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

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Free Radic Res ; 49(9): 1165-72, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25971446

RESUMO

Chloramphenicol (CAP) was an old antimicrobial agent. However, the use of CAP is limited because of its harmful side effects, such as leukemia. The molecular mechanism through which CAP has been strongly correlated with leukemogenesis is still unclear. To elucidate the mechanism of genotoxicity, we examined DNA damage by CAP and its metabolites, nitroso-CAP (CAP-NO), N-hydroxy-CAP (CAP-NHOH), using isolated DNA. CAP-NHOH have the ability of DNA damage including 8-oxo-7,8-dihydro-2'-deoxyguanosine formation in the presence of Cu(II), which was greatly enhanced by the addition of an endogenous reductant NADH. CAP-NO caused DNA damage in the presence of Cu(II), only when reduced by NADH. NADH can non-enzymatically reduce the nitroso form to hydronitroxide radicals, resulting in enhanced generation of reactive oxygen species followed by DNA damage through the redox cycle. Furthermore, we also studied the site specificity of base lesions in DNA treated with piperidine or formamidopyrimidine-DNA glycosylase, using (32)P-5'-end-labeled DNA fragments obtained from the human tumor suppressor gene. CAP metabolites preferentially caused double base lesion, the G and C of the ACG sequence complementary to codon 273 of the p53 gene, in the presence of NADH and Cu(II). Therefore, we conclude that oxidative double base lesion may play a role in carcinogenicity of CAP.


Assuntos
Antibacterianos/química , Cloranfenicol/química , Dano ao DNA , Oxigênio/química , 8-Hidroxi-2'-Desoxiguanosina , Animais , Bovinos , Cloranfenicol/análogos & derivados , DNA/química , DNA-Formamidopirimidina Glicosilase/química , Desoxiguanosina/análogos & derivados , Desoxiguanosina/química , Radicais Livres/química , Genes p53 , Humanos , Hidróxidos , Hidroxilaminas/química , Leucemia/tratamento farmacológico , Piperidinas/química , Espécies Reativas de Oxigênio/química , Espectrofotometria Ultravioleta , Timo/metabolismo
2.
J Med Food ; 18(8): 835-40, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25607850

RESUMO

It remains unclear whether dietary supplementation with coenzyme Q10 (CoQ10) provides beneficial effects for healthy individuals, especially young subjects. This study investigated the effects of dietary supplementation with CoQ10 on oxidative stress in healthy young females. We performed a placebo-controlled trial using a crossover design (n=28) with 100 mg/day CoQ10 in reduced form or placebo, each lasting 2 weeks with a 2-week interval. The urinary levels of 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG), a biomarker of oxidative DNA damage, were determined by high-performance liquid chromatography (HPLC) coupled to an electrochemical detector. Levels of malondialdehyde (MDA), a biomarker of lipid peroxidation, and antioxidant vitamin C in urine were also measured using a thiobarbituric acid-reactive substance method with a commercial kit and by the 2,4-dinitrophenylhydrazine method with HPLC, respectively. Urinary 8-oxodG levels during supplementation with reduced form of CoQ10 (median [first and third quartiles]: 1.76 [1.24-2.08] nmol/mmol creatinine) were significantly lower than those with placebo (2.00 [1.34-2.49] nmol/mmol creatinine, P=.031 by Student's paired t-test using the logarithmically transformed values). In contrast, the urinary levels of MDA and vitamin C were not significantly affected (P=.094 and P=.247 by Student's paired t-test, respectively). There was no evidence of any side effects. Supplementation with CoQ10 in the reduced form showed a slightly protective effect against oxidative DNA damage even in healthy young subjects.


Assuntos
Biomarcadores/urina , Dano ao DNA/efeitos dos fármacos , Desoxiguanosina/análogos & derivados , Estresse Oxidativo/efeitos dos fármacos , Ubiquinona/análogos & derivados , 8-Hidroxi-2'-Desoxiguanosina , Adulto , Antioxidantes/análise , Ácido Ascórbico/urina , Cromatografia Líquida de Alta Pressão , Estudos Cross-Over , Desoxiguanosina/urina , Suplementos Nutricionais , Feminino , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Malondialdeído/urina , Resultado do Tratamento , Ubiquinona/administração & dosagem , Ubiquinona/farmacologia , Vitaminas/farmacologia , Adulto Jovem
3.
Chem Res Toxicol ; 19(10): 1379-85, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17040108

RESUMO

Tetranitromethane (TNM) is used as an oxidizer in rocket propellants and explosives and as an additive to increase the cetane number of diesel fuel. TNM was reported to induce pulmonary adenocarcinomas and squamous cell carcinomas in mice and rats. However, the mechanisms underlying carcinogenesis induced by TNM has not yet been clarified. We previously revealed that nitroTyr and nitroTyr-containing peptides caused Cu(II)-dependent DNA damage in the presence of P450 reductase, which is considered to yield nitroreduction. Since TNM is a reagent for nitration of Tyr in proteins and peptides, we have hypothesized that TNM-treated Tyr and Tyr-containing peptides induce DNA damage by the modification of Tyr. We examined DNA damage induced by TNM-treated amino acids or peptides using (32)P-5'-end-labeled DNA fragments obtained from the human p53 tumor suppressor gene and the c-Ha-ras-1 protooncogene. TNM-treated Tyr and Lys-Tyr-Lys induced DNA damage including the formation of 8-oxo-7,8-dihydro-2'-deoxyguanosine in the presence of Cu(II) and NADH. DNA damage was inhibited by catalase and bathocuproine, indicating the involvement of H(2)O(2) and Cu(I). The cytosine residue of the ACG sequence complementary to codon 273, well-known hotspots of the p53 gene, was cleaved with piperidine and Fpg treatments. On the other hand, nitroTyr and Lys-nitroTyr-Lys did not induce DNA damage in the presence of Cu(II) and NADH. Time-of-flight mass spectrometry confirmed that reactions between Lys-Tyr-Lys and TNM yielded not only Lys-nitroTyr-Lys but also Lys-nitrosoTyr-Lys. Therefore, it is speculated that the nitrosotyrosine residue can induce oxidative DNA damage in the presence of Cu(II) and NADH. It is concluded that Tyr-dependent DNA damage may play an important role in the carcinogenicity of TNM. TNM is a new type of carcinogen that induces DNA damage not by itself but via Tyr modification.


Assuntos
Carcinógenos/farmacologia , Dano ao DNA/genética , Tetranitrometano/farmacologia , Tirosina/farmacologia , 8-Hidroxi-2'-Desoxiguanosina , Animais , Bovinos , Quelantes/farmacologia , Cobre/farmacologia , DNA/genética , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Humanos , Estrutura Molecular , NAD/farmacologia , Oxirredução/efeitos dos fármacos , Peptídeos/farmacologia
4.
Mutat Res ; 607(2): 184-91, 2006 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-16798066

RESUMO

There is an association between occupational exposure to hair dyes and incidence of cancers. Permanent oxidant hair dyes are consisted of many chemical components including ortho-phenylenediamines. To clarify the mechanism of carcinogenesis by hair dyes, we examined DNA damage induced by mutagenic ortho-phenylenediamine (o-PD) and its derivatives, 4-chloro-ortho-phenylenediamine (Cl-PD) and 4-nitro-ortho-phenylenediamine (NO(2)-PD), using (32)P-labeled DNA fragments obtained from the human p16 and the p53 tumor suppressor gene. We also measured the content of 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG), a marker of oxidative DNA damage, in calf thymus DNA with an electrochemical detector coupled to a high performance liquid chromatograph. Carcinogenic o-PD and Cl-PD caused Cu(II)-mediated DNA damage, including 8-oxodG formation, and antioxidant enzyme superoxide dismutase (SOD) enhanced DNA damage. o-PD and Cl-PD caused piperidine-labile and formamidopyrimidine-DNA glycosylase-sensitive lesions at cytosine and guanine residues respectively in the 5'-ACG-3' sequence, complementary to codon 273, a well-known hotspot of the human p53 tumor suppressor gene. UV-vis spectroscopic studies showed that the spectral change of o-PD and Cl-PD required Cu(II), and addition of SOD enhanced it. This suggested that SOD enhanced the rate of Cu(II)-mediated autoxidation of o-PD and Cl-PD, leading to enhancement of DNA damage. On the other hand, mutagenic but non-carcinogenic NO(2)-PD induced no DNA damage. These results suggest that carcinogenicity of ortho-phenylenediamines is associated with ability to cause oxidative DNA damage rather than bacterial mutagenicity.


Assuntos
Dano ao DNA/efeitos dos fármacos , DNA/metabolismo , Tinturas para Cabelo/química , Fenilenodiaminas/toxicidade , Superóxido Dismutase/metabolismo , Animais , Biomarcadores/análise , Biomarcadores/metabolismo , Bovinos , Técnicas de Cultura de Células , Células Cultivadas , Técnicas de Cocultura , Cobre/farmacologia , Desoxiguanosina/análogos & derivados , Desoxiguanosina/análise , Desoxiguanosina/metabolismo , Genes Supressores de Tumor/efeitos dos fármacos , Genes p16 , Genes p53 , Humanos , Queratinócitos/citologia , Oxirredução , Superóxido Dismutase/farmacologia
5.
Free Radic Biol Med ; 40(7): 1242-9, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16545693

RESUMO

3-Nitrobenzanthrone (3-NBA) is an extremely potent mutagen in diesel exhaust. It is a lung carcinogen to rats, and therefore a suspected carcinogen to human. In order to clarify the mechanism of carcinogenicity of 3-NBA, we investigated oxidative DNA damage by N-hydroxy-3-aminobenzanthrone (N-OH-ABA), a metabolite of 3-NBA, using 32P-labeled DNA fragments from the human p53 tumor-suppressor gene. N-OH-ABA caused Cu(II)-mediated DNA damage, and endogenous reductant NADH dramatically enhanced this process. Catalase and a Cu(I)-specific chelator decreased DNA damage, suggesting the involvement of hydrogen peroxide (H2O2) and Cu(I). N-OH-ABA induced DNA damage at cytosine and guanine residues of ACG sequence complementary to codon 273, a well-known hot spot of the p53 gene. N-OH-ABA dose dependently induced 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG) formation in the presence of Cu(II) and NADH. Treatment with N-OH-ABA increased amounts of 8-oxodG in HL-60 cells compared to the H2O2-resistant clone HP100, supporting the involvement of H2O2. The present study has demonstrated that the N-hydroxy metabolite of 3-NBA induces oxidative DNA damage through H2O2 in both a cell-free system and cultured human cells. We conclude that oxidative DNA damage may play an important role in the carcinogenic process of 3-NBA in addition to previously reported DNA adduct formation.


Assuntos
Benzo(a)Antracenos/toxicidade , Carcinógenos/toxicidade , Dano ao DNA/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , 8-Hidroxi-2'-Desoxiguanosina , Catalase/farmacologia , Quelantes/farmacologia , Cobre/farmacologia , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Genes p53/efeitos dos fármacos , Genes ras/efeitos dos fármacos , Células HL-60 , Humanos , NAD/metabolismo , Oxirredução , Consumo de Oxigênio/efeitos dos fármacos , Fenantrolinas/farmacologia , Espectrofotometria Ultravioleta
6.
Antioxid Redox Signal ; 7(11-12): 1728-39, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16356133

RESUMO

Antioxidants are considered as the most promising chemopreventive agents against various human cancers. However, some antioxidants play paradoxical roles, acting as "double-edged sword." A primary property of effective and acceptable chemopreventive agents should be freedom from toxic effects in healthy population. Miscarriage of the intervention by beta-carotene made us realize the necessity for evaluation of safety before recommending use of antioxidant supplements for chemoprevention. We have evaluated the safety of antioxidants on the basis of reactivity with DNA. Our results revealed that phytic acid, luteolin, and retinoic acid did not cause DNA damage under the experimental condition. Furthermore, phytic acid inhibited the formation of 8-oxo-7,8-dihydro-2'-deoxyguanosine, an indicator of oxidative DNA damage, in cultured cells treated with a H(2)O(2)-generating system. Thus, it is expected that these chemopreventive agents can safely protect humans against cancer. On the other hand, some chemopreventive agents with prooxidant properties (alpha-tocopherol, quercetin, catechins, isothiocyanates, N-acetylcysteine) caused DNA damage via generation of reactive oxygen species in the presence of metal ions and endogenous reductants under some circumstances. Furthermore, other chemopreventive agents (beta-carotene, genistein, daidzein, propyl gallate, curcumin) exerted prooxidant properties after metabolic activation. Therefore, further studies on safety should be required when antioxidants are used for cancer prevention.


Assuntos
Antioxidantes/efeitos adversos , Antioxidantes/farmacologia , Neoplasias/metabolismo , Neoplasias/prevenção & controle , Animais , Antioxidantes/química , DNA/efeitos dos fármacos , DNA/metabolismo , Dano ao DNA/efeitos dos fármacos , Humanos
7.
Chem Res Toxicol ; 16(11): 1470-6, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14615974

RESUMO

Benz[a]anthracene (BA) is one of the most abundant polycyclic aromatic hydrocarbons (PAHs) that are ubiquitous environmental pollutants. PAH carcinogenesis is explained by DNA adduct formation by PAH diol epoxide and oxidative DNA damage by PAH o-quinone. Benz[a]anthracene-trans-3,4-dihydrodiol (BA-3,4-dihydrodiol) is a minor metabolite but shows higher mutagenicity and tumorigenicity than parent BA. We confirmed that a BA o-quinone type metabolite, benz[a]anthracene-3,4-dione (BA-3,4-dione), induced oxidative DNA damage in the presence of cytochrome P450 reductase. Interestingly, we found that BA-3,4-dihydrodiol nonenzymatically caused Cu(II)-mediated DNA damage including 8-oxo-7,8-dihydro-2'-deoxyguanosine formation and the addition of NADH enhanced DNA damage. BA-3,4-dihydrodiol induced a double-base lesion of C and G at the 5'-ACG-3' sequence complementary to codon 273 of the human p53 tumor suppressor gene, which is known as a hotspot. The DNA damage was inhibited by catalase and bathocuproine, indicating the involvement of H(2)O(2) and Cu(I). Time-of-flight mass spectroscopic study suggested that BA-3,4-dihydrodiol undergoes Cu(II)-mediated autoxidation leading to the formation of its hydroxylated form of BA-3,4-dihydrodiol, capable of causing oxidative DNA damage. It is noteworthy that BA-3,4-dihydrodiol can nonenzymatically induce DNA damage more efficiently than BA-3,4-dione with metabolic activation. In conclusion, oxidative DNA damage induced by BA-3,4-dihydrodiol not only via quinone-type redox cycle but also via a new type of redox cycle participates in the expression of carcinogenicity of BA and BA-3,4-dihydrodiol.


Assuntos
Benzo(a)Antracenos/efeitos adversos , Benzo(a)Antracenos/metabolismo , Guanina/análogos & derivados , Estresse Oxidativo , Quinonas/efeitos adversos , Animais , Catalase/farmacologia , Cobre/metabolismo , Cobre/farmacologia , Dano ao DNA/fisiologia , Fragmentação do DNA/efeitos dos fármacos , Desoxiadenosinas/biossíntese , Relação Dose-Resposta a Droga , Genes p16/efeitos dos fármacos , Genes ras/efeitos dos fármacos , Guanina/biossíntese , Humanos , NAD/metabolismo , NAD/farmacologia , NADPH-Ferri-Hemoproteína Redutase/metabolismo , Oxirredução , Fenantrolinas/farmacologia , Radioisótopos de Fósforo , Hidrocarbonetos Policíclicos Aromáticos/efeitos adversos , Quinonas/metabolismo , Proteína Supressora de Tumor p53/efeitos dos fármacos
8.
Mutat Res ; 539(1-2): 145-55, 2003 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-12948823

RESUMO

Procarbazine [N-isopropyl-alpha-(2-methylhydrazino)-p-toluamide], a hydrazine derivative, which has been shown to have effective antineoplastic activity, induces cancer in some experimental animals and humans. To clarify a new mechanism for its carcinogenic effect, we examined DNA damage induced by procarbazine in the presence of metal ion, using 32P-5'-end-labeled DNA fragments obtained from the human p53 tumor suppressor gene and the c-Ha-ras-1 protooncogene. Procarbazine plus Cu(II) induced piperidine-labile and formamidopyrimidine-DNA glycosylase-sensitive lesions at the 5'-ACG-3' sequence, complementary to a hotspot of the p53 gene, and the 5'-TG-3' sequence. Catalase partially inhibited DNA damage, suggesting that not only H(2)O(2) but also other reactive species are involved. Procarbazine plus Cu(II) significantly increased the formation of 8-oxo-7,8-dihydro-2'-deoxyguanosine, which was completely inhibited by calatase. Electron spin resonance spin-trapping experiments revealed that methyl radicals were generated from procarbazine and Cu(II). On the basis of these findings, it is considered that procarbazine causes DNA damage through non-enzymatic formation of the Cu(I)-hydroperoxo complex and methyl radicals. In conclusion, in addition to alkylation, oxidative DNA damage may play important roles in not only antitumor effects but also mutagenesis and carcinogenesis induced by procarbazine.


Assuntos
Cobre/toxicidade , Dano ao DNA , Procarbazina/toxicidade , Catalase , Radicais Livres/análise , Genes p53 , Genes ras , Humanos , Estresse Oxidativo , Fenantrolinas/farmacologia
9.
Int J Cancer ; 102(4): 311-7, 2002 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-12402298

RESUMO

Both carcinogenic NF and AAF are metabolized to a common N-hydroxy metabolite, N-OH-AF. We investigated oxidative DNA damage by N-OH-AF, using (32)P-labeled human DNA fragments from the human p53 and p16 tumor-suppressor genes and the c-Ha-ras-1 protooncogene. N-OH-AF caused Cu(II)-mediated DNA damage, and endogenous reductant NADH markedly enhanced this process. Catalase and bathocuproine, a Cu(I)-specific chelator, decreased the DNA damage, suggesting the involvement of H(2)O(2) and Cu(I). N-OH-AF induced piperidine-labile lesions frequently at thymine and cytosine residues. With formamidopyrimidine-DNA glycosylase treatment, N-OH-AF induced cleavage at guanine residues, especially of the ACG sequence complementary to codon 273, a well-known hot spot of the p53 gene. N-OH-AF dose-dependently induced 8-oxodG formation in the presence of Cu(II) and NADH. Treatment with N-OH-AF increased amounts of 8-oxodG in HL-60 cells compared to the H(2)O(2)-resistant clone HP100, supporting the involvement of H(2)O(2). The present study demonstrates that the N-hydroxy metabolite of NF and AAF induces oxidative DNA damage through H(2)O(2) in both a cell-free system and cultured human cells. We conclude that oxidative DNA damage may play an important role in the carcinogenic process of NF and AAF in addition to previously reported DNA adduct formation.


Assuntos
2-Acetilaminofluoreno/farmacologia , Carcinógenos/farmacologia , Dano ao DNA/efeitos dos fármacos , DNA de Neoplasias/efeitos dos fármacos , Desoxiguanosina/análogos & derivados , 2-Acetilaminofluoreno/análogos & derivados , 8-Hidroxi-2'-Desoxiguanosina , Animais , Catalase/farmacologia , Bovinos , Quelantes/farmacologia , Cobre/farmacologia , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Desoxiguanosina/metabolismo , Sequestradores de Radicais Livres/farmacologia , Células HL-60/efeitos dos fármacos , Células HL-60/metabolismo , Humanos , Peróxido de Hidrogênio/farmacologia , NAD/farmacologia , Oxirredução , Fenantrolinas/farmacologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Espectrofotometria Ultravioleta , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA