Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Biochem Biophys Rep ; 28: 101181, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34934826

RESUMEN

The acute liver disease is involved in aberrant release of high-mobility group box 1 (HMGB1). Glycyrrhizin (GL), a traditional Chinese medicine for liver disease, binds to HMGB1, thereby inhibits tissue injury. However the mode of action of GL for chronic liver disease remains unclear. We investigated the effects of glycyrrhizin (GL) and its derivatives on liver differentiation using human iPS cells by using a flow cytometric analysis. GL promoted hepatic differentiation at the hepatoblast formation stage. The GL derivatives, 3-O-mono-glucuronyl 18ß-glycyrrhetinic acid (Mono) and 3-O-[glucosyl (1 â†’ 2)-glucuronyl] 18ß-glycyrrhetinic acid increased AFP+ cell counts and albumin+ cell counts. Glucuronate conjugation seemed to be a requirement for hepatic differentiation. Mono exhibited the most significant hepatic differentiation effect. We evaluated the effects of (±)-2-(2,4-dichlorophenoxy) propionic acid (DP), a T1R3 antagonist, and sucralose, a T1R3 agonist, on hepatic differentiation, and found that DP suppressed Mono-induced hepatic differentiation, while sucralose promoted hepatic differentiation. Thus, GL promoted hepatic differentiation via T1R3 signaling. In addition, Mono increased ß-catenin+ cell count and decreased Hes5+ cell count suggesting the involvement of Wnt and Notch signaling in GL-induced hepatic differentiation. In conclusion, GL exerted a hepatic differentiation effect via sweet receptor (T1R3), canonical Wnt, and Notch signaling.

2.
Free Radic Res ; 49(9): 1165-72, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25971446

RESUMEN

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.


Asunto(s)
Antibacterianos/química , Cloranfenicol/química , Daño del ADN , Oxígeno/química , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Bovinos , Cloranfenicol/análogos & derivados , ADN/química , ADN-Formamidopirimidina Glicosilasa/química , Desoxiguanosina/análogos & derivados , Desoxiguanosina/química , Radicales Libres/química , Genes p53 , Humanos , Hidróxidos , Hidroxilaminas/química , Leucemia/tratamiento farmacológico , Piperidinas/química , Especies Reactivas de Oxígeno/química , Espectrofotometría Ultravioleta , Timo/metabolismo
3.
J Photochem Photobiol B ; 87(3): 209-17, 2007 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-17537641

RESUMEN

The photosensitized DNA damage caused by dihydroxoP(V)tetraphenylporphyrin (P(V)TPP), a cationic water-soluble porphyrin, was examined. The study of near-infrared emission measurements demonstrated the photosensitized singlet oxygen ((1)O(2)) generation by P(V)TPP (quantum yield: 0.28 in ethanol). The fluorescence quenching of P(V)TPP by DNA showed the electron transfer (ET) from nucleobases to photoexcited P(V)TPP. These results have shown that P(V)TPP has ability to damage DNA through dual mechanisms, (1)O(2) generation and ET. Under aerobic conditions, P(V)TPP photosensitized damage was more severe for single-stranded DNA compared to its double-stranded counterpart. Photoexcited P(V)TPP damaged every guanine residue in single-stranded DNA. HPLC measurements confirmed the formation of 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodGuo), an oxidized product of 2'-deoxyguanosine, and showed that the yield of 8-oxodGuo in single-stranded DNA is larger than that in double-stranded DNA. The guanine-specific DNA damage and the enhancement in single-stranded DNA suggest that the (1)O(2) generation mainly contributes to the mechanism of DNA photodamage by P(V)TPP. Absorption spectrum measurements suggested the interaction between P(V)TPP and DNA. This interaction is expected to enhance the (1)O(2)-mediated DNA damage since the lifetime of (1)O(2) is very short. On the other hand, for double-stranded DNA, photosensitized damage at consecutive guanines was much less pronounced. Because the consecutive guanines act as a hole trap, this DNA-damaging pattern suggests the partial involvement of photoinduced ET. However, DNA damage by ET was not a main mechanism, possibly due to the reverse ET. In conclusion, P(V)TPP induces guanine specific photooxidation mainly via (1)O(2) generation. The interaction with DNA and the energy level of the photoexcited porphyrin may be advantageous for (1)O(2)-mediated DNA damage rather than ET mechanism.


Asunto(s)
Daño del ADN , Guanina , Trastornos por Fotosensibilidad , Porfirinas/efectos adversos , Oxígeno Singlete , ADN/metabolismo , Electrones , Oxidación-Reducción , Fósforo
4.
Free Radic Res ; 40(9): 966-73, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17015277

RESUMEN

Although capsaicin exhibits antitumor activity, carcinogenic potential has also been reported. To clarify the mechanism for expression of potential carcinogenicity of capsaicin, we examined DNA damage induced by capsaicin in the presence of metal ion and various kinds of cytochrome P450 (CYP) using 32P-5'-end-labeled DNA fragments. Capsaicin induced Cu(II)-mediated DNA damage efficiently in the presence of CYP1A2 and partially in the presence of 2D6. CYP1A2-treated capsaicin caused double-base lesions at 5'-TG-3', 5'-GC-3' and CG of the 5'-ACG-3' sequence complementary to codon 273, a hotspot of p53 gene. DNA damage was inhibited by catalase and bathocuproine, a Cu(I) chelator, suggesting that reactive species derived from the reaction of H2O2 with Cu(I) participate in DNA damage. Formation of 8-oxo-7,8-dihydro-2'-deoxyguanosine was significantly increased by CYP1A2-treated capsaicin in the presence of Cu(II). Therefore, we conclude that Cu(II)-mediated oxidative DNA damage by CYP-treated capsaicin seems to be relevant for the expression of its carcinogenicity.


Asunto(s)
Capsaicina/química , Capsicum/química , Daño del ADN , ADN/química , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Catalasa/química , Bovinos , Cobre/química , Citocromo P-450 CYP1A2/química , Fragmentación del ADN , Desoxiguanosina/análogos & derivados , Desoxiguanosina/síntesis química , Humanos , Oxidación-Reducción , Estrés Oxidativo , Fenantrolinas/química , Preparaciones de Plantas/química
5.
Chem Res Toxicol ; 19(10): 1379-85, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17040108

RESUMEN

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.


Asunto(s)
Carcinógenos/farmacología , Daño del ADN/genética , Tetranitrometano/farmacología , Tirosina/farmacología , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Bovinos , Quelantes/farmacología , Cobre/farmacología , ADN/genética , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Humanos , Estructura Molecular , NAD/farmacología , Oxidación-Reducción/efectos de los fármacos , Péptidos/farmacología
6.
Mutat Res ; 607(2): 184-91, 2006 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-16798066

RESUMEN

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.


Asunto(s)
Daño del ADN/efectos de los fármacos , ADN/metabolismo , Tinturas para el Cabello/química , Fenilendiaminas/toxicidad , Superóxido Dismutasa/metabolismo , Animales , Biomarcadores/análisis , Biomarcadores/metabolismo , Bovinos , Técnicas de Cultivo de Célula , Células Cultivadas , Técnicas de Cocultivo , Cobre/farmacología , Desoxiguanosina/análogos & derivados , Desoxiguanosina/análisis , Desoxiguanosina/metabolismo , Genes Supresores de Tumor/efectos de los fármacos , Genes p16 , Genes p53 , Humanos , Queratinocitos/citología , Oxidación-Reducción , Superóxido Dismutasa/farmacología
7.
Free Radic Biol Med ; 40(7): 1242-9, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16545693

RESUMEN

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.


Asunto(s)
Benzo(a)Antracenos/toxicidad , Carcinógenos/toxicidad , Daño del ADN/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , 8-Hidroxi-2'-Desoxicoguanosina , Catalasa/farmacología , Quelantes/farmacología , Cobre/farmacología , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Genes p53/efectos de los fármacos , Genes ras/efectos de los fármacos , Células HL-60 , Humanos , NAD/metabolismo , Oxidación-Reducción , Consumo de Oxígeno/efectos de los fármacos , Fenantrolinas/farmacología , Espectrofotometría Ultravioleta
8.
Antioxid Redox Signal ; 7(11-12): 1728-39, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16356133

RESUMEN

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.


Asunto(s)
Antioxidantes/efectos adversos , Antioxidantes/farmacología , Neoplasias/metabolismo , Neoplasias/prevención & control , Animales , Antioxidantes/química , ADN/efectos de los fármacos , ADN/metabolismo , Daño del ADN/efectos de los fármacos , Humanos
9.
Mutat Res ; 565(1): 35-44, 2004 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-15576237

RESUMEN

Eugenol used as a flavor has potential carcinogenicity. DNA adduct formation via 2,3-epoxidation pathway has been thought to be a major mechanism of DNA damage by carcinogenic allylbenzene analogs including eugenol. We examined whether eugenol can induce oxidative DNA damage in the presence of cytochrome P450 using [32P]-5'-end-labeled DNA fragments obtained from human genes relevant to cancer. Eugenol induced Cu(II)-mediated DNA damage in the presence of cytochrome P450 (CYP)1A1, 1A2, 2C9, 2D6, or 2E1. CYP2D6 mediated eugenol-dependent DNA damage most efficiently. Piperidine and formamidopyrimidine-DNA glycosylase treatment induced cleavage sites mainly at T and G residues of the 5'-TG-3' sequence, respectively. Interestingly, CYP2D6-treated eugenol strongly damaged C and G of the 5'-ACG-3' sequence complementary to codon 273 of the p53 gene. These results suggest that CYP2D6-treated eugenol can cause double base lesions. DNA damage was inhibited by both catalase and bathocuproine, suggesting that H2O2 and Cu(I) are involved. These results suggest that Cu(I)-hydroperoxo complex is primary reactive species causing DNA damage. Formation of 8-oxo-7,8-dihydro-2'-deoxyguanosine was significantly increased by CYP2D6-treated eugenol in the presence of Cu(II). Time-of-flight-mass spectrometry demonstrated that CYP2D6 catalyzed O-demethylation of eugenol to produce hydroxychavicol, capable of causing DNA damage. Therefore, it is concluded that eugenol may express carcinogenicity through oxidative DNA damage by its metabolite.


Asunto(s)
Cobre/metabolismo , Citocromo P-450 CYP2D6/metabolismo , Daño del ADN , Metilación de ADN , Desoxiguanosina/análogos & derivados , Eugenol/metabolismo , Isoenzimas/metabolismo , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Quelantes/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Desoxiguanosina/metabolismo , Eugenol/química , Depuradores de Radicales Libres/metabolismo , Humanos , Estructura Molecular , Oxidación-Reducción , Proteína p53 Supresora de Tumor/genética
10.
Biochem Pharmacol ; 68(9): 1869-78, 2004 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-15450952

RESUMEN

Melatonin, an indolic pineal hormone, is produced primarily at night in mammals and is important in controlling biological rhythms. Although melatonin is known to be effective as a free radical scavenger and has an anti-cancer effect, carcinogenic properties have also been reported. In relation to its carcinogenic potential, we have examined whether 6-hydroxymelatonin, a major melatonin metabolite, can induce DNA damage in the presence of metal ion using [32P]-5'-end-labeled DNA fragments obtained from genes relevant to human cancer. 6-Hydroxymelatonin induced site-specific DNA damage in the presence of Cu(II). Formamidopyrimidine-DNA glycosylase treatment induced cleavage sites mainly at G residues of the 5'-TG-3' sequence, whereas piperidine treatment induced cleavage sites at T mainly of 5'-TG-3'. Interestingly, 6-hydroxymelatonin strongly damaged G and C of the 5'-ACG-3' sequence complementary to codon 273 of the p53 gene. These results suggest that 6-hydroxymelatonin can cause double-base lesions. DNA damage was inhibited by both catalase and bathocuproine, Cu(I)-specific stabilizer, suggesting that reactive species derived from the reaction of H2O2 with Cu(I) participate in DNA damage. Cytochrome P450 reductase efficiently enhanced 6-hydroxymelatonin-induced oxidative DNA damage and oxygen consumption, suggesting the formation of redox cycle. It is noteworthy that 6-hydroxymelatonin can efficiently induce DNA damage via non-o-quinone type of redox cycle. Formation of 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG), a characteristic oxidative DNA lesion, in calf thymus DNA was significantly increased by 6-hydroxymelatonin in the presence of Cu(II). Furthermore, 6-hydroxymelatonin significantly increased the formation of 8-oxodG in human leukemia cell line HL-60 but not in HP100, a hydrogen peroxide (H2O2)-resistant cell line derived from HL-60. The 6-hydroxymelatonin-induced 8-oxodG formation in HL-60 cells significantly decreased by the addition of bathocuproine or o-phenanthroline. Therefore, it is concluded that melatonin may exhibit carcinogenic potential through oxidative DNA damage by its metabolite.


Asunto(s)
Daño del ADN/efectos de los fármacos , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Melatonina/análogos & derivados , Melatonina/metabolismo , Melatonina/farmacología , 8-Hidroxi-2'-Desoxicoguanosina , Quelantes/farmacología , ADN/efectos de los fármacos , ADN/metabolismo , Interacciones Farmacológicas , Activación Enzimática/efectos de los fármacos , Depuradores de Radicales Libres/farmacología , Células HL-60 , Humanos , Peróxido de Hidrógeno/metabolismo , NADPH-Ferrihemoproteína Reductasa/metabolismo , Oxidación-Reducción , Consumo de Oxígeno/efectos de los fármacos
11.
Chem Res Toxicol ; 16(11): 1470-6, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14615974

RESUMEN

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.


Asunto(s)
Benzo(a)Antracenos/efectos adversos , Benzo(a)Antracenos/metabolismo , Guanina/análogos & derivados , Estrés Oxidativo , Quinonas/efectos adversos , Animales , Catalasa/farmacología , Cobre/metabolismo , Cobre/farmacología , Daño del ADN/fisiología , Fragmentación del ADN/efectos de los fármacos , Desoxiadenosinas/biosíntesis , Relación Dosis-Respuesta a Droga , Genes p16/efectos de los fármacos , Genes ras/efectos de los fármacos , Guanina/biosíntesis , Humanos , NAD/metabolismo , NAD/farmacología , NADPH-Ferrihemoproteína Reductasa/metabolismo , Oxidación-Reducción , Fenantrolinas/farmacología , Radioisótopos de Fósforo , Hidrocarburos Policíclicos Aromáticos/efectos adversos , Quinonas/metabolismo , Proteína p53 Supresora de Tumor/efectos de los fármacos
12.
Mutat Res ; 539(1-2): 145-55, 2003 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-12948823

RESUMEN

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.


Asunto(s)
Cobre/toxicidad , Daño del ADN , Procarbazina/toxicidad , Catalasa , Radicales Libres/análisis , Genes p53 , Genes ras , Humanos , Estrés Oxidativo , Fenantrolinas/farmacología
13.
Arch Biochem Biophys ; 410(2): 261-8, 2003 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-12573286

RESUMEN

DNA damage mediated by photosensitizers participates in solar carcinogenesis. Fluorescence measurement and high-performance liquid chromatography analysis demonstrated that photoirradiated folic acid, one of the photosensitizers in cells, generates pterine-6-carboxylic acid (PCA). Experiments using 32P-labeled DNA fragments obtained from a human gene showed that ultraviolet A-irradiated folic acid or PCA caused DNA cleavage specifically at consecutive G residues in double-stranded DNA after Escherichia coli formamidopyrimidine-DNA glycosylase or piperidine treatment. The amount of 8-oxo-7,8-dihydro-2(')-deoxyguanosine formed through this DNA photoreaction in double-stranded DNA exceeded that in single-stranded DNA. Kinetic studies suggested that DNA damage is caused mainly by photoexcited PCA generated from folic acid rather than by folic acid itself. In conclusion, photoirradiated folic acid generates PCA, which induces DNA photooxidation specifically at consecutive G residues through electron transfer. Excess intake of folic acid supplements may increase a risk of skin cancer by solar ultraviolet light.


Asunto(s)
Daño del ADN , ADN/efectos de los fármacos , Desoxiguanosina/análogos & derivados , Ácido Fólico/metabolismo , 8-Hidroxi-2'-Desoxicoguanosina , Cromatografía Líquida de Alta Presión , ADN/metabolismo , ADN/efectos de la radiación , Desoxiguanosina/metabolismo , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Espectroscopía de Resonancia por Spin del Electrón , Escherichia coli/metabolismo , Humanos , Cinética , Luz , Modelos Químicos , Estructura Terciaria de Proteína , Rayos Ultravioleta
14.
Int J Cancer ; 102(4): 311-7, 2002 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-12402298

RESUMEN

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.


Asunto(s)
2-Acetilaminofluoreno/farmacología , Carcinógenos/farmacología , Daño del ADN/efectos de los fármacos , ADN de Neoplasias/efectos de los fármacos , Desoxiguanosina/análogos & derivados , 2-Acetilaminofluoreno/análogos & derivados , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Catalasa/farmacología , Bovinos , Quelantes/farmacología , Cobre/farmacología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Desoxiguanosina/metabolismo , Depuradores de Radicales Libres/farmacología , Células HL-60/efectos de los fármacos , Células HL-60/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , NAD/farmacología , Oxidación-Reducción , Fenantrolinas/farmacología , Proteínas Proto-Oncogénicas p21(ras)/genética , Espectrofotometría Ultravioleta , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
15.
Biochem Biophys Res Commun ; 290(2): 778-82, 2002 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-11785968

RESUMEN

Carcinogenic benzo[a]pyrene (BP) is generally considered to show genotoxicity by forming DNA adducts of its metabolite, BP-7,8-diol-9,10-epoxide. We investigated oxidative DNA damage and its sequence specificity induced by BP-7,8-dione, another metabolite of BP, using (32)P-5'-end-labeled DNA. Formamidopyrimidine-DNA glycosylase treatment induced cleavage sites mainly at G residues of 5'-TG-3' sequence and at poly(C) sequences, in DNA incubated with BP-7,8-dione in the presence of NADH and Cu(II), whereas piperidine treatment induced cleavage sites at T mainly of 5'-TG-3'. BP-7,8-dione strongly damaged the G and C of the ACG sequence complementary to codon 273 of the p53 gene. Catalase and a Cu(I)-specific chelator attenuated the DNA damage, indicating the involvement of H(2)O(2) and Cu(I). BP-7,8-dione with NADH and Cu(II) also increased 8-oxo-7,8-dihydro-2'-deoxyguanosine formation. We conclude that oxidative DNA damage, especially double base lesions, may participate in the expression of carcinogenicity of BP in addition to DNA adduct formation.


Asunto(s)
Benzopirenos/química , Carcinógenos/química , ADN/química , 8-Hidroxi-2'-Desoxicoguanosina , Aldehídos/química , Aldehídos/farmacología , Animales , Benzopirenos/farmacología , Sitios de Unión/efectos de los fármacos , Carcinógenos/farmacología , Bovinos , Cobre/química , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , ADN/efectos de los fármacos , Daño del ADN , ADN-Formamidopirimidina Glicosilasa , Desoxiguanosina/análogos & derivados , Desoxiguanosina/análisis , Depuradores de Radicales Libres/química , Genes p53/efectos de los fármacos , Genes ras/efectos de los fármacos , Humanos , N-Glicosil Hidrolasas/química , NAD/química , Oxidación-Reducción/efectos de los fármacos , Piperidinas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA