Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Biopolymers ; 112(1): e23405, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33098572

ABSTRACT

Cellular exposure to tobacco-specific nitrosamines causes formation of promutagenic O6 -[4-oxo-4-(3-pyridyl)but-1-yl]guanine (O6 -POB-G) and O6 -methylguanine (O6 -Me-G) adducts in DNA. These adducts can be directly repaired by O6 -alkylguanine-DNA alkyltransferase (AGT). Repair begins by flipping the damaged base out of the DNA helix. AGT binding and base-flipping have been previously studied using pyrrolocytosine as a fluorescent probe paired to the O6 -alkylguanine lesion, but low fluorescence yield limited the resolution of steps in the repair process. Here, we utilize the highly fluorescent 6-phenylpyrrolo-2'-deoxycytidine (6-phenylpyrrolo-C) to investigate AGT-DNA interactions. Synthetic oligodeoxynucleotide duplexes containing O6 -POB-G and O6 -Me-G adducts were placed within the CpG sites of codons 158, 245, and 248 of the p53 tumor suppressor gene and base-paired to 6-phenylpyrrolo-C in the opposite strand. Neighboring cytosine was either unmethylated or methylated. Stopped-flow fluorescence measurements were performed by mixing the DNA duplexes with C145A or R128G AGT variants. We observe a rapid, two-step, nearly irreversible binding of AGT to DNA followed by two slower steps, one of which is base-flipping. Placing 5-methylcytosine immediately 5' to the alkylated guanosine causes a reduction in rate constant of nucleotide flipping. O6 -POB-G at codon 158 decreased the base flipping rate constant by 3.5-fold compared with O6 -Me-G at the same position. A similar effect was not observed at other codons.


Subject(s)
Cytosine/chemistry , DNA Repair , Fluorescent Dyes/chemistry , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Alkylation , Base Pairing , Biocatalysis , CpG Islands/genetics , Cytidine/analogs & derivatives , Cytidine/chemistry , DNA Adducts/chemistry , DNA Adducts/metabolism , Kinetics , Mutagenesis, Site-Directed , Pyrroles/chemistry , Tumor Suppressor Protein p53/genetics
2.
Biochemistry ; 52(23): 4075-88, 2013 Jun 11.
Article in English | MEDLINE | ID: mdl-23683164

ABSTRACT

Tobacco-specific nitrosamines 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) and N-nitrosonicotine (NNN) are potent carcinogens believed to contribute to the development of lung tumors in smokers. NNK and NNN are metabolized to DNA-reactive species that form a range of nucleobase adducts, including bulky O(6)-[4-oxo-4-(3-pyridyl)but-1-yl]deoxyguanosine (O(6)-POB-dG) lesions. If not repaired, O(6)-POB-dG adducts induce large numbers of G → A and G → T mutations. Previous studies have shown that O(6)-POB-dG can be directly repaired by O(6)-alkylguanine-DNA alkyltransferase (AGT), which transfers the pyridyloxobutyl group from O(6)-alkylguanines in DNA to an active site cysteine residue within the protein. In the present study, we investigated the influence of DNA sequence context and endogenous cytosine methylation on the kinetics of AGT-dependent repair of O(6)-POB-dG in duplex DNA. Synthetic oligodeoxynucleotide duplexes containing site-specific O(6)-POB-dG adducts within K-ras and p53 gene-derived DNA sequences were incubated with recombinant human AGT protein, and the kinetics of POB group transfer was monitored by isotope dilution HPLC-ESI(+)-MS/MS analysis of O(6)-POB-dG remaining in DNA over time. We found that the second-order rates of AGT-mediated repair were influenced by DNA sequence context (10-fold differences) but were only weakly affected by the methylation status of neighboring cytosines. Overall, AGT-mediated repair of O(6)-POB-dG was 2-7 times slower than that of O(6)-Me-dG adducts. To evaluate the contribution of AGT to O(6)-POB-dG repair in human lung, normal human bronchial epithelial cells (HBEC) were treated with model pyridyloxobutylating agent, and O(6)-POB-dG adduct repair over time was monitored by HPLC-ESI(+)-MS/MS. We found that HBEC cells were capable of removing O(6)-POB-dG lesions, and the repair rates were significantly reduced in the presence of an AGT inhibitor (O(6)-benzylguanine). Taken together, our results suggest that AGT plays an important role in protecting human lung against tobacco nitrosamine-mediated DNA damage and that inefficient AGT repair of O(6)-POB-dG at a specific sequences contributes to mutational spectra observed in smoking-induced lung cancer.


Subject(s)
DNA Adducts/chemistry , Deoxyguanosine/analogs & derivatives , O(6)-Methylguanine-DNA Methyltransferase/chemistry , Pyridines/chemistry , Base Sequence , Bronchi/cytology , Carcinogens/chemistry , Carcinogens/pharmacology , Cells, Cultured , DNA Adducts/metabolism , DNA Methylation , DNA Repair , Deoxyguanosine/chemistry , Deoxyguanosine/metabolism , Electrophoretic Mobility Shift Assay , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Guanine/analogs & derivatives , Humans , Kinetics , Nitrosamines/chemistry , Nitrosamines/pharmacology , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Polynucleotides/chemistry , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Pyridines/metabolism , Respiratory Mucosa/enzymology , Transition Temperature , ras Proteins/genetics
3.
Biochemistry ; 52(18): 3171-81, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23566219

ABSTRACT

Although cytotoxic alkylating agents possessing two electrophilic reactive groups are thought to act by cross-linking cellular biomolecules, their exact mechanisms of action have not been established. In cells, these compounds form a mixture of DNA lesions, including nucleobase monoadducts, interstrand and intrastrand cross-links, and DNA-protein cross-links (DPCs). Interstrand DNA-DNA cross-links block replication and transcription by preventing DNA strand separation, contributing to toxicity and mutagenesis. In contrast, potential contributions of drug-induced DPCs are poorly understood. To gain insight into the biological consequences of DPC formation, we generated DNA-reactive protein reagents and examined their toxicity and mutagenesis in mammalian cells. Recombinant human O(6)-alkylguanine DNA alkyltransferase (AGT) protein or its variants (C145A and K125L) were treated with 1,2,3,4-diepoxybutane to yield proteins containing 2-hydroxy-3,4-epoxybutyl groups on cysteine residues. Gel shift and mass spectrometry experiments confirmed that epoxide-functionalized AGT proteins formed covalent DPC but no other types of nucleobase damage when incubated with duplex DNA. Introduction of purified AGT monoepoxides into mammalian cells via electroporation generated AGT-DNA cross-links and induced cell death and mutations at the hypoxanthine-guanine phosphoribosyltransferase gene. Smaller numbers of DPC lesions and reduced levels of cell death were observed when using protein monoepoxides generated from an AGT variant that fails to accumulate in the cell nucleus (K125L), suggesting that nuclear DNA damage is required for toxicity. Taken together, these results indicate that AGT protein monoepoxides produce cytotoxic and mutagenic DPC lesions within chromosomal DNA. More generally, these data suggest that covalent DPC lesions contribute to the cytotoxic and mutagenic effects of bis-electrophiles.


Subject(s)
Cell Death , DNA-Binding Proteins/metabolism , DNA/metabolism , Epoxy Compounds/pharmacology , Mutagenesis , Alkylation , Amino Acid Sequence , Cell Line, Tumor , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Humans , Molecular Sequence Data , Recombinant Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization
4.
Chem Res Toxicol ; 24(11): 1966-75, 2011 Nov 21.
Article in English | MEDLINE | ID: mdl-21913712

ABSTRACT

O(6)-POB-dG (O(6)-[4-oxo-4-(3-pyridyl)but-1-yl]deoxyguanosine) are promutagenic nucleobase adducts that arise from DNA alkylation by metabolically activated tobacco-specific nitrosamines such as 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) and N-nitrosonicotine (NNN). If not repaired, O(6)-POB-dG adducts cause mispairing during DNA replication, leading to G → A and G → T mutations. A specialized DNA repair protein, O(6)-alkylguanine-DNA-alkyltransferase (AGT), transfers the POB group from O(6)-POB-dG in DNA to a cysteine residue within the protein (Cys145), thus restoring normal guanine and preventing mutagenesis. The rates of AGT-mediated repair of O(6)-POB-dG may be affected by local DNA sequence context, potentially leading to adduct accumulation and increased mutagenesis at specific sites within the genome. In the present work, isotope dilution high performance liquid chromatography-electrospray ionization-tandem mass spectrometry (HPLC-ESI(+)-MS/MS)-based methodology was developed to investigate the influence of DNA sequence on the kinetics of AGT-mediated repair of O(6)-POB-dG adducts. In our approach, synthetic DNA duplexes containing O(6)-POB-dG at a specified site are incubated with recombinant human AGT protein for defined periods of time. Following spiking with D(4)-O(6)-POB-dG internal standard and mild acid hydrolysis to release O(6)-POB-guanine (O(6)-POB-G) and D(4)-O(6)-POB-guanine (D(4)-O(6)-POB-G), samples are purified by solid phase extraction (SPE), and O(6)-POB-G adducts remaining in DNA are quantified by capillary HPLC-ESI(+)-MS/MS. The new method was validated by analyzing mixtures containing known amounts of O(6)-POB-G-containig DNA and the corresponding unmodified DNA duplexes and by examining the kinetics of alkyl transfer in the presence of increasing amounts of AGT protein. The disappearance of O(6)-POB-dG from DNA was accompanied by pyridyloxobutylation of AGT Cys-145 as determined by HPLC-ESI(+)-MS/MS of tryptic peptides. The applicability of the new approach was shown by determining the second order kinetics of AGT-mediated repair of O(6)-POB-dG adducts placed within a DNA duplex representing modified rat H-ras sequence (5'-AATAGTATCT[O(6)-POB-G]GAGCC-3') opposite either C or T. Faster rates of alkyl transfer were observed when O(6)-POB-dG was paired with T rather than with C (k = 1.74 × 10(6) M(-1) s(-1) vs 1.17 × 10(6) M(-1) s(-1)).


Subject(s)
DNA Adducts/metabolism , DNA Repair , Deoxyguanosine/metabolism , Genes, ras , Nicotiana/metabolism , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Recombinant Proteins/metabolism , Tandem Mass Spectrometry/methods , Animals , Carcinogens/chemistry , Carcinogens/metabolism , DNA/chemistry , DNA/metabolism , DNA Adducts/chemistry , DNA Damage , Deoxyguanosine/chemistry , Humans , Kinetics , Nitrosamines/chemistry , Nitrosamines/metabolism , Peptide Fragments/analysis , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Radioisotope Dilution Technique , Rats , Spectrometry, Mass, Electrospray Ionization , Nicotiana/chemistry
5.
Mol Microbiol ; 81(4): 1109-24, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21762220

ABSTRACT

We have identified gene fusions of polyamine biosynthetic enzymes S-adenosylmethionine decarboxylase (AdoMetDC, speD) and aminopropyltransferase (speE) orthologues in diverse bacterial phyla. Both domains are functionally active and we demonstrate the novel de novo synthesis of the triamine spermidine from the diamine putrescine by fusion enzymes from ß-proteobacterium Delftia acidovorans and δ-proteobacterium Syntrophus aciditrophicus, in a ΔspeDE gene deletion strain of Salmonella enterica sv. Typhimurium. Fusion proteins from marine α-proteobacterium Candidatus Pelagibacter ubique, actinobacterium Nocardia farcinica, chlorobi species Chloroherpeton thalassium, and ß-proteobacterium D. acidovorans each produce a different profile of non-native polyamines including sym-norspermidine when expressed in Escherichia coli. The different aminopropyltransferase activities together with phylogenetic analysis confirm independent evolutionary origins for some fusions. Comparative genomic analysis strongly indicates that gene fusions arose by merger of adjacent open reading frames. Independent fusion events, and horizontal and vertical gene transfer contributed to the scattered phyletic distribution of the gene fusions. Surprisingly, expression of fusion genes in E. coli and S. Typhimurium revealed novel latent spermidine catabolic activity producing non-native 1,3-diaminopropane in these species. We have also identified fusions of polyamine biosynthetic enzymes agmatine deiminase and N-carbamoylputrescine amidohydrolase in archaea, and of S-adenosylmethionine decarboxylase and ornithine decarboxylase in the single-celled green alga Micromonas.


Subject(s)
Adenosylmethionine Decarboxylase/genetics , Biosynthetic Pathways/genetics , Evolution, Molecular , Gene Fusion , Putrescine/metabolism , Spermidine Synthase/genetics , Spermidine/metabolism , Adenosylmethionine Decarboxylase/metabolism , Archaea/genetics , Archaea/metabolism , Bacteria/genetics , Bacteria/metabolism , Spermidine Synthase/metabolism
6.
J Biol Chem ; 285(18): 13736-41, 2010 Apr 30.
Article in English | MEDLINE | ID: mdl-20212037

ABSTRACT

Alkyltransferase-like proteins (ATLs) are a novel class of DNA repair proteins related to O(6)-alkylguanine-DNA alkyltransferases (AGTs) that tightly bind alkylated DNA and shunt the damaged DNA into the nucleotide excision repair pathway. Here, we present the first structure of a bacterial ATL, from Vibrio parahaemolyticus (vpAtl). We demonstrate that vpAtl adopts an AGT-like fold and that the protein is capable of tightly binding to O(6)-methylguanine-containing DNA and disrupting its repair by human AGT, a hallmark of ATLs. Mutation of highly conserved residues Tyr(23) and Arg(37) demonstrate their critical roles in a conserved mechanism of ATL binding to alkylated DNA. NMR relaxation data reveal a role for conformational plasticity in the guanine-lesion recognition cavity. Our results provide further evidence for the conserved role of ATLs in this primordial mechanism of DNA repair.


Subject(s)
Alkyl and Aryl Transferases/chemistry , DNA Repair/physiology , DNA/chemistry , Guanine/analogs & derivatives , Protein Folding , Vibrio parahaemolyticus/enzymology , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , Amino Acid Substitution , DNA/genetics , DNA/metabolism , Guanine/chemistry , Guanine/metabolism , Humans , Mutation, Missense , Vibrio parahaemolyticus/genetics
7.
J Biol Chem ; 285(11): 8185-95, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20026607

ABSTRACT

O(6)-Alkylguanine-DNA alkyltransferase (AGT) plays a major role in repair of the cytotoxic and mutagenic lesion O(6)-methylguanine (m(6)G) in DNA. Unlike the Escherichia coli alkyltransferase Ogt that also repairs O(4)-methylthymine (m(4)T) efficiently, the human AGT (hAGT) acts poorly on m(4)T. Here we made several hAGT mutants in which residues near the cysteine acceptor site were replaced by corresponding residues from Ogt to investigate the basis for the inefficiency of hAGT in repair of m(4)T. Construct hAGT-03 (where hAGT sequence -V(149)CSSGAVGN(157)- was replaced with the corresponding Ogt -I(143)GRNGTMTG(151)-) exhibited enhanced m(4)T repair activity in vitro compared with hAGT. Three AGT proteins (hAGT, hAGT-03, and Ogt) exhibited similar protection from killing by N-methyl-N'-nitro-N-nitrosoguanidine and caused a reduction in m(6)G-induced G:C to A:T mutations in both nucleotide excision repair (NER)-proficient and -deficient Escherichia coli strains that lack endogenous AGTs. hAGT-03 resembled Ogt in totally reducing the m(4)T-induced T:A to C:G mutations in NER-proficient and -deficient strains. Surprisingly, wild type hAGT expression caused a significant but incomplete decrease in NER-deficient strains but a slight increase in T:A to C:G mutation frequency in NER-proficient strains. The T:A to C:G mutations due to O(4)-alkylthymine formed by ethylating and propylating agents were also efficiently reduced by either hAGT-03 or Ogt, whereas hAGT had little effect irrespective of NER status. These results show that specific alterations in the hAGT active site facilitate efficient recognition and repair of O(4)-alkylthymines and reveal damage-dependent interactions of base and nucleotide excision repair.


Subject(s)
DNA Adducts/metabolism , DNA Repair/physiology , O(6)-Methylguanine-DNA Methyltransferase/genetics , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Thymine/metabolism , Alkylation/physiology , Catalytic Domain , DNA Damage/physiology , Escherichia coli/genetics , Ethane/metabolism , Humans , Methane/metabolism , Mutagenesis/physiology , O(6)-Methylguanine-DNA Methyltransferase/chemistry , Propane/metabolism , Protein Structure, Tertiary
8.
Nature ; 459(7248): 808-13, 2009 Jun 11.
Article in English | MEDLINE | ID: mdl-19516334

ABSTRACT

Alkyltransferase-like proteins (ATLs) share functional motifs with the cancer chemotherapy target O(6)-alkylguanine-DNA alkyltransferase (AGT) and paradoxically protect cells from the biological effects of DNA alkylation damage, despite lacking the reactive cysteine and alkyltransferase activity of AGT. Here we determine Schizosaccharomyces pombe ATL structures without and with damaged DNA containing the endogenous lesion O(6)-methylguanine or cigarette-smoke-derived O(6)-4-(3-pyridyl)-4-oxobutylguanine. These results reveal non-enzymatic DNA nucleotide flipping plus increased DNA distortion and binding pocket size compared to AGT. Our analysis of lesion-binding site conservation identifies new ATLs in sea anemone and ancestral archaea, indicating that ATL interactions are ancestral to present-day repair pathways in all domains of life. Genetic connections to mammalian XPG (also known as ERCC5) and ERCC1 in S. pombe homologues Rad13 and Swi10 and biochemical interactions with Escherichia coli UvrA and UvrC combined with structural results reveal that ATLs sculpt alkylated DNA to create a genetic and structural intersection of base damage processing with nucleotide excision repair.


Subject(s)
Alkyl and Aryl Transferases/chemistry , Alkyl and Aryl Transferases/metabolism , DNA Damage , DNA Repair , Alkylation , Binding Sites , Crystallography, X-Ray , DNA/chemistry , DNA/metabolism , Guanine/analogs & derivatives , Guanine/chemistry , Guanine/metabolism , Humans , Models, Molecular , Protein Binding , Protein Conformation
9.
Cancer Res ; 68(23): 9973-81, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19047179

ABSTRACT

Germ-line mutations in BRCA2 have been linked to early-onset familial breast cancer. BRCA2 is known to play a key role in repairing double-strand breaks. Here, we describe the involvement of BRCA2 in O6-alkylguanine DNA alkyltransferase (AGT)-mediated repair of O6-methylguanine adducts. We show that BRCA2 physically associates and undergoes repair-mediated degradation with AGT. In contrast, BRCA2 with a 29-amino-acid deletion in an evolutionarily conserved domain does not bind to alkylated AGT; the two proteins are not degraded; and mouse embryonic fibroblasts are specifically sensitive to alkylating agents that result in O6-methylguanine adducts. We show that O6-benzylguanine (O6BG), a nontoxic inhibitor of AGT, can also induce BRCA2 degradation. BRCA2 is a viable target for cancer therapy because BRCA2-deficient cells are hypersensitive to chemotherapeutic DNA-damaging agents. We show a marked effect of O6BG pretreatment on cell sensitivity to cisplatin. We also show the efficacy of this approach on a wide range of human tumor cell lines, which suggests that chemosensitization of tumors by targeted degradation of BRCA2 may be an important consideration when devising cancer therapeutics.


Subject(s)
BRCA2 Protein/metabolism , DNA Repair/physiology , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Alkylating Agents , Amino Acid Sequence , Animals , BRCA2 Protein/genetics , Gene Deletion , Guanine/analogs & derivatives , Guanine/metabolism , Guanine/pharmacology , Humans , Methylnitronitrosoguanidine , Mice , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors
10.
Cancer Res ; 66(9): 4968-74, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16651455

ABSTRACT

The repair protein O(6)-alkylguanine-DNA alkyltransferase (AGT) protects cells from the mutagenic and carcinogenic effects of alkylating agents by removing O(6)-alkylguanine adducts from DNA. Recently, we established that AGT protects against the mutagenic effects of pyridyloxobutylation resulting from the metabolic activation of the tobacco-specific nitrosamines (TSNA) 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone and N-nitrosonornicotine by repairing O(6)-[4-oxo-4-(3-pyridyl)butyl]guanine (O(6)-pobG). There have been several epidemiologic studies examining the association between the I143V/K178R AGT genotype and lung cancer risk. Two studies have found positive associations, suggesting that AGT proteins differ in their repair of DNA damage caused by TSNA. However, it is not known how this genotype alters the biochemical activity of AGT. We proposed that AGT proteins may differ in their ability to remove large O(6)-alkylguanine adducts, such as O(6)-pobG, from DNA. Therefore, we examined the repair of O(6)-pobG by wild-type (WT) human, I143V/K178R, and L84F AGT proteins when contained in multiple sequence contexts, including the twelfth codon of H-ras, a mutational hotspot within this oncogene. The AGT-mediated repair of O(6)-pobG was more profoundly influenced by sequence context than that of O(6)-methylguanine. These differences are not the result of secondary structure (hairpin) formation in DNA. In addition, the I143V/K178R variant seems less sensitive to the effects of sequence context than the WT or L84F proteins. These studies indicate that the sequence dependence of O(6)-pobG repair by human AGT (hAGT) varies with subtle changes in protein structure. These data establish a novel functional difference between the I143V/K178R protein and other hAGTs in the repair of a toxicologically relevant substrate, O(6)-pobG.


Subject(s)
DNA Adducts/metabolism , DNA Repair/physiology , Guanine/analogs & derivatives , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Animals , Base Sequence , DNA Adducts/genetics , DNA Repair/genetics , Genes, ras , Guanine/metabolism , Humans , Mice , Oligonucleotides/genetics , Rats
11.
Biochemistry ; 44(46): 15396-405, 2005 Nov 22.
Article in English | MEDLINE | ID: mdl-16285744

ABSTRACT

O6-Alkylguanine-DNA alkyltransferase (AGT) is an important DNA repair protein that protects from alkylating agents by converting O6-alkylguanine to guanine forming S-methylcysteine in the AGT protein. The crystal structure of human AGT shows clearly the presence of two domains. The N-terminal domain contains a bound zinc atom, and zinc binding confers a mechanistic enhancement to repair activity, but this domain has no known function. The C-terminal domain contains all residues so far implicated in alkyl transfer including the cysteine acceptor site (Cys145), the O6-alkylguanine binding pocket, and a DNA binding domain. We have expressed and purified the two domains of human AGT separately. The C-terminal domain was totally inactive in vitro, but good activity forming S-alkylcysteine at Cys145 was obtained after recombination with the N-terminal domain via a freeze-thawing procedure. This suggests that the N-terminal domain plays a critical structural role in maintaining an active configuration of the C-terminal domain. However, this C-terminal domain alone had activity in protecting against the cytotoxic and mutagenic activity of the methylating agent, N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) when expressed in Escherichia coli cells lacking endogenous AGT, suggesting that other proteins can fulfill this function. Remarkably, the free N-terminal domain of hAGT was able to repair O6-alkylguanine in vitro via alkyl transfer provided that zinc ions were present. The N-terminal domain was also able to produce moderate protection from MNNG when expressed in E. coli. This cryptic Zn2+-dependent DNA repair activity may be relevant to the evolution and function of AGTs.


Subject(s)
O(6)-Methylguanine-DNA Methyltransferase/chemistry , Protein Structure, Tertiary , DNA Repair , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Escherichia coli/drug effects , Escherichia coli/genetics , Humans , Methylnitronitrosoguanidine/pharmacology , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Protein Folding , Protein Renaturation , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
12.
Biochemistry ; 44(6): 2143-52, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15697240

ABSTRACT

The value of reducing the activity of ornithine decarboxylase (ODC), a key enzyme in the biosynthesis of polyamines, is well-appreciated. Polyamines are necessary components for cell growth, and manipulation of polyamine homeostasis may be an effective strategy for the treatment of a number of disorders, including neoplastic diseases. An approach to develop an effective DNAzyme, using the 10-23 model, against ODC is described in these studies. DNAzymes able to cleave the target ODC RNA were identified in vitro and further characterized by the effect each had on ODC protein and activity levels using in vitro translated ODC RNA. ODC protein levels and activity correlated well with the RNA cleavage activity of the DNAzyme. One of the DNAzymes, DZ IV, which exhibited good activity, was optimized for use in cell culture studies. The DNAzyme hybridization arms were altered from equal length arms varying in length (8, 9, 10, or 11 nucleotides) or to unequal length arms (7/11 nucleotides), and kinetic analyses were performed to identify the most catalytically efficient configuration. DZ IV with equal arms nine nucleotides in length proved to be the most catalytically efficient. In HEK 293 cells, DZ IV was able to reduce the amount of translated ODC protein, resulting in approximately 80% reduction in ODC activity-a statistically significant enhancement over the apparent antisense effect of a catalytically inactive DNAzyme. These results indicate that this DNAzyme may be a useful tool to study the function of ODC and may have potential therapeutic uses.


Subject(s)
DNA, Catalytic/chemistry , Gene Silencing , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase/metabolism , Animals , Catalytic Domain/genetics , Cell Line , DNA, Catalytic/chemical synthesis , Enzyme Activation/genetics , Humans , Hydrolysis , Kinetics , Mice , Oligodeoxyribonucleotides/chemical synthesis , Oligodeoxyribonucleotides/chemistry , Ornithine Decarboxylase Inhibitors , Oxidation-Reduction , Protein Biosynthesis/genetics , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/chemical synthesis , RNA, Messenger/metabolism , Transcription, Genetic/genetics , Transfection
13.
Proc Natl Acad Sci U S A ; 102(10): 3617-22, 2005 Mar 08.
Article in English | MEDLINE | ID: mdl-15731349

ABSTRACT

A recently discovered DNA repair protein of 303 aa from the archaeal organism Ferroplasma acidarmanus was studied. This protein (AGTendoV) consists of a fusion of the C-terminal active site domain of O(6)-alkylguanine-DNA alkyltransferase (AGT) with an endonuclease V domain. The AGTendoV recombinant protein expressed in Escherichia coli and purified to homogeneity repaired O(6)-methylguanine lesions in DNA via alkyl transfer action despite the complete absence of the N-terminal domain and some differences in key active site residues present in known AGTs. The AGTendoV recombinant protein also cleaved DNA substrates that contained the deaminated bases uracil, hypoxanthine, or xanthine in a similar manner to E. coli endonuclease V. Expression of AGTendoV in E. coli GWR109, a strain that lacks endogenous AGT activity, protected against both the killing and mutagenic activity of N-methyl-N'-nitro-N-nitrosoguanidine and was more effective in preventing mutations than human alkyltransferase, suggesting that the endonuclease V activity may also repair a promutagenic lesion produced by this alkylating agent. Expression of AGTendoV in a DNA repair-deficient E. coli nfi(-)alkA(-) strain protected from spontaneous mutations arising in saturated cultures and restored the mutation frequency to that found in the nfi(+) alkA(+) strain. These results demonstrate the physiological occurrence of two completely different but functional DNA repair activities in a single polypeptide chain.


Subject(s)
Archaeal Proteins/metabolism , DNA Repair , Deoxyribonuclease (Pyrimidine Dimer)/metabolism , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Thermoplasmales/chemistry , Amino Acid Sequence , Archaeal Proteins/chemistry , Molecular Sequence Data
14.
Chem Res Toxicol ; 17(3): 424-34, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15025514

ABSTRACT

The tobacco specific nitrosamine, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), a potent pulmonary carcinogen, both methylates and pyridyloxobutylates DNA. Both reaction pathways generate promutagenic O6-alkylguanine adducts. These adducts, O6-methylguanine (O6-mG) and O6-[4-oxo-4-(3-pyridyl)butyl]guanine (O6-pobG), are repaired by O6-alkylguanine-DNA alkyltransferase (AGT). In this report, we demonstrate that pyridyloxobutyl DNA adducts are repaired by AGT in a reaction that results in pyridyloxobutyl transfer to the active site cysteine. Because minor changes within the binding pocket of AGT can alter the ability of this protein to repair bulky O6-alkylguanine adducts relative to O6-mG, we explored the ability of AGTs from different species as well as several human AGT variants and mutants to discriminate between O6-mG or O6-pobG adducts. We incubated proteins with equal molar amounts of oligodeoxynucleotides containing site specifically incorporated O6-mG or O6-pobG and measured repair. Bacterial AGTs poorly repaired O6-pobG. Mouse and rat AGT repaired both adducts at comparable rates. Wild-type human AGT, variant I143V/K178R, and mutant N157H repaired O6-mG approximately twice as fast as O6-pobG. Human variant G160R and mutants P140K, Y158H, G156A, and E166G did not repair O6-pobG until all of the O6-mG was removed. To understand the role of adduct structure on relative repair rates, the competition experiments were repeated with two other bulky O6-alkylguanine adducts, O6-butylguanine (O6-buG) and O6-benzylguanine (O6-bzG). The proteins displayed similar repair preference of O6-mG relative to O6-buG as observed with O6-pobG. In contrast, all of the mammalian proteins, except the mutant P140K, preferentially repaired O6-bzG. These studies indicate that the rate of repair of O6-pobG is highly dependent on protein structure. Inefficient repair of O6-pobG by bacterial AGT explains the high mutagenic activity of this adduct in bacterial systems. In addition, differences observed in the repair of this adduct by mammalian proteins may translate into differences in sensitivity to the mutagenic and carcinogenic effects of NNK or other pyridyloxobutylating nitrosamines.


Subject(s)
Carcinogens/chemistry , DNA Adducts/chemistry , Guanine/analogs & derivatives , Guanine/chemistry , Nitrosamines/chemistry , O(6)-Methylguanine-DNA Methyltransferase/chemistry , Animals , Cattle , DNA Repair , Humans , Kinetics , Mice , O(6)-Methylguanine-DNA Methyltransferase/classification , Rats , Species Specificity
15.
J Biol Chem ; 279(6): 4250-9, 2004 Feb 06.
Article in English | MEDLINE | ID: mdl-14645247

ABSTRACT

It has been proposed that the DNA repair protein O6-alkylguanine-DNA alkyltransferase increases the mutagenicity of 1,2-dibromoethane by reacting with it at its cysteine acceptor site to form a highly reactive half-mustard, which can then react with DNA (Liu, L., Pegg, A. E., Williams, K. M., and Guengerich, F. P. (2002) J. Biol. Chem. 277, 37920-37928). Incubation of Escherichia coli-expressed human alkyltransferase with 1,2-dibromoethane and single-stranded oligodeoxyribonucleotides led to the formation of covalent transferaseoligo complexes. The order of reaction determined was Gua>Thy>Cyt>Ade. Mass spectrometry analysis of the tryptic digest of the reaction product indicated that some of the adducts led to depurination with the release of the Gly136-Arg147 peptide cross-linked to a Gua at the N7 position, with the site of reaction being the active site Cys145 as established by chromatographic retention time and the fragmentation pattern determined by tandem mass spectrometry of a synthetic peptide adduct. The alkyltransferase-mediated mutations produced by 1,2-dibromoethane were predominantly Gua to Ade transitions but, in the spectrum of such rifampicin-resistant mutations in the RpoB gene, 20% were Gua to Thy transversions. The latter are likely to have arisen from the apurinic site generated from the Gua-N7 adduct. Support exists for an additional adduct/mutagenic pathway because evidence was obtained for DNA adducts other than at the Gua N7 atom and for mutations other than those attributable to depurination. Thus, chemical and biological evidence supports the existence of at least two alkyltransferase-dependent pathways for 1,2-dibromoethane-induced mutagenicity, one involving Gua N7-alkylation by alkyltransferase-S-CH2CH2Br and depurination, plus another as yet uncharacterized system(s).


Subject(s)
DNA Adducts/metabolism , Ethylene Dibromide/metabolism , Mutagens/metabolism , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Animals , Cattle , DNA Adducts/chemistry , DNA Adducts/toxicity , Ethylene Dibromide/chemistry , Ethylene Dibromide/toxicity , Humans , In Vitro Techniques , Molecular Structure , Mutagens/chemistry , Mutagens/toxicity , O(6)-Methylguanine-DNA Methyltransferase/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
16.
Biochem J ; 375(Pt 2): 449-55, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-12892560

ABSTRACT

AGT (O6-alkylguanine DNA alkyltransferase) is an important DNA-repair protein that protects cells from killing and mutagenesis by alkylating agents. The AGT genes from two extremely thermophilic organisms, the bacterium Aquifex aeolicus and the archaeon Archaeoglobus fulgidus were PCR-derived and cloned into an expression vector. The nucleotide sequence of the Aq. aeolicus AGT encodes a 201-amino-acid protein with a molecular mass of 23000 Da and Ar. fulgidus AGT codes for a 147-amino-acid protein with a molecular mass of 16718 Da. The Aq. aeolicus and Ar. fulgidus AGTs were expressed at high levels in Escherichia coli fused to an N-terminal polyhistidine tag that allowed single-step isolation and purification by metal-affinity chromatography. Both AGTs formed inclusion bodies and were not soluble under native purification conditions. Therefore AGT isolation was performed under protein-denaturation conditions in the presence of 8.0 M urea. Soluble AGT was obtained by refolding the AGT in the presence of calf thymus DNA. Both AGTs were active in repairing O6-methylguanine and, at a lower rate, O4-methylthymine in DNA. They exhibited thermostability and optimum activity at high temperature. The thermostable AGTs, particularly that from Aq. aeolicus, were readily inactivated by the low-molecular-mass inhibitor O6-benzylguanine, which is currently in clinical trials to enhance cancer chemotherapy.


Subject(s)
Archaeoglobus fulgidus/enzymology , Bacteria/enzymology , Guanine/analogs & derivatives , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Alkylating Agents/pharmacology , Archaeoglobus fulgidus/genetics , Bacteria/genetics , DNA/drug effects , DNA/genetics , DNA/metabolism , DNA Damage , DNA Repair , Enzyme Stability/drug effects , Guanine/metabolism , Guanine/pharmacology , Hydrogen-Ion Concentration , O(6)-Methylguanine-DNA Methyltransferase/chemistry , O(6)-Methylguanine-DNA Methyltransferase/genetics , Protein Folding , Protein Renaturation , Temperature
17.
Biochemistry ; 42(4): 980-90, 2003 Feb 04.
Article in English | MEDLINE | ID: mdl-12549918

ABSTRACT

A recent crystallographic study of recombinant human O(6)-alkylguanine-DNA alkyltransferase (hAGT) revealed a previously unknown zinc atom [Daniels et al., (2000) EMBO J. 19, 1719-1730]. The effects of zinc on the properties of hAGT are reported here. In bacterial expression systems, recombinant hAGT was produced in increasingly larger quantities when growth media are supplemented with up to 0.1 mM ZnCl(2). Metal-enriched hAGT samples had a 5-fold increase in repair rate constant over conventionally purified protein samples and a 60-fold increase over metal-stripped hAGT. In addition, mutants of the zinc-binding residues had decreases in zinc occupancy that correlated with reductions in repair rate. Zinc modulation did not abolish the repair capacity of a fraction of the hAGT population, as evidenced by the stoichiometric reaction with an oligodeoxyribonucleotide substrate. Zinc occupancy had a similar effect on the rate of reaction with O(6)-benzylguanine, a free base substrate, as on the repair of methylated DNA. Differentially zinc-treated hAGTs showed the same affinity for binding to native DNA and substrate oligodeoxyribonucleotides. Metal content manipulations had little effect upon the CD spectrum of hAGT, but fluorescence studies revealed a small conformational change based upon metal binding, and zinc occupancy correlated with enhanced hAGT stability as evidenced by resistance to the denaturing effects of urea. These results indicate that the presence of zinc confers a mechanistic enhancement to repair activity that does not result from an increase in substrate binding affinity. Zinc also provides conformational stability to hAGT that may influence its regulation.


Subject(s)
Guanine/analogs & derivatives , O(6)-Methylguanine-DNA Methyltransferase/chemistry , Zinc/chemistry , Binding Sites , Chlorides/chemistry , DNA Repair , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/chemistry , Enzyme Inhibitors/chemistry , Enzyme Stability , Guanine/chemistry , Humans , Mass Spectrometry/methods , Metalloproteins/antagonists & inhibitors , Metalloproteins/biosynthesis , Metalloproteins/chemistry , Mutagenesis, Site-Directed , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , Plasmids , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Spectrometry, Fluorescence , Structure-Activity Relationship , Substrate Specificity , Zinc Compounds/chemistry
18.
Biochemistry ; 41(27): 8689-97, 2002 Jul 09.
Article in English | MEDLINE | ID: mdl-12093287

ABSTRACT

Activity of the DNA repair protein O(6)-alkylguanine-DNA alkyltransferase (AGT) is an important source of tumor cell resistance to alkylating agents. AGT inhibitors may prove useful in enhancing chemotherapy. AGT is inactivated by reacting stoichiometrically with O(6)-benzylguanine (b(6)G), which is currently in clinical trials for this purpose. Short oligodeoxyribonucleotides containing a central b(6)G are more potent inactivators of AGT than b(6)G. We examined whether human AGT could react with oligodeoxyribonucleotides containing multiple b(6)G residues. The single-stranded 7-mer 5'-d[T(b(6)G)(5)G]-3' was an excellent AGT substrate with all five b(6)G adducts repaired although one adduct was repaired much more slowly. The highly b(6)G-resistant Y158H and P140K AGT mutants were also inactivated by 5'-d[T(b(6)G)(5)G]-3'. Studies with 7-mers containing a single b(6)G adduct showed that 5'-d[TGGGG(b(6)G)G]-3' was more poorly repaired by wild-type AGT than 5'-d[T(b(6)G)GGGGG]-3' and 5'-d[TGG(b(6)G)GGG]-3' and was even less repairable by mutants Y158H and P140K. This positional effect was unaffected by interchanging the terminal 5'- or 3'-nucleotides and was also observed with single-stranded 16-mer oligodeoxyribonucleotides containing O(6)-methylguanine, where a minimum of four nucleotides 3' to the lesion was required for the most efficient repair. Annealing with the reverse complementary strands to produce double-stranded substrates increased the ability of AGT to repair adducts at all positions except at positions 2 and 15. Our results suggest that AGT recognizes the polarity of single-stranded DNA, with the best substrates having an adduct adjacent to the 5'-terminal residue. These findings will aid in designing novel AGT inhibitors that incorporate O(6)-alkylguanine adducts in oligodeoxyribonucleotide contexts.


Subject(s)
DNA Repair , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Oligodeoxyribonucleotides/metabolism , Alkaline Phosphatase/metabolism , Amino Acid Substitution , Animals , Base Sequence , Crotalus , Escherichia coli/enzymology , Kinetics , Magnetic Resonance Spectroscopy , Mutagenesis, Site-Directed , Phosphodiesterase I , Phosphoric Diester Hydrolases , Recombinant Proteins/metabolism , Substrate Specificity
19.
Cancer Res ; 62(11): 3037-43, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12036910

ABSTRACT

O(6)-Alkylguanine-DNA alkyltransferase (AGT) plays a critical role in protection from the carcinogenic effects of simple alkylating agents by repairing O(6)-alkylguanine adducts via a direct transfer reaction. Nitric oxide (NO) or species derived from it are known to be able to initiate neoplastic growth and cannot only damage DNA, either directly or via the formation of intermediates such as nitrosamines, but can also inhibit some DNA repair processes. We have studied the inactivation of AGT by NO in detail in vitro and in vivo using wild-type human AGT (hAGT) and mutants at key residues. Our results show that hAGT is readily but reversibly inactivated by the formation of S-nitrosylcysteine at Cys-145, which is the alkyl acceptor site. The facile reaction of this cysteine residue with NO is attributable to its interaction with other residues in hAGT including His-146 and Glu-172 that activate the sulfhydryl group of Cys-145 to allow its nucleophilic attack on DNA adducts. Although the S-nitrosylcysteine adduct in hAGT is readily reversible by reaction with other cellular thiols, the formation of S-nitrosocysteine at Cys-145 was found to lead to the rapid degradation of the hAGT protein in vivo. This degradation is brought about by the ubiquitin/proteasomal system. The formation of an S-nitrosylcysteine at Cys-145 in hAGT in response to NO led to a large increase in the ubiquitination of the protein. This NO-mediated increase did not occur with the C145S or C145A mutants. A conformational change in hAGT, which involves opening of an asparagine hinge, normally occurs after alkylation of the protein in its role in DNA repair and causes degradation of the alkylated hAGT. Our results indicate that a similar effect occurs after reaction of the protein with NO. Thus, exposure to NO causes an irreversible loss of DNA repair capacity for alkylation adducts. This may contribute toward the potential development of tumors in cells upon chronic exposure to NO because of inflammation or infection. This may be of particular importance because such exposure may also lead to the formation of N-nitroso compounds that can act as alkylating agents.


Subject(s)
Nitric Oxide/pharmacology , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Animals , CHO Cells , Cricetinae , DNA Repair/drug effects , Enzyme Activation , Humans , Nitric Oxide Donors/pharmacology , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , O(6)-Methylguanine-DNA Methyltransferase/genetics , Plasmids/genetics , Protein Conformation , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , S-Nitroso-N-Acetylpenicillamine/pharmacology , S-Nitrosoglutathione/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...