首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
Alkylation of guanine at the O6 atom is a highly mutagenic DNA lesion because it alters the coding specificity of the base causing G:C to A:T transversion mutations. Specific DNA repair enzymes, e.g. O6‐alkylguanin‐DNA‐Transferases (AGT), recognize and repair such damage after looping out the damaged base to transfer it into the enzyme active site. The exact mechanism how the repair enzyme identifies a damaged site within a large surplus of undamaged DNA is not fully understood. The O6‐alkylation of guanine may change the deformability of DNA which may facilitate the initial binding of a repair enzyme at the damaged site. In order to characterize the effect of O6‐methyl‐guanine (O6‐MeG) containing base pairs on the DNA deformability extensive comparative molecular dynamics (MD) simulations on duplex DNA with central G:C, O6‐MeG:C or O6‐MeG:T base pairs were performed. The simulations indicate significant differences in the helical deformability due to the presence of O6‐MeG compared to regular undamaged DNA. This includes enhanced base pair opening, shear and stagger motions and alterations in the backbone fine structure caused in part by transient rupture of the base pairing at the damaged site and transient insertion of water molecules. It is likely that the increased opening motions of O6‐MeG:C or O6‐MeG:T base pairs play a decisive role for the induced fit recognition or for the looping out of the damaged base by repair enzymes. © 2014 Wiley Periodicals, Inc. Biopolymers 103: 23–32, 2015.  相似文献   

2.
O6-alkylguanine-DNA alkyltransferase (AGT) is a single-cycle DNA repair enzyme that removes pro-mutagenic O6-alkylguanine adducts from DNA. Its functions with short single-stranded and duplex substrates have been characterized, but its ability to act on other DNA structures remains poorly understood. Here, we examine the functions of this enzyme on O6-methylguanine (6mG) adducts in the four-stranded structure of the human telomeric G-quadruplex. On a folded 22-nt G-quadruplex substrate, binding saturated at 2 AGT:DNA, significantly less than the ∼5 AGT:DNA found with linear single-stranded DNAs of similar length, and less than the value found with the telomere sequence under conditions that inhibit quadruplex formation (4 AGT:DNA). Despite these differences, AGT repaired 6mG adducts located within folded G-quadruplexes, at rates that were comparable to those found for a duplex DNA substrate under analogous conditions. Repair was kinetically biphasic with the amplitudes of rapid and slow phases dependent on the position of the adduct within the G-quadruplex: in general, adducts located in the top or bottom tetrads of a quadruplex stack exhibited more rapid-phase repair than did adducts located in the inner tetrad. This distinction may reflect differences in the conformational dynamics of 6mG residues in G-quadruplex DNAs.  相似文献   

3.
The protein O 6-alkylguanine-DNA alkyltransferase(alkyltransferase) is involved in the repair of O 6-alkylguanine and O 4-alkylthymine in DNA and plays an important role in most organisms in attenuating the cytotoxic and mutagenic effects of certain classes of alkylating agents. A genomic clone encompassing the Drosophila melanogaster alkyltransferase gene ( DmAGT ) was identified on the basis of sequence homology with corresponding genes in Saccharomyces cerevisiae and man. The DmAGT gene is located at position 84A on the third chromosome. The nucleotide sequence of DmAGT cDNA revealed an open reading frame encoding 194 amino acids. The MNNG-hypersensitive phenotype of alkyltransferase-deficient bacteria was rescued by expression of the DmAGT cDNA. Furthermore, alkyltransferase activity was identified in crude extracts of Escherichia coli harbouring DmAGT cDNA and this activity was inhibited by preincubation of the extract with an oligonucleotide containing a single O6-methylguanine lesion. Similar to E.coli Ogt and yeast alkyltransferase but in contrast to the human alkyltransferase, the Drosophila alkyltransferase is resistant to inactivation by O 6-benzylguanine. In an E.coli lac Z reversion assay, expression of DmAGT efficiently suppressed MNNG-induced G:C-->A:T as well as A:T-->G:C transition mutations in vivo. These results demonstrate the presence of an alkyltransferase specific for the repair of O 6-methylguanine and O 4-methylthymine in Drosophila.  相似文献   

4.
The DNA repair protein O6-alkylguanine alkyltransferase (AGT) is responsible for removing promutagenic alkyl lesions from exocyclic oxygens located in the major groove of DNA, i.e. the O6 and O4 positions of guanine and thymine. The protein carries out this repair reaction by transferring the alkyl group to an active site cysteine and in doing so directly repairs the premutagenic lesion in a reaction that inactivates the protein. In order to trap a covalent AGT–DNA complex, oligodeoxyribonucleotides containing the novel nucleoside N1,O6-ethanoxanthosine (eX) have been prepared. The eX nucleoside was prepared by deamination of 3′,5′-protected O6-hydroxyethyl-2′-deoxyguanosine followed by cyclization to produce 3′,5′-protected N1,O6-ethano-2′-deoxyxanthosine, which was converted to the nucleoside phosphoramidite and used in the preparation of oligodeoxyribonucleotides. Incubation of human AGT with a DNA duplex containing eX resulted in the formation of a covalent protein–DNA complex. Formation of this complex was dependent on both active human AGT and eX and could be prevented by chemical inactivation of the AGT with O6-benzylguanine. The crosslinking of AGT to DNA using eX occurs with high yield and the resulting complex appears to be well suited for further biochemical and biophysical characterization.  相似文献   

5.
The O6-alkylguanine DNA alkyltransferase (AGT) is a highly conserved protein responsible for direct repair of alkylated guanine and to a lesser degree thymine bases. While specific DNA lesion-bound complexes in crystal structures consist of monomeric AGT, several solution studies have suggested that cooperative DNA binding plays a role in the physiological activities of AGT. Cooperative AGT–DNA complexes have been described by theoretical models, which can be tested by atomic force microscopy (AFM). Direct access to structural features of AGT–DNA complexes at the single molecule level by AFM imaging revealed non-specifically bound, cooperative complexes with limited cluster length. Implications of cooperative binding in AGT–DNA interactions are discussed.  相似文献   

6.
The cytotoxicity of SN1-type alkylating agents such as N-methyl-N′-nitrosourea (MNU), N-methyl-N′-nitro-N-nitrosoguanidine (MNNG), or the cancer chemotherapeutics temozolomide, dacarbazine and streptozotocin has been ascribed to the persistence of O6-methylguanine (meG) in genomic DNA. One hypothesis posits that meG toxicity is caused by futile attempts of the mismatch repair (MMR) system to process meG/C or meG/T mispairs arising during replication, while an alternative proposal suggests that the latter lesions activate DNA damage signaling, cell cycle arrest and apoptosis directly. Attempts to elucidate the molecular mechanism of meG-induced cell killing in vivo have been hampered by the fact that the above reagents induce several types of modifications in genomic DNA, which are processed by different repair pathways. In contrast, defined substrates studied in vitro did not undergo replication. We set out to re-examine this phenomenon in replication-competent Xenopus laevis egg extracts, using either phagemid substrates containing a single meG residue, or methylated sperm chromatin. Our findings provide further support for the futile cycling hypothesis.  相似文献   

7.
8.
O6-Alkyldeoxyguanine adducts induced by tobacco-specific nitrosamines are repaired by O6-alkylguanine DNA alkyltransferase (AGT), which transfers the O6-alkyl group from the damaged base to a cysteine residue within the protein. In the present study, a mass spectrometry-based approach was used to analyze the effects of cytosine methylation on the kinetics of AGT repair of O6-methyldeoxyguanosine (O6-Me-dG) adducts placed within frequently mutated 5′-CG-3′ dinucleotides of the p53 tumor suppressor gene. O6-Me-dG-containing DNA duplexes were incubated with human recombinant AGT protein, followed by rapid quenching, acid hydrolysis, and isotope dilution high pressure liquid chromatography-electrospray ionization tandem mass spectrometry analysis of unrepaired O6-methylguanine. Second-order rate constants were calculated in the absence or presence of the C-5 methyl group at neighboring cytosine residues. We found that the kinetics of AGT-mediated repair of O6-Me-dG were affected by neighboring 5-methylcytosine (MeC) in a sequence-dependent manner. AGT repair of O6-Me-dG adducts placed within 5′-CG-3′ dinucleotides of p53 codons 245 and 248 was hindered when MeC was present in both DNA strands. In contrast, cytosine methylation within p53 codon 158 slightly increased the rate of O6-Me-dG repair by AGT. The effects of MeC located immediately 5′ and in the base paired position to O6-Me-dG were not additive as revealed by experiments with hypomethylated sequences. Furthermore, differences in dealkylation rates did not correlate with AGT protein affinity for cytosine-methylated and unmethylated DNA duplexes or with the rates of AGT-mediated nucleotide flipping, suggesting that MeC influences other kinetic steps involved in repair, e.g. the rate of alkyl transfer from DNA to AGT.Metabolic activation of the tobacco carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)3 produces methyl- and pyridyloxobutyldiazonium ions that can react with DNA to give O6-methyldeoxyguanosine (O6-Me-dG) and O6- pyridyloxobutyl deoxyguanosine (O6-POB-dG) lesions (1). Both adducts are strongly mutagenic because DNA polymerases preferentially misinsert thymine opposite O6-alkylguanines, resulting in G → A transitions (2). Studies in laboratory animals have provided evidence for a direct involvement of O6-Me-dG in NNK-mediated carcinogenesis (3).A specialized repair protein, O6-alkylguanine DNA alkyltransferase (AGT), removes the alkyl group from the O6 position of modified guanine bases, such as O6-Me-dG and O6-POB-dG, restoring normal guanine bases and preventing mutagenesis. AGT preferentially binds double-stranded DNA through a helix-turn-helix motif (4). In the resulting AGT-DNA complex, one recognition helix of the protein is found within the minor groove of the DNA, whereas the other one interacts with the phosphodiester backbone (4). The adducted nucleotide is flipped into a binding pocket within the protein, whereas Arg-128 takes its place in the double helix (4). A hydrogen bonding network around the active site involving His-148, Glu-172, and a water molecule promotes the deprotonation of the active site cysteine (Cys-145) (4, 5). The resulting thiolate anion acts as a nucleophile, displacing the O6 substituent of O6-alk-G and regenerating normal guanine (Fig. 1) (4, 5). The alkylated protein is inactive and is rapidly degraded by the ubiquitin proteolytic pathway (68).Open in a separate windowFIGURE 1.Direct repair of O6-alkyl-guanine adducts by AGT.AGT-mediated repair of O6-Me-dG lesions includes multiple kinetic steps (9). First, the AGT protein must bind adducted DNA in a reactive conformation. The alkylated nucleotide is flipped out of the DNA base stack to enter the hydrophobic pocket within AGT, and the methyl group is transferred from DNA to the protein. Finally, alkylated AGT protein dissociates from the repaired DNA. Zang et al. (9) reported that the chemical step of alkyl transfer is rate-limiting in the case of O6-Me-dG, but not O6-benzyl-dG. Furthermore, previous studies have shown that the repair of O6-Me-dG by mammalian AGT is influenced by the nature of the O6-alkyl group, the length of oligonucleotide duplex, the placement of the adduct, and the identities of neighboring nucleotides (1014).Removing the alkyl group from O6-Me-dG by AGT regenerates normal guanine and protects the genome from G → A transition mutations. For example, Wolf et al. (15) examined the relationship between the inactivation of the AGT gene by promoter hypermethylation and the mutational spectrum of the p53 tumor suppressor gene in non-small cell lung cancer. These authors found that only 8% of lung tumors had G → A transition mutations in the p53 gene when the promoter region of the gene coding for AGT was not methylated, thereby allowing protein expression (15). In contrast, 33% of tumors with a methylated AGT promoter had G → A mutations within the p53 gene (15). The p53 gene is mutated in over 50% of non-small cell lung cancer tumors (16).All CpG sites within the coding sequence of the p53 gene are endogenously methylated (17). Importantly, the same sites are among the major p53 mutational “hotspots” in smoking-induced lung cancer, e.g. codons 157, 158, 245, 248, 249, and 273 (18). Of all p53 mutations, G → A transitions account for 18–24% of genetic changes observed in lung cancer, including 35% of mutations at the CG dinucleotides (15, 19). Given the established role of NNK-induced O6-alkylguanine lesions in tobacco carcinogenesis and mutagenesis (20), they are likely to be involved in the induction of smoking-associated G → A transitions in the p53 gene.The presence of MeC residues may hinder the repair of O6-Me-dG lesions within endogenously methylated CG dinucleotides (14). For example, Bentivenga and Bresnick (14) showed that the repair of O6-Me-dG by recombinant AGT in the context of codon 248 of the p53 gene was reduced by 75% when MeC was placed immediately 5′ to the O6-Me-dG lesion. However, the effects of cytosine methylation on AGT repair of O6-Me-dG in other sequence contexts have not been previously investigated, and it is not known which individual steps in the removal of O6-methyl group are affected by neighboring MeC.Cytosine methylation leads to small structural changes of DNA duplex, including an increase in the base pair rise, roll, and local curvature angles, narrowing of the DNA minor groove, and decreased depth of the major groove (2123). These structural alterations may influence the affinity of the AGT protein for alkylated DNA. Furthermore, MeC enhances base stacking (24) and stabilizes the DNA duplex by increasing the molecular polarizability of the cytosine base (25), which can have an effect on the rate of AGT-mediated nucleotide flipping. The alkyl transfer step itself may be mediated by the presence of MeC through its effects on transition state geometry.The goal of the present study was to systematically examine the effects of cytosine methylation on AGT-mediated repair of O6-Me-dG lesions placed within 5′-CG-3′ dinucleotides representing major p53 mutational hotspots observed in lung cancer. The kinetics of alkyl transfer were analyzed using rapid-quench methods coupled with quantitative analyses of O6-Me-dG by isotope dilution-high performance liquid chromatography-electrospray ionization tandem mass spectrometry (HPLC-ESI-MS/MS) (26). Furthermore, we examined the effects of cytosine methylation on AGT binding to O6-Me-dG-containing DNA and its influence on the rate of O6-Me-dG nucleotide flipping in the presence of AGT protein.  相似文献   

9.
10.
The pro-mutagenicity of chemically-induced methylation of DNA at the O6 position of dexoyguanosine was studied in cultured adult rat liver epithelial cells. To modify the level of O6-methyldeoxyguanosine (O6-medGuo) resulting from exposure to an alkylating agent, partial depletion of the O6-alkylguanine-DNA alkyltransferase (AGT) repair system was produced by pretreatment of ARL 18 cells with a non-toxic dose of exogenous O6-methylguanine (O6-meG). Exposure of cells to 0.6 mM O6-meG for 4 h depleted AGT activity by about 40%. Intact and pretreated cells were exposed to a range of doses of N-methyl-N′-nitro-N-nitrosoguanidine (MNNG), and mutagenesis at the hypoxanthine-guanine phosphoribosyl transferase (HGPRT) locus was quantified by measurement of 6-thioguanine-resistant mutants. The mutagenicity of MNNG was dose dependent and was greater in O6-meG pretreated cultures than in intact cultures. Immunoslot blot measurement of O6-medGuo employing a mouse monoclonal antibody demonstrated that MNNG produced O[su6-medGuo and that the intact liver cells were efficient in eliminating this lesion from their DNA. Since depletion of AGT would be expected to affect the rate of elimination of only O6-medGuo, it is concluded that this lesion is highly pro-mutagenic.  相似文献   

11.
T E Spratt  J D Wu  D E Levy  S Kanugula  A E Pegg 《Biochemistry》1999,38(21):6801-6806
O6-Alkylguanine-DNA alkyltransferase (AGT) repairs DNA by transferring the methyl group from the 6-position of guanine to a cysteine residue on the protein. We previously found that the Escherichia coli Ada protein makes critical interactions with O6-methylguanine (O6mG) at the N1- and O6-positions. Human AGT has a different specificity than the bacterial protein. We reacted hAGT with double-stranded pentadecadeoxynucleotides containing analogues of O6mG. The second-order rate constants were in the following order (x10(-)5 M-1 s-1): O6mG (1.4), O6-methylhypoxanthine (1.6) > Se6-methyl-6-selenoguanine (0.1) > S6-methyl-6-thioguanine (S6mG) (0.02) > S6-methyl-6-thiohypoxanthine (S6mH), O6-methyl-1-deazaguanine (O6m1DG), O6-methyl-3-deazaguanine (O6m3DG), and O6-methyl-7-deazaguanine (O6m7DG) (all <0.0001). Electrophoretic mobility shift assays were carried out to determine the binding affinity to hAGT. Oligodeoxynucleotides containing O6mG, S6mG and O6m3DG bound to AGT in the presence of competitor DNA with Kd values from 5 to 20 microM, while those containing G, S6mH, O6m1DG, and O6m7DG did not (Kd > 200 microM). These results indicate that the 1-, N2-, and 7- positions of O6mG are critical in binding to hAGT, while the 3- and O6-positions are involved in methyl transfer. These results suggest that the active site of ada AGT is more flexible than hAGT and may be the reason ada AGT reacts with O4mT faster than hAGT.  相似文献   

12.
13.
14.
The predominant pathway for the repair of O6-methylguanine in DNA is via the activity of an alkyltransferase protein that transfers the methyl group to a cysteine acceptor site on the protein itself. This review article describes recent studies on this alkyltransferase. The protein repairs not only methyl groups but also 2-chloroethyl-, benzyl- and pyridyloxobutyl-adducts. It acts on double-stranded DNA by flipping the O6-guanine adduct out of the DNA helix and into a binding pocket. The free base, O6-benzylguanine, is able to bind in this pocket and react with the cysteine, rendering it an effective inactivator of mammalian alkyltransferases. The alkylated form of the protein is rapidly degraded by the ubiquitin/proteasomal system. Some tumor cells do not express alkyltransferase despite having an intact gene. Methylation of key sites in CpG-rich islands in the promoter region are involved in this silencing and a change in the nuclear localization of an enhancer binding protein may also contribute. The alkyltransferase promoter contains Sp1, GRE and AP-1 sites and is slightly inducible by glucocorticoids and protein kinase C activators. There is a complex relationship between p53 and alkyltransferase expression with p53 mediating a rise in alkyltransferase in response to ionizing radiation but having no clear effect on basal levels. DNA adducts at the O6-position of guanine are a major factor in the carcinogenic, mutagenic, apoptopic and clastogenic actions of methylating agents and chloroethylating agents. Studies with transgenic mice in which alkyltransferase levels are increased or decreased confirm the importance of this repair pathway in protecting against carcinogenesis. Alkyltransferase activity in tumors protects them from therapeutic agents such as temozolomide and BCNU. This resistance is abolished by O6-benzylguanine and this drug is currently in clinical trials to enhance cancer chemotherapy by these agents. Studies are in progress to reduce the toxicity of such therapy towards the bone marrow by gene therapy to express alkyltransferases with mutations imparting resistance to O6-benzylguanine at high levels in marrow stem cells. Several polymorphisms in the human alkyltransferase gene have been identified but the significance of these in terms of alkyltransferase action is currently unknown.  相似文献   

15.
Binding experiments with alkyl-transfer-active and -inactive mutants of human O6-alkylguanine DNA alkyltransferase (AGT) show that it forms an O6-methylguanine (6mG)-specific complex on duplex DNA that is distinct from non-specific assemblies previously studied. Specific complexes with duplex DNA have a 2:1 stoichiometry that is formed without accumulation of a 1:1 intermediate. This establishes a role for cooperative interactions in lesion binding. Similar specific complexes could not be detected with single-stranded DNA. The small difference between specific and non-specific binding affinities strongly limits the roles that specific binding can play in the lesion search process. Alkyl-transfer kinetics with a single-stranded substrate indicate that two or more AGT monomers participate in the rate-limiting step, showing for the first time a functional link between cooperative binding and the repair reaction. Alkyl-transfer kinetics with a duplex substrate suggest that two pathways contribute to the formation of the specific 6mG-complex; one at least first order in AGT, we interpret as direct lesion binding. The second, independent of [AGT], is likely to include AGT transfer from distal sites to the lesion in a relatively slow unimolecular step. We propose that transfer between distal and lesion sites is a critical step in the repair process.  相似文献   

16.
17.
Although it is known that (i) O6-alkylguanine-DNA alkyltransferase (AGT) confers tumor cell resistance to guanine O6-targeting drugs such as cloretazine, carmustine, and temozolomide and that (ii) AGT levels in tumors are highly variable, measurement of AGT activity in tumors before treatment is not a routine clinical practice. This derives in part from the lack of a reliable clinical AGT assay; therefore, a simple AGT assay was devised based on transfer of radioactive benzyl residues from [benzene-3H]O6-benzylguanine ([3H]BG) to AGT. The assay involves incubation of intact cells or cell homogenates with [3H]BG and measurement of radioactivity in a 70% methanol precipitable fraction. Approximately 85% of AGT in intact cells was recovered in cell homogenates. Accuracy of the AGT assay was confirmed by examination of AGT levels by Western blot analysis with the exception of false-positive results in melanin-containing cells due to [3H]BG binding to melanin. Second-order kinetic constants for human and murine AGT were 1100 and 380 M−1 s−1, respectively. AGT levels in various human cell lines ranged from less than 500 molecules/cell (detection limit) to 45,000 molecules/cell. Rodent cell lines frequently lacked AGT expression, and AGT levels in rodent cells were much lower than in human cells.  相似文献   

18.
J C Delaney  J M Essigmann 《Biochemistry》2001,40(49):14968-14975
Understanding the origins of mutational hotspots is complicated by the intertwining of several variables. The selective formation, repair, and replication of a DNA lesion, such as O(6)-methylguanine (m(6)G), can, in principle, be influenced by the surrounding nucleotide environment. A nearest-neighbor analysis was used to address the contribution of sequence context on m(6)G repair by the Escherichia coli methyltransferases Ada or Ogt, and on DNA polymerase infidelity in vivo. Sixteen M13 viral genomes with m(6)G flanked by all permutations of G, A, T, and C were constructed and individually transformed into repair-deficient and repair-proficient isogenic cell strains. The 16 genomes were introduced in duplicate into 5 different cellular backgrounds for a total of 160 independent experiments, for which mutations were scored using a recently developed assay. The Ada methyltransferase demonstrated strong 5' and 3' sequence-specific repair of m(6)G in vivo. The Ada 5' preference decreased in the general order: GXN > CXN > TXN > AXN (X = m(6)G, N = any base), while the Ada 3' preference decreased in the order: NX(T/C) > NX(G/A), with mutation frequencies (MFs) ranging from 35% to 90%. The Ogt methyltransferase provided MFs ranging from 10% to 25%. As was demonstrated by Ada, the Ogt methyltransferase repaired m(6)G poorly in an AXN context. When both methyltransferases were removed, the MF was nearly 100% for all sequence contexts, consistent with the view that the replicative DNA polymerase places T opposite m(6)G during replication irrespective of the local sequence environment.  相似文献   

19.
Two new agents based upon the structure of the clinically active prodrug laromustine were synthesized. These agents, 2-(2-chloroethyl)-N-methyl-1,2-bis(methylsulfonyl)-N-nitrosohydrazinecarboxamide (1) and N-(2-chloroethyl)-2-methyl-1,2-bis(methylsulfonyl)-N-nitrosohydrazinecarboxamide (2), were designed to retain the potent chloroethylating and DNA cross-linking functions of laromustine, and gain the ability to methylate DNA at the O-6 position of guanine, while lacking the carbamoylating activity of laromustine. The methylating arm was introduced with the intent of depleting the DNA repair protein O6-alkylguanine-DNA alkyltransferase (AGT). Compound 1 is markedly more cytotoxic than laromustine in both AGT minus EMT6 mouse mammary carcinoma cells and high AGT expressing DU145 human prostate carcinoma cells. DNA cross-linking studies indicated that its cross-linking efficiency is nearly identical to its predicted active decomposition product, 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine (90CE), which is also produced by laromustine. AGT ablation studies in DU145 cells demonstrated that 1 can efficiently deplete AGT. Studies assaying methanol and 2-chloroethanol production as a consequence of the methylation and chloroethylation of water by 1 and 2 confirmed their ability to function as methylating and chloroethylating agents and provided insights into the superior activity of 1.  相似文献   

20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号