesis with modified bases often requires modified synthetic or deprotection conditions. Furthermore, several modified bases of biological interest are prone to further damage during synthesis and oligonucleotide isolation. In this article, we describe the application of matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS) to the characterization of a series of modified synthetic oligonucleotides. The potential for and limits in obtaining high mass accuracy for confirming oligonucleotide composition are discussed. Examination of the isotope cluster is also proposed as a method for confirming oligonucleotide elemental composition. MALDI-TOF-MS analysis of the unpurified reaction mixture can be used to confirm synthetic sequence and to reveal potential problems during synthesis. Analysis during and after purification can yield important information on depurination and base oxidation. It can also reveal unexpected problems that can occur with nonstandard synthesis, deprotection, or purification strategies. Proper characterization of modified oligonucleotides is essential for the correct interpretation of experiments performed with these substrates, and MALDI-TOF-MS analysis provides a simple yet extensive method of characterization that can be used at multiple stages of oligonucleotide production and use.


Subject(s)
Oligonucleotides/chemistry , Oligonucleotides/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Artifacts , Base Composition , Base Sequence , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA Damage , Exodeoxyribonucleases/metabolism , Hydrolysis , Nucleic Acid Denaturation , Oligonucleotides/chemical synthesis , Oligonucleotides/genetics , Oxidation-Reduction , Purines/chemistry , Thermodynamics
4.
Chem Res Toxicol ; 20(12): 1787-96, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17914883

ABSTRACT

The DNA of all organisms is constantly damaged by oxidation. Among the array of damage products is 5-hydroxymethyluracil, derived from oxidation of the thymine methyl group. Previous studies have established that HmU can be a sensitive and valuable marker of DNA damage. More recently, the corresponding deoxynucleoside, 5-hydroxymethyl-2'-deoxyuridine (HmdU), has proven to be valuable for the introduction of controlled amounts of a single type of damage lesion into the DNA of replicating cells, which is subsequently repaired by the base excision repair pathway. Complicating the study of HmU formation and repair, however, is the known chemical reactivity of the hydroxymethyl group of HmU under conditions used to hydrolyze DNA. In the work reported here, this chemical property has been exploited by creating conditions that convert HmU to the corresponding methoxymethyluracil (MmU) derivative that can be further derivatized to the 3,5-bis-(trifluoromethyl)benzyl analogue. This derivatized compound can be detected by gas chromatography-negative chemical ionization-mass spectrometry (GC-NCI-MS) with good sensitivity. Using isotopically enriched exogenous HmdU and human osteosarcoma cells (U2OS) in culture, we demonstrate that this method allows for the measurement of HmU in DNA formed from the incorporation of exogenous HmdU. We further demonstrate that the addition of isotopically enriched uridine to the culture medium allows for the simultaneous measurement of DNA replication and repair kinetics. This sensitive and facile method should prove valuable for studies on DNA oxidation damage and repair in living cells.


Subject(s)
DNA Damage , DNA Repair , Gas Chromatography-Mass Spectrometry/methods , Thymidine/analogs & derivatives , Biomarkers/analysis , Cell Line, Tumor , Cell Survival/drug effects , Formates/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Hydrolysis , Sensitivity and Specificity , Thymidine/analysis , Thymidine/pharmacology , Thymine/metabolism
5.
Nucleic Acids Res ; 33(9): 3057-64, 2005.
Article in English | MEDLINE | ID: mdl-15917437

ABSTRACT

Perturbations in cytosine methylation signals are observed in the majority of human tumors; however, it is as yet unknown how methylation patterns become altered. Epigenetic changes can result in the activation of transforming genes as well as in the silencing of tumor suppressor genes. We report that methyl-CpG-binding proteins (MBPs), specific for methyl-CpG dinucleotides, bind with high affinity to halogenated pyrimidine lesions, previously shown to result from peroxidase-mediated inflammatory processes. Emerging data suggest that the initial binding of MBPs to methyl-CpG sequences may be a seeding event that recruits chromatin-modifying enzymes and DNA methyltransferase, initiating a cascade of events that result in gene silencing. MBD4, a protein with both methyl-binding and glycosylase activity demonstrated repair activity against a series of 5-substituted pyrimidines, with the greatest efficiency against 5-chlorouracil, but undetectable activity against 5-chlorocytosine. The data presented here suggest that halogenated pyrimidine damage products can potentially accumulate and mimic endogenous methylation signals.


Subject(s)
Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , CpG Islands , DNA Damage , DNA Methylation , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Pyrimidines/chemistry , Repressor Proteins/chemistry , Repressor Proteins/metabolism , 5-Methylcytosine/chemistry , Animals , Binding Sites , DNA Glycosylases/metabolism , Electrophoretic Mobility Shift Assay , Endodeoxyribonucleases/metabolism , Halogens/chemistry , Humans , Methyl-CpG-Binding Protein 2 , Mice , Protein Binding , Protein Structure, Tertiary
6.
Chem Res Toxicol ; 17(9): 1236-44, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15377157

ABSTRACT

Recent studies have shown that reactive chlorine species, derived from myeloperoxidase-mediated inflammation responses, can modify DNA bases, generating 5-chloropyrimidines. The chlorinated adducts could be mutagenic or perturb DNA-protein interactions; however, the biological significance of these adducts is as yet unknown. We report here a method for the synthesis of 5-chlorocytosine- (ClC-) containing oligonucleotides that will be used in subsequent biochemical and biophysical studies to determine the consequences of pyrimidine chlorination. The ClC-phosphoramidite synthon is obtained by chlorination of 2'-deoxyuridine followed by conversion to the O(4)-ethyl analogue. The amino group needed to form the corresponding cytosine derivative is added by displacement of the O(4)-ethyl group during ammonia deprotection. A battery of methods, including mass spectrometry, has been used to characterize oligonucleotides containing ClC. Following oligonucleotide synthesis and deprotection, only trace amounts of the deamination product 5-chlorouracil can be detected by enzymatic cleavage of duplex oligonucleotides with the mispaired uracil glycosylase, MUG. In contrast to previous reports, we find that ClC is more stable in DNA than anticipated. Approximately 20% ClC is lost under standard formic acid hydrolysis conditions (88% formic acid, 140 degrees C, 30 min), while only 5% is recovered as 5-chlorouracil (ClU).


Subject(s)
Cytosine/analogs & derivatives , Cytosine/chemistry , Mutagens/chemical synthesis , Oligonucleotides/chemical synthesis , Uracil/analogs & derivatives , Deamination , Formates , Hydrolysis , Oligonucleotides/chemistry , Uracil/chemical synthesis
7.
Nucleic Acids Res ; 32(14): 4100-8, 2004.
Article in English | MEDLINE | ID: mdl-15302911

ABSTRACT

Cytosine methylation in CpG dinucleotides is believed to be important in gene regulation, and is generally associated with reduced levels of transcription. Methylation-mediated gene silencing involves a series of DNA-protein and protein-protein interactions that begins with the binding of methyl-CpG binding proteins (MBPs) followed by the recruitment of histone-modifying enzymes that together promote chromatin condensation and inactivation. It is widely known that alterations in methylation patterns, and associated gene activities, are often found in human tumors. However, the mechanisms by which methylation patterns are altered are not currently understood. In this paper, we investigate the impact of oxidative damage to a methyl-CpG site on MBP binding by the selective placement of 8-oxoguanine (8-oxoG) and 5-hydroxymethylcytosine (HmC) in a MBP recognition sequence. Duplexes containing these specific modifications were assayed for binding to the methyl-CpG binding domain (MBD) of one member of the MBP family, methyl-CpG binding protein 2 (MeCP2). Our results reveal that oxidation of either a single guanine to 8-oxoG or of a single 5mC to HmC, significantly inhibits binding of the MBD to the oligonucleotide duplex, reducing the binding affinity by at least an order of magnitude. Oxidative damage to DNA could therefore result in heritable, epigenetic changes in chromatin organization.


Subject(s)
5-Methylcytosine/chemistry , Chromosomal Proteins, Non-Histone , CpG Islands , Cytosine/analogs & derivatives , Cytosine/chemistry , DNA-Binding Proteins/chemistry , Guanosine/analogs & derivatives , Guanosine/chemistry , Repressor Proteins , Base Sequence , Binding Sites , DNA Damage , DNA Methylation , DNA-Binding Proteins/metabolism , Electrophoretic Mobility Shift Assay , Methyl-CpG-Binding Protein 2 , Models, Molecular , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/metabolism , Oxidative Stress , Protein Structure, Tertiary
8.
Nucleic Acids Res ; 32(15): 4503-11, 2004.
Article in English | MEDLINE | ID: mdl-15328364

ABSTRACT

DNA ligases, found in both prokaryotes and eukaryotes, covalently link the 3'-hydroxyl and 5'-phosphate ends of duplex DNA segments. This reaction represents a completion step for DNA replication, repair and recombination. It is well established that ligases are sensitive to mispairs present on the 3' side of the ligase junction, but tolerant of mispairs on the 5' side. While such discrimination would increase the overall accuracy of DNA replication and repair, the mechanisms by which this fidelity is accomplished are as yet unknown. In this paper, we present the results of experiments with Tth ligase from Thermus thermophilus HB8 and a series of nucleoside analogs in which the mechanism of discrimination has been probed. Using a series of purine analogs substituted in the 2 and 6 positions, we establish that the apparent base pair geometry is much more important than relative base pair stability and that major groove contacts are of little importance. This result is further confirmed using 5-fluorouracil (FU) mispaired with guanine. At neutral pH, the FU:G mispair on the 3' side of a ligase junction is predominantly in a neutral wobble configuration and is poorly ligated. Increasing the solution pH increases the proportion of an ionized base pair approximating Watson-Crick geometry, substantially increasing the relative ligation efficiency. These results suggest that the ligase could distinguish Watson-Crick from mispaired geometry by probing the hydrogen bond acceptors present in the minor groove as has been proposed for DNA polymerases. The significance of minor groove hydrogen bonding interactions is confirmed with both Tth and T4 DNA ligases upon examination of base pairs containing the pyrimidine shape analog, difluorotoluene (DFT). Although DFT paired with adenine approximates Watson-Crick geometry, a minor groove hydrogen bond acceptor is lost. Consistent with this hypothesis, we observe that DFT-containing base pairs inhibit ligation when on the 3' side of the ligase junction. The NAD+-dependent ligase, Tth, is more sensitive to the DFT analog on the unligated strand whereas the ATP-dependent T4 ligase is more sensitive to substitutions in the template strand. Electrophoretic gel mobility-shift assays demonstrate that the Tth ligase binds poorly to oligonucleotide substrates containing analogs with altered minor groove contacts.


Subject(s)
DNA Ligases/metabolism , DNA/metabolism , Base Pairing , Binding Sites , DNA/chemistry , Fluorouracil/chemistry , Guanine/chemistry , Hydrogen Bonding , Hydrogen-Ion Concentration , Nucleic Acid Conformation , Uracil/chemistry
9.
Biochemistry ; 43(19): 5688-97, 2004 May 18.
Article in English | MEDLINE | ID: mdl-15134443

ABSTRACT

Oxidation of the thymine methyl group can generate 5-formyluracil (FoU), which is known to be both mutagenic and chemically unstable in DNA. Synthetic oligonucleotides containing FoU at defined sites have been prepared to investigate potential mechanisms by which FoU might perturb DNA function. The half-life of the glycosidic bond of an FoU residue in single-stranded DNA under physiological conditions of temperature and pH is estimated to be approximately 148 days, orders of magnitude shorter than the parent pyrimidine, thymine. This reduced stability of FoU residues in DNA is attributed to the inductive properties of the 5-formyl substituent. Oxidative modification of the thymine methyl group could also inhibit association with sequence-specific DNA-binding proteins. Alternatively, the 5-formyl substituent of FoU could cross-link nonspecifically with protein amino groups. Transcription factor AP-1 is known to make specific contacts with thymine methyl groups of DNA in its recognition sequence. Substitution of T by FoU is shown to inhibit AP-1 (c-Jun homodimer) binding with a DeltaDeltaG of approximately 0.6 kcal/mol. No evidence of cross-link formation is observed with either AP-1 or polylysine. Molecular modeling studies on the FoU-containing oligonucleotide sequence corresponding to the duplex used in the experimental studies demonstrate that the 5-formyl substituent of an FoU residue paired with adenine lies in the plane of the pyrimidine base and is well protected from solvent on one face and only partially accessible on the other. The results of this study suggest that although FoU residues in DNA are considerably more labile than thymine, they are likely to be present long enough to miscode as well as interfere with DNA-protein interactions.


Subject(s)
DNA/chemistry , DNA/metabolism , Mutagens/chemistry , Uracil/analogs & derivatives , Uracil/chemistry , Binding, Competitive , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/metabolism , DNA/drug effects , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/drug effects , DNA, Single-Stranded/metabolism , Glycosides/chemistry , Glycosides/metabolism , Hydrolysis , Models, Molecular , Mutagens/isolation & purification , Mutagens/metabolism , Nucleic Acid Heteroduplexes/chemistry , Nucleic Acid Heteroduplexes/metabolism , Peptides/chemistry , Peptides/metabolism , Polylysine/chemistry , Polylysine/metabolism , Protein Binding/drug effects , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Proto-Oncogene Proteins c-jun/metabolism , Thionucleotides/chemical synthesis , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/metabolism , Uracil/isolation & purification , Uracil/metabolism
10.
DNA Repair (Amst) ; 2(2): 199-210, 2003 Feb 03.
Article in English | MEDLINE | ID: mdl-12531390

ABSTRACT

The oxidation of the thymine methyl group can generate 5-formyluracil (FoU). Template FoU residues are known to miscode, generating base substitution mutations. The repair of the FoU lesion is therefore important in minimizing mutations induced by DNA oxidation. We have studied the repair of FoU in synthetic oligonucleotides when paired with A and G. In E. coli cell extract, the repair of FoU is four orders of magnitude lower than the repair of U and is similar for both FoU:A and FoU:G base pairs. In HeLa nuclear extract, the repair of FoU:A is similarly four orders of magnitude lower than the repair of uracil, although the FoU:G lesion is repaired 10 times more efficiently than FoU:A. The FoU:G lesion is shown to be repaired by E. coli mismatch uracil DNA glycosylase (Mug), thermophile mismatch thymine DNA glycosylase (Tdg), mouse mismatch thymine DNA glycosylase (mTDG) and human methyl-CpG-binding thymine DNA glycosylase (MBD4), whereas the FoU:A lesion is repaired only by Mug and mTDG. The repair of FoU relative to the other pyrimidines examined here in human cell extract differs from the substrate preferences of the known glycosylases, suggesting that additional, and as yet unidentified glycosylases exist in human cells to repair the FoU lesion. Indeed, as observed in HeLa nuclear extract, the repair of mispaired FoU derived from misincorporation of dGMP across from template FoU could promote rather than minimize mutagenesis. The pathways by which this important lesion is repaired in human cells are as yet unexplained, and are likely to be complex.


Subject(s)
DNA Repair , Uracil/analogs & derivatives , Uracil/metabolism , Escherichia coli/physiology , HeLa Cells , Humans
11.
Chem Res Toxicol ; 15(12): 1595-601, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12482242

ABSTRACT

To maintain genomic integrity, DNA repair enzymes continually remove damaged bases and lesions resulting from endogenous and exogenous processes. These repair enzymes must distinguish damaged bases from normal bases to prevent the inadvertent removal of normal bases, which would promote genomic instability. The mechanisms by which this high level of specificity is accomplished are as yet unresolved. One member of the uracil-DNA glycosylase family of repair enzymes, Escherichia coli mismatch-specific uracil-DNA glycosylase (Mug), is reported to distinguish U:G mispairs from U:A base pairs based upon specific contacts with the mispaired guanine after flipping the target uracil out of the duplex. However, recent studies suggest other mechanisms for base selection, including local duplex stability. In this study, we used the modified base N6-methyladenine to probe the effect of local helix perturbation on Mug recognition of uracil. N6-Methyladenine is found in E. coli as part of both the mismatch repair and restriction-modification systems. In its cis isomer, N6-methyladenine destabilizes hydrogen bonding by interfering with pseudo-Watson-Crick base pairing. It is observed that the selection of uracil by Mug is sequence dependent and that uracil residues in sequences of reduced thermostability are preferentially removed. The replacement of adenine by N6-methyladenine increases the frequency of removal of the uracil residue paired opposite the modified adenine. These results are in accord with suggestions that local helix stability is an important determinant of base recognition by some DNA repair enzymes and provide a potential strategy for identifying the sequence location of modified bases in DNA.


Subject(s)
Adenine/analogs & derivatives , Adenine/metabolism , Base Pair Mismatch , DNA Glycosylases , DNA/metabolism , N-Glycosyl Hydrolases/metabolism , Oligonucleotides/metabolism , Uracil/metabolism , Adenine/chemistry , Base Composition , Base Sequence , DNA/chemistry , DNA Repair , Drug Stability , Escherichia coli/enzymology , Nucleic Acid Denaturation , Oligonucleotides/chemical synthesis , Oligonucleotides/chemistry , Phosphorus Isotopes , Substrate Specificity , Temperature , Thymine/chemistry , Thymine/metabolism , Uracil/chemistry , Uracil-DNA Glycosidase
12.
Nucleic Acids Res ; 30(18): 4068-74, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12235391

ABSTRACT

A specific and efficient method is presented for the conversion of 2'-deoxyuridine to thymidine via formation and reduction of the intermediate 5-hydroxymethyl derivative. The method has been used to generate both thymidine and 5-methyl-2'-deoxycytidine containing the stable isotopes 2H, 13C and 15N. Oligodeoxyribonucleotides have been constructed with these mass-tagged bases to investigate sequence-selectivity in hydroxyl radical reactions of pyrimidine methyl groups monitored by mass spectrometry. Studying the reactivity of 5-methylcytosine (5mC) is difficult as the reaction products can deaminate to the corresponding thymine derivatives, making the origin of the reaction products ambiguous. The method reported here can distinguish products derived from 5mC and thymine as well as investigate differences in reactivity for either base in different sequence contexts. The efficiency of formation of 5-hydroxymethyluracil from thymine is observed to be similar in magnitude in two different sequence contexts and when present in a mispair with guanine. The oxidation of 5mC proceeds slightly more efficiently than that of thymine and generates both 5-hydroxymethylcytosine and 5-formylcytosine but not the deaminated products. Thymine glycol is generated by both thymine and 5mC, although with reduced efficiency for 5mC. The method presented here should be widely applicable, enabling the examination of the reactivity of selected bases in DNA.


Subject(s)
DNA/chemistry , Pyrimidines/chemical synthesis , Base Sequence , Carbon Isotopes , DNA Probes/chemistry , Deuterium , Gas Chromatography-Mass Spectrometry , Hydroxyl Radical/chemistry , Nitrogen Isotopes , Oligonucleotides/chemistry , Reactive Oxygen Species/chemistry , Thymidine/chemistry , Thymine/chemistry
13.
Chem Res Toxicol ; 15(8): 1001-9, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12184783

ABSTRACT

In response to continuous hydrolytic and oxidative DNA damage, cells of all organisms have a complex network of repair systems that recognize, remove, and rebuild the injured sites. Damaged pyrimidines are generally removed by glycosylases that must scan the entire genome to locate lesions with sufficient fidelity to selectively remove the damage without inadvertent removal of normal bases. We report here studies conducted with a series of base analogues designed to test mechanisms of base recognition suggested by structural studies of glycosylase complexes. The oligonucleotide series examined here includes 5-halouracils with increasing substituent size and purine analogues placed opposite the target uracil with hydrogen, amino, and keto substituents in the 2- and 6-positions. The glycosylases studied here include Escherichia coli uracil-DNA glycosylase (UNG), E. coli mismatch uracil-DNA glycosylase (MUG), and the Methanobacterium thermoautotrophicum mismatch thymine-DNA glycosylase (TDG). The results of this study suggest that these glycosylases utilize several strategies for base identification, including (1) steric limitations on the size of the 5-substituent, (2) electronic-inductive properties of the 5-substituent, (3) reduced thermal stability of mispairs, and (4) specific functional groups on the purine base in the opposing strand. Contrary to predictions based upon the crystal structure, the preference of MUG for mispaired uracil over thymine is not based upon steric exclusion. Furthermore, the preference for mispaired uracil over uracil paired with adenine is more likely due to reduced thermal stability as opposed to specific recognition of the mispaired guanine. On the other hand, TDG, which exhibits modest discrimination among various pyrimidines, shows strong interactions with functional groups present on the purine opposite the target pyrimidine. These results provide new insights into the mechanisms of base selection by DNA repair glycosylases.


Subject(s)
Base Pair Mismatch , DNA Glycosylases , DNA Repair , N-Glycosyl Hydrolases/metabolism , Thymine DNA Glycosylase , Base Sequence , Electrophoresis, Polyacrylamide Gel , Escherichia coli/enzymology , Methanobacterium/enzymology , Molecular Sequence Data , Oligonucleotides/chemistry , Substrate Specificity , Uracil-DNA Glycosidase
14.
Biochemistry ; 41(25): 8093-102, 2002 Jun 25.
Article in English | MEDLINE | ID: mdl-12069602

ABSTRACT

The repair of DNA damage, caused by both endogenous and exogenous sources, is necessary to remove lesions that either miscode or block DNA or RNA polymerases. We propose that damage also must be repaired to maintain sequence-specific DNA-protein interactions. In this paper, we have systematically studied two lesions that interfere with one important DNA landmark, the thymine methyl group. Oxidation of the thymine methyl group in DNA generates 5-hydroxymethyluracil (HmU) whereas the misincorporation of dUMP into DNA generates uracil (U), replacing the methyl group with a hydrogen. Both substitutions are shown to inhibit binding of the AP-1 (c-Jun) transcription factor. The energy cost of the perturbation, approximately 0.4 kcal/mol, is similar in magnitude for both U and HmU substitutions and is additive when multiple substitutions are present. A third lesion, substitution of the central C:G base pair of the AP-1 DNA binding domain with the pro-mutagenic U:G mispair, unexpectedly increases AP-1 binding, allowing the transcription factor to interfere with uracil DNA glycosylase activity. Our results support the hypothesis that an additional role for DNA repair systems is to maintain the integrity of sequence-specific DNA-protein interactions, a role of particular importance in long-lived organisms.


Subject(s)
Base Pair Mismatch , DNA Damage , DNA Glycosylases , Pentoxyl/analogs & derivatives , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/metabolism , Adenine/metabolism , Binding, Competitive , Cytosine/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Humans , N-Glycosyl Hydrolases/metabolism , Nucleic Acid Heteroduplexes/metabolism , Oligonucleotides/metabolism , Pentoxyl/metabolism , Protein Binding , Thymine/metabolism , Transcription Factor AP-1/physiology , Uracil/metabolism , Uracil-DNA Glycosidase
15.
Chem Res Toxicol ; 15(1): 33-9, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11800595

ABSTRACT

The oxidation of pyrimidine 5-methyl groups, derived from either thymine or 5-methylcytosine, can generate 5-hydroxymethyluracil (HmU) in DNA. An activity from HeLa cells that removes 5-hydroxymethyluracil (HmU) from DNA has been partially purified and characterized using a battery of oligonucleotides containing modified bases. This partially purified activity preferentially removes HmU mispaired with guanine. The HmU repair activity also acts on uracil and fluorouracil but not 5-substituted uracil derivatives with halogens larger than fluorine. However, neither mispaired thymine nor ethenocytosine are substrates. HmU is readily removed when paired with guanine, hypoxanthine (deoxyinosine), and purine (deoxynebularine), but not from single-stranded substrates. Upon the basis of these substrate preferences, we conclude that (1) the mispaired HmU repair activity is distinct from previously reported glycosylases including UDG, TDG, MUG, and SMUG1 activities, (2) the binding pocket is highly selective for the 5-hydroxymethyl group, and (3) the preference for mispaired HmU derives from reduced thermal stability of the mispair, as opposed to selective recognition of the mispaired guanine residue in the opposing DNA strand.


Subject(s)
DNA Glycosylases , N-Glycosyl Hydrolases/metabolism , Oligonucleotides/metabolism , HeLa Cells , Humans , Molecular Weight , N-Glycosyl Hydrolases/analysis , Oligonucleotide Probes/chemistry , Oligonucleotides/chemistry , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...