Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Gut ; 59(1): 88-97, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19850960

RESUMEN

BACKGROUND AND AIMS: Hepatocellular carcinoma (HCC) frequently results from synergism between chemical and infectious liver carcinogens. Worldwide, the highest incidence of HCC is in regions endemic for the foodborne contaminant aflatoxin B1 (AFB1) and hepatitis B virus (HBV) infection. Recently, gut microbes have been implicated in multisystemic diseases including obesity and diabetes. Here, the hypothesis that specific intestinal bacteria promote liver cancer was tested in chemical and viral transgenic mouse models. METHODS: Helicobacter-free C3H/HeN mice were inoculated with AFB1 and/or Helicobacter hepaticus. The incidence, multiplicity and surface area of liver tumours were quantitated at 40 weeks. Molecular pathways involved in tumourigenesis were analysed by microarray, quantitative real-time PCR, liquid chromatography/mass spectrometry, ELISA, western blot and immunohistochemistry. In a separate experiment, C57BL/6 FL-N/35 mice harbouring a full-length hepatitis C virus (HCV) transgene were crossed with C3H/HeN mice and cancer rates compared between offspring with and without H hepaticus. RESULTS: Intestinal colonisation by H hepaticus was sufficient to promote aflatoxin- and HCV transgene-induced HCC. Neither bacterial translocation to the liver nor induction of hepatitis was necessary. From its preferred niche in the intestinal mucus layer, H hepaticus activated nuclear factor-kappaB (NF-kappaB)-regulated networks associated with innate and T helper 1 (Th1)-type adaptive immunity both in the lower bowel and liver. Biomarkers indicative of tumour progression included hepatocyte turnover, Wnt/beta-catenin activation and oxidative injury with decreased phagocytic clearance of damaged cells. CONCLUSIONS: Enteric microbiota define HCC risk in mice exposed to carcinogenic chemicals or hepatitis virus transgenes. These results have implications for human liver cancer risk assessment and prevention.


Asunto(s)
Aflatoxina B1/toxicidad , Hepatitis B/complicaciones , Intestinos/microbiología , Neoplasias Hepáticas Experimentales/etiología , Inmunidad Adaptativa , Animales , Proliferación Celular , Transformación Celular Neoplásica/genética , Quimiocinas/sangre , Cocarcinogénesis , Femenino , Infecciones por Helicobacter/complicaciones , Helicobacter hepaticus , Hepatitis B/inmunología , Inmunidad Innata , Subunidad p40 de la Interleucina-12/sangre , Neoplasias Hepáticas Experimentales/microbiología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Estrés Oxidativo/fisiología , Factores Sexuales , Transducción de Señal/fisiología , Células TH1/inmunología
2.
Bioorg Med Chem Lett ; 14(14): 3829-33, 2004 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-15203171

RESUMEN

A series of bifunctional compounds was prepared consisting of 17beta estradiol linked to a DNA damaging N,N-bis-(2-chloroethyl)aniline. The objective of our studies was to determine the characteristics of the linker that permitted both reaction with DNA and binding of the resultant covalent adducts to the estrogen receptor. Linker characteristics were pivotal determinants underlying the ability of the compounds to kill selectively breast cancer cells that express the estrogen receptor.


Asunto(s)
Compuestos de Anilina/uso terapéutico , Antineoplásicos Alquilantes/síntesis química , Neoplasias de la Mama/tratamiento farmacológico , Supervivencia Celular/efectos de los fármacos , Estradiol/uso terapéutico , Compuestos de Anilina/química , Compuestos de Anilina/farmacología , Mostaza de Anilina , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/uso terapéutico , Sitios de Unión , Neoplasias de la Mama/metabolismo , Aductos de ADN/metabolismo , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Estradiol/química , Estradiol/farmacología , Estudios de Evaluación como Asunto , Femenino , Humanos , Receptores de Estrógenos/metabolismo , Células Tumorales Cultivadas
3.
Biochemistry ; 40(49): 14968-75, 2001 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-11732917

RESUMEN

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


Asunto(s)
Proteínas Bacterianas/metabolismo , Reparación del ADN , Replicación del ADN , Proteínas de Escherichia coli , Escherichia coli/enzimología , Guanosina/análogos & derivados , Guanosina/metabolismo , Metiltransferasas , Oligodesoxirribonucleótidos/metabolismo , Secuencia de Bases , Escherichia coli/genética , Genes Virales , Mutación , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Oligodesoxirribonucleótidos/genética , Factores de Transcripción
4.
Biochemistry ; 40(22): 6653-9, 2001 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-11380260

RESUMEN

The human endonuclease III homologue (hNTH1) removes premutagenic cytosine damage from DNA. This includes 5-hydroxycytosine, which has increased potential for pairing with adenine, resulting in C --> T transition mutations. Here we report that hNTH1 acts on both 5-hydroxycytosine and abasic sites preferentially when these are situated opposite guanines in DNA. Discrimination against other opposite bases is strongly dependent on the presence of magnesium. To further elucidate this effect, we have introduced mutations in the helix-hairpin-helix domain of hNTH1 (K212S, P211R, +G212, and DeltaP211), and measured the kinetics of 5-hydroxycytosine removal of the mutants relative to wild type. The K212S and DeltaP211 (truncated hairpin) mutant proteins were both inactive, whereas the extended hairpin in the +G212 mutant diminished recognition and binding to 5-hydroxycytosine-containing DNA. The P211R mutant resembled native hNTH1, except for decreased specificity of binding. Despite the altered kinetic parameters, the active mutants retained the ability to discriminate against the pairing base, indicating that enzyme interactions with the opposite strand relies on other domains than the active site helix-hairpin-helix motif.


Asunto(s)
Citosina/análogos & derivados , Daño del ADN , Reparación del ADN , ADN/química , Desoxirribonucleasa (Dímero de Pirimidina) , Endodesoxirribonucleasas/química , Proteínas de Escherichia coli , Guanina/química , Secuencia de Aminoácidos , Disparidad de Par Base , Tampones (Química) , Liasas de Carbono-Oxígeno/química , Liasas de Carbono-Oxígeno/metabolismo , Citosina/química , ADN/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa , ADN-Formamidopirimidina Glicosilasa , Desoxirribonucleasa IV (Fago T4-Inducido) , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/metabolismo , Guanina/metabolismo , Secuencias Hélice-Giro-Hélice/genética , Humanos , Cinética , Datos de Secuencia Molecular , N-Glicosil Hidrolasas/genética , N-Glicosil Hidrolasas/metabolismo , Homología de Secuencia de Ácido Nucleico , Especificidad por Sustrato/genética
5.
Mutat Res ; 478(1-2): 1-21, 2001 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-11406166

RESUMEN

Cisplatin is a widely used chemotherapeutic agent. It reacts with nucleophilic bases in DNA and forms 1,2-d(ApG), 1,2-d(GpG) and 1,3-d(GpTpG) intrastrand crosslinks, interstrand crosslinks and monofunctional adducts. The presence of these adducts in DNA is through to be responsible for the therapeutic efficacy of cisplatin. The exact signal transduction pathway that leads to cell cycle arrest and cell death following treatment with the drug is not known but cell death is believed to be mediated by the recognition of the adducts by cellular proteins. Here we describe the structural information available for cisplatin and related platinum adducts, the interactions of the adducts with cellular proteins and the implications of these interactions for cell survival.


Asunto(s)
Antígenos Nucleares , Antineoplásicos/farmacología , Cisplatino/farmacología , Aductos de ADN/efectos de los fármacos , ADN Helicasas , Proteínas/metabolismo , Animales , Antineoplásicos/química , Cisplatino/química , Aductos de ADN/química , Aductos de ADN/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Desoxirribodipirimidina Fotoliasa/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Humanos , Autoantígeno Ku , Proteínas Nucleares/metabolismo , Unión Proteica
6.
Mutat Res ; 478(1-2): 23-43, 2001 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-11406167

RESUMEN

The use of cisplatin in cancer chemotherapy is limited by acquired or intrinsic resistance of cells to the drug. Cisplatin enters the cells and its chloride ligands are replaced by water, forming aquated species that react with nucleophilic sites in cellular macromolecules. The presence of the cisplatin adducts in DNA is thought to trigger cell cycle arrest and apoptosis. Knowledge of the mechanism of action of cisplatin has improved our understanding of resistance. Decreased intracellular concentration due to decreased drug uptake, increased reflux or increased inactivation by sulfhydryl molecules such as glutathione can cause resistance to cisplatin. Increased excision of the adducts from DNA by repair pathways or increased lesion bypass can also result in resistance. Finally, altered expression of regulatory proteins involved in signal transduction pathways that control the apoptotic pathway can also affect sensitivity to the drug. An improved understanding of the mechanisms of resistance operative in vivo has identified targets for intervention and may increase the utility of cisplatin for the treatment of cancer.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Resistencia a Antineoplásicos , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Cisplatino/química , Aductos de ADN/química , Aductos de ADN/efectos de los fármacos , Reparación del ADN , Humanos
7.
Carcinogenesis ; 22(4): 535-45, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11285186

RESUMEN

Dietary exposure to aflatoxin B(1) (AFB(1)) is associated with an increased incidence of hepatocellular carcinoma (HCC), especially in populations in which exposure to hepatitis B virus (HBV) is a common occurrence. Most HCC samples from people living where HBV is prevalent have one striking mutational hotspot: a GC-->TA transversion at the third position of codon 249 of the p53 gene. In this review, the chemical reaction of an electrophilic derivative of aflatoxin with specific DNA sequences is examined, along with the types of mutations caused by AFB(1) and the sequence context dependence of those mutations. An attempt is made to assign the source of these mutations to specific chemical forms of AFB(1)-DNA damage. In addition, epidemiological and experimental data are examined regarding the synergistic effects of AFB(1) and HBV on HCC formation and the predominance of one hotspot GC-->TA transversion in the p53 gene of affected individuals.


Asunto(s)
Aflatoxina B1/química , Aflatoxina B1/metabolismo , Carcinoma Hepatocelular/inducido químicamente , Neoplasias Hepáticas/inducido químicamente , Mutación , Animales , Carcinógenos , Carcinoma Hepatocelular/genética , Peces , Genes p53/genética , Virus de la Hepatitis B/metabolismo , Humanos , Neoplasias Hepáticas/genética , Modelos Biológicos , Modelos Químicos , Mutágenos , Ratas , Especificidad de la Especie
9.
Biochemistry ; 39(27): 8032-8, 2000 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-10891085

RESUMEN

The human 3-methyladenine DNA glycosylase (AAG) is a repair enzyme that removes a number of damaged bases from DNA, including adducts formed by some chemotherapeutic agents. Cisplatin is one of the most widely used anticancer drugs. Its success in killing tumor cells results from its ability to form DNA adducts and the cellular processes triggered by the presence of those adducts in DNA. Variations in tumor response to cisplatin may result from altered expression of cellular proteins that recognize cisplatin adducts. The present study focuses on the interaction between the cisplatin intrastrand cross-links and human AAG. Using site-specifically modified oligonucleotides containing each of the cisplatin intrastrand cross-links, we found that AAG readily recognized cisplatin adducts. The apparent dissociation constants for the 1, 2-d(GpG), the 1,2-d(ApG), and the 1,3-d(GpTpG) oligonucleotides were 115 nM, 71 nM, and 144 nM, respectively. For comparison, the apparent dissociation constant for an oligonucleotide containing a single 1,N(6)-ethenoadenine (epsilonA), which is repaired efficiently by AAG, was 26 nM. Despite the affinity of AAG for cisplatin adducts, AAG was not able to release any of these adducts from DNA. Furthermore, it was demonstrated that the presence of cisplatin adducts in the reactions inhibited the excision of epsilonA by AAG. These data suggest a previously unexplored dimension to the toxicological response of cells to cisplatin. We suggest that cisplatin adducts could titrate AAG away from its natural substrates, resulting in higher mutagenesis and/or cell death because of the persistence of AAG substrates in DNA.


Asunto(s)
Adenina/análogos & derivados , Cisplatino/farmacología , Aductos de ADN/farmacología , ADN Glicosilasas , Reparación del ADN/efectos de los fármacos , N-Glicosil Hidrolasas/metabolismo , Adenina/metabolismo , Secuencia de Bases , Cisplatino/metabolismo , Aductos de ADN/metabolismo , Humanos , Unión Proteica
10.
J Biol Chem ; 275(34): 26632-6, 2000 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-10882712

RESUMEN

Interstrand DNA cross-link damage is a severe challenge to genomic integrity. Nucleotide excision repair plays some role in the repair of DNA cross-links caused by psoralens and other agents. However, in mammalian cells there is evidence that the ERCC1-XPF nuclease has a specialized additional function during interstrand DNA cross-link repair, beyond its role in nucleotide excision repair. We placed a psoralen monoadduct or interstrand cross-link in a duplex, 4-6 bases from a junction with unpaired DNA. ERCC1-XPF endonucleolytically cleaved within the duplex on either side of the adduct, on the strand having an unpaired 3' tail. Cross-links that were cleaved only on the 5' side were purified and reincubated with ERCC1-XPF. A second cleavage was then observed on the 3' side. Relevant partially unwound structures near a cross-link may be expected to arise frequently, for example at stalled DNA replication forks. The results show that the single enzyme ERCC1-XPF can release one arm of a cross-link and suggest a novel mechanism for interstrand cross-link repair.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Endonucleasas , Conformación de Ácido Nucleico , Proteínas/metabolismo , Secuencia de Bases , Reactivos de Enlaces Cruzados/farmacología , Reparación del ADN , Ficusina/farmacología , Datos de Secuencia Molecular , Oligonucleótidos/metabolismo
11.
Chem Biol ; 7(1): 39-50, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10662689

RESUMEN

BACKGROUND: Cisplatin is a DNA-damaging drug used for treatment of testicular tumors. The toxicity of cisplatin probably results from its ability to form DNA adducts that inhibit polymerases. Blocked replication represents a particular challenge for tumor cells, which are committed to unremitting division. Recombination provides a mechanism by which replication can proceed despite the presence of lesions and therefore could be significant for managing cisplatin toxicity. RESULTS: Recombination-deficient Escherichia coli mutants were strikingly sensitive to cisplatin when compared with the parental strain. Our data identified both daughter-strand gap and double-strand break recombination pathways as critical for survival following treatment with cisplatin. Although it is established that nucleotide excision repair (NER) significantly protects against cisplatin toxicity, most recombination-deficient strains were as sensitive to the drug as the NER-deficient uvrA mutant. Recombination/NER deficient double mutants were more sensitive to cisplatin than the corresponding single mutants, suggesting that recombination and NER pathways play independent roles in countering cisplatin toxicity. Cisplatin was a potent recombinogen in comparison with the trans isomer and canonical alkylating agents. Mitomycin C, which like cisplatin, forms DNA cross-links, was also recombinogenic at minimally toxic doses. CONCLUSIONS: We have demonstrated that all of the major recombination pathways are critical for E. coli survival following treatment with cisplatin. Moreover, recombination pathways act independently of NER and are of equal importance to NER as genoprotective systems against cisplatin toxicity. Taken together, these results shed new light on how cells survive and succumb to this widely used anticancer drug.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Recombinación Genética/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Reactivos de Enlaces Cruzados/farmacología , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Reparación del ADN/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Genotipo , Mutación/fisiología , Recombinación Genética/genética
12.
Chem Biol ; 6(10): 743-53, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10508678

RESUMEN

BACKGROUND: Detailed analyses of mutational hotspots following DNA damage provide an understanding of oncogene activation and tumor suppressor gene inactivation, and hence provide an insight into the earliest steps in the induction of cancer. A mutational hotspot might be created by preferential lesion formation, decreased lesion repair, or increased misinsertion past the lesion during DNA replication. The respective contribution of these factors might be influenced by the DNA sequence context of the hotspot. RESULTS: As a prelude to addressing the contribution of all possible nearest-neighbor contexts on the replication past O6-methylguanine (m6G) and repair of m6G in vivo, we have devised a mutation frequency (MF) detection strategy on the basis of the properties of type IIs restriction enzymes. We also report a method for constructing site-specific single-stranded viral DNA genomes that should yield identical ligation efficiencies regardless of the lesion or its surrounding sequence context. Using repair-deficient Escherichia coli, we discovered that m6G in three sequence contexts was nearly 100% mutagenic in vivo, showing that the DNA polymerase holoenzyme almost always placed a thymine base opposite m6G during replication. In partially repair-proficient cells, the Ada O6-methylguanine-DNA methyltransferase repair protein was twice as efficient on m6G when a guanine base rather than an adenine base was 5' to the lesion. CONCLUSIONS: The system allows the mutagenic potential of, theoretically, any DNA lesion that exhibits point mutations, in any varied local sequence context, to be rapidly determined. The assay demonstrates low background, high throughput, and does not require phenotypic selection, making it possible to discern the effects of sequence context on the processing of m6G.


Asunto(s)
Guanina/análogos & derivados , Mutagénesis , Análisis Mutacional de ADN , Reparación del ADN , ADN de Cadena Simple/síntesis química , ADN Viral/síntesis química , Electroporación , Escherichia coli , Guanina/química , Mutación Puntual
13.
Biochemistry ; 38(19): 6204-12, 1999 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-10320349

RESUMEN

The anticancer drug cisplatin induces a spectrum of lesions in DNA. The effect of such DNA damage on transcription by RNA polymerase II (RNA pol II) in human cell extracts was investigated at the level of initiation and elongation. RNA pol II transcription directed from the adenovirus major late promoter was inhibited following treatment of the promoter-containing template with increasing concentrations of cisplatin. Furthermore, transcription from an undamaged promoter fragment was depleted in the presence of increasing amounts of cisplatin DNA damage on an exogenous plasmid, suggesting such damage may hijack an essential factor for transcription initiation. The effect of cisplatin damage on RNA pol II elongation was investigated using site-specifically-placed cisplatin adducts. The GTG adduct was an effective block to RNA pol II elongation, inhibiting the polymerase by 80%. In contrast, RNA pol II completely bypassed the cisplatin GG intrastrand adduct. These studies suggest that the inhibition of RNA pol II transcription observed following the treatment of cells with cisplatin is likely to reflect the combined effects of DNA damage at the level of both transcription initiation and elongation.


Asunto(s)
Cisplatino/farmacología , Daño del ADN , ARN Polimerasa II/metabolismo , Transcripción Genética , Adenoviridae/efectos de los fármacos , Adenoviridae/genética , Extractos Celulares , Reactivos de Enlaces Cruzados/farmacología , Aductos de ADN , Daño del ADN/genética , Humanos , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , ARN Polimerasa II/antagonistas & inhibidores , Moldes Genéticos , Transcripción Genética/efectos de los fármacos
14.
Nucleic Acids Res ; 27(11): 2256-64, 1999 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-10325412

RESUMEN

Schizosaccharomyces pombe ultraviolet DNA endonuclease (UVDE or Uve1p) has been shown to cleave 5' to UV light-induced cyclobutane pyrimidine dimers (CPDs) and pyrimidine-pyrimidone (6-4) photoproducts (6-4PP). This endonuclease is believed to function in the initial step in an alternative excision repair pathway for the removal of DNA damage caused by exposure to UV light. An active truncated form of this protein, Delta228-Uve1p, has been successfully overexpressed, affinity purified and partially characterized. In the present study we present data from a detailed substrate specificity trial. We have determined that the substrate range of Uve1p is much greater than was originally believed. We demonstrate that this DNA damage repair protein is capable of recognizing an array of UV-induced DNA photoproducts (cis-syn-, trans-syn I- and trans-syn II CPDs, 6-4PP and Dewar isomers) that cause varying degrees of distortion in a duplex DNA molecule. We also demonstrate that Uve1p recognizes non-UV-induced DNA damage, such as platinum-DNA GG diadducts, uracil, dihydrouracil and abasic sites. This is the first time that a single DNA repair endonuclease with the ability to recognize such a diverse range of lesions has been described. This study suggests that Uve1p and the alternative excision repair pathway may participate broadly in the repair of DNA damage.


Asunto(s)
Reparación del ADN , Endodesoxirribonucleasas/metabolismo , Dímeros de Pirimidina/metabolismo , Proteínas de Schizosaccharomyces pombe , Schizosaccharomyces/enzimología , Aductos de ADN/efectos de la radiación , Endodesoxirribonucleasas/genética , Conformación de Ácido Nucleico , Oligodesoxirribonucleótidos/metabolismo , Platino (Metal) , Dímeros de Pirimidina/efectos de la radiación , Pirimidinas/efectos de la radiación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Especificidad por Sustrato , Rayos Ultravioleta
15.
J Biol Chem ; 274(13): 8516-23, 1999 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-10085084

RESUMEN

We recently showed that abasic sites, uracil mismatches, nicks, and gaps can trap DNA topoisomerase I (top1) when these lesions are introduced in the vicinity of a top1 cleavage site (Pourquier, P., Ueng, L.-M., Kohlhagen, G., Mazumder, A., Gupta, M., Kohn, K. W., and Pommier, Y. (1997) J. Biol. Chem. 272, 7792-7796; Pourquier, P., Pilon, A. A., Kohlhagen, G., Mazumder, A., Sharma, A., and Pommier, Y. (1997) J. Biol. Chem. 26441-26447). In this study, we investigated the effects on top1 of an abundant base damage generated by various oxidative stresses: 7,8-dihydro-8-oxoguanine (8-oxoG). Using purified eukaryotic top1 and oligonucleotides containing the 8-oxoG modification, we found a 3-7-fold increase in top1-mediated DNA cleavage when 8-oxoG was present at the +1 or +2 position relative to the cleavage site. Another oxidative lesion, 5-hydroxycytosine, also enhanced top1 cleavage by 2-fold when incorporated at the +1 position of the scissile strand. 8-oxoG at the +1 position enhanced noncovalent top1 DNA binding and had no detectable effect on DNA religation or on the incision step. top1 trapping by 8-oxoG was markedly enhanced when asparagine adjacent to the catalytic tyrosine was mutated to histidine, suggesting a direct interaction between this residue and the DNA major groove immediately downstream from the top1 cleavage site. Altogether, these results demonstrate that oxidative base lesions can increase top1 binding to DNA and induce top1 cleavage complexes.


Asunto(s)
Citosina/análogos & derivados , Daño del ADN/genética , ADN-Topoisomerasas de Tipo I/metabolismo , Guanosina/análogos & derivados , Camptotecina/farmacología , Citosina/farmacología , ADN/metabolismo , ADN-Topoisomerasas de Tipo I/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Guanosina/farmacología , Humanos , Cinética , Estructura Molecular , Mutación/genética , Oligonucleótidos/metabolismo , Estrés Oxidativo
16.
Proc Natl Acad Sci U S A ; 96(4): 1492-7, 1999 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-9990051

RESUMEN

The human immunodeficiency virus (HIV) replicates its genome and mutates at exceptionally high rates. As a result, the virus is able to evade immunological and chemical antiviral agents. We tested the hypothesis that a further increase in the mutation rate by promutagenic nucleoside analogs would abolish viral replication. We evaluated deoxynucleoside analogs for lack of toxicity to human cells, incorporation by HIV reverse transcriptase, resistance to repair when incorporated into the DNA strand of an RNA.DNA hybrid, and mispairing at high frequency. Among the candidates tested, 5-hydroxydeoxycytidine (5-OH-dC) fulfilled these criteria. In seven of nine experiments, the presence of this analog resulted in the loss of viral replicative potential after 9-24 sequential passages of HIV in human CEM cells. In contrast, loss of viral replication was not observed in 28 control cultures passaged in the absence of the nucleoside analog, nor with other analogs tested. Sequence analysis of a portion of the HIV reverse transcriptase gene demonstrated a disproportionate increase in G --> A substitutions, mutations predicted to result from misincorporation of 5-OH-dC into the cDNA during reverse transcription. Thus, "lethal mutagenesis" driven by the class of deoxynucleoside analogs represented by 5-OH-dC could provide a new approach to treating HIV infections and, potentially, other viral infections.


Asunto(s)
Antimetabolitos/farmacología , Antivirales/farmacología , VIH-1/genética , Mutagénesis , Nucleósidos/farmacología , Antimetabolitos/farmacocinética , Antivirales/farmacocinética , Línea Celular , Supervivencia Celular/efectos de los fármacos , Transcriptasa Inversa del VIH/metabolismo , VIH-1/efectos de los fármacos , Humanos , Hibridación de Ácido Nucleico , Nucleósidos/farmacocinética , ARN Viral/efectos de los fármacos , ARN Viral/genética
17.
Essays Biochem ; 34: 191-211, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10730196

RESUMEN

Cisplatin is a widely used anti-cancer drug that is exceptionally effective against testicular cancer. trans-DDP, the geometric isomer of cisplatin, is ineffective as a chemotherapeutic agent. The anti-tumour activity of cisplatin is generally attributed to its formation of DNA adducts, both intrastrand and interstrand crosslinks, which induce structural distortions in DNA. The DNA adducts of cisplatin are thought to mediate its cytotoxic effects by inhibiting DNA replication and transcription and, ultimately, by inducing programmed cell death, or apoptosis. The adducts of both cis- and trans-DDP are removed from DNA by the nucleotide-excision-repair pathway. Cellular proteins possessing certain DNA-binding motifs, including the HMG domain, bind selectively to DNA modified by cisplatin, but not to DNA adducts of trans-DDP; evidence suggests a possible role for these proteins in modulating cisplatin cytotoxicity. Both intrinsic and drug-induced resistance often limit the success of cisplatin; several specific mechanisms of cisplatin resistance have been identified.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Antineoplásicos/química , Antineoplásicos/metabolismo , Cisplatino/química , Cisplatino/metabolismo , Aductos de ADN/química , Aductos de ADN/metabolismo , Reparación del ADN , Replicación del ADN/efectos de los fármacos , Resistencia a Medicamentos , Humanos , Masculino , Modelos Moleculares , Proteínas Nucleares/metabolismo , Neoplasias Testiculares/tratamiento farmacológico , Transcripción Genética/efectos de los fármacos
18.
Biochemistry ; 37(46): 16307-15, 1998 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-9819223

RESUMEN

Several eukaryotic cellular proteins recognize DNA modified by the anticancer drug cisplatin (cis-diamminedichloroplatinum(II) or cis-DDP); among these proteins is a class of DNA-binding molecules containing the HMG (high-mobility group) box DNA recognition motif. We have previously reported the extraordinarily high binding activity to cisplatin adducts by human upstream binding factor (hUBF), an HMG box containing transcription factor that stimulates ribosomal RNA synthesis (Treiber et al. (1994) Proc. Natl. Acad. Sci. U.S.A. 91, 5672-5676). In the present study, we discovered that (1) hUBF interacted selectively with DNA lesions formed by therapeutically effective platinum compounds [Pt(en)Cl2] and [Pt(dach)Cl2], in addition to the lesions formed by cis-DDP, suggesting a possible association with their anticancer effect; (2) multiple HMG boxes contributed additively to the hUBF-adduct interaction, providing a possible explanation for the unusually high affinity of hUBF for cis-DDP adducts as compared to the lower affinities of other HMG box proteins; and (3) ribosomal RNA transcription in a reconstituted system is specifically inhibited in the presence of cis-DDP adducts. In this third experiment, a ratio of adducts/promoter of approximately 4:1 completely abolished the transcription activated by hUBF. Taken together, these data lend support to the view that transcription factors involved in cellular growth regulation, such as ribosomal RNA transcription, may be hijacked by cis-DDP adducts resulting in functional inhibition.


Asunto(s)
Cisplatino/farmacología , Aductos de ADN/farmacología , Proteínas de Unión al ADN/metabolismo , Proteínas del Complejo de Iniciación de Transcripción Pol1 , ARN/antagonistas & inhibidores , ARN/biosíntesis , Ribosomas/efectos de los fármacos , Factores de Transcripción/metabolismo , Antineoplásicos/metabolismo , Cisplatino/análogos & derivados , Cisplatino/metabolismo , ADN/farmacología , Etilenodiaminas/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Humanos , Compuestos Organoplatinos/metabolismo , Unión Proteica/efectos de los fármacos
19.
Mutat Res ; 400(1-2): 99-115, 1998 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-9685598

RESUMEN

Oxidative DNA damage has been implicated in mutagenesis, carcinogenesis and aging. Endogenous cellular processes such as aerobic metabolism generate reactive oxygen species (ROS) that interact with DNA to form dozens of DNA lesions. If unrepaired, these lesions can exert a number of deleterious effects including the induction of mutations. In an effort to understand the genetic consequences of cellular oxidative damage, many laboratories have determined the patterns of mutations generated by the interaction of ROS with DNA. Compilation of these mutational spectra has revealed that GC-->AT transitions and GC-->TA transversions are the most commonly observed mutations resulting from oxidative damage to DNA. Since mutational spectra convey only the end result of a complex cascade of events, which includes formation of multiple adducts, repair processing, and polymerase errors, it is difficult if not impossible to assess the mutational specificity of individual DNA lesions directly from these spectra. This problem is especially complicated in the case of oxidative DNA damage owing to the multiplicity of lesions formed by a single damaging agent. The task of assigning specific features of mutational spectra to individual DNA lesions has been made possible with the advent of a technology to analyze the mutational properties of single defined adducts, in vitro and in vivo. At the same time, parallel progress in the discovery and cloning of repair enzymes has advanced understanding of the biochemical mechanisms by which cells excise DNA damage. This combination of tools has brought our understanding of DNA lesions to a new level of sophistication. In this review, we summarize the known properties of individual oxidative lesions in terms of their structure, mutagenicity and repairability.


Asunto(s)
Daño del ADN/genética , Reparación del ADN/genética , Mutagénesis Sitio-Dirigida/genética , Animales , Aductos de ADN/genética , Análisis Mutacional de ADN , Humanos , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo
20.
Proc Natl Acad Sci U S A ; 95(7): 3578-82, 1998 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-9520408

RESUMEN

The most common base substitution arising from oxidative damage of DNA is a GC --> AT transition. In an effort to determine the oxidized lesion(s) that gives rise to this mutation, the mutagenicity of three oxidized cytosines, 5-hydroxycytosine, 5-hydroxyuracil, and uracil glycol, were investigated in Escherichia coli. An M13 viral genome was constructed to contain a single oxidized cytosine at a specific site. Replication in vivo of the single-stranded genomes yielded mutation frequencies of 0.05%, 83%, and 80% for 5-hydroxycytosine, 5-hydroxyuracil, and uracil glycol, respectively. The predominant mutation observed was C --> T. A model for C --> T oxidative mutagenesis is suggested in which initial cytosine oxidation is followed by deamination to a poorly repaired uracil derivative that is strongly miscoding during replication.


Asunto(s)
Citosina , Daño del ADN , Mutación , Estrés Oxidativo , Replicación del ADN , ADN Viral/genética , ADN Viral/metabolismo , Escherichia coli , Genoma Viral , Mutagénesis , Oxidación-Reducción , Estrés Oxidativo/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...