Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Environ Mol Mutagen ; 60(9): 792-806, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31374128

RESUMEN

Aristolochic acids (AAs) are human nephrotoxins and carcinogens found in concoctions of Aristolochia plants used in traditional medicinal practices worldwide. Genotoxicity of AAs is associated with the formation of active species catalyzed by metabolic enzymes, the full repertoire of which is unknown. Recently, we provided evidence that sulfonation is important for bioactivation of AAs. Here, we employ Salmonella typhimurium umu tester strains expressing human N-acetyltransferases (NATs) and sulfotransferases (SULTs), to study the role of conjugation reactions in the genotoxicities of N-hydroxyaristolactams (AL-I-NOH and AL-II-NOH), metabolites of AA-I and AA-II. Both N-hydroxyaristolactams show stronger genotoxic effects in umu strains expressing human NAT1 and NAT2, than in the parent strain. Additionally, AL-I-NOH displays increased genotoxicity in strains expressing human SULT1A1 and SULT1A2, whereas AL-II-NOH shows enhanced genotoxicity in SULT1A1/2 and SULT1A3 strains. 2,6-Dichloro-4-nitrophenol, SULTs inhibitor, reduced umuC gene expression induced by N-hydroxyaristolactams in SULT1A2 strain. N-hydroxyaristolactams are also mutagenic in parent strains, suggesting that an additional mechanism(s) may contribute to their genotoxicities. Accordingly, using putative SULT substrates and inhibitors, we found that cytosols obtained from human kidney HK-2 cells activate N-hydroxyaristolactams in aristolactam-DNA adducts with the limited involvement of SULTs. Removal of low-molecular-weight reactants in the 3.5-10 kDa range inhibits the formation of aristolactam-DNA by 500-fold, which could not be prevented by the addition of cofactors for SULTs and NATs. In conclusion, our results demonstrate that the genotoxicities of N-hydroxyaristolactams depend on the cell type and involve not only sulfonation but also N,O-acetyltransfer and an additional yet unknown mechanism(s). Environ. Mol. Mutagen. 2019. © 2019 Wiley Periodicals, Inc.


Asunto(s)
Ácidos Aristolóquicos/metabolismo , Ácidos Aristolóquicos/toxicidad , Acetiltransferasas/metabolismo , Arilamina N-Acetiltransferasa/metabolismo , Arilsulfotransferasa/metabolismo , Carcinógenos/toxicidad , Línea Celular , ADN/efectos de los fármacos , Aductos de ADN/genética , Humanos , Mutágenos/toxicidad , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/metabolismo , Sulfotransferasas/metabolismo
2.
Carcinogenesis ; 37(7): 647-655, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27207664

RESUMEN

Aristolochic acids (AA) are implicated in the development of chronic renal disease and upper urinary tract carcinoma in humans. Using in vitro approaches, we demonstrated that N-hydroxyaristolactams, metabolites derived from partial nitroreduction of AA, require sulfotransferase (SULT)-catalyzed conjugation with a sulfonyl group to form aristolactam-DNA adducts. Following up on this observation, bioactivation of AA-I and N-hydroxyaristolactam I (AL-I-NOH) was studied in human kidney (HK-2) and skin fibroblast (GM00637) cell lines. Pentachlorophenol, a known SULT inhibitor, significantly reduced cell death and aristolactam-DNA adduct levels in HK-2 cells following exposure to AA-I and AL-I-NOH, suggesting a role for Phase II metabolism in AA activation. A gene knockdown, siRNA approach was employed to establish the involvement of selected SULTs and nitroreductases in AA-I bioactivation. Silencing of SULT1A1 and PAPSS2 led to a significant decrease in aristolactam-DNA levels in both cell lines following exposure to AA-I, indicating the critical role for sulfonation in the activation of AA-I in vivo Since HK-2 cells proved relatively resistant to knockdown with siRNAs, gene silencing of xanthine oxidoreductase, cytochrome P450 oxidoreductase and NADPH:quinone oxidoreductase was conducted in GM00637 cells, showing a significant increase, decrease and no effect on aristolactam-DNA levels, respectively. In GM00637 cells exposed to AL-I-NOH, suppressing the SULT pathway led to a significant decrease in aristolactam-DNA formation, mirroring data obtained for AA-I. We conclude from these studies that SULT1A1 is involved in the bioactivation of AA-I through the sulfonation of AL-I-NOH, contributing significantly to the toxicities of AA observed in vivo.


Asunto(s)
Ácidos Aristolóquicos/metabolismo , Arilsulfotransferasa/genética , Complejos Multienzimáticos/genética , Sulfato Adenililtransferasa/genética , Arilsulfotransferasa/antagonistas & inhibidores , Arilsulfotransferasa/metabolismo , Carcinógenos/metabolismo , Carcinógenos/toxicidad , ADN/genética , ADN/metabolismo , Fibroblastos/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Riñón/metabolismo , Riñón/patología , Complejos Multienzimáticos/metabolismo , NADPH-Ferrihemoproteína Reductasa/metabolismo , Pentaclorofenol/farmacología , ARN Interferente Pequeño , Sulfato Adenililtransferasa/metabolismo , Xantina Deshidrogenasa/metabolismo
3.
Chem Res Toxicol ; 27(7): 1236-42, 2014 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-24877584

RESUMEN

Plants from the Aristolochia genus have been recommended for the treatment of a variety of human ailments since the time of Hippocrates. However, many species produce the highly toxic aristolochic acids (AAs), which are both nephrotoxic and carcinogenic. For the purposes of extensive biological studies, a versatile approach to the synthesis of the AAs and their major metabolites was devised based primarily on a Suzuki-Miyaura coupling reaction. The key to success lies in the preparation of a common ring-A precursor, namely, the tetrahydropyranyl ether of 2-nitromethyl-3-iodo-4,5-methylendioxybenzyl alcohol (27), which was generated in excellent yield by oxidation of the aldoxime precursor 26. Suzuki-Miyaura coupling of 27 with a variety of benzaldehyde 2-boronates was accompanied by an aldol condensation/elimination reaction to give the desired phenanthrene intermediate directly. Deprotection of the benzyl alcohol followed by two sequential oxidation steps gave the desired phenanthrene nitrocarboxylic acids. This approach was used to synthesize AAs I-IV and several other related compounds, including AA I and AA II bearing an aminopropyloxy group at position-6, which were required for further conversion to fluorescent biological probes. Further successful application of the Suzuki-Miyaura coupling reaction to the synthesis of the N-hydroxyaristolactams of AA I and AA II then allowed the synthesis of the putative, but until now elusive, N-acetoxy- and N-sulfonyloxy-aristolactam metabolites.


Asunto(s)
Ácidos Aristolóquicos/síntesis química , Ácidos Aristolóquicos/química , Aductos de ADN , Lactamas/síntesis química
4.
Carcinogenesis ; 35(8): 1814-22, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24743514

RESUMEN

Aristolochic acids are potent human carcinogens; the role of phase II metabolism in their bioactivation is unclear. Accordingly, we tested the ability of the partially reduced metabolites, N-hydroxyaristolactams (AL-NOHs), and their N-O-sulfonated and N-O-acetylated derivatives to react with DNA to form aristolactam-DNA adducts. AL-NOHs displayed little or no activity in this regard while the sulfo- and acetyl compounds readily form DNA adducts, as detected by (32)P-post-labeling analysis. Mouse hepatic and renal cytosols stimulated binding of AL-NOHs to DNA in the presence of adenosine 3'-phosphate 5'-phosphosulfate (PAPS) but not of acetyl-CoA. Using Time of Flight liquid chromatography/mass spectrometry, N-hydroxyaristolactam I formed the sulfated compound in the presence of PAPS and certain human sulfotransferases, SULT1B1 >>> SULT1A2 > SULT1A1 >>> SULT1A3. The same pattern of SULT reactivity was observed when N-hydroxyaristolactam I was incubated with these enzymes and PAPS and the reaction was monitored by formation of aristolactam-DNA adducts. In the presence of human NAD(P)H: quinone oxidoreductase, the ability of aristolochic acid I to bind DNA covalently was increased significantly by addition of PAPS and SULT1B1. We conclude from these studies that AL-NOHs, formed following partial nitroreduction of aristolochic acids, serve as substrates for SULT1B1, producing N-sulfated esters, which, in turn, are converted to highly active species that react with DNA and, potentially, cellular proteins, resulting in the genotoxicity and nephrotoxicity associated with ingestion of aristolochic acids by humans.


Asunto(s)
Ácidos Aristolóquicos/farmacología , Carcinógenos/farmacología , Aductos de ADN/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Animales , Arilsulfotransferasa/metabolismo , Western Blotting , Proliferación Celular , Células Cultivadas , Citosol/metabolismo , Aductos de ADN/metabolismo , Etanolaminas , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Riñón/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C3H , Modelos Moleculares , Estructura Molecular , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Oxidorreductasas/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Ácidos Esteáricos , Sulfotransferasas/metabolismo
5.
Chem Res Toxicol ; 25(11): 2423-31, 2012 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-22897814

RESUMEN

The addition of hydroxyl radicals to the C8 position of guanine can lead to the formation of a 2,6-diamino-4-hydroxy-5-formamido-2'-deoxypyrimidine (Fapy-dG) lesion, whose endogenous levels in cellular DNA rival those of 8-oxo-7,8-dihydroxy-2'-deoxyguanosine. Despite its prevalence, the structure of duplex DNA containing Fapy-dG is unknown. We have prepared an undecameric duplex containing a centrally located ß-cFapy-dG residue paired to dC and determined its solution structure by high-resolution NMR spectroscopy and restrained molecular dynamic simulations. The damaged duplex adopts a right-handed helical structure with all residues in an anti conformation, forming Watson-Crick base pair alignments, and 2-deoxyribose conformations in the C2'-endo/C1'-exo range. The formamido group of Fapy rotates out of the pyrimidine plane and is present in the Z and E configurations that equilibrate with an approximate 2:1 population ratio. The two isomeric duplexes show similar lesion-induced deviations from a canonical B-from DNA conformation that are minor and limited to the central three-base-pair segment of the duplex, affecting the stacking interactions with the 5-lesion-neighboring residue. We discuss the implications of our observations for translesion synthesis during DNA replication and the recognition of Fapy-dG by DNA glycosylases.


Asunto(s)
ADN/química , Desoxiguanosina/química , Modelos Moleculares , Simulación de Dinámica Molecular , Resonancia Magnética Nuclear Biomolecular , Conformación de Ácido Nucleico , Soluciones
6.
Nucleic Acids Res ; 39(13): 5776-89, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21415012

RESUMEN

Accumulation of damaged guanine nucleobases within genomic DNA, including the imidazole ring opened N(6)-(2-Deoxy-α,ß-D-erythro-pentafuranosyl)-2,6-diamino-4-hydroxy-5-formylamidopyrimidine (Fapy-dG), is associated with progression of age-related diseases and cancer. To evaluate the impact of this mutagenic lesion on DNA structure and energetics, we have developed a novel synthetic strategy to incorporate cognate Fapy-dG site-specifically within any oligodeoxynucleotide sequence. The scheme involves the synthesis of an oligonucleotide precursor containing a 5-nitropyrimidine moiety at the desired lesion site via standard solid-phase procedures. Following deprotection and isolation, the Fapy-dG lesion is generated by catalytic hydrogenation and subsequent formylation. NMR assignment of the Fapy-dG lesion (X) embedded within a TXT trimer reveals the presence of rotameric and anomeric species. The latter have been characterized by synthesizing the tridecamer oligodeoxynucleotide d(GCGTACXCATGCG) harboring Fapy-dG as the central residue and developing a protocol to resolve the isomeric components. Hybridization of the chromatographically isolated fractions with their complementary d(CGCATGCGTACGC) counterpart yields two Fapy-dG·C duplexes that are differentially destabilized relative to the canonical G·C parent. The resultant duplexes exhibit distinct thermal and thermodynamic profiles that are characteristic of α- and ß-anomers, the former more destabilizing than the latter. These anomer-specific impacts are discussed in terms of differential repair enzyme recognition, processing and translesion synthesis.


Asunto(s)
Daño del ADN , Formamidas/química , Furanos/química , Oligodesoxirribonucleótidos/química , Pirimidinas/química , Cromatografía por Intercambio Iónico , ADN/química , ADN de Cadena Simple/química , Isomerismo , Mutágenos/química , Conformación de Ácido Nucleico , Oligodesoxirribonucleótidos/síntesis química , Oligodesoxirribonucleótidos/aislamiento & purificación , Termodinámica
7.
Nucleic Acids Res ; 38(1): 339-52, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19854934

RESUMEN

Aristolochic acids I and II (AA-I, AA-II) are found in all Aristolochia species. Ingestion of these acids either in the form of herbal remedies or as contaminated wheat flour causes a dose-dependent chronic kidney failure characterized by renal tubulointerstitial fibrosis. In approximately 50% of these cases, the condition is accompanied by an upper urinary tract malignancy. The disease is now termed aristolochic acid nephropathy (AAN). AA-I is largely responsible for the nephrotoxicity while both AA-I and AA-II are genotoxic. DNA adducts derived from AA-I and AA-II have been isolated from renal tissues of patients suffering from AAN. We describe the total synthesis, de novo, of the dA and dG adducts derived from AA-II, their incorporation site-specifically into DNA oligomers and the splicing of these modified oligomers into a plasmid construct followed by transfection into mouse embryonic fibroblasts. Analysis of the plasmid progeny revealed that both adducts blocked replication but were still partly processed by DNA polymerase(s). Although the majority of coding events involved insertion of correct nucleotides, substantial misincorporation of bases also was noted. The dA adduct is significantly more mutagenic than the dG adduct; both adducts give rise, almost exclusively, to misincorporation of dA, which leads to AL-II-dA-->T and AL-II-dG-->T transversions.


Asunto(s)
Ácidos Aristolóquicos/síntesis química , Aductos de ADN/síntesis química , Mutagénesis , Animales , Ácidos Aristolóquicos/química , Células Cultivadas , ADN/biosíntesis , Aductos de ADN/química , Ratones , Mutagénesis Sitio-Dirigida , Oligodesoxirribonucleótidos/síntesis química , Oligodesoxirribonucleótidos/química
8.
Nucleic Acids Res ; 37(7): 2153-63, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19223332

RESUMEN

Acrolein is a cell metabolic product and a main component of cigarette smoke. Its reaction with DNA produces two guanine lesions gamma-OH-PdG, a major adduct that is nonmutagenic in mammalian cells, and the positional isomer alpha-OH-PdG. We describe here the solution structure of a short DNA duplex containing a single alpha-OH-PdG lesion, as determined by solution NMR spectroscopy and restrained molecular dynamics simulations. The spectroscopic data show a mostly regular right-handed helix, locally perturbed at its center by the presence of the lesion. All undamaged residues of the duplex are in anti orientation, forming standard Watson-Crick base-pair alignments. Duplication of proton signals near the damaged site differentiates two enantiomeric duplexes, thus establishing the exocyclic nature of the lesion. At the lesion site, alpha-OH-PdG rotates to a syn conformation, pairing to its counter cytosine residue that is protonated at pH 5.9. Three-dimensional models produced by restrained molecular dynamics simulations show different hydrogen-bonding patterns between the lesion and its cytosine partner and identify further stabilization of alpha-OH-PdG in a syn conformation by intra-residue hydrogen bonds. We compare the alpha-OH-PdG.dC duplex structure with that of duplexes containing the analogous lesion propano-dG and discuss the implications of our findings for the mutagenic bypass of acrolein lesions.


Asunto(s)
Acroleína/toxicidad , Aductos de ADN/química , Contaminantes Ambientales/toxicidad , Guanosina/análogos & derivados , Desoxicitidina/química , Guanosina/química , Modelos Moleculares , Mutagénesis , Resonancia Magnética Nuclear Biomolecular , Protones , Soluciones
9.
Chem Res Toxicol ; 18(3): 457-65, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15777085

RESUMEN

In previous work we described an efficient procedure for the synthesis of the respective N2 and N6 adducts of 2'-deoxyguanosine (dG) and 2'-deoxyadenosine (dA) derived from a series of aminoaryl compounds. We now outline methods for the site-specific introduction into oligomeric DNA of the adducts dG-N2-AN (6), dG-N2-AAN (7), dG-N2-AF (8), and dG-N2-AAF (9) derived from 2-aminonaphthalene (2-AN) or 2-aminofluorene (2-AF). For the 2-AN adduct 7, containing an acetylamino group, the 5'-O-4,4'-dimethoxytrityl- (DMT-) 3'-O-phosphoramidite (14) required for automated DNA synthesis was synthesized in high yield via the sequence 10-->11-->14. On the other hand, introduction of the desacetyl adduct 6 into oligomeric DNA was accomplished via the N-trifluoroacetyl-DMT-phosphoramidite derivative 18. This involved a similar sequence (10-->15-->18) except that the order of the reactions was changed to avoid a decomposition that occurred when the silyl-protected amino derivative 11 was treated with trifluoroacetic anhydride. In the 2-AF series the 5'-O-DMT-3'-O-phosphoramidites 27a and 27b, related to 8 and 9, were prepared by similar methods. Again, however, the order of the reactions was changed to avoid the extreme insolubility associated with the N2-[3-(2-acetylaminofluoren-3-yl)]dG (dG-N2-AAF, 9) adduct that we had noted previously. The incorporation into oligomeric DNA of the acetylamino compounds 7 and 9 proceeded smoothly and in high yield (95-100%). By contrast, the trifluoroacetyl analogues led in both the naphthyl and fluorenyl series to a mixture of oligomers containing the desired free amino adduct (6 or 8) accompanied by the N-acetyl adduct (7 or 9, respectively, after the deprotection step), indicating secondary acetylation by the capping agent acetic anhydride.


Asunto(s)
2-Naftilamina/química , Aductos de ADN/síntesis química , Desoxiguanosina/química , Fluorenos/química , Oligonucleótidos/química , Aductos de ADN/química , Oligonucleótidos/síntesis química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...