Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 4: e731, 2013 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-23868064

RESUMO

Sirtuin 3 (Sirt3), a major mitochondrial NAD(+)-dependent deacetylase, targets various mitochondrial proteins for lysine deacetylation and regulates important cellular functions such as energy metabolism, aging, and stress response. In this study, we identified the human 8-oxoguanine-DNA glycosylase 1 (OGG1), a DNA repair enzyme that excises 7,8-dihydro-8-oxoguanine (8-oxoG) from damaged genome, as a new target protein for Sirt3. We found that Sirt3 physically associated with OGG1 and deacetylated this DNA glycosylase and that deacetylation by Sirt3 prevented the degradation of the OGG1 protein and controlled its incision activity. We further showed that regulation of the acetylation and turnover of OGG1 by Sirt3 played a critical role in repairing mitochondrial DNA (mtDNA) damage, protecting mitochondrial integrity, and preventing apoptotic cell death under oxidative stress. We observed that following ionizing radiation, human tumor cells with silencing of Sirt3 expression exhibited deteriorated oxidative damage of mtDNA, as measured by the accumulation of 8-oxoG and 4977 common deletion, and showed more severe mitochondrial dysfunction and underwent greater apoptosis in comparison with the cells without silencing of Sirt3 expression. The results reported here not only reveal a new function and mechanism for Sirt3 in defending the mitochondrial genome against oxidative damage and protecting from the genotoxic stress-induced apoptotic cell death but also provide evidence supporting a new mtDNA repair pathway.


Assuntos
Apoptose , DNA Glicosilases/metabolismo , DNA Mitocondrial/genética , Estresse Oxidativo , Sirtuína 3/metabolismo , Acetilação , Calpaína/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Dano ao DNA , Reparo do DNA , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Potencial da Membrana Mitocondrial , Ligação Proteica , Mapeamento de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Proteólise , Sirtuína 3/genética
2.
Biochemistry ; 40(9): 2647-52, 2001 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-11258875

RESUMO

DNA polymerases replicate DNA with high fidelity despite the small differences in energy between correct and incorrect base pairs. X-ray crystallographic and structure-activity kinetic experiments have implicated interactions with the minor groove of the DNA as being crucial for catalysis and fidelity. The current hypothesis is that polymerases check the geometry of the base pairs through hydrogen bonds and steric interactions with the minor groove of the DNA. The mechanisms by which these interactions are related to catalysis and fidelity are not known. In this manuscript, we have studied these interactions using a combination of site-specific mutagenesis of Escherichia coli DNA polymerase I (Klenow fragment) and atomic substitution of the DNA. Crystal structures have predicted hydrogen bonds from Arg668 to the terminal base on the primer (P1) and Gln849 to its base pair partner (T1). Kinetic studies, however, have implicated the minor groove of the primer terminus but not its base pair partner as being important to catalysis and fidelity. Hydrogen bonds between Arg668 and Gln849 to the DNA were probed with the site specific mutants, R668A and Q849A. Hydrogen bonds from the DNA were probed with three oligodeoxynucleotides which have a guanine or 3-deazaguanine (3DG) at P1, T1, or T2. We found that the pre-steady-state parameter k(pol) was decreased with R668A (40-fold) and Q849A (150-fold) or with 3DG at P1 (300-fold) or T2 (25-fold). When R668A was combined with 3DG at P1 the decrease in rate was only 80-fold, consistent with a hydrogen bond between Arg668 and P1. In contrast, when the 3DG substitution at P1 was combined with Q849A the rate reduction was 15000-fold. Similar reactions between R668A or Q849A and T2 showed that there are interactions between these sites although the interactions are not as strong as between P1 and R668.


Assuntos
Substituição de Aminoácidos/genética , DNA Polimerase I/química , DNA/química , Escherichia coli/enzimologia , Conformação de Ácido Nucleico , DNA Polimerase I/genética , Primers do DNA/química , Escherichia coli/genética , Ligação de Hidrogênio , Cinética , Mutagênese Sítio-Dirigida , Ácidos Nucleicos Heteroduplexes/química , Especificidade por Substrato/genética
3.
Chem Res Toxicol ; 12(9): 809-15, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10490502

RESUMO

Several fluoroquinolone antibacterial agents exhibit an adverse phototoxic effect in humans and are photo-cocarcinogenic in mice. The UV-induced production of reactive oxygen species plays a role in the toxicity and may be involved in carcinogenicity. Four fluoroquinolones were examined for the ability to photochemically produce oxidative damage in naked DNA. The major structural difference in the fluoroquinolones that would have an effect on their photostability is the functionality at the 8-position. At this position, 1-cyclopropyl-7-(2,8-diazbicyclo[4.3.0]non-8-yl)-6, 8-difluoro-1,4-dihydro-4-oxo-3-quinolinecarboxylic acid (BAY y3118) contains a chlorine atom, lomefloxacin a fluorine atom, ciprofloxacin a proton, and moxifloxacin a methoxy group. The formation of 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodGuo) in calf thymus DNA was assessed by HPLC with electrochemical detection, and strand breaks were measured in pBR322 with agarose gel electrophoresis. The relative photolability of the fluoroquinolones correlated to the extent of production of 8-oxodGuo and strand breaks, with both UVA and UVB irradiation, in the following order: BAY y3118 approximately lomefloxacin > ciprofloxacin > moxifloxacin. Experiments were performed to determine whether the mechanism of damage was due to a type I (radical) or type II (singlet oxygen) pathway. Nitrogen depletion of oxygen resulted in a decrease in the extent of formation of 8-oxodGuo, suggesting that oxygen was involved. The use of selective radical or singlet oxygen inhibitors was inconclusive with respect to which pathway was involved. The use of D(2)O as a solvent, which would extend the lifetime of singlet oxygen, suggested that this species is involved in the formation of 8-oxodGuo by moxifloxacin and ciprofloxacin, but not by lomefloxacin and BAY y3118. Similarly, it was found that singlet oxygen was not involved in strand break formation. Thus, the evidence suggests that fluoroquinolones can photochemically produce DNA damage by both type I and type II mechanisms.


Assuntos
Anti-Infecciosos/farmacologia , Dano ao DNA , DNA/efeitos dos fármacos , Desoxiguanosina/análogos & derivados , Fármacos Fotossensibilizantes/farmacologia , 8-Hidroxi-2'-Desoxiguanosina , Animais , Anti-Infecciosos/química , Bovinos , Cromatografia Líquida de Alta Pressão , Desoxiguanosina/biossíntese , Estabilidade de Medicamentos , Eletroforese em Gel de Ágar , Fluoroquinolonas , Oxirredução , Fármacos Fotossensibilizantes/química , Espécies Reativas de Oxigênio/metabolismo , Relação Estrutura-Atividade , Raios Ultravioleta
4.
Biochemistry ; 38(21): 6801-6, 1999 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-10346901

RESUMO

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.


Assuntos
Guanina/análogos & derivados , O(6)-Metilguanina-DNA Metiltransferase/química , O(6)-Metilguanina-DNA Metiltransferase/genética , Oligodesoxirribonucleotídeos/química , Sítios de Ligação/genética , Reparo do DNA , Guanina/química , Guanina/metabolismo , Humanos , Cinética , Mutagênese , Ácidos Nucleicos Heteroduplexes/química , Ácidos Nucleicos Heteroduplexes/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Oligodesoxirribonucleotídeos/metabolismo , Ligação Proteica/genética , Relação Estrutura-Atividade , Especificidade por Substrato/genética
5.
Chem Res Toxicol ; 12(2): 127-31, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10027788

RESUMO

DNA pyridyloxobutylation occurs during the metabolic activation of the tobacco-specific nitrosamines, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) and N'-nitrosonornicotine (NNN). This pathway contributes significantly to the carcinogenic and mutagenic activity of these nitrosamines. In general, the chemical structure of pyridyloxobutyl adducts are not well understood. Recently, an AGT reactive pyridyloxobutyl adduct was identified as O6-[4-oxo-4-(3-pyridyl)butyl]guanine (O6-pobG). To better understand the importance of this adduct to the biological activity of pyridyloxobutylating agents, we developed a method for site-specifically incorporating O6-pobG into DNA oligonucleotides. They were synthesized using the phosphoramidite of the precursor 2'-deoxy-O6-{3-[2-(3-pyridyl)-1,3-dithian-2-yl]propyl}guanosine. The dithiane group was oxidatively removed with N-chlorosuccinimide in a final postoligomerization reaction to generate the desired product. Human AGT with a polyhistidine tag was able to repair the O6-pobG-containing DNA oligonucleotide, generating unmodified oligonucleotide. These results are consistent with an alkyl group transfer mechanism for the repair of O6-pobG by AGT.


Assuntos
DNA/síntese química , Guanina/análogos & derivados , O(6)-Metilguanina-DNA Metiltransferase/química , Oligonucleotídeos/síntese química , Piridinas/química , Adutos de DNA/química , Guanina/química , Humanos , Mutagênicos/química , Proteínas Recombinantes/química
6.
Cancer Lett ; 129(2): 131-8, 1998 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-9719453

RESUMO

The tobacco-specific 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) is a potent lung carcinogen in mice, rats and Syrian golden hamsters and a suspected human lung carcinogen. We have reported earlier that structural analogs of the naturally occurring pulmonary toxin 4-ipomeanol (IPO) were non toxic up to 50 micromol/mouse. Because these analogs are in part structurally similar to NNK, they are expected to compete for the same enzymes and/or reactive sites within DNA. Both NNK and IPO are primarily metabolized by cytochrome P450 enzymes in the Clara cells of the lung but also in the liver. We describe here the optimal conditions for the study of NNK metabolism in human liver microsomes and our investigation of four non-toxic IPO analogs as potential inhibitors of NNK activation. The IPO analogs studied were 4-hydroxy-1-phenyl-1-octanone (4-HPO), 1,4-diphenyl-4-hydroxy-1-butanone (DPHB), 4-hydroxy-1-phenylpentane (HPPentane) and amyl benzene (AB). When added to microsomal incubations of human liver cells at a concentration of 100 microM, all of these compounds were strong inhibitors of NNK activation, decreasing the total alpha-hydroxylation of NNK, which is the main pathway of activation, by 60-70% and preventing N-oxidation by 78-86%.


Assuntos
Microssomos Hepáticos/metabolismo , Nitrosaminas/metabolismo , Terpenos/farmacologia , Antineoplásicos , Biomarcadores/análise , Biotransformação/efeitos dos fármacos , Butanonas/metabolismo , Butanonas/farmacologia , Humanos , Hidroxilação/efeitos dos fármacos , Cetonas/farmacocinética , Cetonas/farmacologia , Microssomos Hepáticos/efeitos dos fármacos , Pentanos/farmacologia , Piridinas/metabolismo
7.
Carcinogenesis ; 18(9): 1851-4, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9328186

RESUMO

4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL) is an important metabolite of the tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). Using the chiral derivatizing agent, (R)-(+)-alpha-methylbenzyl isocyanate [(R)-(+)-MBIC], previous work has shown that the enantiomeric ratio of metabolically formed NNAL and its glucuronide derivative may be species dependent. However, the absolute configuration of such NNAL has not been previously reported. Synthetically prepared racemic NNAL was converted to diastereomeric esters by reaction with (R)-(+)- and (S)-(-)-alpha-methoxy-alpha-(trifluoromethyl)phenylacetic acid (MTPA) chloride (Mosher's reagent) and the products were characterized by 1H-NMR. Based on chemical shift data, the absolute configuration of NNAL in each diastereomeric ester was assigned. Hydrolysis of (R)-NNAL-(R)-MTPA gave (R)-NNAL. This was converted to the corresponding carbamate by reaction with (R)-(+)-alpha-MBIC and the absolute configurations of the diastereomeric carbamates formed by reaction of (R)- and (S)-NNAL with (R)-(+)-MBIC were thereby assigned. Conversion of metabolically produced NNAL to the same carbamates allowed us to assign the NNAL formed from NNK by rat liver microsomes as (R)-NNAL. The major and minor NNAL-glucuronide diastereomers found in the urine of patas monkeys and humans exposed to NNK were similarly assigned; they were formed from (R)-NNAL and (S)-NNAL, respectively.


Assuntos
Microssomos Hepáticos/metabolismo , Nitrosaminas/química , Nitrosaminas/farmacocinética , Animais , Conformação Molecular , Ratos , Estereoisomerismo
8.
Biochemistry ; 36(43): 13292-7, 1997 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-9341220

RESUMO

A mechanism by which the Klenow fragment of DNA polymerase I monitors the geometry of the base pairs may involve hydrogen bonds between the polymerase and the minor groove of the nascent base pair. The involvement of the 3-position of guanine in the template strand was examined by synthesizing oligodeoxynucleotides containing guanine and 3-deazaguanine and comparing the steady-state kinetics of the incorporation of all four dNTPs. The Vmax/Km decreased a significant amount (170-fold) only when dCTP was the co-substrate suggesting that a hydrogen bond exists only when the correct base pair is being replicated. This approach was also used to examine how the Klenow fragment interacts with the 3-position of the mutagenic base O6-methylguanine (O6mG). The Vmax/Km for the incorporation of dTTP opposite O6-methyl-3-deazaguanine (O6m3DG) was 1700-fold less than opposite O6mG. In contrast, a small 6-fold increase in Vmax/Km occurred for the incorporation of dCTP opposite O6m3DG relative to O6mG. This result suggests that the hydrogen bond between the Klenow fragment and O6mG is more important in the incorporation of dTTP opposite O6mG and may contribute to the mutagenicity of O6mG.


Assuntos
DNA Polimerase I/metabolismo , Desoxirribonucleotídeos/metabolismo , Guanina/metabolismo , Antimetabólitos/metabolismo , Nucleotídeos de Desoxiadenina/metabolismo , Nucleotídeos de Desoxicitosina/metabolismo , Nucleotídeos de Desoxiguanina/metabolismo , Guanina/análogos & derivados , Cinética , Nucleotídeos de Timina/metabolismo
9.
Nucleic Acids Res ; 25(16): 3354-61, 1997 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-9241252

RESUMO

During DNA replication, mutations occur when an incorrect dNTP is incorporated opposite a carcinogen-modified nucleotide. We have probed the structures of the interaction between O 6-methylguanine ( O 6mG) and cytosine and thymine during replication by kinetic means in order to examine the structure during the rate determining step. The kinetics of incorporation of dCTP and dTTP opposite O 6mG and three analogs, S 6-methyl-6-thioguanine, O 6-methyl-1-deazaguanine and O 6-methylhypoxanthine, have been measured with four polymerases, the Klenow fragment of DNA polymerase I, the Klenow fragment with the proof-reading exonuclease inactivated, Taq and Tth polymerases. In the insertion of dTTP opposite O 6mG, a large decrease in V max/ K m was observed only upon modification of the N1 position. This result is consistent with a Watson-Crick type configuration. For the incorporation of dCTP, the V max/ K m was significantly decreased only with removal of the exocyclic amino group at the 2 position. The pH dependence of the ratio of incorporation of dCTP and dTTP was independent of pH at physiological pH. This result suggests that dCTP is incorporated via an uncharged complex such as the wobble configuration.


Assuntos
Citosina/química , Replicação do DNA , Guanina/análogos & derivados , Ligação de Hidrogênio , Timina/química , Guanina/química , Concentração de Íons de Hidrogênio , Relação Estrutura-Atividade
10.
Toxicol Appl Pharmacol ; 145(2): 381-7, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9266812

RESUMO

We have previously reported that two fluoroquinolone antibiotics gave rise to 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxo-dG) in DNA of cells concurrently exposed to UV-A and that this correlated with clinical phototoxicity. To determine the structural basis for generation of oxidative damage, the ability of two synthetic fluoroquinolone candidate antibiotics, Bayer 12-8039 (12-8039) and Bayer Y3118 (Y3118), to give rise to 8-oxo-dG in cultured liver epithelial cells was compared. 12-8039 contains a methoxy group at the 8 position of the quinolone nucleus, whereas Y3118 contains a chlorine group at the same position. Y3118 produced dose-dependent increases in 8-oxo-dG formation in cultured cells after UVA irradiation, whereas the 8-OCH3-substituted 12-8039 produced no increase. Also, after exposure to 20 J/cm2 UVA, UV spectral scans of both compounds revealed that Y3118 underwent photodegradation whereas 12-8039 was stable. These results demonstrate that the presence of an 8-OCH3 group on the quinolone nucleus is important for the reduction of photogeneration of oxidative DNA damage and photodegradation in the presence of UVA irradiation. From this, we suggest that 12-8039 has little phototoxic potential.


Assuntos
Anti-Infecciosos/farmacologia , Compostos Aza , Desoxiguanosina/análogos & derivados , Fluoroquinolonas , Quinolinas , Raios Ultravioleta/efeitos adversos , 8-Hidroxi-2'-Desoxiguanosina , Animais , Anti-Infecciosos/química , Anti-Infecciosos/efeitos da radiação , Linhagem Celular , Desoxiguanosina/antagonistas & inibidores , Desoxiguanosina/biossíntese , Desoxiguanosina/efeitos da radiação , Epitélio/efeitos dos fármacos , Epitélio/efeitos da radiação , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/efeitos da radiação , Moxifloxacina , Quinolonas/química , Quinolonas/farmacologia , Quinolonas/efeitos da radiação , Ratos
11.
Chem Res Toxicol ; 10(5): 562-7, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9168254

RESUMO

The lung carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) is activated to reactive metabolites that methylate or pyridyloxobutylate DNA. Previous studies demonstrated that pyridyloxobutylated DNA interferes with the repair of O6-methylguanine (O6-mG) by O6-alkylguanine-DNA alkyltransferase (AGT). The AGT reactivity of pyridyloxobutylated DNA was attributed to (pyridyloxobutyl)guanine adducts. One potential AGT substrate adduct, 2'-deoxy-O6-[4-oxo-4-(3-pyridyl)butyl]guanosine (O6-pobdG), was prepared. This adduct was stable at pH 7.0 for greater than 13 days and to neutral thermal hydrolysis conditions (pH 7.0, 100 degrees C, 30 min). Under mild acid hydrolysis conditions (0.1 N HCl, 80 degrees C), O6-pobdG was depurinated to yield O6-[4-oxo-4-(3-pyridyl)butyl]guanine (O6-pobG). O6-pobdG was hydrolyzed to 4-hydroxy-1-(3-pyridyl)-1-butanone and guanine under strong acid hydrolysis conditions (0.8 N HCl, 80 degrees C). O6-pobG was detected in 0.1 N HCl hydrolysates of DNA alkylated with the model pyridyloxobutylating agent 4-(acetoxymethylnitrosamino)-1-(3-[5-3H]pyridyl)-1-butanone ([5-3H]NNKOAc). When [5-3H]NNKOAc-treated DNA was incubated with either rat liver or recombinant human AGT, O6-pobG was removed, presumably a result of transfer of the pyridyloxobutyl group from the O6-position of guanine to AGT's active site.


Assuntos
Adutos de DNA/análise , DNA/efeitos dos fármacos , Guanina/análogos & derivados , Metiltransferases/metabolismo , Nitrosaminas/toxicidade , Piridinas/toxicidade , Animais , Guanina/análise , Humanos , Fígado/enzimologia , O(6)-Metilguanina-DNA Metiltransferase , Ratos , Especificidade por Substrato
12.
Chem Res Toxicol ; 10(12): 1412-9, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9437533

RESUMO

Reaction of DNA with the carcinogens N-methyl-N-nitrosourea and N-nitroso-N,N-dimethylamine produces several methylated species including the premutagenic O6-methylguanine. The mechanism of methylation is believed to be through a methanediazonium ion. We have studied the mechanism of methylation of DNA by these carcinogens by analyzing the stereochemistry of the methyl transfer. DNA was methylated in vitro by (R)- and (S)-N-[2H1,3H]methyl-N-nitrosourea and in vivo by (R)- and (S)-N-[2H1,3H]methyl-N-methyl-N-nitrosamine and (R)- and (S)-N-[2H1,3H]methyl-N-nitrosourea. 7-Methylguanine, 3-methyladenine, O6-methylguanine, and the methylated phosphate backbone were isolated. The methyl groups were converted into acetic acid, and the stereochemistry was analyzed. The identity of the nucleophile did not influence the stereochemistry of the methylation reaction. It was found that the methyl group was transferred with an average of 73% inversion and 27% retention of configuration. The most likely mechanism for the retention of configuration is through multiple methylation events in which nucleophiles which initially react with the methanediazonium ion react as electrophiles with DNA.


Assuntos
Alquilantes/farmacologia , Metilação de DNA , DNA/efeitos dos fármacos , Metilnitrosoureia/farmacologia , Compostos Nitrosos/farmacologia , Alquilantes/síntese química , Alquilantes/química , Animais , Diazometano/química , Feminino , Metilnitrosoureia/síntese química , Metilnitrosoureia/química , Conformação Molecular , Compostos Nitrosos/síntese química , Compostos Nitrosos/química , Ratos , Ratos Sprague-Dawley , Estereoisomerismo
13.
Chem Res Toxicol ; 9(6): 949-53, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8870981

RESUMO

Pyridyloxobutylation of DNA yields adducts that react with O6-alkylguanine-DNA alkyl-transferase (AGT) to prevent the repair of O6-methylguanine (O6-mG). The chemical characterization of pyridyloxobutyl adducts has been confounded by their instability under DNA hydrolysis conditions. They decompose to 4-hydroxy-1-(3-pyridyl)-1-butanone (HPB) during the chemical or enzymatic hydrolysis of DNA. The goal of these studies was to determine which bases are pyridyloxobutylated to form AGT-reactive adducts. The model pyridyloxobutylating agent, 4-[(acetoxymethyl)nitrosamino]-1-(3-pyridyl)-1-butanone (NNKOAc), was reacted with either poly(dAdT) or poly(dGdC) to generate DNA substrates for reaction with AGT. Only the pyridyloxobutylated poly(dGdC) was able to prevent the ability of partially purified rat liver AGT to repair O6-mG. These results paralleled those obtained for the corresponding methylated substrates. These studies are consistent with the pyridyloxobutylation of GC base pairs and not AT base pairs in the DNA to generate a substrate for AGT. In order to distinguish between the formation of reactive adducts at C residues versus G residues, two oligomers were designed that were complementary to one another. One oligomer contained A, T, and G residues, whereas its complement contained T, A, and C residues. Only the dG-containing oligomer reacted with NNKOAc to generate an AGT-reactive adduct, again paralleling the results obtained for a methylating agent. These results demonstrate that pyridyloxobutylation of only guanine residues produces adducts that react with AGT. These AGT-reactive guanine adducts are relatively stable within DNA, with a half-life of 1-2 weeks at 37 degrees C. They represent up to 70% of the total HPB-releasing adducts in the NNKOAc-treated DNA. We postulate that a potential AGT-reactive adduct is an O6-(pyridyloxobutyl)guanine adduct.


Assuntos
Guanina/química , Metiltransferases/química , Nitrosaminas , Piridinas , Animais , Adutos de DNA/química , Reparo do DNA , Guanina/análogos & derivados , Guanina/metabolismo , Técnicas In Vitro , Fígado/metabolismo , Metiltransferases/metabolismo , O(6)-Metilguanina-DNA Metiltransferase , Poli dA-dT/química , Polidesoxirribonucleotídeos/química , Ratos
14.
Chem Res Toxicol ; 9(1): 58-66, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8924617

RESUMO

The customary salting and pickling of fish in high risk gastric cancer regions were modeled to explore the relevant causative chemicals. The fish Sanma hiraki was treated with sodium chloride and sodium nitrite at pH 3. Previously, it had been found that an extract of the treated fish was mutagenic in Salmonella typhimurium TA 1535 without S9 and also that it induced glandular stomach cancer upon gavage to rats. We now demonstrate that the mutagenicity was enhanced by preincubation of the raw meat for several days before salt-nitrite treatment. HPLC techniques showed that three mutagens were present in the fish extract. One of the mutagens was found to be stable over the pH range of 1.0-9.0. This mutagen was purified by silica gel solid phase extraction, followed by a series of reverse phase HPLC steps, and was characterized by low and high resolution MS, NMR, and FT-IR. While N-nitroso compounds were generally believed to be associated with gastric carcinogenesis, it was unexpectedly found that the mutagen has the novel structure 2-chloro-4-methylthiobutanoic acid (CMBA). Based on the structure, it seemed likely that methionine might be the precursor, and this was, indeed, proven. Both salt and nitrite are essential factors for forming this mutagen. The yield of CMBA was linear for chloride concentrations from 0 to 800 mM NaCl. Of 20 amino acids reacted with nitrite and chloride at pH 3, only methionine generated a mutagen for S. typhimurium TA 1535. Tryptophan gave a product mutagenic in S. typhimurium TA 100 and TA 98, but not TA 1535, and in the case of tyrosine, the mutagen was active only for TA 100. These results suggest an important role for salt in gastric carcinogenesis and provide new approaches for exploring the formation of mutagens/carcinogens for specific target organs.


Assuntos
Butiratos/toxicidade , Peixes , Conservação de Alimentos , Metionina/toxicidade , Mutagênicos/toxicidade , Neoplasias Gástricas/induzido quimicamente , Aminoácidos/efeitos dos fármacos , Animais , Butiratos/isolamento & purificação , Estabilidade de Medicamentos , Concentração de Íons de Hidrogênio , Metionina/efeitos dos fármacos , Testes de Mutagenicidade , Mutagênicos/isolamento & purificação , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/genética , Nitrito de Sódio/toxicidade , Compostos de Sulfidrila
16.
Biochemistry ; 33(37): 11364-71, 1994 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-7727387

RESUMO

O6-Alkylguanine-DNA alkyltransferase (AGT) repairs the mutagenic O6-methylguanine (O6-mG) lesion by transferring a methyl group from the 6-position of guanine to a cysteine residue on the protein. The simplest possible mechanism is an SN2 process in which the cysteine displaces the methyl group off of the guanine in a concerted reaction. To probe the interactions between the protein and guanine leaving group, oligodeoxynucleotide duplexes containing analogs of O6mG were synthesized and then reacted with AGT. The analogs, which were incorporated into deoxynucleotides include O6-methylhypoxanthine (O6-mH),S6-methyl-6-thioguanine (S6mG),S6-methyl-6-thiohypoxanthine (S6mH),Se6-methyl-6-selenoguanine (Se6mG),Se6-methyl-6-selenohypoxanthine (Se6mH), O6-methyl-1-deazaguanine (O6m1DG), O6-methyl-3-deazaguanine (O6m3DG), and O6-methyl-7-deazaguanine (O6m7DG), differ from O6mG in that the heteroatoms have been replaced so that they are poorer hydrogen bond participants and proton acceptors. AGT was reacted with oligonucleotide duplexes of the sequence 5'-GGC GCT XGA GGC GTG-3' in which X was O6mG or an analog in which X was paired with C. The reactions in 50 mM Tris-HCl and 1 mM EDTA, pH7.6 and 37 degrees C, were followed by anion-exchange HPLC in 10 mM NaOH with a NaCl gradient. All detected reactions were demethylations of the oligodeoxynucleotides except for O6m3DG, which reacted in an unknown manner.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Guanina/análogos & derivados , Metiltransferases/metabolismo , Oligodesoxirribonucleotídeos/síntese química , Sequência de Bases , Sítios de Ligação , Glicosilação , Ligação de Hidrogênio , Hidrólise , Indicadores e Reagentes , Espectroscopia de Ressonância Magnética , Dados de Sequência Molecular , Conformação de Ácido Nucleico , O(6)-Metilguanina-DNA Metiltransferase , Oligodesoxirribonucleotídeos/química , Oligodesoxirribonucleotídeos/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato
18.
Cell Growth Differ ; 4(7): 595-602, 1993 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8398900

RESUMO

Nuclear proteins from human melanoma cells exhibit strong binding activity to the UV response element (TGACAACA); however, this binding is inhibited following UV-C irradiation. In contrast, the binding of nuclear proteins from rodent fibroblasts and human keratinocytes to the UV-responsive element is initially weak and increases significantly upon UV irradiation. The addition of nuclear proteins from UV-irradiated melanoma cells to those prepared from nonirradiated cells inhibited the binding to the UV-responsive element in a concentration-dependent manner. Fast protein liquid chromatographic analysis of nuclear proteins from UV-irradiated melanoma cells revealed 12 and 14 kilodalton proteins within a fraction which also contained the inhibitory activity. The inhibitor blocks the binding of proteins to three other target sequences, AP1, CREB, and PEBP2, as well as the in vitro transcription of SV40 promoter sequences. The inhibitor was also found in UV-irradiated melanocytes, suggesting that it is tissue specific. The induction of a transcriptional inhibitor in response to UV irradiation represents a regulatory event that may play an important role in the transcriptional response of both normal and malignant melanocytes to UV irradiation.


Assuntos
Melanócitos/efeitos da radiação , Melanoma/metabolismo , Proteínas Nucleares/metabolismo , Transcrição Gênica/efeitos da radiação , Raios Ultravioleta , Sequência de Bases , Fracionamento Químico , Humanos , Melanócitos/metabolismo , Dados de Sequência Molecular , Ligação Proteica , Células Tumorais Cultivadas
19.
Biochemistry ; 31(14): 3688-94, 1992 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-1314648

RESUMO

The DNA repair protein O6-alkylguanine-DNA alkyltransferase (AGT) repairs the promutagenic O6-methylguanine lesion by transferring the methyl group to a cysteine residue on the protein. A mechanism in which AGT activates the guanyl moiety as a leaving group by protonation of a heteroatom on guanine was probed by reacting AGT with analogues of O6-methylguanine in which the heteroatoms were changed. The initial rates of reaction were measured at various substrate concentrations in 50 mM Hepes, 1 mM EDTA, 1 mM DTT, and 10% glycerol, pH 7.8 at 37 degrees C. The kinact (h-1) and Kin (mM) were determined for O6-methylguanine (1.66 +/- 0.19, 1.51 +/- 0.32), 6-methoxypurine (1.07 +/- 0.25, 10.6 +/- 4.2), S6-methyl-6-thioguanine (0.63 +/- 0.04, 1.17 +/- 0.18), 6-methylthiopurine (no reaction), Se6-methyl-6-selenoguanine (1.76 +/- 0.28, 10.6 +/- 5.0), 6-methylselenopurine (2.51 +/- 0.62, 15.7 +/- 6.3), O6-methyl-1-deazaguanine (1.71 +/- 0.34, 14.8 +/- 4.4), O6-methyl-3-deazaguanine (1.90 +/- 0.24, 2.54 +/- 0.59), and O6-methyl-7-deazaguanine (1.97 +/- 0.26, 2.56 +/- 0.72). These results indicate that replacement of the nitrogens does not affect the kinact parameter but the Kin is increased upon removal of the exocyclic amino group and the nitrogen at the 1-position. Replacement of the oxygen with sulfur decreases the kinact, and replacement with selenium increases the Kin. The results are consistent with a mechanism in which O6-methylguanine binds to the active site of AGT with hydrogen bonds to the oxygen, the exocyclic amino group, and the nitrogen at the 1-position of the substrate.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Guanina/análogos & derivados , Metiltransferases/química , Oxigênio/química , Escherichia coli/enzimologia , Guanina/química , Cinética , Espectroscopia de Ressonância Magnética , Metilação , O(6)-Metilguanina-DNA Metiltransferase , Prótons
20.
Cancer Res ; 50(17): 5453-9, 1990 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-2201436

RESUMO

The tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) forms hemoglobin adducts in rats. Upon mild base treatment, 4-hydroxy-1-(3-pyridyl)-1-butanone (HPB) is released from this globin. HPB has been suggested as a dosimeter for exposure to and metabolic activation of tobacco-specific nitrosamines. The purpose of this study was 2-fold: (a) to determine whether cysteine adducts of NNK were precursors to HPB, and (b) to determine to what extent cysteine adducts accounted for the material bound to globin that is not released upon mild base hydrolysis. The chemistry of cysteine adduct formation was investigated by reacting N-acetyl-L-cysteine with three model compounds for pyridyloxobutylation by metabolically activated NNK: 4-(carbethoxynitrosamino)-1-(3-pyridyl)-1-butanone (1); 4-oxo-4-(3-pyridyl)-1-butylmethanesulfonate (2); and 4-iodo-1-(3-pyridyl)-1-butanone (3). Five adducts were isolated and characterized by their spectral properties and by independent syntheses: two diastereomers of N-acetyl-S-[1-methyl-3-oxo-3-(3-pyridyl)propyl]-L-cysteine (7a,b), N-acetyl-S-[4-oxo-4-(3-pyridyl)-1-butyl]-L-cysteine (9), and two diastereomers of N-acetyl-S-(2-[2-(3-pyridyl)]-2,3,4,5-tetrahydrofuranyl)-L-cystein e (11a,b). Only 11a,b produced HPB upon mild base treatment; however, the chemistry of this adduct did not support its role as a major precursor to HPB released upon base treatment of globin. The formation of adducts in rat hemoglobin was then examined by reacting it with tritium-labeled 1 [( 5-3H]1) or tritium-labeled 4-oxo-4-(3-pyridyl)-1-butyl p-toluenesulfonate [( 5-3H]4). The results demonstrated that the amino acids corresponding to 7a,b were present in hemoglobin reacted with [5-3H]1, accounting for 72% of the bound tritium. Amino acids corresponding to 9 were not detected in this globin. In contrast, hemoglobin reacted with [5-3H]4 contained the amino acid corresponding to 9 (15% of bound tritium), but not those corresponding to 7a,b. These results indicated that the alpha, beta-unsaturated ketone, 1-(3-pyridyl)-2-buten-1-one (5), played a major role in the hemoglobin binding of 1, but not of 4. Cysteine adducts were not detected in globin isolated from rats treated with [5-3H]NNK. The results of this study provide insights into the mechanisms of cysteine adduct formation in vitro by pryidyloxobutylating agents and indicate that these adducts are not formed in NNK-treated rats.


Assuntos
Carcinógenos/metabolismo , Cisteína , Hemoglobinas/metabolismo , Nitrosaminas/metabolismo , Animais , Biotransformação , Cromatografia Líquida de Alta Pressão , Espectroscopia de Ressonância Magnética , Masculino , Estrutura Molecular , Ligação Proteica , Técnica de Diluição de Radioisótopos , Ratos , Ratos Endogâmicos F344 , Relação Estrutura-Atividade , Trítio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...