Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(15)2021 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-34361004

RESUMEN

This article reviews evidence suggesting that a common mechanism of initiation leads to the development of many prevalent types of cancer. Endogenous estrogens, in the form of catechol estrogen-3,4-quinones, play a central role in this pathway of cancer initiation. The catechol estrogen-3,4-quinones react with specific purine bases in DNA to form depurinating estrogen-DNA adducts that generate apurinic sites. The apurinic sites can then lead to cancer-causing mutations. The process of cancer initiation has been demonstrated using results from test tube reactions, cultured mammalian cells, and human subjects. Increased amounts of estrogen-DNA adducts are found not only in people with several different types of cancer but also in women at high risk for breast cancer, indicating that the formation of adducts is on the pathway to cancer initiation. Two compounds, resveratrol, and N-acetylcysteine, are particularly good at preventing the formation of estrogen-DNA adducts in humans and are, thus, potential cancer-prevention compounds.


Asunto(s)
Acetilcisteína/farmacología , Carcinogénesis/efectos de los fármacos , Estradiol/farmacología , Estrona/farmacología , Quinonas/farmacología , Resveratrol/farmacología , Animales , Antioxidantes/farmacología , Carcinogénesis/genética , Aductos de ADN , Estradiol/toxicidad , Estrógenos/farmacología , Estrógenos/toxicidad , Estrona/toxicidad , Humanos , Quinonas/toxicidad
2.
Int J Cancer ; 141(6): 1078-1090, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28388839

RESUMEN

Endogenous estrogens become carcinogens when dangerous metabolites, the catechol estrogen quinones, are formed. In particular, the catechol estrogen-3,4-quinones can react with DNA to produce an excess of specific depurinating estrogen-DNA adducts. Loss of these adducts leaves apurinic sites in the DNA, generating subsequent cancer-initiating mutations. Unbalanced estrogen metabolism yields excessive catechol estrogen-3,4-quinones, increasing formation of depurinating estrogen-DNA adducts and the risk of initiating cancer. Evidence for this mechanism of cancer initiation comes from various types of studies. High levels of depurinating estrogen-DNA adducts have been observed in women with breast, ovarian or thyroid cancer, as well as in men with prostate cancer or non-Hodgkin lymphoma. Observation of high levels of depurinating estrogen-DNA adducts in high risk women before the presence of breast cancer indicates that adduct formation is a critical factor in breast cancer initiation. Formation of analogous depurinating dopamine-DNA adducts is hypothesized to initiate Parkinson's disease by affecting dopaminergic neurons. Two dietary supplements, N-acetylcysteine and resveratrol complement each other in reducing formation of catechol estrogen-3,4-quinones and inhibiting formation of estrogen-DNA adducts in cultured human and mouse breast epithelial cells. They also inhibit malignant transformation of these cells. In addition, formation of adducts was reduced in women who followed a Healthy Breast Protocol that includes N-acetylcysteine and resveratrol. When initiation of cancer is blocked, promotion, progression and development of the disease cannot occur. These results suggest that reducing formation of depurinating estrogen-DNA adducts can reduce the risk of developing a variety of types of human cancer.


Asunto(s)
Aductos de ADN/metabolismo , Estrógenos/metabolismo , Neoplasias/etiología , Neoplasias/prevención & control , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/prevención & control , Animales , Dopamina/metabolismo , Humanos , Neoplasias/metabolismo , Enfermedad de Parkinson/metabolismo , Purinas/metabolismo
3.
Int J Cancer ; 134(10): 2414-23, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24170413

RESUMEN

Greater exposure to estrogens is a risk factor for ovarian cancer. To investigate the role of estrogens in ovarian cancer, a spot urine sample and a saliva sample were obtained from 33 women with ovarian cancer and 34 age-matched controls. Thirty-eight estrogen metabolites, conjugates and DNA adducts were analyzed in the urine samples using ultraperformance liquid chromatography/tandem mass spectrometry, and the ratio of adducts to metabolites and conjugates was calculated for each sample. The ratio of depurinating estrogen-DNA adducts to estrogen metabolites and conjugates was significantly higher in cases compared to controls (p < 0.0001), demonstrating high specificity and sensitivity. DNA was purified from the saliva samples and analyzed for genetic polymorphisms in the genes for two estrogen-metabolizing enzymes. Women with two low-activity alleles of catechol-O-methyltransferase plus one or two high-activity alleles of cytochrome P450 1B1 had higher levels of estrogen-DNA adducts and were more likely to have ovarian cancer. These findings indicate that estrogen metabolism is unbalanced in ovarian cancer and suggest that formation of estrogen-DNA adducts plays a critical role in the initiation of ovarian cancer.


Asunto(s)
Aductos de ADN/orina , ADN de Neoplasias/orina , Estrógenos/orina , Anciano , Hidrocarburo de Aril Hidroxilasas/genética , Hidrocarburo de Aril Hidroxilasas/metabolismo , Catecol O-Metiltransferasa/genética , Catecol O-Metiltransferasa/metabolismo , Cromatografía Líquida de Alta Presión , Citocromo P-450 CYP1B1 , Aductos de ADN/química , Aductos de ADN/metabolismo , ADN de Neoplasias/química , ADN de Neoplasias/metabolismo , Estrógenos/química , Estrógenos/metabolismo , Femenino , Frecuencia de los Genes , Genotipo , Humanos , Modelos Lineales , Persona de Mediana Edad , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/orina , Polimorfismo de Nucleótido Simple , Factores de Riesgo , Saliva/química , Saliva/metabolismo , Espectrometría de Masas en Tándem
4.
Carcinogenesis ; 34(11): 2587-92, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23843041

RESUMEN

Sulforaphane (SFN) is a potent inducer of detoxication enzymes such as NAD(P)H:quinone oxidoreductase 1 (NQO1) and glutathione-S-transferase (GST) via the Kelch-like erythroid-derived protein with CNC homology-associated protein 1 (Keap1)-NF-E2-related factor 2 (Nrf2) signaling pathway. NQO1 reduces the carcinogenic estrogen metabolite, catechol estrogen-3,4-quinone, whereas GSTs detoxify it through conjugation with glutathione. These 3,4-quinones can react with DNA to form depurinating DNA adducts. Thus, SFN may alter estrogen metabolism and thus protect against estrogen-mediated DNA damage and carcinogenesis. Human breast epithelial MCF-10A cells were treated with either vehicle or SFN and either estradiol (E2) or its metabolite 4-hydroxyestradiol (4-OHE2). 4-Hydroxy-derived estrogen metabolites and depurinating DNA adducts formed from E2 and its interconvertable metabolite estrone (E1) were analyzed by mass spectrometry. Levels of the depurinated adducts, 4-OHE1/2-1-N3Adenine and 4-OHE1/2-1-N7Guanine, were reduced by 60% in SFN-treated cells, whereas levels of 4-OCH3E1/2 and 4-OHE1/2-glutathione conjugates increased. To constitutively enhance the expression of Nrf2-regulated genes, cells were treated with either scrambled or siKEAP1 RNA. Following E2 or 4-OHE2 treatments, levels of the adenine and guanine adducts dropped 60-70% in siKEAP1-treated cells, whereas 4-OHE1/2-glutathione conjugates increased. However, 4-OCH3E1/2 decreased 50% after siKEAP1 treatment. Thus, treatment with SFN or siKEAP1 has similar effects on reduction of depurinating estrogen-DNA adduct levels following estrogen challenge. However, these pharmacologic and genetic approaches have different effects on estrogen metabolism to O-methyl and glutathione conjugates. Activation of the Nrf2 pathway, especially elevated NQO1, may account for some but not all of the protective effects of SFN against estrogen-mediated DNA damage.


Asunto(s)
Anticarcinógenos/farmacología , Mama/efectos de los fármacos , Aductos de ADN/efectos de los fármacos , Estrógenos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Isotiocianatos/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Apoptosis , Western Blotting , Mama/citología , Mama/metabolismo , Proliferación Celular , Células Cultivadas , Cromatografía Líquida de Alta Presión , Femenino , Humanos , Técnicas para Inmunoenzimas , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Proteína 1 Asociada A ECH Tipo Kelch , Factor 2 Relacionado con NF-E2/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Sulfóxidos , Espectrometría de Masas en Tándem
5.
Int J Cancer ; 133(11): 2642-9, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23686454

RESUMEN

Well-differentiated thyroid cancer most frequently occurs in premenopausal women. Greater exposure to estrogens may be a risk factor for thyroid cancer. To investigate the role of estrogens in thyroid cancer, a spot urine sample was obtained from 40 women with thyroid cancer and 40 age-matched controls. Thirty-eight estrogen metabolites, conjugates and DNA adducts were analyzed by using ultraperformance liquid chromatography/tandem mass spectrometry and the ratio of adducts to metabolites and conjugates was calculated for each sample. The ratio of depurinating estrogen-DNA adducts to estrogen metabolites and conjugates significantly differed between cases and controls (p < 0.0001), demonstrating high specificity and sensitivity. These findings indicate that estrogen metabolism is unbalanced in thyroid cancer and suggest that formation of estrogen-DNA adducts might play a role in the initiation of thyroid cancer.


Asunto(s)
Aductos de ADN/orina , Estrógenos/metabolismo , Estrógenos/orina , Neoplasias de la Tiroides/orina , Adulto , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/orina , Cromatografía Liquida , Femenino , Humanos , Persona de Mediana Edad , Estadificación de Neoplasias , Factores de Riesgo , Espectrometría de Masas en Tándem , Neoplasias de la Tiroides/patología
6.
IUBMB Life ; 64(2): 169-79, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22162200

RESUMEN

Depurinating DNA adducts formed by aromatic hydrocarbons and catechol estrogen quinones play a major role in cancer initiation. Most of these adducts depurinate instantaneously, but some guanine adducts depurinate from DNA with half-lives of hours. We report here, that after 10 h at 37 °C, reaction of estradiol-3,4-quinone (E(2)-3,4-Q) with ds-DNA to yield N7Gua and N3Ade adducts was complete and more efficient than with ss-DNA. When E(2)-3,4-Q reacted with t-RNA, no adducts were detected after 10 h, and the level of N3Ade and N7Gua adducts after 10 days was less than half that with ss-DNA after 10 h. Reaction of E(2)-3,4-Q and dG yielded 4-OHE(2)-1-N7dG, which spontaneously depurinated to yield 4-OHE(2)-1-N7Gua. To investigate the mechanism of depurination, E(2)-3,4-Q was reacted with carbocyclicdeoxyguanosine, in which the ring oxygen of the deoxyribose moiety is substituted with CH(2) , and depurination was observed. The results from this experiment demonstrate that the oxocarbenium ion mechanism plays the major role in depurination and provides the first experimental evidence for this mechanism. A newly discovered ß-elimination mechanism also plays a minor role in depurination. Understanding why the depurinating estrogen-DNA adducts come from DNA, and not from RNA, underscores the critical role that these adducts play in initiating cancer.


Asunto(s)
Carcinógenos/química , Transformación Celular Neoplásica , Aductos de ADN/química , Daño del ADN , Estradiol/análogos & derivados , Ciclopentanos/química , Aductos de ADN/síntesis química , ADN de Cadena Simple/química , Desoxiguanosina/análogos & derivados , Desoxiguanosina/química , Estradiol/química , Humanos , Concentración de Iones de Hidrógeno , Cinética , Purinas/química , ARN de Transferencia/química
7.
Drug Discov Today Dis Mech ; 9(1-2): e55-e69, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-26246832

RESUMEN

Metabolism of estrogens via the catechol estrogen pathway is characterized by a balanced set of activating and protective enzymes (homeostasis). Disruption of homeostasis, with excessive production of catechol estrogen quinones, can lead to reaction of these quinones with DNA to form depurinating estrogen-DNA adducts. Some of the mutations generated by these events can lead to initiation of breast cancer. A wealth of evidence, from studies of metabolism, mutagenicity, cell transformation and carcinogenicity, demonstrates that estrogens are genotoxic. Women at high risk for breast cancer, or diagnosed with the disease, have relatively high levels of depurinating estrogen-DNA adducts compared to normal-risk women. The dietary supplements N-acetylcysteine and resveratrol can inhibit formation of catechol estrogen quinones and their reaction with DNA to form estrogen-DNA adducts, thereby preventing initiation of breast cancer.

8.
Breast Cancer Res Treat ; 126(3): 779-89, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21170676

RESUMEN

Extensive data suggest that estradiol contributes to the development of breast cancer by acting as a mitogen and exerting direct genotoxic effects after enzymatic conversion to 4-hydroxyestradiol (4-OHE2) via cytochrome P450 1B1 (CYP1B1). The mammary gland, ovary, and uterus all express CYP1B1. Overexpression of this enzyme has been associated with an increased risk of breast cancer and blockade might reduce this carcinogenic effect. For this reason, we conducted systematic in vitro and in vivo studies of a CYP1B1 inhibitor, TMS (2,3',4,5'-tetramethoxystilbene). We found that TMS blocked the enzymatic conversion of radiolabeled estradiol to both 2-hydroxyestradiol (2-OHE2) and 4-OHE2, but did not inhibit Cyp1b1 message formation. In vivo studies using mass spectrometry showed that TMS inhibited formation of 2-OHE2 and 4-OHE2 and the resulting estrogen-DNA adducts. To examine its biologic actions in vivo, we investigated whether TMS could block the hyperplastic changes that occur in the developing breast of aromatase-transfected mice. We found that TMS induced a significant reduction of ductal structures in mice less than 6 months in age. In older mice, no reduction in breast morphology occurred. These latter studies uncovered unexpected estrogen agonistic actions of TMS at high doses, including a paradoxical stimulation of breast ductal structures and the endometrium. These studies suggest that the enzyme inhibitory properties of TMS, as well as the effects on developing breast, could implicate a role for TMS in breast cancer prevention, but only in low doses and on developing breast.


Asunto(s)
Glándulas Mamarias Animales/embriología , Estilbenos/farmacología , Animales , Aromatasa/metabolismo , Hidrocarburo de Aril Hidroxilasas/biosíntesis , Proliferación Celular , Citocromo P-450 CYP1A1/biosíntesis , Citocromo P-450 CYP1B1 , Estrógenos/metabolismo , Femenino , Letrozol , Glándulas Mamarias Animales/efectos de los fármacos , Ratones , Nitrilos/farmacología , Ovariectomía/métodos , Triazoles/farmacología
9.
IUBMB Life ; 63(12): 1087-93, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22045657

RESUMEN

The neurotransmitter dopamine is oxidized to its quinone (DA-Q), which at neutral pH undergoes intramolecular cyclization by 1,4-Michael addition, followed by oxidation to form leukochrome, then aminochrome, and finally neuromelanin. At lower pH, the amino group of DA is partially protonated, allowing the competitive intermolecular 1,4-Michael addition with nucleophiles in DNA to form the depurinating adducts, DA-6-N3Ade and DA-6-N7Gua. Catechol estrogen-3,4-quinones react by 1,4-Michael addition to form the depurinating 4-hydroxyestrone(estradiol)-1-N3Ade [4-OHE1(E2)-1-N3Ade] and 4-OHE1(E2)-1-N7Gua adducts, which are implicated in the initiation of breast and other human cancers. The effect of pH was studied by reacting tyrosinase-activated DA with DNA and measuring the formation of depurinating adducts. The most adducts were formed at pH 4, 5, and 6, and their level was nominal at pH 7 and 8. The N3Ade adduct depurinated instantaneously, but N7Gua had a half-life of 3 H. The slow loss of the N7Gua adduct is analogous to that observed in previous studies of natural and synthetic estrogens. The antioxidants N-acetylcysteine and resveratrol efficiently blocked formation of the DA-DNA adducts. Thus, slightly acidic conditions render competitive the reaction of DA-Q with DNA to form depurinating adducts. We hypothesize that formation of these adducts could lead to mutations that initiate Parkinson's disease. If so, use of N-acetylcysteine and resveratrol as dietary supplements may prevent initiation of this disease.


Asunto(s)
Antioxidantes/química , Aductos de ADN/química , Aductos de ADN/genética , Dopamina/química , Monofenol Monooxigenasa/química , Enfermedad de Parkinson/genética , Acetilcisteína/química , Benzoquinonas/química , Benzoquinonas/metabolismo , Aductos de ADN/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Cinética , Melatonina/química , Enfermedad de Parkinson/patología , Resveratrol , Estilbenos/química , Ácido Tióctico/química
10.
Biomarkers ; 16(5): 434-44, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21692648

RESUMEN

Damage to DNA by dopamine quinone and/or catechol estrogen quinones may play a significant role in the initiation of Parkinson's disease (PD). Depurinating estrogen-DNA adducts are shed from cells and excreted in urine. The aim of this study was to discover whether higher levels of estrogen-DNA adducts are associated with PD. Forty estrogen metabolites, conjugates, and DNA adducts were analyzed in urine samples from 20 PD cases and 40 matched controls by using ultra performance liquid chromatography/tandem mass spectrometry. The levels of adducts in cases versus controls (P < 0.005) suggest that unbalanced estrogen metabolism could play a causal role in the initiation of PD.


Asunto(s)
Encéfalo/metabolismo , Estrógenos/metabolismo , Enfermedad de Parkinson/etiología , Adulto , Anciano , Anciano de 80 o más Años , Cromatografía Liquida/métodos , Aductos de ADN/metabolismo , Estrógenos/orina , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedad de Parkinson/metabolismo , Extracción en Fase Sólida , Espectrometría de Masas en Tándem
11.
IUBMB Life ; 62(10): 746-51, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20945454

RESUMEN

Bisphenol A (BPA) displays weak estrogenic properties and could be a weak carcinogen by a mechanism similar to that of estrone (E(1)), estradiol (E(2)) and the synthetic estrogen diethylstilbestrol, a human carcinogen. A wide variety of scientific evidence supports the hypothesis that certain estrogen metabolites, predominantly catechol estrogen-3,4-quinones, react with DNA to cause mutations that can lead to the initiation of cancer. One of the major pathways of estrogen metabolism leads to the 4-catechol estrogens, 4-OHE(1)(E(2)), which are oxidized to their quinones, E(1)(E(2))-3,4-Q. The quinones react with DNA to form predominantly the depurinating adducts 4-OHE(1)(E(2))-1-N3Ade and 4-OHE(1)(E(2))-1-N7Gua. This process constitutes the predominant pathway in the initiation of cancer by estrogens. One pathway of BPA metabolism is hydroxylation of one of its symmetric benzene rings to form its catechol, 3-OHBPA. Subsequent oxidation to BPA-3,4-quinone would lead to reaction with DNA to form predominantly the depurinating adducts 3-OHBPA-6-N3Ade and 3-OHBPA-6-N7Gua. The resulting apurinic sites in the DNA could generate mutations in critical genes that can initiate human cancers. The catechol of BPA may also alter expression of estrogen-activating and deactivating enzymes, and/or compete with methoxylation of 4-OHE(1)(E(2)) by catechol-O-methyltransferase, thereby unbalancing the metabolism of estrogens to increase formation of E(1)(E(2))-3,4-Q and the depurinating estrogen-DNA adducts leading to cancer initiation. Thus, exposure to BPA could increase the risk of developing cancer by direct and/or indirect mechanisms. Knowledge of these mechanisms would allow us to begin to understand how BPA may act as a weak carcinogen and would be useful for regulating its use.


Asunto(s)
Biomarcadores de Tumor/orina , Estrógenos de Catecol/metabolismo , Estrógenos/metabolismo , Fenoles/orina , Compuestos de Bencidrilo , Carcinógenos/metabolismo , Catecol O-Metiltransferasa/metabolismo , ADN/genética , ADN/metabolismo , Aductos de ADN/metabolismo , Dietilestilbestrol/metabolismo , Estradiol/metabolismo , Estrógenos no Esteroides/metabolismo , Humanos , Mutación , Fenoles/metabolismo , Quinonas/metabolismo
12.
Future Oncol ; 6(1): 75-91, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20021210

RESUMEN

Experiments on estrogen metabolism, formation of DNA adducts, mutagenicity, cell transformation and carcinogenicity have led to and supported the hypothesis that the reaction of specific estrogen metabolites, mostly the electrophilic catechol estrogen-3,4-quinones, with DNA can generate the critical mutations to initiate breast and other human cancers. Analysis of depurinating estrogen-DNA adducts in urine demonstrates that women at high risk of, or with breast cancer, have high levels of the adducts, indicating a critical role for adduct formation in breast cancer initiation. Men with prostate cancer or non-Hodgkin lymphoma also have high levels of estrogen-DNA adducts. This knowledge of the first step in cancer initiation suggests the use of specific antioxidants that can block formation of the adducts by chemical and biochemical mechanisms. Two antioxidants, N-acetylcysteine and resveratrol, are prime candidates to prevent breast and other human cancers because in various M in vitro and in vivo experiments, they reduce the formation of estrogen-DNA adducts.


Asunto(s)
Neoplasias de la Mama/etiología , Neoplasias de la Mama/prevención & control , Aductos de ADN/metabolismo , Estrógenos/metabolismo , Acetilcisteína/farmacología , Animales , Antineoplásicos/farmacología , Antioxidantes/farmacología , Aductos de ADN/efectos adversos , Femenino , Humanos , Masculino , Neoplasias/etiología , Neoplasias/prevención & control , Resveratrol , Estilbenos/farmacología
13.
Int J Cancer ; 124(6): 1276-84, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19089919

RESUMEN

Diethylstilbestrol (DES) is a human carcinogen, based on sufficient epidemiological evidence. DES is mainly metabolized to its catechol, 3'-hydroxyDES (3'-OH-DES), which can further oxidize to DES-3',4'-quinone (DES-3',4'-Q). Similarly to estradiol-3,4-quinone, the reaction of DES-3',4'-Q with DNA would form the depurinating 3'-OH-DES-6'-N3Ade and 3'-OH-DES-6'-N7Gua adducts. To prove this hypothesis, synthesis of DES-3',4'-Q by oxidation of 3'-OH-DES with Ag(2)O was tried; this failed due to instantaneous formation of a spiro-quinone. Oxidation of 3'-OH-DES by lactoperoxidase or tyrosinase in the presence of DNA led to the formation of 3'-OH-DES-6'-N3Ade and 3'-OH-DES-6'-N7Gua adducts. These adducts were tentatively identified by LC-MS/MS as 3'-OH-DES-6'-N3Ade, m/z = 418 [M+H](+), and 3'-OH-DES-6'-N7Gua, m/z = 434 [M+H](+). Demonstration of their structures derived from their oxidation by MnO(2) to the DES quinone adducts and subsequent tautomerization to the dienestrol (DIES) catechol adducts, which are identical to the standard 3'-OH-DIES-6'-N3Ade, m/z = 416 [M+H](+), and 3'-OH-DIES-6'-N7Gua, m/z = 432 [M+H](+), adducts. The reaction of DIES-3',4'-Q or lactoperoxidase-activated 3'-OH-DIES with DNA did not produce any depurinating adducts, due to the dienic chain being perpendicular to the phenyl planes, which impedes the intercalation of DIES into the DNA. Enzymic oxidation of 3'-OH-DES suggests that the catechol of DES intercalates into DNA and is then oxidized to its quinone to yield N3Ade and N7Gua adducts. These results suggest that the common denominator of tumor initiation by the synthetic estrogen DES and the natural estrogen estradiol is formation of their catechol quinones, which react with DNA to afford the depurinating N3Ade and N7Gua adducts.


Asunto(s)
Aductos de ADN , Dietilestilbestrol/toxicidad , Carcinógenos/toxicidad , Catecoles/metabolismo , ADN/metabolismo , Dietilestilbestrol/química , Dietilestilbestrol/metabolismo , Estrógenos/síntesis química , Estrógenos/química , Modelos Moleculares , Oxidación-Reducción , Quinonas/síntesis química , Quinonas/química , Quinonas/metabolismo , Espectrometría de Masa Bombardeada por Átomos Veloces
14.
Prostate ; 69(1): 41-8, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18816637

RESUMEN

BACKGROUND: Although the estrogens estrone and estradiol are recognized to play very important roles in the risk of developing prostate cancer (Pca), the molecular mechanism by which estrogens initiate and/or promote Pca is still unknown. Substantial evidence supports that specific metabolites of estrogens, catechol estrogen quinones, can react with DNA to form depurinating estrogen-DNA adducts. Apurinic sites derived from depurination of these adducts can induce mutations leading to cancer. Once released from DNA, depurinating estrogen-DNA adducts are shed from cells into the bloodstream and excreted in urine. By analyzing profiles of estrogen metabolites, conjugates, and depurinating DNA adducts in urine from men with and without prostate cancer, potential biomarkers of Pca can be detected. The goal of this case-control study was to detect and identify potential biomarkers of Pca. METHODS: Urine samples from fourteen cases, men diagnosed with Pca, and 125 controls, men who had not been diagnosed with Pca, were partially purified by solid phase extraction and analyzed by ultraperformance liquid chromatography/tandem mass spectrometry. The urinary levels of androgens, estrogens, estrogen metabolites, conjugates and depurinating DNA adducts were measured. RESULTS: The ratio of depurinating estrogen-DNA adducts to the sum of the corresponding estrogen metabolites and conjugates was significantly higher in cases (median: 57.34) compared to controls (median: 23.39) (P < 0.001). CONCLUSIONS: This study suggests that depurinating estrogen-DNA adducts could serve as potential biomarkers to predict risk of Pca. They also could be useful tools for early clinical diagnosis and development of suitable strategies to prevent Pca.


Asunto(s)
Andrógenos/orina , Biomarcadores de Tumor/orina , Aductos de ADN/orina , Estrógenos/orina , Neoplasias de la Próstata/epidemiología , Neoplasias de la Próstata/orina , Anciano , Anciano de 80 o más Años , Andrógenos/biosíntesis , Estudios de Casos y Controles , Aductos de ADN/biosíntesis , Diagnóstico Precoz , Estrógenos/biosíntesis , Humanos , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Neoplasias de la Próstata/diagnóstico , Factores de Riesgo
15.
Biomarkers ; 14(7): 502-12, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19863189

RESUMEN

A variety of evidence suggests that estrogens may induce non-Hodgkin lymphoma (NHL). The reaction of catechol estrogen quinones with DNA to form depurinating estrogen-DNA adducts is hypothesized to initiate this process. These adducts are released from DNA, shed from cells into the bloodstream and excreted in urine. The aim of this study was to determine whether or not the depurinating estrogen-DNA adducts might be involved in the aetiology of human NHL. Estrogen metabolites, conjugates and depurinating DNA adducts were identified and quantified in spot urine samples from 15 men with NHL and 30 healthy control men by using ultraperformance liquid chromatography/tandem mass spectrometry. The levels of estrogen-DNA adducts were significantly higher in the men with NHL than in the healthy control men. Thus, formation of estrogen-DNA adducts may play a critical role in the aetiology of NHL, and these adducts could be potential biomarkers of NHL risk.


Asunto(s)
Biomarcadores de Tumor/orina , Aductos de ADN/orina , Estrógenos/orina , Linfoma no Hodgkin/diagnóstico , Anciano , Cromatografía Líquida de Alta Presión , ADN/química , ADN/orina , Aductos de ADN/química , Estrógenos/química , Humanos , Linfoma no Hodgkin/tratamiento farmacológico , Linfoma no Hodgkin/orina , Masculino , Persona de Mediana Edad , Estructura Molecular , Espectrometría de Masas en Tándem
16.
Mol Cancer Ther ; 7(1): 19-26, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18187806

RESUMEN

Using a single nucleotide polymorphism association study in 52 men with prostate cancer receiving docetaxel, we found that individuals carrying two copies of the CYP1B1*3 polymorphic variant had a poor prognosis after docetaxel-based therapies compared with individuals carrying at least one copy of the CYP1B1*1 allele (30.6 versus 12.8 months; P=0.0004). The association between CYP1B1*3 and response to therapy was not observed in similar subjects receiving non-taxane-based therapy (P=0.18). The systemic clearance of docetaxel was also unrelated to CYP1B1 genotype status (P=0.39), indicating that the association of CYP1B1*3 with clinical response is not due to docetaxel metabolism. To explain these results, we hypothesized that an indirect gene-drug interaction was interfering with the primary mechanism of action of docetaxel, tubulin polymerization. We therefore conducted tubulin polymerization experiments with taxanes in the presence or absence of certain CYP1B1 estrogen metabolites, which are known to bind to nucleophilic sites in proteins and DNA, that revealed the primary estrogen metabolite of CYP1B1, 4-hydroxyestradiol (4-OHE2), when oxidized to estradiol-3,4-quinone strongly inhibits tubulin polymerization. The 4-OHE2 is also formed more readily by the protein encoded by the CYP1B1*3 allele, validating further our data in patients. Furthermore, estradiol-3,4-quinone reacted in vitro with docetaxel to form the 4-OHE2-docetaxel adduct. This pilot study provides evidence that CYP1B1*3 may be an important marker for estimating docetaxel efficacy in patients with prostate cancer. This link is likely associated with CYP1B1*3 genotype-dependent estrogen metabolism.


Asunto(s)
Alelos , Hidrocarburo de Aril Hidroxilasas/genética , Hidrocarburo de Aril Hidroxilasas/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Taxoides/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Citocromo P-450 CYP1B1 , Docetaxel , Estrógenos/química , Estrógenos/metabolismo , Genotipo , Humanos , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Tasa de Supervivencia , Taxoides/química , Taxoides/farmacocinética , Tubulina (Proteína)/metabolismo
17.
Horm Cancer ; 10(2-3): 77-88, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30877616

RESUMEN

Androgens are thought to cause prostate cancer, but the underlying mechanisms are unclear. Data from animal studies suggest that for androgens to cause prostate cancer, they must be aromatized to estrogen and act in concert with estrogen metabolites. We tested the hypothesis that androgen-receptor and estrogen receptor-mediated effects of androgen and estrogen are necessary, as well as genotoxicity of estrogen metabolites. NBL rats were treated with androgenic and estrogenic compounds for 16-75 weeks through slow-release silastic implants or pellets. Testosterone alone induced cancer in the prostate of 37% of rats. 5α-Dihydrotestosterone, which cannot be converted to estradiol or testosterone, did not cause a significant prostate cancer incidence (4%). Addition of estradiol to 5α-dihydrotestosterone treatment did not markedly enhance prostate cancer incidence (14%), unlike adding estradiol to testosterone treatment which induced a 100% tumor incidence. Testosterone plus estradiol treatment induced a DNA adduct detectable by 32P-postlabeling, oxidative DNA damage (8-hydroxyguanosine), and lipid peroxidation at the site within the prostate where this treatment causes cancers, preceding later cancer formation. The non-estrogenic 4-hydroxy metabolite of estradiol, when combined with testosterone, induced prostatic dysplasia within 16 weeks and, after long-term treatment, a very low incidence of prostate cancer (21%). When an estrogen that cannot be hydroxylated (2-fluoroestradiol) was added to this combined treatment with testosterone and 4-hydroxyestradiol, dysplasia frequency after 16 weeks was doubled. These results strongly support the hypothesis, but additional definitive studies are needed which may identify new targets to interfere with these mechanisms that are clinically feasible in humans.


Asunto(s)
Andrógenos/efectos adversos , Carcinogénesis , Estrógenos/efectos adversos , Neoplasias de la Próstata/inducido químicamente , Animales , Carcinoma , Aductos de ADN , Daño del ADN , Dihidrotestosterona/metabolismo , Estradiol/metabolismo , Estrógenos de Catecol/química , Guanosina/análogos & derivados , Guanosina/farmacología , Humanos , Incidencia , Masculino , Próstata , Ratas , Receptores de Estrógenos/metabolismo , Testosterona/metabolismo
18.
Int J Cancer ; 122(9): 1949-57, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18098283

RESUMEN

Estrogens can become endogenous carcinogens via formation of catechol estrogen quinones, which react with DNA to form specific depurinating estrogen-DNA adducts. The mutations resulting from these adducts can lead to cell transformation and the initiation of breast cancer. Estrogen metabolites, conjugates and depurinating DNA adducts in urine samples from 46 healthy control women, 12 high-risk women and 17 women with breast cancer were analyzed. The estrogen metabolites, conjugates and depurinating DNA adducts were identified and quantified by using ultraperformance liquid chromatography/tandem mass spectrometry. The levels of the ratios of depurinating DNA adducts to their respective estrogen metabolites and conjugates were significantly higher in high-risk women (p < 0.001) and women with breast cancer (p < 0.001) than in control subjects. The high-risk and breast cancer groups were not significantly different (p = 0.62). After adjusting for patient characteristics, these ratios were still significantly associated with health status. Thus, the depurinating estrogen-DNA adducts are possible biomarkers for early detection of breast cancer risk and response to preventive treatment.


Asunto(s)
Biomarcadores de Tumor/orina , Neoplasias de la Mama/orina , Aductos de ADN/orina , Estrógenos Conjugados (USP)/orina , Adulto , Anciano , Análisis de Varianza , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/etiología , Neoplasias de la Mama/metabolismo , Cromatografía Liquida , Estudios Transversales , Aductos de ADN/metabolismo , Estrógenos Conjugados (USP)/metabolismo , Femenino , Humanos , Italia , Modelos Lineales , Persona de Mediana Edad , Modelos Estadísticos , Valores de Referencia , Medición de Riesgo , Factores de Riesgo , Espectrometría de Masas en Tándem , Estados Unidos
19.
Free Radic Biol Med ; 45(2): 136-45, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18423413

RESUMEN

Resveratrol (Resv), a natural occurring phytolexin present in grapes and other foods, possesses chemopreventive effects revealed by its striking modulation of diverse cellular events associated with tumor initiation, promotion, and progression. Catechol estrogens generated in the metabolism of estrogens are oxidized to catechol quinones that react with DNA to form predominantly depurinating estrogen-DNA adducts. This event can generate the mutations responsible for cancer initiation. In this regard, Resv acts as both an antioxidant and an inducer of the phase II enzyme NAD(P)H:quinone oxidoreductase 1 (NQO1). In this report, we present the effects of Resv on the metabolism of estrogens in normal breast epithelial cells (MCF-10F) treated with 4-hydroxyestradiol (4-OHE(2)) or estradiol-3,4-quinone (E(2)-3,4-Q). Resv induced NQO1 in a dose- and time-dependent manner, but did not affect the expression of catechol-O-methyltransferase. Ultraperformance liquid chromatography/tandem mass spectrometry was used to determine the effects of Resv on estrogen metabolism. Preincubation of the cells with Resv for 48 h decreased the formation of depurinating estrogen-DNA adducts from 4-OHE(2) or E(2)-3,4-Q and increased formation of methoxycatechol estrogens. When Resv was also present with the 4-OHE(2) or E(2)-3,4-Q, even greater increases in methoxycatechol estrogens were observed, and the DNA adducts were undetectable. We conclude that Resv can protect breast cells from carcinogenic estrogen metabolites, suggesting that it could be used in breast cancer prevention.


Asunto(s)
Antioxidantes/farmacología , Neoplasias de la Mama/prevención & control , Aductos de ADN/efectos de los fármacos , Estrógenos/metabolismo , Estilbenos/farmacología , Línea Celular Tumoral , Cromatografía Liquida , Femenino , Humanos , NAD(P)H Deshidrogenasa (Quinona)/efectos de los fármacos , Resveratrol , Espectrometría de Masas en Tándem
20.
J Steroid Biochem Mol Biol ; 109(1-2): 22-30, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18226522

RESUMEN

A growing number of studies indicate that breast cancer initiation is related to abnormal estrogen oxidation to form an excess of estrogen-3,4-quinones, which react with DNA to form depurinating adducts and induce mutations. This mechanism is often called estrogen genotoxicity. 4-Catechol estrogens, precursors of the estrogen-3,4-quinones, were previously shown to account for most of the transforming and tumorigenic activity. We examined whether estrogen-induced transformation can be reduced by inhibiting the oxidation of a 4-catechol estrogen to its quinone. We demonstrate that E6 cells (a normal mouse epithelial cell line) can be transformed by a single treatment with a catechol estrogen or its quinone. The transforming activities of 4-hydroxyestradiol and estradiol-3,4-quinone were comparable. N-Acetylcysteine, a common antioxidant, inhibited the oxidation of 4-hydroxyestradiol to the quinone and consequent formation of DNA adducts. It also drastically reduced estrogen-induced transformation of E6 cells. These results strongly implicate estrogen genotoxicity in mammary cell transformation. Since N-acetylcysteine is well tolerated in clinical studies, it may be a promising candidate for breast cancer prevention.


Asunto(s)
Acetilcisteína/farmacología , Transformación Celular Neoplásica/efectos de los fármacos , Estrógenos de Catecol/toxicidad , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/patología , Animales , Línea Celular , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Ensayo de Unidades Formadoras de Colonias , Aductos de ADN/efectos de los fármacos , Aductos de ADN/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Estradiol/análogos & derivados , Estradiol/toxicidad , Estrógenos de Catecol/antagonistas & inhibidores , Femenino , Genes ras , Glándulas Mamarias Animales/metabolismo , Ratones , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA