Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cell Endocrinol ; 587: 112200, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38518841

RESUMEN

OBJECTIVE: Myocardial injuries resulting from hypoxia are a significant concern, and this study aimed to explore potential protective strategies against such damage. Specifically, we sought to investigate the cardioprotective effects of 16α-hydroxyestrone (16α-OHE1). METHODS: Male Sprague‒Dawley (SD) rats were subjected to hypoxic conditions simulating high-altitude exposure at 6000 m in a low-pressure chamber for 7 days. Before and during hypoxic exposure, estradiol (E2) and various doses of 16α-OHE1 were administered for 14 days. Heart weight/body weight (HW/BW), myocardial structure, Myocardial injury indicators and inflammatory infiltration in rats were measured. H9C2 cells cultured under 5% O2 conditions received E2 and varying doses of 16α-OHE1; Cell viability, apoptosis, inflammatory infiltration, and Myocardial injury indicators were determined. Expression levels of ß2AR were determined in rat hearts and H9C2 cells. The ß2AR inhibitor, ICI 118,551, was employed to investigate ß2AR's role in 16α-OHE1's cardioprotective effects. RESULTS: Hypoxia led to substantial myocardial damage, evident in increased heart HW, CK-MB, cTnT, ANP, BNP, structural myocardial changes, inflammatory infiltration, and apoptosis. Pre-treatment with E2 and 16α-OHE1 significantly mitigated these adverse changes. Importantly, the protective effects of E2 and 16α-OHE1 were associated with the upregulation of ß2AR expression in both rat hearts and H9C2 cells. However, inhibition of ß2AR by ICI 118,551 in H9C2 cells nullified the protective effect of 16α-OHE1 on myocardium. CONCLUSION: Our findings suggest that 16α-OHE1 can effectively reduce hypoxia-induced myocardial injury in rats through ß2ARs, indicating a promising avenue for cardioprotection.


Asunto(s)
Hidroxiestronas , Inflamación , Propanolaminas , Masculino , Animales , Ratas , Hidroxiestronas/farmacología , Ratas Sprague-Dawley , Miocardio , Receptores Adrenérgicos
2.
Molecules ; 25(21)2020 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-33182506

RESUMEN

Ovarian cancer is the leading cause of death from gynecologic malignancies. Some estrogens, as well as xenoestrogens, such as chromium (VI) (Cr(VI)), are indicated as important pathogenic agents. The objective of this study was to evaluate the role of estradiol and some its metabolites upon exposure to the metalloestrogen Cr(VI) in an in vitro model. The changes in cell viability of malignant ovarian cancer cells (SKOV-3 resistant to cisplatin) exposed to 17ß-estradiol (E2) and its two metabolites, 2-methoxyestradiol (2-MeOE2) and 16α-hydroxyestrone (16α-OHE1), upon exposure to potassium chromate (VI) and its interactions were examined. The single and mixed models of action, during short and long times of incubation with estrogens, were applied. The different effects (synergism and antagonism) of estrogens on cell viability in the presence of Cr(VI) was observed. E2 and 16α-OHE1 caused a synergistic effect after exposure to Cr(VI). 2-MeOE2 showed an antagonistic effect on Cr(VI). The examined estrogens could be ranked according to the most protective effect or least toxicity in the order: 2-MeOE2 > E2 > 16α-OHE1. Early pre-incubation (24 h or 7 days) of cells with estrogens caused mostly an antagonistic effect-protective against the toxic action of Cr(VI). The beneficial action of estrogens on the toxic effect of Cr(VI), in the context of the risk of ovarian cancer, seems to be important and further studies are needed.


Asunto(s)
2-Metoxiestradiol/farmacología , Cromatos/farmacología , Cromo/farmacología , Estradiol/farmacología , Hidroxiestronas/farmacología , Neoplasias Ováricas/patología , Compuestos de Potasio/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Antagonismo de Drogas , Sinergismo Farmacológico , Femenino , Humanos , Neoplasias Ováricas/tratamiento farmacológico
3.
Sci Rep ; 10(1): 7283, 2020 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-32350290

RESUMEN

Earlier studies showed that endogenous estrogens have neuroprotective effect against oxidative damage. The present study seeks to investigate the protective effect of various endogenous estrogen metabolites against oxidative neurotoxicity in vitro and in vivo. Using immortalized mouse hippocampal neuronal cells as an in vitro model, 4-hydroxyestrone, an estrone metabolite with little estrogenic activity, is found to have the strongest neuroprotective effect against oxidative neurotoxicity among 25 endogenous estrogen metabolites tested, and its protective effect is stronger than 17ß-estradiol. Similarly, 4-Hydroxyestrone also exerts a stronger protective effect than 17ß-estradiol against kanic acid-induced hippocampal oxidative damage in rats. Neuroprotection by 4-hydroxyestrone involves increased cytoplasmic translocation of p53 resulting from SIRT1-mediated deacetylation of p53. Analysis of brain microsomal enzymes shows that estrogen 4-hydroxylation is the main metabolic pathway in the central nervous system. Together, these results show that 4-hydroxyestrone is an endogenous neuroestrogen that can strongly protect against oxidative neuronal damage.


Asunto(s)
Hipocampo/metabolismo , Hidroxiestronas/farmacología , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Animales , Línea Celular Transformada , Estradiol/farmacología , Hipocampo/patología , Masculino , Ratones , Neuronas/patología , Síndromes de Neurotoxicidad/tratamiento farmacológico , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/patología , Ratas , Ratas Sprague-Dawley
4.
Adv Clin Exp Med ; 26(2): 215-221, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28791837

RESUMEN

BACKGROUND: The increasing incidence of estrogen-dependent breast cancer and the presence in the environment of a large number of factors that interact with estrogen receptors have sparked interest in chemical influences on estrogen-dependent processes. In a previous work, the authors examined the interaction of estradiol with chromium. In the present article the importance of estradiol biotransformation in these interactions is investigated. There is no information in the available literature about the role of metabolites in exposure to chromium. It seems important because estradiol metabolites have various carcinogenic abilities and their formation during biotransformation could be increased or decreased by environmental enzyme inducers or inhibitors. The metabolites could play a detoxifying role or create a toxic synergism in free radical processes induced by chromium VI (CrVI). OBJECTIVES: The aim of this study was to evaluate the influence of 2 17ß-estradiol metabolites - 4-hydroxyestradiol (4-OHE2) and 16α-hydroxyestrone (16α-OHE1) - in conditions of oxidative stress caused by CrVI. MATERIAL AND METHODS: Human blood, erythrocytes or mitochondria isolated from human placentas after natural deliveries were used in the experiments. The influence of CrVI, 4-OHE2 and 16-OHE1 on thiobarbituric acid reactive substances (TBARS), the hydroxyl radical (•OH), superoxide dismutase (SOD), glutathione peroxidase (GPx) and glutathione-S-transferase (GST), and the interactions of the metabolites exposed to chromium expressed by these factors were examined. RESULTS: 4-OHE2 reduced the level of TBARS induced by CrVI in mitochondria (p < 0.05) and in erythrocytes (p < 0.05), and increased SOD activity (p < 0.05). 16α-OHE1 increased the activity of GST in erythrocytes exposed to CrVI (p < 0.05). CONCLUSIONS: The metabolites do not have toxic interactions with CrVI. On the contrary, they exhibited a protective effect. The mechanism of protection varied: 4-OHE2 decreased TBARS and increased SOD activity, while 16α-OHE1 increased GST activity.


Asunto(s)
Cromo/farmacología , Estrógenos de Catecol/farmacología , Hidroxiestronas/farmacología , Estrés Oxidativo/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Estradiol/metabolismo , Estrógenos de Catecol/metabolismo , Femenino , Glutatión Peroxidasa/metabolismo , Glutatión Transferasa/metabolismo , Humanos , Hidroxiestronas/metabolismo , Radical Hidroxilo/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Superóxido Dismutasa/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
5.
Hypertension ; 68(3): 796-808, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27402919

RESUMEN

Estrogen and oxidative stress have been implicated in pulmonary arterial hypertension (PAH). Mechanisms linking these systems are elusive. We hypothesized that estrogen metabolite, 16α-hydroxyestrone (16αOHE1), stimulates nicotinamide adenine dinucleotide phosphate oxidase (Nox)-induced reactive oxygen species (ROS) generation and proliferative responses in human pulmonary artery smooth muscle cells (hPASMCs) and that in PAH aberrant growth signaling promotes vascular remodeling. The pathophysiological significance of estrogen-Nox-dependent processes was studied in female Nox1(-/-) and Nox4(-/-) mice with PAH. PASMCs from control subjects (control hPASMCs) and PAH patients (PAH-hPASMCs) were exposed to estrogen and 16αOHE1 in the presence/absence of inhibitors of Nox, cytochrome P450 1B1, and estrogen receptors. Estrogen, through estrogen receptor-α, increased Nox-derived ROS and redox-sensitive growth in hPASMCs, with greater effects in PAH-hPASMCs versus control hPASMCs. Estrogen effects were inhibited by cytochrome P450 1B1 blockade. 16αOHE1 stimulated transient ROS production in hPASMCs, with sustained responses in PAH-hPASMCs. Basal expression of Nox1/Nox4 was potentiated in PAH-hPASMCs. In hPASMCs, 16αOHE1 increased Nox1 expression, stimulated irreversible oxidation of protein tyrosine phosphatases, decreased nuclear factor erythroid-related factor 2 activity and expression of nuclear factor erythroid-related factor 2-regulated antioxidant genes, and promoted proliferation. This was further amplified in PAH-hPASMCs. Nox1(-/-) but not Nox4(-/-) mice were protected against PAH and vascular remodeling. Our findings demonstrate that in PAH-hPASMCs, 16αOHE1 stimulates redox-sensitive cell growth primarily through Nox1. Supporting this, in vivo studies exhibited protection against pulmonary hypertension and remodeling in Nox1(-/-) mice. This study provides new insights through Nox1/ROS and nuclear factor erythroid-related factor 2 whereby 16αOHE1 influences hPASMC function, which when upregulated may contribute to vascular injury in PAH, particularly important in women.


Asunto(s)
Hidroxiestronas/farmacología , Hipertensión Pulmonar/fisiopatología , NADPH Oxidasas/metabolismo , NADP/metabolismo , Oxidación-Reducción/efectos de los fármacos , Animales , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Femenino , Humanos , Ratones , NADPH Oxidasa 1 , NADPH Oxidasas/genética , Arteria Pulmonar/citología , Arteria Pulmonar/metabolismo , Transducción de Señal , Remodelación Vascular/efectos de los fármacos , Remodelación Vascular/genética , Remodelación Vascular/fisiología
6.
Circulation ; 126(9): 1087-98, 2012 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-22859684

RESUMEN

BACKGROUND: Pulmonary arterial hypertension (PAH) is a hyperproliferative vascular disorder observed predominantly in women. Estrogen is a potent mitogen in human pulmonary artery smooth muscle cells and contributes to PAH in vivo; however, the mechanisms attributed to this causation remain obscure. Curiously, heightened expression of the estrogen-metabolizing enzyme cytochrome P450 1B1 (CYP1B1) is reported in idiopathic PAH and murine models of PAH. METHODS AND RESULTS: Here, we investigated the putative pathogenic role of CYP1B1 in PAH. Quantitative reverse transcription-polymerase chain reaction, immunoblotting, and in situ analysis revealed that pulmonary CYP1B1 is increased in hypoxic PAH, hypoxic+SU5416 PAH, and human PAH and is highly expressed within the pulmonary vascular wall. PAH was assessed in mice via measurement of right ventricular hypertrophy, pulmonary vascular remodeling, and right ventricular systolic pressure. Hypoxic PAH was attenuated in CYP1B1(-/-) mice, and the potent CYP1B1 inhibitor 2,3',4,5'-tetramethoxystilbene (TMS; 3 mg · kg(-1) · d(-1) IP) significantly attenuated hypoxic PAH and hypoxic+SU5416 PAH in vivo. TMS also abolished estrogen-induced proliferation in human pulmonary artery smooth muscle cells and PAH-pulmonary artery smooth muscle cells. The estrogen metabolite 16α-hydroxyestrone provoked human pulmonary artery smooth muscle cell proliferation, and this mitogenic effect was greatly pronounced in PAH-pulmonary artery smooth muscle cells. ELISA analysis revealed that 16α-hydroxyestrone concentration was elevated in PAH, consistent with CYP1B1 overexpression and activity. Finally, administration of the CYP1B1 metabolite 16α-hydroxyestrone (1.5 mg · kg(-1) · d(-1) IP) caused the development of PAH in mice. CONCLUSIONS: Increased CYP1B1-mediated estrogen metabolism promotes the development of PAH, likely via the formation of mitogens, including 16α-hydroxyestrone. Collectively, this study reveals a possible novel therapeutic target in clinical PAH.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/fisiología , Estrógenos/metabolismo , Hipertensión Pulmonar/enzimología , Arteria Pulmonar/enzimología , Animales , Hidrocarburo de Aril Hidroxilasas/antagonistas & inhibidores , Hidrocarburo de Aril Hidroxilasas/biosíntesis , Hidrocarburo de Aril Hidroxilasas/deficiencia , Hidrocarburo de Aril Hidroxilasas/genética , Hipoxia de la Célula , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Enfermedad Crónica , Citocromo P-450 CYP1B1 , Inducción Enzimática , Estradiol/farmacología , Femenino , Humanos , Hidroxiestronas/metabolismo , Hidroxiestronas/farmacología , Hidroxiestronas/toxicidad , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/patología , Hipertrofia Ventricular Derecha/enzimología , Hipoxia/complicaciones , Pulmón/enzimología , Pulmón/patología , Masculino , Ratones , Ratones Noqueados , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Arteria Pulmonar/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estilbenos/farmacología , Regulación hacia Arriba
7.
Ann N Y Acad Sci ; 1155: 237-41, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19250210

RESUMEN

In this study we investigated the impact of estrogen antagonists and of 16alpha-OHE1 (an estrogen derivative that binds to and induces transactivation of estrogen receptors) on estrogen metabolism in malignant HepG2 human liver cells featured by high estrogen sulfotransferase (EST); our aim was to clarify the potential correlation of EST and ER. As expected, the HepG2 cells exhibited a very high EST activity, with the majority of estrogen metabolites (over 86%) being detected as sulfates by 24 h. The coincubation of E2 and the antiestrogen tamoxifen induced a weak inhibition of EST activity (from 85.4% to 81.5%), while the coincubation with the pure antagonist ICI-182 and with 16alpha-OHE1 produced a 50% and 90% decrease of EST, respectively. Interestingly, both selective estrogen receptor modulators (SERMs) TAM and ICI-182, along with the same 16alpha-OHE1, gave rise respectively to a 2.8%, 3.2%, and 4.6% of de novo 16alpha-OHE1 formation. The inhibition of EST and the increase of 16alpha-OHE1 formation were both time- and dose-dependent. Our results suggest that EST activity is tightly associated with ER transactivation and can be regulated by selective estrogen receptor modulators (SERMs), including antiestrogens and 16alpha-OHE1. In this framework, 16alpha-OHE1 may have a potential role in human liver carcinogenesis, also through the inhibition of EST and the production of unconjugated, bioavailable estrogens.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Hidroxiestronas/farmacología , Neoplasias Hepáticas/enzimología , Sulfotransferasas/antagonistas & inhibidores , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Humanos , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Sulfotransferasas/metabolismo
8.
Cell Biol Int ; 32(4): 401-5, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18272409

RESUMEN

A variety of investigations on peroxiredoxins (Prxs) in different types of cancer have been carried out, but the estrogen-related function of Prxs in breast cancer has not yet been studied. In order to study the involvement of Prxs in the growth of breast cancer cells by estrogen, we evaluated the effect of mitogenic estrogen metabolites on the expression of Prx isoforms (I to VI) in MCF-7 cells and found that the transcript/protein expression of Prx IV was significantly induced by 16alpha-hydroxyestrone (OHE1) under both serum-free and serum conditions. In addition, treatment with Prx IV-specific siRNA significantly inhibited the 16alpha-OHE1-induced proliferation of MCF-7 cells. These results suggested that Prx IV involved in the 16alpha-OHE1-induced proliferation of MCF-7 cells has a proliferative effect and may be related to cancer development or progression.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Hidroxiestronas/farmacología , Peroxirredoxinas/metabolismo , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Peroxirredoxinas/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
9.
Int J Cancer ; 120(8): 1627-33, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17230533

RESUMEN

To examine a possible mechanism of endogenous estrogen-induced carcinogenesis, we studied the effect of the catechol-O-methyltransferase (COMT) inhibitor Ro 41-0960 on cell transforming and clastogenic activities of 2 catechol estrogens 2- and 4-hydroxyestrone (2- or 4-OHE1) using Syrian hamster embryo (SHE) cells. COMT activity was assayed by determining the methylation of 2- or 4-OHE1 using gas chromatography. The production of 2-methoxyestrone in cultures treated with 2-OHE1 was approximately 2-fold that of 4-methoxyestrone in cultures treated with 4-OHE1. 4-OHE1 induced morphological transformation at a higher frequency than 2-OHE1 did and the frequencies of cell transformation and chromosome aberrations were not significantly changed in cells treated with 4-OHE1 in the presence of Ro 41-0960. In contrast, the frequencies of cell transformation and chromosome aberrations were markedly increased in cells treated with 2-OHE1 along with Ro 41-0960 when compared to cells treated with 2-OHE1 alone. In addition, both catechol estrogens induced P53 protein expression and apoptosis. The frequencies of apoptotic cells induced by the catechol estrogens were modified by the COMT inhibition in a manner similar to those observed with the chromosome aberrations assay and the cell transformation assay, indicating that each effect by the catechol estrogens at the three measured endpoints might be caused by a mechanism similar to the others. Our findings indicate that COMT activity has an influence on cell transforming activity and its related genetic effects of catechol estrogens in SHE cells, which implies that an individual activity of COMT may be one of the etiological factors in endogenous estrogen-induced carcinogenesis.


Asunto(s)
Apoptosis/efectos de los fármacos , Benzofenonas/farmacología , Inhibidores de Catecol O-Metiltransferasa , Transformación Celular Neoplásica/efectos de los fármacos , Aberraciones Cromosómicas , Hidroxiestronas/farmacología , Animales , Catecol O-Metiltransferasa/genética , Catecol O-Metiltransferasa/metabolismo , Células Cultivadas/efectos de los fármacos , Cricetinae , Mesocricetus/embriología , Reacción en Cadena de la Polimerasa , Proteína p53 Supresora de Tumor/metabolismo
10.
J Mol Endocrinol ; 34(1): 91-105, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15691880

RESUMEN

We studied the effects of 2-methoxyestradiol (2-ME2) and 16alpha-hydroxyestrone (16alpha-OHE1), two metabolites of estradiol (E2), on DNA synthesis, cell cycle progression and cyclin D1 protein levels in estrogen receptor-positive MCF-7 cells. E2 and 16alpha-OHE1 stimulated DNA synthesis, and 2-ME2 inhibited the stimulatory effects of these agents. E2 and 16alpha-OHE1 stimulated the progression of cells from G1 to S phase and this effect was attenuated by 2-ME2. Western blot analysis showed that E2 and 16alpha-OHE1 increased cyclin D1 protein level by about fourfold compared with control. 2-ME2 had no significant effect on cyclin D1; however, it prevented the accumulation of cyclin D1 in the presence of E2 and 16alpha-OHE1. Cells transfected with a cyclin D1 reporter gene and treated with E2 or 16alpha-OHE1 showed 7- and 9.5-fold increase respectively in promoter activity compared with control. This activity was significantly inhibited by 2-ME2. Cyclin D1 transactivation was mediated by the cAMP response element (CRE) region, which binds activating transcription factor 2 (ATF-2). DNA affinity assay showed 2.5- and 3.5-fold increases in ATF-2 binding to CRE in the presence of E2 and 16alpha-OHE1 respectively. The binding of ATF-2 was inhibited by the presence of 2-ME2. These results show that 2-ME2 can downregulate cyclin D1 and thereby cell cycle progression by a mechanism involving the disruption of ATF-2 binding to cyclin D1 promoter.


Asunto(s)
Neoplasias de la Mama/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Ciclina D1/efectos de los fármacos , Estradiol/análogos & derivados , Estradiol/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Hidroxiestronas/farmacología , Factores de Transcripción/metabolismo , 2-Metoxiestradiol , Factor de Transcripción Activador 2 , Western Blotting , Neoplasias de la Mama/metabolismo , Ciclina D1/metabolismo , Replicación del ADN/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
J Mol Endocrinol ; 27(3): 293-307, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11719282

RESUMEN

It has been suggested that alterations in estradiol (E(2)) metabolism, resulting in increased production of 16alpha-hydroxyestrone (16alpha-OHE(1)), is associated with an increased risk of breast cancer. In the present study, we examined the effects of 16alpha-OHE(1)on DNA synthesis, cell cycle progression, and the expression of cell cycle regulatory genes in MCF-7 breast cancer cells. G(1) synchronized cells were treated with 1 to 25 nM 16alpha-OHE(1) for 24 and 48 h. [(3)H]Thymidine incorporation assay showed that 16alpha-OHE(1) caused an 8-fold increase in DNA synthesis compared with that of control cells, whereas E(2) caused a 4-fold increase. Flow cytometric analysis of cell cycle progression also demonstrated the potency of 16alpha-OHE(1) in stimulating cell growth. When G(1) synchronized cells were treated with 10 nM 16alpha-OHE(1) for 24 h, 62+/-3% of cells were in S phase compared with 14+/-3% and 52+/-2% of cells in the control and E(2)-treated groups respectively. In order to explore the role of 16alpha-OHE(1) in cell cycle regulation, we examined its effects on cyclins (D1, E, A, B1), cyclin dependent kinases (Cdk4, Cdk2), and retinoblastoma protein (pRB) using Western and Northern blot analysis. Treatment of cells with 10 nM 16alpha-OHE(1) resulted in 4- and 3-fold increases in cyclin D1 and cyclin A, respectively, at the protein level. There was also a significant increase in pRB phosphorylation and Cdk2 activation. In addition, transient transfection assay using an estrogen response element-driven luciferase reporter vector showed a 15-fold increase in estrogen receptor-mediated transactivation compared with control. These results show that 16alpha-OHE(1) is a potent estrogen capable of accelerating cell cycle kinetics and stimulating the expression of cell cycle regulatory proteins.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Quinasas CDC2-CDC28 , Ciclo Celular/efectos de los fármacos , Ciclinas/metabolismo , Hidroxiestronas/farmacología , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias Hormono-Dependientes/patología , Neoplasias de la Mama/genética , División Celular/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/genética , ADN de Neoplasias/biosíntesis , Estradiol/metabolismo , Femenino , Genes Reporteros , Humanos , Luciferasas/genética , Neoplasias Hormono-Dependientes/genética , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/genética , Receptores de Estrógenos/genética , Proteína de Retinoblastoma/metabolismo , Activación Transcripcional/efectos de los fármacos , Células Tumorales Cultivadas
12.
J Endocrinol ; 170(1): 165-74, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11431149

RESUMEN

2-Hydroxyestrone (2-OHE(1)) and 16alpha-hydroxyestrone (16alpha-OHE(1)) have been reported to be risk factors for negative bone balance and breast cancer, respectively. The roles of these two metabolites of estrone as estrogen agonists or antagonists with respect to estrogen target tissues, or both, are poorly defined. The purpose of this study was to characterize metabolite and tissue-specific differences between the actions of hydroxylated estrones on selected reproductive and non-reproductive estrogen target tissues in growing rats. First, the effects of ovariectomy were determined. Ovariectomy had the expected effects, including increases in all dynamic bone measurements at the proximal tibial epiphysis, without induction of bone loss. Second, ovariectomized growing rats were continuously treated for 3 weeks with 2-OHE(1), 16alpha-OHE(1), 17beta-estradiol (E(2)), a combination of E(2) and 2-OHE(1) (E(2)+2-OHE(1)), or a combination of E(2) and 16alpha-OHE(1) (E(2)+16alpha-OHE(1)), using controlled release subcutaneous implanted pellets containing 5 mg 2-OHE(1), 5 mg 16alpha-OHE(1), 0.05 mg E(2) or placebo. E(2) reduced body weight gain and radial and longitudinal bone growth as well as indices of cancellous bone turnover, and increased serum cholesterol, uterine wet weight and epithelial cell height, and proliferative cell nuclear antigen labeling in mammary gland. The hydroxylated estrones did not alter uterine wet weight and 16alpha-OHE(1) antagonized the E(2)-stimulated increase in epithelial cell height. 2-OHE(1) had no effect on cortical bone, whereas 16alpha-OHE(1) was an estrogen agonist with respect to all cortical bone measurements. 16alpha-OHE(1) also behaved as an estrogen agonist with respect to serum cholesterol and cancellous bone measurements. 2-OHE(1) had no effect on most E(2)-regulated indices of cancellous bone growth and turnover, but was a weak estrogen agonist with respect to mineral apposition rate and bone formation rate. Neither estrogen metabolite influenced body weight gain. Third, weanling rats were treated for 1 week with vehicle, E(2) (200 microg/kg per day) or 16alpha-OHE(1) (30, 100, 300, 1000 and 3000 microg/kg per day) to confirm uterotropic effects of daily subcutaneous (s.c.) administration of 16alpha-OHE(1). 16alpha-OHE(1) increased uterine weight in a dose-response manner to values that did not differ from rats treated with E(2). We conclude that the estrogen metabolites 2-OHE(1) and 16alpha-OHE(1) have target tissue-specific biological activities which differ from one another as well as from E(2). These findings add further support to the concept that there are several classes of estrogens with distinct biological activities. Furthermore, differences in the route of administration could influence the tissue specificity of estrogen metabolites.


Asunto(s)
Remodelación Ósea/efectos de los fármacos , Estrógenos/agonistas , Hidroxiestronas/farmacología , Glándulas Mamarias Animales/efectos de los fármacos , Animales , Colesterol/sangre , Relación Dosis-Respuesta a Droga , Implantes de Medicamentos , Estradiol/farmacología , Terapia de Reemplazo de Estrógeno , Femenino , Humanos , Glándulas Mamarias Animales/metabolismo , Modelos Animales , Tamaño de los Órganos/efectos de los fármacos , Ovariectomía , Antígeno Nuclear de Célula en Proliferación/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Útero/anatomía & histología , Útero/efectos de los fármacos
13.
J Gen Virol ; 82(Pt 1): 201-213, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11125173

RESUMEN

Human papillomavirus (HPV) type 16 infects the genital tract and is generally acknowledged to be a causative agent of cervical cancer. HPV infection alone is not sufficient to induce cervical cancer and other factors such as steroid hormones are thought to play a role in the establishment and/or progression of this disease. The HPV-16 E2 protein is required for virus replication and modulates viral gene expression whereas the HPV-16 E7 protein is required for cell transformation. We and others have shown that both the E2 and E7 proteins can induce apoptotic cell death in HPV-transformed and non-HPV transformed cell lines. Here we show that the steroid hormones oestrogen and progesterone can both increase the levels of E2- and E7-induced apoptosis. The oestrogen metabolite 16 alpha-hydroxyoestrone also increases E2- and E7-induced cell death and the dietary component indole-3-carbinol, which reduces the formation of 16alpha-hydroxyoestrone from oestrogen, blocks the effects of oestrogen. Thus the metabolism of oestrogen to 16 alpha-hydroxyoestrone appears to be required for the effects of this hormone on E2- and E7-induced cell death. We also show that the oestrogen receptor antagonist 3-hydroxytamoxifen blocks the effects of oestrogen on E2- and E7-induced cell death, whereas the anti-progesterone RU486 blocks the effects of both progesterone and oestrogen. We discuss these results in terms of the origin and progression of cervical cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas de Unión al ADN , Estrógenos/farmacología , Proteínas Oncogénicas Virales/farmacología , Papillomaviridae/química , Progesterona/farmacología , Tamoxifeno/análogos & derivados , Relación Dosis-Respuesta a Droga , Células HeLa , Humanos , Hidroxiestronas/farmacología , Indoles/farmacología , Mifepristona/farmacología , Proteínas E7 de Papillomavirus , Progesterona/agonistas , Receptores de Estrógenos/agonistas , Tamoxifeno/farmacología , Células Tumorales Cultivadas
14.
Cancer Res ; 60(19): 5441-50, 2000 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-11034086

RESUMEN

There is considerable interest in the potential use of estrogen derivatives for the treatment and prevention of breast cancer. We demonstrated previously that the sulfamoylated estrone derivative 2-methoxyestrone-3-O-sulfamate (2-MeOEMATE) induced G2-M cell cycle arrest and modest levels of apoptosis in breast cancer cells in vitro, whereas the parent estrone derivative, 2-methoxyestrone, did not. 2-MeOEMATE also induced breast tumor regression in vivo in intact rats. To further explore the significance of sulfamoylation on the anticancer activity of estrone derivatives and to elucidate their mechanism of action, we synthesized two additional agents, 2-ethylestrone and 2-ethylestrone-3-O-sulfamate (2EtEMATE). 2-MeOEMATE and 2-EtEMATE inhibited the growth of a panel of estrogen receptor-negative and -positive breast cancer cell lines in vitro, induced mitotic arrest and apoptosis, and suppressed the long-term clonogenic potential of MCF7 and CAL51 breast cancer cells. In each assay, the sulfamoylated estrone derivatives were >10-fold more potent than their parent compounds. The sulfamoylated estrone derivatives were also significantly more potent inhibitors of cell growth than the previously studied endogenous estradiol metabolite 2-methoxyestradiol. 2-MeOEMATE and 2-EtEMATE functioned as antimicrotubule agents and inhibited the ability of paclitaxel to promote tubulin assembly in vitro. Like other antimicrotubule agents, the sulfamoylated estrone derivatives induced BCL-2 and BCL-XL phosphorylation and increased p53 expression. 2-MeOEMATE and 2-EtEMATE are novel antimicrotubule agents that have potent anticancer activity in breast cancer cells in vitro and may be beneficial as anticancer agents in vivo.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Estrona/análogos & derivados , Microtúbulos/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Estrona/farmacología , Humanos , Hidroxiestronas/farmacología , Mitosis/efectos de los fármacos , Tubulina (Proteína)/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos
15.
J Endocrinol ; 167(2): 281-7, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11054642

RESUMEN

Recent data indicate that the catechol estrogen, 2-hydroxyestrone (2-OHE(1)), has no effect on any target tissue including bone, whereas 16 alpha-hydroxyestrone (16 alpha-OHE(1)) exerts tissue-selective estrogen agonist activity. The effect of the catechol estrogen, 4-hydroxyestrone (4-OHE(1)), putatively associated with tumorigenesis, has not been studied in the skeleton. The purpose of this study was to assess the effect of 4-OHE(1) on tibia, uterine and mammary gland histology and blood cholesterol in ovariectomized (OVX'd) growing rats. Ten-week-old female Sprague-Dawley rats were injected subcutaneously with 200 microg/kg BW per day with 4-OHE(1), 17 beta-estradiol (E(2)) or vehicle for three weeks. OVX resulted in uterine atrophy, increased body weight, radial bone growth and cancellous bone turnover, and hypercholesterolemia. E(2) prevented these changes with the expected exception that the subcutaneous infusion of this high dose of estrogen did not prevent the hypercholesterolemia. 4-OHE(1) prevented the increase in blood cholesterol and the increase in body weight. 4-OHE(1) appeared to have partial estrogen activity in the uterus; uterine weight and epithelial cell height were significantly greater than the OVX rats but significantly less (twofold) than the E(2) animals. Analysis of variance indicated that 4-OHE(1) slightly decreased the periosteal mineral apposition rate (P<0.05) compared with vehicle-treated rats but had no effect on double-labeled perimeter or bone formation rate. Similarly, 4-OHE(1) was a partial estrogen agonist on cancellous bone turnover. The data suggest that the catechol estrogen, 4-OHE(1), unlike 2-OHE(1), has estrogen activity. Furthermore, the profile of activity differs from that of 16 alpha-OHE(1). Our results suggest that estrogen metabolites may selectively influence estrogen-target tissues and, concomitantly, modulate estrogen-associated disease risk.


Asunto(s)
Hidroxiestronas/farmacología , Animales , Peso Corporal/efectos de los fármacos , Remodelación Ósea/efectos de los fármacos , Huesos/efectos de los fármacos , Colesterol/sangre , Estradiol/farmacología , Femenino , Crecimiento/efectos de los fármacos , Glándulas Mamarias Animales/efectos de los fármacos , Tamaño de los Órganos/efectos de los fármacos , Ovariectomía , Ratas , Ratas Sprague-Dawley , Útero/anatomía & histología , Útero/efectos de los fármacos
16.
J Bone Miner Res ; 15(3): 499-506, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10750564

RESUMEN

This article describes experiments that were performed to examine the direct action of estrogen metabolites on cultured human osteoblast cells. The human fetal osteoblastic cell line, hFOB/ER9, which expresses high levels of the estrogen receptor (ER) alpha, was used to examine the direct effects of 16alpha-hydroxyestrone (16alpha-OHE1) and 2-hydroxyestrone (2-OHE1) on osteoblast differentiation. The 16alpha-OHE1 caused a decrease in osteocalcin (OC) secretion to a maximum of 40% of control values (vehicle-treated cells) at 10(-7) M. Alkaline phosphatase (AP) activity was significantly induced at 10(-7) M 16alpha-OHE1 with greater than 500% of control at 10(-6) M 16alpha-OHE1. Finally, AP steady-state messenger RNA (mRNA) levels were increased within 24 h of 16alpha-OHE1 treatment. In contrast to 16alpha-OHE1, 2-OHE1 had no effects on the secretion of OC, AP activity, or AP gene expression. The 2-OHE1 also did not display any antiestrogen activity because treatment in combination with 17beta-estradiol (E2) and 16alpha-OHE1 had no significant effect on the reduction in OC secretion or induction of AP activity. Similar to E2, 16alpha-OHE1 stimulated the expression of an early response gene, a TGF-beta inducible early gene, designated TIEG, as early as 60 minutes after treatment, whereas treatment with 2-OHE1 displayed no effect. Support that the 16alpha-OHE1 regulation of these osteoblasts (OB) markers was mediated through the ER is shown by the fact that the estrogen antagonist ICI 182,780 abrogated these effects. These data suggest that is a potent estrogen agonist on human osteoblastic hOB/ER9 cells. In contrast, 2-OHE1 displayed no estrogenic or antiestrogenic activity in this human osteoblast cell model.


Asunto(s)
Estrógenos/metabolismo , Hidroxiestronas/farmacología , Osteoblastos/efectos de los fármacos , Fosfatasa Alcalina/biosíntesis , Fosfatasa Alcalina/genética , Matriz Ósea/efectos de los fármacos , Matriz Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Inducción Enzimática , Estradiol/análogos & derivados , Estradiol/farmacología , Receptor alfa de Estrógeno , Fulvestrant , Humanos , Osteoblastos/citología , Osteoblastos/metabolismo , Osteocalcina/metabolismo , ARN Mensajero/biosíntesis , Receptores de Estrógenos/agonistas , Receptores de Estrógenos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo
17.
Mol Cell Endocrinol ; 160(1-2): 61-6, 2000 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-10715539

RESUMEN

Most cancer cells are dependent on glucose uptake to fulfil their energy requirements. In the present investigation we have examined the ability of 2-methoxyestrone (2-MeOE1), 2-methoxyestradiol (2-MeOE2), 2-methoxyestrone-3-O-sulfamate (2-MeOEMATE), and a number of related compounds, to inhibit 2-deoxy-D-[1-(3)H]-glucose uptake in MCF-7 breast cancer cells. Glucose uptake was shown to be linear with respect to cell number and time over a 5-35min period. 2-MeOE2, 2-MeOE1 and 2-MeOEMATE inhibited glucose uptake by 25-49% at 10 microM. 2-Hydroxyestradiol and estrone sulfate had little effect on glucose uptake, whereas estrone glucuronide inhibited uptake by 29%. There is evidence that 2-methoxyestrogens may exert an anti-mitotic effect on cells by stabilizing microtubules in a similar manner to that of paclitaxel. We therefore examined the effect of exposing cells to 2-MeOEMATE or paclitaxel for 24 h on basal or insulin stimulated glucose uptake. Using these conditions, 2-MeOEMATE and paclitaxel inhibited basal glucose uptake by 50 and 22%, respectively, and insulin stimulated uptake by 36 and 51%, respectively. The development of drugs that can inhibit glucose uptake could have therapeutic potential for the treatment of breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Desoxiglucosa/metabolismo , Estrona/análogos & derivados , Hidroxiestronas/farmacología , 2-Metoxiestradiol , Antineoplásicos/farmacología , Transporte Biológico Activo/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Estradiol/análogos & derivados , Estradiol/farmacología , Estrona/farmacología , Femenino , Humanos , Insulina/farmacología , Cinética , Paclitaxel/farmacología , Células Tumorales Cultivadas
18.
Life Sci ; 65(13): PL167-70, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10503964

RESUMEN

Estradiol can stimulate prostacyclin production in the vessel wall, thereby eliciting vasodilatation. In the present work the effect of the estradiol metabolites estrone, 2-methoxyestrone, 2-methoxyestradiol, and 16alpha-hydroxyestrone were investigated to find out if they are also able to stimulate prostacyclin synthesis. All metabolites triggered an increase of prostacyclin synthesis in human endothelial cells starting at a concentration of 10(-9) M. The parent substance, 17beta-estradiol, accomplished this effect only starting at a concentration of 10(-8) M. These results indicate that estradiol metabolites may take part in the estradiol-induced vasodilatation in vivo.


Asunto(s)
Endotelio Vascular/metabolismo , Epoprostenol/biosíntesis , Estradiol/metabolismo , 2-Metoxiestradiol , Células Cultivadas , Estradiol/análogos & derivados , Estradiol/farmacología , Estrona/farmacología , Humanos , Hidroxiestronas/farmacología
19.
Anticancer Res ; 19(3A): 1673-80, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10470100

RESUMEN

BACKGROUND: Cervical cancer constitutes the second most common cancer in women. Estrogen promotes development of cervical cancer in cells infected with high risk human papillomaviruses (HPVs). We asked whether the phytochemical indole-3-carbinol (I3C) has anti-estrogenic activities in cervical cells with the goal of preventing cancer in HPV infected cells. MATERIALS AND METHODS: Using the cervical cancer cell line CaSki, we evaluated expression of HPV and cytochrome p450 (CYP) enzymes by Northern, RNase protection or quantitative RT-PCR. I3C binding to estrogen receptor was measured by competition with estradiol. Estrogen metabolites were measured by gas chromarography-mass spectrometry (GC-MS). RESULTS: Estradiol increased expression of HPV oncogenes whereas I3C and the estrogen metabolite 2-hydroxyestrone (2-OHE) abrogated the estrogen-increased expression of HPV oncogenes. Both I3C and 2-OHE competed with estradiol for estrogen receptor binding. I3C enhanced gene expression of CYP enzymes responsible for 2-hydroxylation of estrogen, and induced the formation of 2-OHE. CONCLUSION: I3C has anti-estrogenic activities which should prevent cancer in cervical cells.


Asunto(s)
Anticarcinógenos/farmacología , Carcinoma de Células Escamosas/prevención & control , Cuello del Útero/efectos de los fármacos , Antagonistas de Estrógenos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Viral de la Expresión Génica/efectos de los fármacos , Indoles/farmacología , Proteínas de Neoplasias/biosíntesis , Proteínas Represoras , Neoplasias del Cuello Uterino/prevención & control , Unión Competitiva , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/virología , Cuello del Útero/enzimología , Cuello del Útero/virología , Citocromo P-450 CYP1A1/biosíntesis , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A2/biosíntesis , Citocromo P-450 CYP1A2/genética , Sistema Enzimático del Citocromo P-450/biosíntesis , Inducción Enzimática/efectos de los fármacos , Estradiol/farmacología , Femenino , Humanos , Hidroxiestronas/farmacología , Proteínas Oncogénicas Virales/biosíntesis , Proteínas Oncogénicas Virales/genética , Oncogenes/efectos de los fármacos , Papillomaviridae/genética , Papillomaviridae/patogenicidad , Papillomaviridae/fisiología , Proteínas E7 de Papillomavirus , Infecciones por Papillomavirus/enzimología , Infecciones por Papillomavirus/patología , ARN Mensajero/biosíntesis , ARN Neoplásico/biosíntesis , ARN Viral/biosíntesis , Receptores de Estrógenos/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Infecciones Tumorales por Virus/enzimología , Infecciones Tumorales por Virus/patología , Infecciones Tumorales por Virus/virología , Neoplasias del Cuello Uterino/enzimología , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/virología
20.
Eur J Pharmacol ; 368(1): 95-102, 1999 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-10096774

RESUMEN

Our earlier studies have identified oligomycin sensitivity-conferring protein (OSCP), a subunit of proton F0F1-ATPase/ATP synthase in the mitochondrial inner membranes, as a new estradiol binding protein. This finding suggests that mitochondrial ATPase/ATP synthase could be a potential target for estradiol or compounds with similar structures. Here, we report that estradiol and several other compounds inhibited F0F1-ATPase activity of detergent-solubilized rat brain mitochondrial preparations in a following decreasing order: diethylstilbestrol (half-inhibition concentration, IC50 of 10-25 microM) > alpha-zearalenol, 4-hydroxyestradiol (1C50 of 55 microM) >2-hydroxyestradiol (IC50 of 110 microM), 17beta-estradiol, 17alpha-estradiol > beta-zearalanol > estriol, testosterone, 16alpha-hydroxyestrone > corticosterone, progesterone, dehydroepiandrosterone, dehydroepiandrosterone 3-sulfate, cholesterol (less than 10% inhibition at 140 microM). On the other hand, Na+, K+ -ATPase of porcine cortex showed different sensitivity to the compounds tested above. At 70 microM, the rank of inhibitory potency in decreasing order was as follows: 2-hydroxyestradiol (IC50 of 70 microM) > diethylstilbestrol> 4-hydroxyestradiol > progesterone > alpha-zearalenol, while other compounds had little effect (less than 5%). The data indicate that the ubiquitous mitochondrial F0F1-ATPase is a specific target site for estradiol and related estrogenic compounds; however, under this in vitro condition, the effect seems to require pharmacological concentrations.


Asunto(s)
Encéfalo/efectos de los fármacos , Estrógenos/farmacología , Mitocondrias/efectos de los fármacos , ATPasas de Translocación de Protón/antagonistas & inhibidores , Animales , Encéfalo/enzimología , Colesterol/farmacología , Corticosterona/farmacología , Deshidroepiandrosterona/farmacología , Detergentes , Dietilestilbestrol/farmacología , Inhibidores Enzimáticos/farmacología , Femenino , Hidroxiestronas/farmacología , Microsomas/efectos de los fármacos , Microsomas/enzimología , Mitocondrias/enzimología , Ouabaína/farmacología , Progesterona/farmacología , ATPasas de Translocación de Protón/metabolismo , Ratas , Ratas Sprague-Dawley , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , Solubilidad , Porcinos , Testosterona/farmacología , Zeranol/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA