Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Type of study
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.
Sci Rep ; 10(1): 11290, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32647312

ABSTRACT

Smoking-related lung tumors are characterized by profound epigenetic changes including scrambled patterns of DNA methylation, deregulated histone acetylation, altered gene expression levels, distorted microRNA profiles, and a global loss of cytosine hydroxymethylation marks. Here, we employed an enhanced version of bisulfite sequencing (RRBS/oxRRBS) followed by next generation sequencing to separately map DNA epigenetic marks 5-methyl-dC and 5-hydroxymethyl-dC in genomic DNA isolated from lungs of A/J mice exposed whole-body to environmental cigarette smoke for 10 weeks. Exposure to cigarette smoke significantly affected the patterns of cytosine methylation and hydroxymethylation in the lungs. Differentially hydroxymethylated regions were associated with inflammatory response/disease, organismal injury, and respiratory diseases and were involved in regulation of cellular development, function, growth, and proliferation. To identify epigenetic changes in the lung associated with exposure to tobacco carcinogens and inflammation, A/J mice were intranasally treated with the tobacco carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), the inflammatory agent lipopolysaccharide (LPS), or both. NNK alone caused minimal epigenetic alterations, while exposure either to LPS or NNK/LPS in combination led to increased levels of global cytosine methylation and formylation, reduced cytosine hydroxymethylation, decreased histone acetylation, and altered expression levels of multiple genes. Our results suggest that inflammatory processes are responsible for epigenetic changes contributing to lung cancer development.


Subject(s)
Epigenesis, Genetic , Inhalation Exposure , Lung Neoplasms/genetics , Lung/drug effects , Smoke/adverse effects , Animals , Carcinogens/metabolism , Cell Proliferation , Chromatography, High Pressure Liquid , CpG Islands , Cytosine/chemistry , DNA/metabolism , DNA Methylation , Female , High-Throughput Nucleotide Sequencing , Histones/chemistry , Histones/metabolism , Inflammation , Mice , Mice, Inbred Strains , Nitrosamines/metabolism , Smoking , Sulfites/pharmacology , Nicotiana , Tobacco Products
3.
Biochemistry ; 57(42): 6061-6069, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30230311

ABSTRACT

A precise balance of DNA methylation and demethylation is required for epigenetic control of cell identity, development, and growth. DNA methylation marks are introduced by de novo DNA methyltransferases DNMT3a/b and are maintained throughout cell divisions by DNA methyltransferase 1 (DNMT1), which adds methyl groups to hemimethylated CpG dinucleotides generated during DNA replication. Ten eleven translocation (TET) dioxygenases oxidize 5-methylcytosine (mC) to 5-hydroxymethylcytosine (hmC), 5-formylcytosine (fC), and 5-carboxylcytosine (caC), a process known to induce DNA demethylation and gene reactivation. In this study, we investigated the catalytic activity of human DNMT1 in the presence of oxidized forms of mC. A mass spectrometry-based assay was employed to study the kinetics of DNMT1-mediated cytosine methylation in CG dinucleotides containing C, mC, hmC, fC, or caC across from the target cytosine. Homology modeling, coupled with molecular dynamics simulations, was used to explore the structural consequences of mC oxidation with regard to the geometry of protein-DNA complexes. The DNMT1 enzymatic activity was strongly affected by the oxidation status of mC, with the catalytic efficiency decreasing in the following order: mC > hmC > fC > caC. Molecular dynamics simulations revealed that DNMT1 forms an unproductive complex with DNA duplexes containing oxidized forms of mC as a consequence of altered interactions of the target recognition domain of the protein with the C-5 substituent on cytosine. Our results provide new structural and mechanistic insight into TET-mediated DNA demethylation.


Subject(s)
5-Methylcytosine/analogs & derivatives , CpG Islands , DNA (Cytosine-5-)-Methyltransferase 1/chemistry , DNA Demethylation , 5-Methylcytosine/chemistry , 5-Methylcytosine/metabolism , Catalysis , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Humans , Oxidation-Reduction
4.
Article in English | MEDLINE | ID: mdl-26595795

ABSTRACT

A liquid chromatography-electrospray ionization-tandem mass spectrometry (HPLC-ESI(+)-MS/MS) method for the analysis of the tobacco-specific carcinogens N'-nitrosonornicotine (NNN) and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL) and their glucuronides (total NNN and total NNAL) in human urine was developed. The method has excellent accuracy and intra-day and inter-day precision, and limits of quantitation of 0.015 and 0.075pmol/mL urine, respectively, for total NNN and total NNAL. A unique aspect of this method is internal assessment of possible artifactual formation of NNN by inclusion of the monitor amine [pyridine-D4]nornicotine. We found that artifactual formation of NNN comprised only 2.5% of the measured amounts of total NNN in urine of cigarette smokers, under our conditions using ammonium sulfamate as an inhibitor of nitrosation. The method was applied to urine samples from cigarette smokers and e-cigarette users. Levels of total NNN and total NNAL in the urine of cigarette smokers averaged 0.060±0.035pmol/mL and 2.41±1.41pmol/mL urine, (N=38), respectively, which were both significantly greater than in the urine of 27 e-cigarette users.


Subject(s)
Electronic Nicotine Delivery Systems , Nitrosamines/urine , Pyridines/urine , Smoking/urine , Chromatography, Liquid , Humans , Limit of Detection , Reference Standards , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
5.
Chem Res Toxicol ; 28(11): 2151-9, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26398225

ABSTRACT

The tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK, 1) is a potent lung carcinogen in laboratory animals and is believed to play a key role in the development of lung cancer in smokers. Metabolic activation of NNK leads to the formation of pyridyloxobutyl DNA adducts, a critical step in its mechanism of carcinogenesis. In addition to DNA nucleobase adducts, DNA phosphate adducts can be formed by pyridyloxobutylation of the oxygen atoms of the internucleotidic phosphodiester linkages. We report the use of a liquid chromatography-nanoelectrospray ionization-high-resolution tandem mass spectrometry technique to characterize 30 novel pyridyloxobutyl DNA phosphate adducts in calf thymus DNA (CT-DNA) treated with 4-(acetoxymethylnitrosamino)-1-(3-pyridyl)-1-butanone (NNKOAc, 2), a regiochemically activated form of NNK. A (15)N3-labeled internal standard was synthesized for one of the most abundant phosphate adducts, dCp[4-oxo-4-(3-pyridyl)butyl]dC (CpopC), and this standard was used to quantify CpopC and to estimate the levels of other adducts in the NNKOAc-treated CT-DNA. Formation of DNA phosphate adducts by NNK in vivo was further investigated in rats treated with NNK acutely (0.1 mmol/kg once daily for 4 days by subcutaneous injection) and chronically (5 ppm in drinking water for 10, 30, 50, and 70 weeks). This study provides the first comprehensive structural identification and quantitation of a panel of DNA phosphate adducts of a structurally complex carcinogen and chemical support for future mechanistic studies of tobacco carcinogenesis in humans.


Subject(s)
Carcinogens/toxicity , DNA Adducts , Nicotiana , Nitrosamines/toxicity , Phosphates/metabolism , Animals , DNA/metabolism , Lung/metabolism , Lung Neoplasms , Male , Rats, Inbred F344 , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
6.
Nicotine Tob Res ; 17(6): 704-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25335945

ABSTRACT

INTRODUCTION: Electronic cigarettes (e-cigarettes) are rapidly increasing in popularity but little information is available on their potential toxic or carcinogenic effects. METHODS: Twenty-eight e-cigarette smokers who had not smoked tobacco cigarettes for at least 2 months provided urine samples which were analyzed by validated methods for a suite of toxicant and carcinogen metabolites including 1-hydroxypyrene (1-HOP), 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol and its glucuronides (total NNAL), 3-hydroxypropylmercapturic acid (3-HPMA), 2-hydroxypropylmercapturic acid (2-HPMA), 3-hydroxy-1-methylpropylmercapturic acid (HMPMA), S-phenylmercapturic acid (SPMA), nicotine, and cotinine. Levels of these compounds were compared to those found in cigarette smokers from three previous studies. RESULTS: Levels of 1-HOP, total NNAL, 3-HPMA, 2-HPMA, HMPMA, and SPMA were significantly lower in the urine of e-cigarette users compared to cigarette smokers. Levels of nicotine and cotinine were significantly lower in e-cigarette users compared to cigarette smokers in one study but not in another. CONCLUSIONS: With respect to the compounds analyzed here, e-cigarettes have a more favorable toxicity profile than tobacco cigarettes.


Subject(s)
Carcinogens/analysis , Electronic Nicotine Delivery Systems , Hazardous Substances/urine , Smoking/urine , Tobacco Products , Acetylcysteine/analogs & derivatives , Acetylcysteine/urine , Adult , Cotinine/urine , Female , Humans , Male , Middle Aged , Nicotine/urine , Nitrosamines/urine , Pyrenes/urine , Pyridines/urine , Young Adult
7.
Anal Chem ; 87(3): 1514-7, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25544129

ABSTRACT

Two of the most widely measured compounds in the urine of people who use tobacco products are cotinine, a major metabolite of the addictive constituent nicotine, and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL), a metabolite of the powerful lung carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). Thousands of analyses have been reported in the literature, carried out exclusively, to the best of our knowledge, by separate methods. In the study reported here, we have developed a sensitive, accurate, and precise liquid chromatography-electrospray ionization-tandem mass spectrometry-selected reaction monitoring method for the combined analysis of total cotinine (the sum of cotinine and its glucuronide) and total NNAL (the sum of NNAL and its glucuronide). The new method quantifies naturally occurring [(13)C]cotinine to minimize problems associated with the vast differences in concentration of total cotinine and total NNAL in urine. This method should greatly facilitate future determinations of these important compounds.


Subject(s)
Cotinine/urine , Nicotine/metabolism , Nitrosamines/urine , Pyridines/urine , Smoking/urine , Chromatography, Liquid , Cotinine/metabolism , Humans , Nitrosamines/metabolism , Pyridines/metabolism , Smoking/metabolism , Tandem Mass Spectrometry , Nicotiana/metabolism
8.
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
9.
Nature ; 494(7437): 366-70, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23389445

ABSTRACT

Several mutations are required for cancer development, and genome sequencing has revealed that many cancers, including breast cancer, have somatic mutation spectra dominated by C-to-T transitions. Most of these mutations occur at hydrolytically disfavoured non-methylated cytosines throughout the genome, and are sometimes clustered. Here we show that the DNA cytosine deaminase APOBEC3B is a probable source of these mutations. APOBEC3B messenger RNA is upregulated in most primary breast tumours and breast cancer cell lines. Tumours that express high levels of APOBEC3B have twice as many mutations as those that express low levels and are more likely to have mutations in TP53. Endogenous APOBEC3B protein is predominantly nuclear and the only detectable source of DNA C-to-U editing activity in breast cancer cell-line extracts. Knockdown experiments show that endogenous APOBEC3B correlates with increased levels of genomic uracil, increased mutation frequencies, and C-to-T transitions. Furthermore, induced APOBEC3B overexpression causes cell cycle deviations, cell death, DNA fragmentation, γ-H2AX accumulation and C-to-T mutations. Our data suggest a model in which APOBEC3B-catalysed deamination provides a chronic source of DNA damage in breast cancers that could select TP53 inactivation and explain how some tumours evolve rapidly and manifest heterogeneity.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cytidine Deaminase/metabolism , Mutagenesis , Point Mutation , Base Sequence , Biocatalysis , Breast Neoplasms/pathology , Cell Death , Cell Line, Tumor , Cytidine Deaminase/genetics , DNA Damage/genetics , DNA Fragmentation , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Deamination , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Histones/metabolism , Humans , Minor Histocompatibility Antigens , Mutagenesis/genetics , Phenotype , Point Mutation/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Uracil/metabolism
10.
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
11.
Nucleic Acids Res ; 39(9): 3988-4006, 2011 May.
Article in English | MEDLINE | ID: mdl-21245046

ABSTRACT

Endogenous 5-methylcytosine ((Me)C) residues are found at all CG dinucleotides of the p53 tumor suppressor gene, including the mutational 'hotspots' for smoking induced lung cancer. (Me)C enhances the reactivity of its base paired guanine towards carcinogenic diolepoxide metabolites of polycyclic aromatic hydrocarbons (PAH) present in cigarette smoke. In the present study, the structural basis for these effects was investigated using a series of unnatural nucleoside analogs and a representative PAH diolepoxide, benzo[a]pyrene diolepoxide (BPDE). Synthetic DNA duplexes derived from a frequently mutated region of the p53 gene (5'-CCCGGCACCC GC[(15)N(3),(13)C(1)-G]TCCGCG-3', + strand) were prepared containing [(15)N(3), (13)C(1)]-guanine opposite unsubstituted cytosine, (Me)C, abasic site, or unnatural nucleobase analogs. Following BPDE treatment and hydrolysis of the modified DNA to 2'-deoxynucleosides, N(2)-BPDE-dG adducts formed at the [(15)N(3), (13)C(1)]-labeled guanine and elsewhere in the sequence were quantified by mass spectrometry. We found that C-5 alkylcytosines and related structural analogs specifically enhance the reactivity of the base paired guanine towards BPDE and modify the diastereomeric composition of N(2)-BPDE-dG adducts. Fluorescence and molecular docking studies revealed that 5-alkylcytosines and unnatural nucleobase analogs with extended aromatic systems facilitate the formation of intercalative BPDE-DNA complexes, placing BPDE in a favorable orientation for nucleophilic attack by the N(2) position of guanine.


Subject(s)
7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/analogs & derivatives , Cytosine/analogs & derivatives , DNA Adducts/chemistry , Deoxyguanosine/analogs & derivatives , 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/chemistry , Base Pairing , Chromatography, High Pressure Liquid , Deoxyguanosine/chemistry , Genes, p53 , Guanine/chemistry , Isotope Labeling , Models, Molecular , Oligodeoxyribonucleotides/chemical synthesis , Oligodeoxyribonucleotides/chemistry , Spectrometry, Fluorescence , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
12.
Chem Res Toxicol ; 17(11): 1399-405, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15540937

ABSTRACT

The compound 3-amino-2-quinoxalinecarbonitrile 1,4-dioxide (4) displays potent hypoxia-selective cytotoxicity in cell culture. This compound is structurally similar to the known hypoxia-selective DNA-damaging agent tirapazamine (1, TPZ), but the ability of 4 to cause DNA damage under low-oxygen conditions has not previously been characterized. The results presented here provide the first evidence that 4 causes reductively activated DNA damage under hypoxic conditions. The findings indicate that one-electron reduction of 4 by NADPH:cytochrome P450 reductase yields an oxygen-sensitive intermediate (5). This activated intermediate is rapidly destroyed by reaction with O2 under aerobic conditions, but goes forward to cause DNA damage under low-oxygen conditions. Analysis of the DNA damage indicates that reductive activation of 4 leads to production of a highly reactive, freely diffusible oxidizing radical that causes sequence-independent cleavage of the deoxyribose backbone and oxidative damage to the heterocyclic bases in duplex DNA. On the basis of the experiments reported here, the chemical nature of the DNA damage caused by redox-activated 4 is analogous to that reported previously for TPZ.


Subject(s)
Antineoplastic Agents/metabolism , DNA Damage , DNA/drug effects , Hypoxia , NADPH-Ferrihemoprotein Reductase/metabolism , Nitriles/metabolism , Oxides/metabolism , Quinoxalines/metabolism , Antineoplastic Agents/pharmacology , Dose-Response Relationship, Drug , Nitriles/pharmacology , Oxides/pharmacology , Quinoxalines/pharmacology
13.
Bioorg Med Chem Lett ; 12(17): 2325-9, 2002 Sep 02.
Article in English | MEDLINE | ID: mdl-12161126

ABSTRACT

Tirapazamine is a bioreductively activated DNA-damaging agent that selectively kills the hypoxic cells found in solid tumors. In this work, base excision repair enzymes were used to provide evidence that tirapazamine causes significant amounts of damage to both purine and pyrimidine residues in double-stranded DNA.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Damage/drug effects , Triazines/pharmacology , DNA/metabolism , DNA Ligases/metabolism , Oxidation-Reduction , Purines/analysis , Pyrimidines/analysis , Restriction Mapping , Substrate Specificity , Tirapazamine
14.
Photochem Photobiol ; 75(4): 339-45, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12003122

ABSTRACT

The absorption and fluorescence spectra of 3-aminobenzo-1,2,4-triazine di-N-oxide (tirapazamine) have been recorded and exhibit a dependence on solvent that correlates with the Dimroth ET30 parameter. Time-dependent density functional theory calculations reveal that the transition of tirapazamine in the visible region is pi-->pi* in nature. The fluorescence lifetime is 98+/-2 ps in water. The fluorescence quantum yield is approximately 0.002 in water. The fluorescence of tirapazamine is efficiently quenched by electron donors via an electron-transfer process. Linear Stern-Volmer fluorescence quenching plots are observed with sodium azide, potassium thiocyanate, guanosine monophosphate and tryptophan (Trp) methyl ester hydrochloride. Guanosine monophosphate, tyrosine (Tyr) methyl ester hydrochloride and Trp methyl ester hydrochloride appear to quench the fluorescence at a rate greater than diffusion control implying that these substrates complex with tirapazamine in its ground state. This complexation was detected by absorption spectroscopy.


Subject(s)
Antineoplastic Agents/chemistry , Triazines/chemistry , Electron Transport , Photochemistry , Spectrophotometry, Ultraviolet , Tirapazamine
SELECTION OF CITATIONS
SEARCH DETAIL
...