Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Placenta ; 33(5): 424-32, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22385826

RESUMEN

Characterization of normal changes in the serum proteome during pregnancy may enhance understanding of maternal physiology and lead to the development of new gestational biomarkers. In 23 Nepalese pregnant women who delivered at term, two-dimensional difference in-gel electrophoresis (DIGE) was used to assess changes in relative protein abundance between paired serum samples collected in the first and third trimesters. One-hundred and forty-five of over 700 protein spots in DIGE gels (pI 4.2-6.8) exhibited nominally significant (p < 0.05) differences in abundance across trimesters. Additional filtering using a Bonferroni correction reduced the number of significant (p < 0.00019) spots to 61. Mass spectrometric analysis detected 38 proteins associated with gestational age, cytoskeletal remodeling, blood pressure regulation, lipid and nutrient transport, and inflammation. One new protein, pregnancy-specific ß-glycoprotein 4 was detected. A follow-up isotope tagging for relative and absolute quantitation (iTRAQ) experiment of six mothers from the DIGE study revealed 111 proteins, of which 11 exhibited significant (p < 0.05) differences between trimesters. Four of these proteins: gelsolin, complement C1r subcomponent, α-1-acid glycoprotein, and α-1B-glycoprotein also changed in the DIGE analysis. Although not previously associated with normal pregnancy, gelsolin decreased in abundance by the third trimester (p < 0.01) in DIGE, iTRAQ and Western analyses. Changes in abundance of proteins in serum that are associated with syncytiotrophoblasts (gelsolin, pregnancy-specific ß-1 glycoprotein 1 and ß-2-glycoprotein I) probably reflect dynamics of a placental proteome shed into maternal circulation during pregnancy. Measurement of changes in the maternal serum proteome, when linked with birth outcomes, may yield biomarkers for tracking reproductive health in resource poor settings in future studies.


Asunto(s)
Primer Trimestre del Embarazo/sangre , Tercer Trimestre del Embarazo/sangre , Proteoma , Western Blotting , Cromatografía Liquida , Femenino , Humanos , Desnutrición , Espectrometría de Masas , Nepal , Embarazo , Población Rural , Electroforesis Bidimensional Diferencial en Gel
2.
Cancer Res ; 61(20): 7488-94, 2001 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-11606384

RESUMEN

Many of the major identified risk factors for breast cancer are associated with exposure to endogenous estrogen. In addition to the effects of estrogen as a growth factor, experimental and epidemiological evidence suggest that catechol metabolites of estrogen also contribute to estrogen carcinogenesis by both direct and indirect genotoxic mechanisms. O-Methylation catalyzed by catechol-O-methyltransferase (COMT) is a Phase II metabolic inactivation pathway for catechol estrogens. We and others have found that a polymorphism in the COMT gene, which codes for a low activity variant of the COMT enzyme, is associated with an increased risk of developing breast cancer; therefore, the goal of the current study was to investigate the role of decreased COMT activity on estrogen catechol levels and on oxidative DNA damage, as measured by 8-hydroxy-2'-deoxyguanosine (8-oxo-dG) levels. MCF-7 cells were pretreated with dioxin as a means to increase estrogen metabolism to catechol estrogens, then treated with estradiol (E2) +/- Ro 41-0960, a COMT-specific inhibitor. After extraction from culture medium, estrogen metabolites were separated using an high-performance liquid chromatography-electrochemical detection method. As expected, dioxin dramatically increased E2 oxidative metabolism, primarily to its 2-OH and 2-methoxy metabolites. The COMT inhibitor blocked 2-methoxy E2 formation. This was associated with increased 2-hydroxy E2 (2-OH E2) and 8-oxo-dG levels. In the presence of COMT inhibition, increased oxidative DNA damage was detected in MCF-7 cells exposed to as low as 0.1 microM E2, whereas in the absence of COMT inhibition, no increase in 8-oxo-dG was detected at E2 concentrations < or =10 microM. This study is the first to show that O-methylation of 2-OH E2 by COMT is protective against oxidative DNA damage caused by 2-OH E2, a major oxidative metabolite of E2.


Asunto(s)
Neoplasias de la Mama/metabolismo , Inhibidores de Catecol O-Metiltransferasa , Daño del ADN , Desoxiguanosina/análogos & derivados , Estradiol/metabolismo , Estradiol/farmacología , Estrógenos de Catecol/metabolismo , 8-Hidroxi-2'-Desoxicoguanosina , Benzofenonas/farmacología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , ADN de Neoplasias/metabolismo , Desoxiguanosina/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Estrés Oxidativo/fisiología , Células Tumorales Cultivadas
3.
Carcinogenesis ; 22(10): 1661-5, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11577006

RESUMEN

Catechol-O-methyltransferase (COMT) catalyzes the O-methylation of catechol estrogens (CEs), using S-adenosylmethionine (SAM) as a methyl donor. Several studies have indicated that the val108met COMT polymorphism, which results in a 3-4-fold decrease in activity, is associated with increased breast cancer risk. Folate, whose intake levels have also been associated with breast cancer risk, and other micronutrients in the folate metabolic pathway influence levels of SAM and S-adenosylhomocysteine (SAH), a COMT inhibitor generated by the demethylation of SAM. Because these micronutrients have been shown to alter SAM and SAH levels, we hypothesized that they could also affect COMT-catalyzed CE methylation. Although measurements of SAM and SAH were not initially collected, a secondary analysis of data from two nested case-control studies was performed to examine whether serum levels of folate, vitamin B12 (B12), pyridoxal 5'-phosphate (PLP), cysteine and homocysteine, in conjunction with COMT genotype, were associated with breast cancer risk. COMT(HH) (high activity COMT homozygote) breast cancer cases had statistically significantly lower levels of homocysteine (P = 0.05) and cysteine (P = 0.04) and higher levels of PLP (P = 0.02) than COMT(HH) controls. In contrast, COMT(LL) (low activity COMT homozygote) cases had higher levels of homocysteine than COMT(LL) controls (P = 0.05). No associations were seen between B12, COMT genotype, and breast cancer risk. An increasing number of COMT(L) alleles was significantly associated with increased breast cancer risk in women with below median levels of folate (P(trend) = 0.05) or above median levels of homocysteine (P(trend) = 0.02). These findings are consistent with a role for certain folate pathway micronutrients in mediating the association between COMT genotype and breast cancer risk.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias de la Mama/etiología , Catecol O-Metiltransferasa/genética , Ácido Fólico/sangre , Micronutrientes/efectos adversos , Estudios de Casos y Controles , Catecol O-Metiltransferasa/metabolismo , Cisteína/sangre , Estrógenos de Catecol/metabolismo , Femenino , Genotipo , Homocisteína/sangre , Humanos , Metilación , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Polimorfismo de Longitud del Fragmento de Restricción , Fosfato de Piridoxal/sangre , Factores de Riesgo , S-Adenosilmetionina/metabolismo , Vitamina B 12/sangre
4.
J Natl Cancer Inst Monogr ; (27): 67-73, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10963620

RESUMEN

A common thread linking the main risks for developing breast cancer in women is cumulative, excessive exposure to estrogen. The standard paradigm to account for this association focuses on increased cell proliferation caused by estrogen through estrogen receptor-mediated signal transduction accompanied by increased probability for mutation to occur during DNA synthesis. This chapter provides an overview of the mounting evidence, provided from cell culture and whole animal experimental studies, in support of a role for the oxidative metabolites of estrogen, in particular, the catechol estrogens, in the development of estrogen carcinogenesis. This provides a paradigm for how estrogens may contribute to the development of human breast cancer. The chapters that follow will fill in the details. Evidence shows that the catechols themselves are signaling molecules that work through the estrogen receptor. In addition, upon further oxidation, the catechols can give rise to reactive quinones capable of forming direct adducts with glutathione and purines in DNA and of redox cycling to generate reactive oxygen species that can cause oxidative damage. Estradiol and estrone, as well as their 4-hydroxy catechols, are carcinogenic in the Syrian golden hamster kidney, and ethinyl estradiol is a strong promoter of hepatocarcinogenesis in the rat. Increased oxidative DNA damage has been detected in target tissues after estrogen treatment in both animal model systems. Furthermore, several recent molecular epidemiologic studies have found that a polymorphism associated with a low-activity form of catechol-O-methyltransferase, an enzyme involved in the inactivation of catechol estrogens, is associated with an increased risk for developing breast cancer. The increased risk is observed in certain women, although the studies are not consistent on which subgroup of women (e.g., premenopausal or postmenopausal) is at increased risk, and one study detected no increased risk. Reasons for such discrepancies are discussed in light of factors, such as genetic polymorphisms and environmental/lifestyle susceptibility factors, which control the tissue-specific balance within cells among the estrogen metabolites. It is concluded that such factors will have to be identified through additional mechanistic studies and that, as they are identified, they can be incorporated into future molecular epidemiologic studies designed to determine their actual impact on cancer risk in human populations.


Asunto(s)
Biotransformación , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Estrógenos de Catecol/metabolismo , Estrógenos/metabolismo , Animales , Neoplasias de la Mama/epidemiología , Cricetinae , Daño del ADN , Femenino , Humanos , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Modelos Biológicos , Epidemiología Molecular , Oxidación-Reducción , Polimorfismo Genético , Ratas , Especies Reactivas de Oxígeno , Factores de Riesgo
5.
J Natl Cancer Inst Monogr ; (27): 95-112, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10963622

RESUMEN

Estrogen exposure represents the major known risk factor for development of breast cancer in women and is implicated in the development of prostate cancer in men. Human breast tissue has been shown to be a site of oxidative metabolism of estrogen due to the presence of specific cytochrome P450 enzymes. The oxidative metabolism of 17beta-estradiol (E2) to E2-3,4-quinone metabolites by an E2-4-hydroxylase in breast tissue provides a rational hypothesis to explain the mammary carcinogenic effects of estrogen in women because this metabolite is directly genotoxic and can undergo redox cycling to form genotoxic reactive oxygen species. In this chapter, evidence in support of this hypothesis and of the role of P4501B1 as the 4-hydroxylase expressed in human breast tissue is reviewed. However, the plausibility of this hypothesis has been questioned on the grounds that insufficient E2 is present in breast tissue to be converted to biologically significant amounts of metabolite. This critique is based on the assumption that plasma and tissue E2 levels are concordant. However, breast cancer tissue E2 levels are 10-fold to 50-fold higher in postmenopausal women than predicted from plasma levels. Consequently, factors must be present to alter breast tissue E2 levels independently of plasma concentrations. One such factor may be the local production of E2 in breast tissue through the enzyme aromatase, and the evidence supporting the expression of aromatase in breast tissue is also reviewed in this chapter. If correct, mutations or environmental factors enhancing aromatase activity might result in high tissue concentrations of E2 that would likely be sufficient to serve as substrates for CYP1B1, given its high affinity for E2. This concept, if verified experimentally, would provide plausibility to the hypothesis that sufficient E2 may be present in tissue for formation of catechol metabolites that are estrogenic and which, upon further oxidative metabolism, form genotoxic species at levels that may contribute to estrogen carcinogenesis.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas , Neoplasias de la Mama/etiología , Neoplasias de la Mama/metabolismo , Mama/metabolismo , Carcinoma/etiología , Carcinoma/metabolismo , Estrógenos/metabolismo , Aromatasa/metabolismo , Aromatasa/fisiología , Mama/enzimología , Neoplasias de la Mama/enzimología , Carcinoma/enzimología , Citocromo P-450 CYP1B1 , Sistema Enzimático del Citocromo P-450/metabolismo , Estrógenos/biosíntesis , Femenino , Humanos , Hidroxilación , Modelos Químicos , Oxidación-Reducción
6.
Carcinogenesis ; 21(6): 1205-11, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10837011

RESUMEN

Ethinyl estradiol (EE) is a strong promoter of hepatocarcinogenesis in the rat. Treatment with EE and other hepatic promoters induces transient growth stimulation followed by growth inhibition (mitosuppression) in hepatocytes. Previously, we identified several genes whose transcript levels were increased during EE-induced mitosuppression, including transforming growth factor beta (TGF-beta), which inhibits growth and induces apoptosis in hepatocytes. Various hepatic promoters, including phenobarbital and several peroxisomal proliferators, have been shown to inhibit TGF-beta-induced apoptosis in rat hepatocytes. The goal of this study was to investigate whether EE is also an inhibitor of TGF-beta-induced apoptosis in rat hepatocytes. Several approaches to detect apoptosis were used, including the TUNEL assay, detection of high molecular weight DNA fragmentation by field inversion gel electrophoresis and determination of cytosolic cytochrome c levels by western analysis. TGF-beta-induced apoptosis in cultured, precision cut liver slices and hepatocytes of female rats. EE (

Asunto(s)
Apoptosis/efectos de los fármacos , Etinilestradiol/farmacología , Hígado/efectos de los fármacos , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Técnicas de Cultivo , Femenino , Hígado/citología , Ratas , Ratas Endogámicas F344
7.
Cancer Epidemiol Biomarkers Prev ; 9(12): 1373-6, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11142424

RESUMEN

A valine-108-methionine polymorphism in exon 4 of the catechol-O-methyltransferase (COMT) gene causes a 3- to 4-fold reduction in enzyme activity and has been associated with an increased risk of breast cancer. This increased risk may be attributable to a decreased ability of the protein encoded by the low-activity allele (COMT(L)) to methylate and inactivate catechol estrogens, which have been implicated in estrogen carcinogenesis. Because estrogens have also been implicated in the etiology of ovarian cancer, we analyzed 108 cases and 106 controls from a case-control study conducted in Mainz, Germany, to test the hypothesis that COMT(L) is associated with ovarian cancer risk. No significant association was found between the COMT genotype and ovarian cancer risk (for the intermediate-activity COMT genotype versus the high-activity COMT genotype, OR, 1.29; 95% CI, 0.63-2.64; for the low-activity COMT genotype versus the high-activity COMT genotype, OR, 1.17; 95% CI, 0.52-2.61). We also hypothesized that women who were both low-activity COMT genotype- and glutathione S-transferase (GST) M1- and/or T1 null would be at higher risk for ovarian cancer because the combination of these genotypes could theoretically lead to higher catechol estrogen exposure. However, the association between the COMT polymorphism and ovarian cancer risk was similar across GSTM1 and GSTT1 genotypes (Ptrend > 0.40, for all strata). Because of the small sample size of this study population, odds ratios of a small magnitude could not be completely ruled out; however, the results presented do not support a strong association between the COMT polymorphism and the risk of ovarian cancer.


Asunto(s)
Catecol O-Metiltransferasa/genética , Glutatión Transferasa/genética , Neoplasias Ováricas/enzimología , Polimorfismo Genético , Estudios de Casos y Controles , Femenino , Genotipo , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/genética
8.
Toxicol Sci ; 51(2): 224-35, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10543024

RESUMEN

Ethinyl estradiol (EE) is a strong promoter of hepatocarcinogenesis. Treatment of rats with EE and other hepatic promoters induces a mitosuppressed state characterized by decreased hepatocyte turnover and reduced growth responsiveness. Previously, we identified several nuclear and mitochondrial genome-encoded mitochondrial genes whose transcripts were increased during EE-induced hepatic mitosuppression in rats and in EE-treated HepG2 cells (Chen et al. Carcinogenesis, 17, 2783-2786, 1996 and Carcinogenesis, 19, 101-107, 1998). In both cultured rat hepatocytes and HepG2 cells, EE increased respiratory chain activity (reflected by increased mitochondrial superoxide production detected as increased lucigenin-derived chemiluminescence (LDCL). In this paper, we provide additional characterizations of these effects. Increased LDCL was detected in mitochondria isolated from EE-treated rats, documenting that these estrogen effects on mitochondrial function are not confined to cells in culture. EE and estradiol (E2) increased LDCL in cultured rat hepatocytes and HepG2 cells in a dose- (beginning at 0.25 microM levels) and time-dependent response. Inhibition of P450-mediated estrogen metabolism inhibited, while direct exposure to E2 catechol metabolites enhanced LDCL. Co-treatment with glutathione ester or with the specific antiestrogen, ICI 182708 inhibited LDCL. In contrast, estrogen-induced LDCL was enhanced by glutathione depletion, and by inhibition of catechol-o-methyltransferase. These results support a working hypothesis that in liver cells, increased respiratory chain activity induced by estrogen treatment requires both metabolism to catechols and an estrogen receptor-mediated signal transduction pathway.


Asunto(s)
Congéneres del Estradiol/toxicidad , Etinilestradiol/toxicidad , Hígado/efectos de los fármacos , Hígado/metabolismo , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Superóxidos/metabolismo , Animales , Carcinoma Hepatocelular/metabolismo , Catecol O-Metiltransferasa/metabolismo , Inhibidores de Catecol O-Metiltransferasa , Células Cultivadas , Estradiol/análogos & derivados , Estradiol/farmacología , Moduladores de los Receptores de Estrógeno/farmacología , Fulvestrant , Glutatión/deficiencia , Glutatión/metabolismo , Hígado/citología , Hígado/enzimología , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/metabolismo , Masculino , Ratas , Ratas Endogámicas F344 , Células Tumorales Cultivadas
9.
Cancer Epidemiol Biomarkers Prev ; 8(6): 525-32, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10385143

RESUMEN

A nested case-control study was conducted to examine the association between serum concentrations of 1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene (DDE), the primary metabolite of 1,1,1-trichloro-2,2-bis(p-chlorophenyl)ethane (DDT), and polychlorinated biphenyls (PCBs) and the development of breast cancer up to 20 years later. Cases (n = 346) and controls (n = 346) were selected from cohorts of women who donated blood in 1974, 1989, or both, and were matched on age, race, menopausal status, and month and year of blood donation. Analyses were stratified by cohort participation because median DDE and PCB concentrations among the controls were 59 and 147% higher in 1974 than 1989, respectively. Median concentrations of DDE were lower among cases than controls in both time periods [11.7% lower in 1974 (P = 0.06) and 8.6% lower in 1989 (P = 0.41)]. Median concentrations of PCBs were similar among cases and controls [P = 0.21 for 1974 and P = 0.37 for 1989 (Wilcoxon signed rank test)]. The risk of developing breast cancer among women with the highest concentrations of DDE was roughly half that among women with the lowest concentrations, whether based on concentrations in 1974 [odds ratio (OR), 0.50; 95% confidence interval (CI), 0.27-0.89; P(trend) = 0.02] or in 1989 (OR, 0.53; 95% CI, 0.24-1.17; P(trend) = 0.08). The associations between circulating concentrations of PCBs and breast cancer were less pronounced but still in the same direction (1974: OR, 0.68; 95% CI, 0.36-12.9; P(trend) = 0.2; and 1989: OR, 0.73; 95% CI, 0.37-1.46; P(trend) = 0.6). Adjustment for family history of breast cancer, body mass index, age at menarche or first birth, and months of lactation did not materially alter these associations. These associations remained consistent regardless of lactation history and length of the follow-up interval, with the strongest inverse association observed among women diagnosed 16-20 years after blood drawing. Results from this prospective, community-based nested case-control study are reassuring. Even after 20 years of follow-up, exposure to relatively high concentrations of DDE or PCBs showed no evidence of contributing to an increased risk of breast cancer.


Asunto(s)
Neoplasias de la Mama/sangre , Neoplasias de la Mama/inducido químicamente , Carcinógenos/efectos adversos , Carcinógenos/metabolismo , DDT/efectos adversos , DDT/sangre , Diclorodifenil Dicloroetileno/efectos adversos , Diclorodifenil Dicloroetileno/sangre , Contaminantes Ambientales/efectos adversos , Contaminantes Ambientales/sangre , Bifenilos Policlorados/efectos adversos , Bifenilos Policlorados/sangre , Adulto , Anciano , Bancos de Sangre , Estudios de Casos y Controles , Femenino , Humanos , Maryland , Persona de Mediana Edad , Estudios Prospectivos , Factores de Riesgo , Encuestas y Cuestionarios , Factores de Tiempo
10.
Biochem Biophys Res Commun ; 244(1): 298-303, 1998 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-9514920

RESUMEN

Reactive oxygen species are known to induce strand breaks and/or base modifications in DNA. DNA strand breaks are associated with many pathologies and programmed cell death. We have examined the ability of the polyamines and their analogues to protect phi X-174 plasmid DNA from strand breakage induced by a oxygen-radical generating system. Spermine and several unsymmetrically substituted polyamine analogues reduced the amount of strand breakage at a physiologically relevant concentration of 1 mM. However, putrescine, spermidine, N1-acetylspermine, N1-acetylspermidine and symmetrically alkylated polyamine analogues were not able to reduce strand breakage at the same concentration. Thus, the unsymmetrically alkylated polyamine analogues and natural spermine can protect DNA against strand breakage induced by Cu(II)/H2O2 generated ROS similar to other more classical antioxidants.


Asunto(s)
Antioxidantes/farmacología , Poliaminas Biogénicas/química , Poliaminas Biogénicas/farmacología , Daño del ADN/efectos de los fármacos , Especies Reactivas de Oxígeno , Antineoplásicos/farmacología , Bacteriófago phi X 174/efectos de los fármacos , Bacteriófago phi X 174/genética , Cobre/farmacología , Peróxido de Hidrógeno/farmacología , Poliaminas/farmacología , Putrescina/farmacología , Espermidina/farmacología , Espermina/análogos & derivados , Espermina/farmacología , Relación Estructura-Actividad
11.
Carcinogenesis ; 19(12): 2187-93, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9886577

RESUMEN

Ethinyl estradiol (EE) is a strong hepatic promoter and weak complete hepatocarcinogen. Among the effects on rat liver caused by chronic exposure to non-hepatotoxic doses of EE is an initial, transient increase in hepatocyte growth followed by a subsequent inhibition (mitosuppression) of basal and/or induced liver growth. To investigate the mechanism of EE-induced mitosuppression, we performed a differential display and identified 10 genes whose expression was increased 2- to 4-fold in EE-induced, mitosuppressed livers (Chen et al., Carcinogenesis, 17, 2783-2786, 1996). We found that one of these clones was homologous to nuclear genome-encoded mitochondrial ATP synthase subunit E. Here, we describe the identification of two additional cDNAs representing transcripts whose levels were elevated during EE-induced mitosuppression as mitochondrial DNA-encoded cytochrome c oxidase subunit III and ATP synthase 6. In addition, we found that EE, estradiol and the estradiol catechol metabolites, 4-OH-estradiol and 2-OH-estradiol, increased the levels of these and other mitochondrial genome-encoded transcripts in human hepatoma HepG2 cells. We also observed that this increase can be blocked by inhibition of cytochrome P450-mediated estrogen metabolism, and that this increase is accompanied by increased mitochondrial superoxide production, which reflects increased respiratory chain activity.


Asunto(s)
Carcinógenos/toxicidad , ADN Mitocondrial/efectos de los fármacos , ADN Mitocondrial/metabolismo , Etinilestradiol/toxicidad , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/metabolismo , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , ARN Mensajero/metabolismo , Superóxidos/metabolismo , Animales , Catecoles/metabolismo , Catecoles/toxicidad , ADN Complementario/efectos de los fármacos , ADN Complementario/genética , ADN Complementario/metabolismo , ADN Mitocondrial/genética , Estradiol/toxicidad , Femenino , Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Hepáticas Experimentales/genética , Mitocondrias Hepáticas/genética , ATPasas de Translocación de Protón/genética , Ratas , Ratas Endogámicas F344 , Células Tumorales Cultivadas
13.
Arch Biochem Biophys ; 347(1): 45-52, 1997 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-9344463

RESUMEN

Catechol estrogens are genotoxic, indirectly through redox cycling mechanisms leading to oxidative DNA damage and directly by formation of quinone-DNA adducts. Previously, we demonstrated that Cu2+ can oxidize estradiol (E2) catechols, establishing a copper redox cycle leading to the formation of DNA strand breaks. The goal of this study was to use electron spin resonance techniques to identify the free radical intermediates formed. The 2- and 4-OH catechols of E2 and ethinyl estradiol (EE) were oxidized to semiquinone intermediates, stabilized by Mg2+, when incubated with Cu2+. The 4-OH-EE semiquinone decayed more slowly than the 2-OH-EE semiquinone. Using the spin trap alpha-(4-pyridyl-1-oxide)-N-tert-butylnitrone, 4-OH-E2 plus Cu2+ generated hydroxyl radicals at a greater rate than 2-OH-E2 plus Cu2+. Formation of hydroxyl and methyl radical adducts was detected, using 5,5-dimethyl-1-pyrroline-N-oxide as the spin trap, when 2-OH-E2 was incubated with Cu2+ and 1% dimethyl sulfoxide. This was inhibited by the Cu1+ chelator bathocuproinedisulfonic acid and catalase. These data demonstrate that the oxidation of estrogen catechols by Cu2+ leads to a Cu-dependent mechanism of hydroxyl radical production via a hydrogen peroxide intermediate and suggest a mechanism for estrogen-associated site-specific DNA damage and mutagenesis.


Asunto(s)
Cobre/metabolismo , Estrógenos de Catecol/metabolismo , Radical Hidroxilo/metabolismo , Benzoquinonas/metabolismo , Óxidos N-Cíclicos/metabolismo , Daño del ADN , Espectroscopía de Resonancia por Spin del Electrón , Estradiol/análogos & derivados , Estradiol/metabolismo , Etinilestradiol/análogos & derivados , Etinilestradiol/metabolismo , Radicales Libres/metabolismo , Glutatión/farmacología , Peróxido de Hidrógeno/metabolismo , Estructura Molecular , Mutagénesis , Óxidos de Nitrógeno/metabolismo , Oxidación-Reducción , Fenantrolinas/metabolismo , Piridinas , Marcadores de Spin
14.
Proc Natl Acad Sci U S A ; 94(21): 11557-62, 1997 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-9326648

RESUMEN

N1-ethyl-N11-[(cyclopropyl)methyl]-4,8,-diazaundecane (CPENSpm) is a polyamine analogue that represents a new class of antitumor agents that demonstrate phenotype-specific cytotoxic activity. However, the precise mechanism of its selective cytotoxic activity is not known. CPENSpm treatment results in the superinduction of the polyamine catabolic enzyme spermidine/spermine N1-acetyltransferase (SSAT) in sensitive cell types and has been demonstrated to induce programmed cell death (PCD). The catalysis of polyamines by the SSAT/polyamine oxidase (PAO) pathway produces H2O2 as one product, suggesting that PCD produced by CPENSpm may be, in part, due to oxidative stress as a result of H2O2 production. In the sensitive human nonsmall cell line H157, the coaddition of catalase significantly reduces high molecular weight (HMW) DNA (>/=50 kb) and nuclear fragmentation. Important to note, specific inhibition of PAO by N,N'-bis(2, 3-butadienyl)-1,4-butane-diamine results in a significant reduction of the formation of HMW DNA and nuclear fragmentation. In contrast, the coaddition of catalase or PAO inhibitor has no effect on reducing HMW DNA fragmentation induced by N1-ethyl-N11-[(cycloheptyl)methyl]-4,8,-diazaundecane, which does not induce SSAT and does not deplete intracellular polyamines. These results strongly suggest that H2O2 production by PAO has a role in CPENSpm cytotoxicity in sensitive cells via PCD and demonstrate a potential basis for differential sensitivity to this promising new class of antineoplastic agents. Furthermore, the data suggest a general mechanism by which, under certain stimuli, cells can commit suicide through catabolism of the ubiquitous intracellular polyamines.


Asunto(s)
Antineoplásicos/toxicidad , Apoptosis/fisiología , Poliaminas/metabolismo , Poliaminas/toxicidad , Acetiltransferasas/antagonistas & inhibidores , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas , Catalasa/farmacología , Fragmentación del ADN , ADN de Neoplasias/efectos de los fármacos , Humanos , Neoplasias Pulmonares , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/antagonistas & inhibidores , Putrescina/análogos & derivados , Putrescina/farmacología , Células Tumorales Cultivadas
15.
Xenobiotica ; 27(4): 341-55, 1997 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9149374

RESUMEN

1. The objective of this study was to determine the basal and inducible activities of several cytochrome P450 (CYP) isozymes and monitor the acinar and hepatocyte morphology in precision cut, cultured rat and mouse liver slices. 2. The slices were cultured up to 96 h in Chee's essential medium supplemented with insulin, transferrin, selenium, DMSO, dexamethasone and epidermal growth factor. A dynamic roller system was used to incubate the slices at 37 degrees C in an atmosphere of 95% O2:5% CO2. 3. Histopathology of the liver slices revealed maintenance of normal hepatic lobular architecture with time in culture. 4. CYP isozyme activities were measured at various times of culture. In rat liver slices, at 72 h, CYP1A1/1A2 activity was induced 4-fold by beta NF and 37-fold by dioxin (TCDD) whereas in mouse liver slices, 1A1/1A2 activity was not inducible by beta NF but was induced 19-fold by TCDD. At 72 h, CYP2A5 (coumarin-7-hydroxylase) activity was not detected in rat liver slices but in mouse liver slices, 2A5 was induced 2-fold by beta NF, 11-fold by phenobarbital (PB) and 3-fold by TCDD. 5. Hydroxylation of testosterone at specific positions was used as an indication of the activities of various P450 isoforms. Testosterone was added to the cultures at 0 and 72 h and the metabolites were measured at 24 and 96 h respectively by hplc analysis. Depending upon the species, the treatment and the time in culture, CYP1A, 2A, 3A, 2B and 2C activities were detectable. 3A activity was highly induced by PB in both rat and mouse liver slices. These results demonstrate that this culture system can be used to assess and compare xenobiotic metabolism in liver slices from rodent species.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Isoenzimas/metabolismo , Hígado/enzimología , Xenobióticos/metabolismo , Animales , Medios de Cultivo , Sistema Enzimático del Citocromo P-450/biosíntesis , Inducción Enzimática/efectos de los fármacos , Isoenzimas/biosíntesis , Hígado/anatomía & histología , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Proteínas/metabolismo , Ratas , Ratas Endogámicas F344 , Especificidad de la Especie , Testosterona/metabolismo
16.
Cancer Res ; 57(24): 5493-7, 1997 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-9407957

RESUMEN

Mounting evidence suggests that catechol metabolites of estradiol may contribute to the development of estrogen-induced cancers. O-Methylation, catalyzed by catechol-O-methyltransferase (COMT), inactivates catechol estrogens. COMT is polymorphic in the human population, with 25% of Caucasians being homozygous for a low activity allele of the enzyme (COMT(LL)). We hypothesized that low activity COMT may be a risk factor for human breast cancer and designed a PCR-based RFLP assay to determine COMT genotype in a cohort of 112 matched, nested case-control samples. In the total study population, the odds ratios for the association of breast cancer risk with COMT(HL) and COMT(LL) genotypes were 1.30 [confidence interval (CI), 0.66-2.58] and 1.45 (CI, 0.69-3.07), respectively. Postmenopausal COMT(LL) women had a greater than 2-fold increased risk of developing breast cancer [odds ratio (OR), 2.18; CI, 0.93-5.11]. The association of COMT(LL) with the development of postmenopausal breast cancer was stronger and statistically significant in those women with a body mass index >24.47 kg/m2 (OR, 3.58; CI, 1.07-11.98). When COMT(LL) was combined with either glutathione S-transferase (GST) M1 null or with GSTP1 Ile-105-Val/Val-105-Val (intermediate/low activity, respectively) genotypes, the risk for developing postmenopausal breast cancer was also significantly increased. Our findings suggest that the allele encoding low activity COMT may be an important contributor to the postmenopausal development of breast cancer in certain women.


Asunto(s)
Alelos , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Catecol O-Metiltransferasa/genética , Catecol O-Metiltransferasa/metabolismo , Anciano , Neoplasias de la Mama/epidemiología , Estudios de Casos y Controles , ADN/genética , ADN de Neoplasias/genética , Femenino , Genotipo , Humanos , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Polimorfismo Genético , Polimorfismo de Longitud del Fragmento de Restricción , Factores de Riesgo
17.
Toxicol In Vitro ; 11(6): 753-9, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20654380

RESUMEN

The objective of this study was to determine whether medium supplementation by antioxidants and fatty acids would improve the viability of cultured rat hepatocytes and protect them against doxorubicin toxicity. We examined the effects of three agents: vitamin E, sodium pyruvate and egg yolk (the combination of vitamin E, sodium pyruvate and fatty acids is a proprietary, patented technology of Warner Lambert called CRT) 0.3% (v/v) as a source of fatty acids, on cell viability measured by the dehydrogenase-dependent bioreduction of a tetrazolium salt (MTS). Untreated hepatocytes and hepatocytes treated with carbon tetrachloride (CCl(4), EC(50) 5.7 mm) or doxorubicin (1 and 30 mum) were exposed to different amounts of a mixture of antioxidants and fatty acids. The mixture, identified as 1X, provided a final concentration of 5 units of vitamin E, 0.1% egg yolk and 10 mm sodium pyruvate while the 3X and 5X mixtures contained proportionately higher concentrations of these components. The mixtures were added 18 hr prior to, simultaneously with or following treatment with doxorubicin and just simultaneously with CCl(4). Neither vitamin E, sodium pyruvate nor egg yolk alone improved viability. However, the viability of untreated hepatocytes improved significantly when the 3X mixture was added after 18 hr as indicated by determination of MTS reduction activity 24 hr later. The viability of doxorubicin treated cultures (1 and 30 mum) increased significantly when exposed either to the 3X or 5X mixtures simultaneously. A significant increase in viability was also seen when cells were exposed to the 3X mixture following doxorubicin (1 mum). The mixtures did not protect against toxicity caused by CCl(4), perhaps due to the overwhelming level of damage at its EC(50) concentration. It is proposed that the antioxidant properties of vitamin E and sodium pyruvate protect the cells from low levels of reactive oxygen species generated spontaneously in culture and by doxorubicin metabolism while the fatty acids help to maintain the integrity of hepatocyte membranes, resulting in greater viability of the hepatocytes.

18.
Carcinogenesis ; 17(12): 2783-6, 1996 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9006120

RESUMEN

In this study, our goal was to identify genes whose expression in liver is altered in female F-344 rats during mitosuppression induced by 42 days of ethinyl estradiol (EE) treatment (Yager et al., Carcinogenesis, 15, 2117-2123, 1994). Northern analysis demonstrated that the mRNA levels for transforming growth factor-beta1 (TGF-beta1) and the mannose 6-phosphate/insulin-like growth factor II receptor were significantly increased by EE treatment. Ten cDNA clones representing mRNAs whose expression was increased two- to four-fold in the mitosuppressed livers were identified by differential display. Sequence analysis revealed that one was homologous to the S-24 ribosomal protein and another to mitochondrial ATPase subunit e. The remaining clones showed no homology to known genes in GenBank. However, the expression of clones 15, 16 and 17 was increased in HepG2 cells following treatment with doxorubicin suggesting their induction by oxidative DNA damage. These results suggest that two independent but interrelated signalling pathways, one mediated through transforming growth factor-beta and the other through oxidative DNA damage, may contribute to hepatic mitosuppression caused by EE, perhaps through activation of cyclin-dependent kinase inhibitors.


Asunto(s)
Etinilestradiol/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Hígado/efectos de los fármacos , Animales , Femenino , Ratas , Ratas Endogámicas F344 , Receptor IGF Tipo 2/genética , Factor de Crecimiento Transformador beta/genética
19.
Annu Rev Pharmacol Toxicol ; 36: 203-32, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-8725388

RESUMEN

In western society, the causes of several cancers--including breast, endometrium, ovary, liver, and prostate--have been linked to inappropriate and/or prolonged exposure to synthetic or endogenous steroidal hormones. In this review, we discuss the mechanisms of estrogen carcinogenesis with a focus on estrogen metabolism to 16 alpha-hydroxy estrone and 2- and 4-hydroxy catechol estrogens and the potential effects of these metabolites in vitro and in vivo on hamster liver and kidney and rat liver carcinogenesis models. The examples demonstrate that the parent compounds and their metabolites cause both nongenotoxic cell proliferative effects as well as direct and indirect genotoxic effects, which illustrates the complex nature of estrogen carcinogenesis. These effects, in combination with the metabolic state of the tissue and the timing of its exposure, may determine the cell type (organ) of tumor development and the severity of disease.


Asunto(s)
Estrógenos/efectos adversos , Neoplasias Renales/inducido químicamente , Neoplasias Hepáticas/inducido químicamente , Animales , Neoplasias de la Mama/inducido químicamente , Neoplasias de la Mama/genética , Daño del ADN , Modelos Animales de Enfermedad , Estrógenos/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias de los Genitales Femeninos/inducido químicamente , Neoplasias de los Genitales Femeninos/genética , Humanos , Neoplasias Renales/genética , Neoplasias Hepáticas/genética , Masculino , Oxidación-Reducción , Neoplasias de la Próstata/inducido químicamente , Neoplasias de la Próstata/genética
20.
Cancer Lett ; 98(2): 219-25, 1996 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-8556712

RESUMEN

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a more potent hepatocarcinogen in female than in male or ovariectomized rats. A possible mechanism for this increased sensitivity is through enhanced metabolic activation of estrogens by TCDD-induced enzymes leading to oxidative damage in the cell. As a marker for oxidative DNA damage, 8-oxo-deoxyguanosine (8-oxo-dG) was quantitated in livers of intact and ovariectomized Sprague-Dawley rats chronically treated with TCDD (125 ng/kg per day) with and without diethylnitrosamine as initiator. Elevated levels of 8-oxo-dG were detected in a significantly greater number of the intact compared to ovariectomized TCDD-treated rats. Expression of CYP1B1 mRNA, a newly identified cytochrome P450 with proposed estrogen hydroxylase activity, was highly induced by TCDD. The results are consistent with the hypothesis that increased metabolism of endogenous estrogens to catechols by TCDD-induced enzymes may lead to increased oxidative DNA damage and hence contribute to TCDD-mediated hepatocarcinogenicity in female rats.


Asunto(s)
Cocarcinogénesis , Daño del ADN , ADN/metabolismo , Neoplasias Hepáticas Experimentales/inducido químicamente , Hígado/efectos de los fármacos , Ovario/fisiología , Dibenzodioxinas Policloradas/toxicidad , Animales , Carcinógenos/toxicidad , ADN/efectos de los fármacos , Desoxiguanosina/metabolismo , Dietilnitrosamina/toxicidad , Estradiol/fisiología , Femenino , Hígado/metabolismo , Neoplasias Hepáticas Experimentales/metabolismo , Ovariectomía , Oxidación-Reducción , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA