Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
2.
Exp Hematol ; 65: 29-33, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29964089

RESUMEN

Abcg2, a member of the ATP-binding cassette transporter family, is expressed in adult hematopoietic stem cells (HSCs) and is required for the side population phenotype of adult bone marrow HSCs and other adult tissue-specific stem cells. Lineage tracing in adult mice using the Abcg2-Cre mouse model showed that Abcg2 marks HSCs, intestinal stem cells, and spermatogonial stem cells. It is unclear whether definitive HSCs or their precursors in early embryonic development can be marked by Abcg2 expression. Here, we treated pregnant Abcg2 Cre/Cre RosaLSL-YFP mice with a single injection of 4-hydroxytamoxifen at embryonic day 7.5. Four months after birth, a small yellow fluorescent protein-positive (YFP+) cell population could be detected in all of the major white blood cell lineages and this was stable for 8 months. Transplant of bone marrow cells or Sca1+YFP+ cells from these mice showed continued multilineage marking in recipient mice at 4 months. These results demonstrate that Abcg2 expression marks precursors to adult long-term repopulating HSCs at E7.5 to E8.5 and contributes to a stable subpopulation of HSCs well into adulthood.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Factor de Transcripción Activador 2 , Linaje de la Célula , Ratones/embriología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Factor de Transcripción Activador 2/metabolismo , Animales , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Hidroxitestosteronas/farmacología , Modelos Animales , Técnicas de Cultivo de Órganos
3.
Toxicol Lett ; 292: 39-45, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29702199

RESUMEN

4-Hydroxyandrost-4-ene-3,17-dione, also named formestane, is an irreversible aromatase inhibitor and therapeutically used as anti-breast cancer medication in post-menopausal women. Currently, no therapeutical indication led to approval of its 17-hydroxylated analog 4-hydroxytestosterone, an anabolic steroid. However, it is currently investigated in a clinical trial for breast cancer. In context with sports doping, aromatase inhibitors are administered to reduce estrogenic side effects of misused anabolic substances or their metabolites. Therefore, both substances are prohibited in sports by the World Anti-Doping Agency (WADA). Analysis of urinary phase I and phase II metabolites showed similar results for both compounds. In the current investigation, 4-hydroxyandrost-4-ene-3,17-dione, 4-hydroxytestosterone and seven of their described urinary metabolites as well as 2α-hydroxyandrostenedione were tested in the yeast androgen screen and the yeast estrogen screen. Androgenic effects were observed for all tested substances, except for one, which showed anti-androgenic properties. With regard to the yeast estrogen screen, estrogenic effects were observed for only two metabolites at rather high concentrations, while six out of the ten substances tested showed anti-estrogenic properties. In terms of the strong androgenic effect observed for 4-hydroxytestosterone (10-8 M), 4-hydroxyandrost-4-ene-3,17-dione (10-8 M) and two more urinary metabolites, the yeast androgen assay may also be used to trace abuse in urine samples.


Asunto(s)
Andrógenos/farmacología , Androstenodiona/análogos & derivados , Doping en los Deportes , Receptor alfa de Estrógeno/agonistas , Estrógenos/farmacología , Hidroxitestosteronas/farmacología , Sustancias para Mejorar el Rendimiento/farmacología , Receptores Androgénicos/efectos de los fármacos , Detección de Abuso de Sustancias/métodos , Congéneres de la Testosterona/farmacología , Levaduras/efectos de los fármacos , Andrógenos/química , Androstenodiona/química , Androstenodiona/metabolismo , Androstenodiona/farmacología , Biotransformación , Relación Dosis-Respuesta a Droga , Receptor alfa de Estrógeno/química , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Estrógenos/química , Estrógenos/metabolismo , Humanos , Hidroxitestosteronas/química , Hidroxitestosteronas/metabolismo , Simulación del Acoplamiento Molecular , Sustancias para Mejorar el Rendimiento/química , Sustancias para Mejorar el Rendimiento/metabolismo , Conformación Proteica , Receptores Androgénicos/química , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Congéneres de la Testosterona/química , Congéneres de la Testosterona/metabolismo , Levaduras/genética , Levaduras/metabolismo
4.
Oncotarget ; 8(18): 30252-30264, 2017 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-28415819

RESUMEN

Tamoxifen is still the most commonly used endocrine therapy drug for estrogen receptor (ER)-positive breast cancer patients and has an excellent outcome, but tamoxifen resistance remains a great impediment to successful treatment. Recent studies have prompted an anti-tumor effect of aspirin. Here, we demonstrated that aspirin not only inhibits the growth of ER-positive breast cancer cell line MCF-7, especially when combined with tamoxifen, but also has a potential function to overcome tamoxifen resistance in MCF-7/TAM. Aspirin combined with tamoxifen can down regulate cyclinD1 and block cell cycle in G0/G1 phase. Besides, tamoxifen alone represses c-myc, progesterone receptor (PR) and cyclinD1 in MCF-7 cell line but not in MCF-7/TAM, while aspirin combined with tamoxifen can inhibit the expression of these proteins in the resistant cell line. When knocking down c-myc in MCF-7/TAM, cells become more sensitive to tamoxifen, cell cycle is blocked as well, indicating that aspirin can regulate c-myc and cyclinD1 proteins to overcome tamoxifen resistance. Our study discovered a novel role of aspirin based on its anti-tumor effect, and put forward some kinds of possible mechanisms of tamoxifen resistance in ER-positive breast cancer cells, providing a new strategy for the treatment of ER-positive breast carcinoma.


Asunto(s)
Aspirina/farmacología , Neoplasias de la Mama/metabolismo , Ciclina D1/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores de Estrógenos/metabolismo , Tamoxifeno/farmacología , Antineoplásicos Hormonales/farmacología , Biomarcadores , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Humanos , Hidroxitestosteronas/farmacología
5.
Hypertension ; 67(5): 916-26, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26928804

RESUMEN

6ß-Hydroxytestosterone, a cytochrome P450 1B1-derived metabolite of testosterone, contributes to the development of angiotensin II-induced hypertension and associated cardiovascular pathophysiology. In view of the critical role of angiotensin II in the maintenance of renal homeostasis, development of hypertension, and end-organ damage, this study was conducted to determine the contribution of 6ß-hydroxytestosterone to angiotensin II actions on water consumption and renal function in male Cyp1b1(+/+) and Cyp1b1(-/-) mice. Castration of Cyp1b1(+/+) mice or Cyp1b1(-/-) gene disruption minimized the angiotensin II-induced increase in water consumption, urine output, proteinuria, and sodium excretion and decreases in urine osmolality. 6ß-Hydroxytestosterone did not alter angiotensin II-induced increases in water intake, urine output, proteinuria, and sodium excretion or decreases in osmolality in Cyp1b1(+/+) mice, but restored these effects of angiotensin II in Cyp1b1(-/-) or castrated Cyp1b1(+/+) mice. Cyp1b1 gene disruption or castration prevented angiotensin II-induced renal fibrosis, oxidative stress, inflammation, urinary excretion of angiotensinogen, expression of angiotensin II type 1 receptor, and angiotensin-converting enzyme. 6ß-Hydroxytestosterone did not alter angiotensin II-induced renal fibrosis, inflammation, oxidative stress, urinary excretion of angiotensinogen, expression of angiotensin II type 1 receptor, or angiotensin-converting enzyme in Cyp1b1(+/+)mice. However, in Cyp1b1(-/-) or castrated Cyp1b1(+/+) mice, it restored these effects of angiotensin II. These data indicate that 6ß-hydroxytestosterone contributes to increased thirst, impairment of renal function, and end-organ injury associated with angiotensin II-induced hypertension in male mice and that cytochrome P450 1B1 could serve as a novel target for treating renal disease and hypertension in male mice.


Asunto(s)
Angiotensina II/farmacología , Citocromo P-450 CYP1B1/genética , Hidroxitestosteronas/farmacología , Enfermedades Renales/metabolismo , Estrés Oxidativo/efectos de los fármacos , Análisis de Varianza , Animales , Biopsia con Aguja , Castración , Modelos Animales de Enfermedad , Hipertensión/fisiopatología , Inmunohistoquímica , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/patología , Pruebas de Función Renal , Masculino , Ratones , ARN Mensajero/análisis , Distribución Aleatoria , Especies Reactivas de Oxígeno/metabolismo , Valores de Referencia , Factores de Riesgo
6.
Mol Cell Endocrinol ; 422: 160-171, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26690777

RESUMEN

Tamoxifen, a selective estrogen receptor modulator, is a commonly prescribed adjuvant therapy for estrogen receptor-α (ERα)-positive breast cancer patients. To determine if extracellular factors contribute to the modulation of IGF-1 signaling after tamoxifen treatment, MCF-7 cells were treated with IGF-1 in conditioned medium (CM) obtained from 4-OHT-treated MCF-7 cells and the accumulation of phospho-Akt (S473) was measured. CM inhibited IGF-1-dependent cell signaling and suggesting the involvement of extracellular factors (ie. IGFBPs). A significant increase in IGFBP-1 mRNA and extracellular IGFBP-1 protein was observed in 4-OHT-treated MCF-7 cells. Knockdown experiments demonstrated that both GPER1 and CREB mediate IGFBP-1 induction. Furthermore, experiments showed that 4-OHT-dependent IGFBP-1 transcription is downstream of GPER1-activation in breast cancer cells. Additionally, neutralization and knockdown experiments demonstrated a role for IGFBP-1 in the observed inhibition of IGF-1 signaling. These results suggested that 4-OHT inhibits IGF-1 signaling via GPER1 and CREB mediated extracellular IGFBP-1 accumulation in breast cancer cells.


Asunto(s)
Neoplasias de la Mama/genética , Hidroxitestosteronas/farmacología , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/farmacología , Receptores de Estrógenos/genética , Receptores Acoplados a Proteínas G/genética , Tamoxifeno/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Medios de Cultivo Condicionados/química , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Femenino , Humanos , Células MCF-7 , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos
7.
Hypertension ; 65(6): 1279-87, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25870196

RESUMEN

Previously, we showed that Cyp1b1 gene disruption minimizes angiotensin II-induced hypertension and associated pathophysiological changes in male mice. This study was conducted to test the hypothesis that cytochrome P450 1B1-generated metabolites of testosterone, 6ß-hydroxytestosterone and 16α-hydroxytestosterone, contribute to angiotensin II-induced hypertension and its pathogenesis. Angiotensin II infusion for 2 weeks increased cardiac cytochrome P450 1B1 activity and plasma levels of 6ß-hydroxytestosterone, but not 16α-hydroxytestosterone, in Cyp1b1(+/+) mice without altering Cyp1b1 gene expression; these effects of angiotensin II were not observed in Cyp1b1(-/-) mice. Angiotensin II-induced increase in systolic blood pressure and associated cardiac hypertrophy, and fibrosis, measured by intracardiac accumulation of α-smooth muscle actin, collagen, and transforming growth factor-ß, and increased nicotinamide adenine dinucleotide phosphate oxidase activity and production of reactive oxygen species; these changes were minimized in Cyp1b1(-/-) or castrated Cyp1b1(+/+) mice, and restored by treatment with 6ß-hydroxytestoterone. In Cyp1b1(+/+) mice, 6ß-hydroxytestosterone did not alter the angiotensin II-induced increase in systolic blood pressure; the basal systolic blood pressure was also not affected by this agent in either genotype. Angiotensin II or castration did not alter cardiac, angiotensin II type 1 receptor, angiotensin-converting enzyme, Mas receptor, or androgen receptor mRNA levels in Cyp1b1(+/+) or in Cyp1b1(-/-) mice. These data suggest that the testosterone metabolite, 6ß-hydroxytestosterone, contributes to angiotensin II-induced hypertension and associated cardiac pathogenesis in male mice, most probably by acting as a permissive factor. Moreover, cytochrome P450 1B1 could serve as a novel target for developing agents for treating renin-angiotensin and testosterone-dependent hypertension and associated pathogenesis in males.


Asunto(s)
Angiotensina II/farmacología , Cardiomegalia/fisiopatología , Citocromo P-450 CYP1B1/genética , Hidroxitestosteronas/farmacología , Hipertensión/fisiopatología , Animales , Castración , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Hidroxitestosteronas/metabolismo , Hipertensión/tratamiento farmacológico , Masculino , Ratones , Distribución Aleatoria , Especies Reactivas de Oxígeno/metabolismo , Valores de Referencia
8.
Neuron ; 78(5): 773-84, 2013 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-23764283

RESUMEN

Targeting genetically encoded tools for neural circuit dissection to relevant cellular populations is a major challenge in neurobiology. We developed an approach, targeted recombination in active populations (TRAP), to obtain genetic access to neurons that were activated by defined stimuli. This method utilizes mice in which the tamoxifen-dependent recombinase CreER(T2) is expressed in an activity-dependent manner from the loci of the immediate early genes Arc and Fos. Active cells that express CreER(T2) can only undergo recombination when tamoxifen is present, allowing genetic access to neurons that are active during a time window of less than 12 hr. We show that TRAP can provide selective access to neurons activated by specific somatosensory, visual, and auditory stimuli and by experience in a novel environment. When combined with tools for labeling, tracing, recording, and manipulating neurons, TRAP offers a powerful approach for understanding how the brain processes information and generates behavior.


Asunto(s)
Corteza Cerebral/citología , Regulación de la Expresión Génica/genética , Neuronas/fisiología , Recombinación Genética/genética , Análisis de Varianza , Animales , Antineoplásicos Hormonales/farmacología , Recuento de Células , Proteínas del Citoesqueleto/genética , Relación Dosis-Respuesta a Droga , Lateralidad Funcional/genética , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/citología , Hidroxitestosteronas/farmacología , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas Oncogénicas v-fos/genética , Estimulación Luminosa , ARN Mensajero/metabolismo , Recombinasas/genética , Privación Sensorial , Tamoxifeno/farmacología , Factores de Tiempo , Vibrisas/inervación
9.
J Steroid Biochem Mol Biol ; 138: 54-62, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23562642

RESUMEN

About 70% of breast tumors express androgen receptors. In addition, there is clinical evidence suggesting that androgens can inhibit mammary epithelial proliferation. Vice versa, there is also significant evidence indicating that androgens can increase the risk of breast cancer via multiple mechanisms, e.g. direct conversion to estrogens that can bind to the estrogen receptor and thereby stimulate cell proliferation. We examined the effect of testosterone (T) and dihydroxytestosterone (DHT) on cell proliferation, pS2 and Ki-67 expression in three different breast cancer cell lines alone or in co-culture with primary human breast adipose fibroblasts (BAFs) obtained from breast cancer patients. In the co-cultures, T induced cell proliferation, pS2 and Ki-67 expression in the estrogen receptor positive (ER(+)) MCF-7 and T47D cells. This was not observed in the (ER(-)) MDA-MB-231 cells. The differences might be explained by the high expression of aromatase, which converts androgens to estrogens in BAFs followed by ER-mediated cell proliferation. In line with this absence of increased cell proliferation, pS2 and Ki-67 expression was observed in the presence of DHT, which is not a substrate for aromatase. In contrast, DHT caused a significant suppression of cell proliferation (68% and 38%), pS2 and Ki-67 expression in the (ER(+)) MCF-7 and T47D cells. More importantly, DHT decreased cell proliferation in (ER(-)) MDA-MB-231 cells by 38%. The results suggest that androgens that cannot be aromatized, like DHT, may provide a perspective for treatment of breast cancer patients, especially those with triple negative breast cancer.


Asunto(s)
Andrógenos/farmacología , Técnicas de Cocultivo/métodos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Glándulas Mamarias Humanas/citología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Estradiol/metabolismo , Femenino , Fibroblastos/citología , Humanos , Hidroxitestosteronas/metabolismo , Hidroxitestosteronas/farmacología , Receptores de Estrógenos/metabolismo , Testosterona/metabolismo , Testosterona/farmacología
10.
PLoS One ; 8(4): e62334, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23626803

RESUMEN

INTRODUCTION: The role of miRNAs in acquired endocrine-resistant breast cancer is not fully understood. One hallmark of tumor progression is epithelial-to-mesenchymal transition (EMT), characterized by a loss of cell adhesion resulting from reduced E-cadherin and increased cell mobility. miR-200 family members regulate EMT by suppressing expression of transcriptional repressors ZEB1/2. Previously we reported that the expression of miR-200a, miR-200b, and miR-200c was lower in LY2 endocrine-resistant, mesenchymal breast cancer cells compared to parental, endocrine sensitive, epithelial MCF-7 breast cancer cells. Here we investigated the regulation of miR-200 family members and their role in endocrine-sensitivity in breast cancer cells. RESULTS: miR-200 family expression was progressively reduced in a breast cancer cell line model of advancing endocrine/tamoxifen (TAM) resistance. Concomitant with miR-200 decrease, there was an increase in ZEB1 mRNA expression. Overexpression of miR-200b or miR-200c in LY2 cells altered cell morphology to a more epithelial appearance and inhibited cell migration. Further, miR-200b and miR-200c overexpression sensitized LY2 cells to growth inhibition by estrogen receptor (ER) antagonists TAM and fulvestrant. Knockdown of ZEB1 in LY2 cells recapitulated the effect of miR-200b and miR-200c overexpression resulting in inhibition of LY2 cell proliferation by TAM and fulvestrant, but not the aromatase inhibitor exemestane. Demethylating agent 5-aza-2'-deoxycytidine (5-aza-dC) in combination with histone deacetylase inhibitor trichostatin A (TSA) increased miR-200b and miR-200c in LY2 cells. Concomitant with the increase in miR-200b and miR-200c, ZEB1 expression was decreased and cells appeared more epithelial in morphology and were sensitized to TAM and fulvestrant inhibition. Likewise, knockdown of ZEB1 increased antiestrogen sensitivity of LY2 cells resulting in inhibition of cell proliferation. CONCLUSIONS: Our data indicate that reduced miRNA-200b and miR-200c expression contributes to endocrine resistance in breast cancer cells and that the reduced expression of these miR-200 family members in endocrine-resistant cells can be reversed by 5-aza-dC+TSA.


Asunto(s)
Neoplasias de la Mama/genética , Resistencia a Antineoplásicos/genética , Expresión Génica , MicroARNs/genética , Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Epigénesis Genética/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Estradiol/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/genética , Humanos , Hidroxitestosteronas/farmacología , Células MCF-7 , Factores de Transcripción/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
11.
Carcinogenesis ; 34(3): 703-12, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23222814

RESUMEN

Widespread distribution of bisphenol-A (BPA) complicates epidemiological studies of possible carcinogenic effects on the breast because there are few unexposed controls. To address this challenge, we previously developed non-cancerous human high-risk donor breast epithelial cell (HRBEC) cultures, wherein BPA exposure could be controlled experimentally. BPA consistently induced activation of the mammalian target of rapamycin (mTOR) pathway--accompanied by dose-dependent evasion of apoptosis and increased proliferation--in HRBECs from multiple donors. Here, we demonstrate key molecular changes underlying BPA-induced cellular reprogramming. In 3/3 BPA-exposed HRBEC cell lines, and in T47D breast cancer cells, proapoptotic negative regulators of the cell cycle (p53, p21(WAF1) and BAX) were markedly reduced, with concomitant increases in proliferation-initiating gene products (proliferating cell nuclear antigen, cyclins, CDKs and phosphorylated pRb). However, simultaneous exposure to BPA and the polyphenol, curcumin, partially or fully reduced the spectrum of effects associated with BPA alone, including mTOR pathway proteins (AKT1, RPS6, pRPS6 and 4EBP1). BPA exposure induced an increase in the ERα (Estrogen Receptor): ERß ratio--an effect also reversed by curcumin (analysis of variance, P < 0.02 for all test proteins). At the functional level, concurrent curcumin exposure reduced BPA-induced apoptosis evasion and rapid growth kinetics in all cell lines to varying degrees. Moreover, BPA extended the proliferation potential of 6/6 primary finite-life HRBEC cultures--another effect reduced by curcumin. Even after removal of BPA, 1/6 samples maintained continuous growth--a hallmark of cancer. We show that BPA exposure induces aberrant expression of multiple checkpoints that regulate cell survival, proliferation and apoptosis and that such changes can be effectively ameliorated.


Asunto(s)
Apoptosis , Proliferación Celular/efectos de los fármacos , Células Epiteliales/fisiología , Glándulas Mamarias Humanas/patología , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Compuestos de Bencidrilo/farmacología , Neoplasias de la Mama , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Curcumina/farmacología , Regulación hacia Abajo/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Receptor alfa de Estrógeno/agonistas , Receptor alfa de Estrógeno/metabolismo , Estrógenos no Esteroides/farmacología , Femenino , Humanos , Hidroxitestosteronas/farmacología , Cinética , Fenoles/farmacología , Cultivo Primario de Células , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Proteína p53 Supresora de Tumor/genética
12.
PLoS One ; 7(1): e30174, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22276155

RESUMEN

It is prevailingly thought that the antiestrogens tamoxifen and ICI 182, 780 are competitive antagonists of the estrogen-binding site of the estrogen receptor-alpha (ER-α). However, a plethora of evidence demonstrated both antiestrogens exhibit agonist activities in different systems such as activation of the membrane-initiated signaling pathways. The mechanisms by which antiestrogens mediate estrogen-like activities have not been fully established. Previously, a variant of ER-α, EP-α36, has been cloned and showed to mediate membrane-initiated estrogen and antiestrogen signaling in cells only expressing ER-α36. Here, we investigated the molecular mechanisms underlying the antiestrogen signaling in ER-negative breast cancer MDA-MB-231 and MDA-MB-436 cells that express high levels of endogenous ER-α36. We found that the effects of both 4-hydoxytamoxifen (4-OHT) and ICI 182, 780 (ICI) exhibited a non-monotonic, or biphasic dose response curve; antiestrogens at low concentrations, elicited a mitogenic signaling pathway to stimulate cell proliferation while at high concentrations, antiestrogens inhibited cell growth. Antiestrogens at l nM induced the phosphorylation of the Src-Y416 residue, an event to activate Src, while at 5 µM induced Src-Y527 phosphorylation that inactivates Src. Antiestrogens at 1 nM also induced phosphorylation of the MAPK/ERK and activated the Cyclin D1 promoter activity through the Src/EGFR/STAT5 pathways but not at 5 µM. Knock-down of ER-α36 abrogated the biphasic antiestrogen signaling in these cells. Our results thus indicated that ER-α36 mediates biphasic antiestrogen signaling in the ER-negative breast cancer cells and Src functions as a switch of antiestrogen signaling dependent on concentrations of antiestrogens through the EGFR/STAT5 pathway.


Asunto(s)
Neoplasias de la Mama/metabolismo , Moduladores de los Receptores de Estrógeno/farmacología , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor alfa de Estrógeno/metabolismo , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromonas/farmacología , Estradiol/análogos & derivados , Estradiol/farmacología , Femenino , Fulvestrant , Humanos , Hidroxitestosteronas/farmacología , Immunoblotting , Inmunoprecipitación , Morfolinas/farmacología , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
13.
Virchows Arch ; 456(4): 403-9, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20300773

RESUMEN

Histo-blood group antigens CD173 (H2) and CD174 (Lewis Y) are known to be developmentally regulated carbohydrate antigens which are expressed to a varying degree on many human carcinomas. We hypothesized that they might represent markers of cancer-initiating cells (or cancer stem cells, CSC). In order to test this hypothesis, we examined the co-expression of CD173 and CD174 with stem cell markers CD44 and CD133 by flow cytometry analysis, immunocytochemistry, and immunohistochemistry on cell lines and tissue sections from breast cancer. In three breast cancer cell lines, the percentage of CD173(+)/CD44(+) cells ranged from 17% to >60% and of CD174(+)/CD44(+) from 21% to 57%. In breast cancer tissue sections from 15 patients, up to 50% of tumor cells simultaneously expressed CD173, CD174, and CD44 antigens. Co-expression of CD173 and CD174 with CD133 was also observed, but to a lesser percentage. Co-immunoprecipitation and sandwich ELISA experiments on breast cancer cell lines suggested that CD173 and CD174 are carried on the CD44 molecule. The results show that in these tissues CD173 (H2) and CD174 (LeY) are associated with CD44 expression, suggesting that these carbohydrate antigens are markers of cancer-initiating cells or of early progenitors of breast carcinomas.


Asunto(s)
Adenocarcinoma/inmunología , Biomarcadores de Tumor/metabolismo , Antígenos de Grupos Sanguíneos/metabolismo , Neoplasias de la Mama/inmunología , Fucosiltransferasas/metabolismo , Receptores de Hialuranos/metabolismo , Antígeno AC133 , Adenocarcinoma/patología , Antígenos CD/metabolismo , Neoplasias de la Mama/patología , Carcinoma Intraductal no Infiltrante/inmunología , Carcinoma Intraductal no Infiltrante/patología , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glicoproteínas/metabolismo , Humanos , Hidroxitestosteronas/farmacología , Péptidos/metabolismo , Células Madre/efectos de los fármacos , Células Madre/inmunología , Células Madre/patología
14.
Endocrinology ; 151(2): 748-54, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20016029

RESUMEN

The cytochrome P450 2A1 (CYP2A1) is a P450 enzyme that catalyzes the metabolism of testosterone. CYP2A1 has been reported to be present in rat testis. However, its developmental changes and function have not been well characterized. The purpose of this study was to measure the abundance of CYP2A1 (Cyp2a1) mRNA in the developing rat testis and Leydig cells and examine the effects of its product, 7 alpha-hydroxytestosterone (7HT), on an important enzyme, 11 beta-hydroxysteroid dehydrogenase type 1 (11 beta-HSD1) that interconverts active corticosterone and inactive 11-dehydrocorticosterone. As detected by real-time PCR, Cyp2a1 was found to be present exclusively in the Leydig cell. CYP2A1 activity in adult Leydig cells was 5-fold higher than those in progenitor or immature Leydig cells. 7HT competitively suppressed 11 beta-HSD1 oxidase and reductase activities in rat testis microsome with inhibitory constant of 1.2 and 2.9 mum, respectively. In intact Leydig cells, 7HT did not inhibit 11 beta-HSD1 reductase activity, but it stimulated its reductase activity. Thus, at 100 nm and higher concentrations, 7HT significantly switched 11 beta-HSD1 oxidoreductase activities toward reductase. The present data shows that 7HT, the product formed by CYP2A1 from testosterone, regulates the direction of 11 beta-HSD1 activity in rat Leydig cells.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/genética , Hidroxitestosteronas/farmacología , Células Intersticiales del Testículo/enzimología , Esteroide Hidroxilasas/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/efectos de los fármacos , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Animales , Animales Recién Nacidos , Separación Celular , Familia 2 del Citocromo P450 , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/fisiología , Masculino , NADP/metabolismo , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , Ratas , Regeneración , Testículo/enzimología , Testículo/crecimiento & desarrollo
15.
Mol Cell Endocrinol ; 305(1-2): 12-21, 2009 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-19433257

RESUMEN

Gender differences in lung disease and cancer are well-established. We reported estrogenic transcriptional responses in lung adenocarcinoma cells from females but not males despite similar estrogen receptor (ER) expression. Here we tested the hypothesis that normal human bronchial epithelial cells (HBECs) show gender-independent estrogenic responses. We report that a small sample of HBECs express approximately twice as much ERbeta as ERalpha. ERalpha and ERbeta were located in the cytoplasm, nucleus, and mitochondria. In contrast to lung adenocarcinoma cells, estradiol (E2) induced estrogen response element (ERE)-mediated luciferase reporter activity in transiently transfected HBECs regardless of donor gender. Overexpression of ERalpha-VP16 increased ERE-mediated transcriptional activity in all HBECs. E2 increased and 4-hydroxytamoxifen and ICI 182,780 inhibited HBEC proliferation and cyclin D1 expression in a cell line-specific manner. In conclusion, the response of HBECs to ER ligands is gender-independent suggesting that estrogenic sensitivity may be acquired during lung carcinogenesis.


Asunto(s)
Adenocarcinoma/metabolismo , Bronquios/metabolismo , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Neoplasias Pulmonares/metabolismo , Adenocarcinoma/patología , Bronquios/citología , Bronquios/efectos de los fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/análisis , Receptor beta de Estrógeno/análisis , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Reporteros , Humanos , Hidroxitestosteronas/farmacología , Neoplasias Pulmonares/patología , Masculino , Mitocondrias/metabolismo , Factores Sexuales
16.
Mol Cancer Ther ; 7(7): 2096-108, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18645020

RESUMEN

Breast cancer resistance to the antiestrogens tamoxifen (OHT) and fulvestrant is accompanied by alterations in both estrogen-dependent and estrogen-independent signaling pathways. Consequently, effective inhibition of both pathways may be necessary to block proliferation of antiestrogen-resistant breast cancer cells. In this study, we examined the effects of apigenin, a dietary plant flavonoid with potential anticancer properties, on estrogen-responsive, antiestrogen-sensitive MCF7 breast cancer cells and two MCF7 sublines with acquired resistance to either OHT or fulvestrant. We found that apigenin can function as both an estrogen and an antiestrogen in a dose-dependent manner. At low concentrations (1 mumol/L), apigenin stimulated MCF7 cell growth but had no effect on the antiestrogen-resistant MCF7 sublines. In contrast, at high concentrations (>10 mumol/L), the drug inhibited growth of MCF7 cells and the antiestrogen-resistant sublines, and the combination of apigenin with either OHT or fulvestrant showed synergistic, growth-inhibitory effects on both antiestrogen-sensitive and antiestrogen-resistant breast cancer cells. To further elucidate the molecular mechanism of apigenin as either an estrogen or an antiestrogen, effects of the drug on estrogen receptor-alpha (ERalpha); transactivation activity, mobility, stability, and ERalpha-coactivator interactions were investigated. Low-dose apigenin enhanced receptor transcriptional activity by promoting interaction between ERalpha and its coactivator amplified in breast cancer-1. However, higher doses (>10 mumol/L) of apigenin inhibited ERalpha mobility (as determined by fluorescence recovery after photobleaching assays), down-regulated ERalpha and amplified in breast cancer-1 expression levels, and inhibited multiple protein kinases, including p38, protein kinase A, mitogen-activated protein kinase, and AKT. Collectively, these results show that apigenin can function as both an antiestrogen and a protein kinase inhibitor with activity against breast cancer cells with acquired resistance to OHT or fulvestrant. We conclude that apigenin, through its ability to target both ERalpha-dependent and ERalpha-independent pathways, holds promise as a new therapeutic agent against antiestrogen-resistant breast cancer.


Asunto(s)
Apigenina/farmacología , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Moduladores de los Receptores de Estrógeno/farmacología , Receptor alfa de Estrógeno/metabolismo , Neoplasias de la Mama/enzimología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Estradiol/análogos & derivados , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Femenino , Recuperación de Fluorescencia tras Fotoblanqueo , Fulvestrant , Genes Reporteros , Histona Acetiltransferasas/metabolismo , Humanos , Hidroxitestosteronas/farmacología , Coactivador 3 de Receptor Nuclear , Unión Proteica/efectos de los fármacos , Proteínas Quinasas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Transactivadores/metabolismo , Transcripción Genética/efectos de los fármacos
17.
Proc Natl Acad Sci U S A ; 104(52): 20793-8, 2007 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-18087039

RESUMEN

Posttranslational modification by small ubiquitin-like modifier (SUMO) controls diverse cellular functions of transcription factors and coregulators and participates in various cellular processes including signal transduction and transcriptional regulation. Here, we report that pontin, a component of chromatin-remodeling complexes, is SUMO-modified, and that SUMOylation of pontin is an active control mechanism for the transcriptional regulation of pontin on androgen-receptor target genes in prostate cancer cells. Biochemical purification of pontin-containing complexes revealed the presence of the Ubc9 SUMO-conjugating enzyme that underlies its function as an activator. Intriguingly, 5alpha-dihydroxytestosterone treatments significantly increased the SUMOylation of pontin, and SUMOylated pontin showed further activation of a subset of nuclear receptor-dependent transcription and led to an increase in proliferation and growth of prostate cancer cells. These data clearly define a functional model and provide a link between SUMO modification and prostate cancer progression.


Asunto(s)
Proteínas Portadoras/química , Cromatina/química , ADN Helicasas/química , Neoplasias de la Próstata/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/química , ATPasas Asociadas con Actividades Celulares Diversas , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica , Humanos , Hidroxitestosteronas/farmacología , Lisina/química , Masculino , Modelos Biológicos , Proteína SUMO-1 , Transducción de Señal , Transcripción Genética , Enzimas Ubiquitina-Conjugadoras/metabolismo
18.
Exp Neurol ; 199(1): 143-55, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16464451

RESUMEN

Transplantation of neural stem cells into the brain is a novel approach to the treatment of chronic stroke disability. For clinical application, safety and efficacy of defined, stable cell lines produced under GMP conditions are required. To this end, a human neural stem cell line, CTX0E03, was derived from human somatic stem cells following genetic modification with a conditional immortalizing gene, c-mycER(TAM). This transgene generates a fusion protein that stimulates cell proliferation in the presence of a synthetic drug 4-hydroxy-tamoxifen (4-OHT). The cell line is clonal, expands rapidly in culture (doubling time 50-60 h) and has a normal human karyotype (46 XY). In the absence of growth factors and 4-OHT, the cells undergo growth arrest and differentiate into neurons and astrocytes. Transplantation of CTX0E03 in a rat model of stroke (MCAo) caused statistically significant improvements in both sensorimotor function and gross motor asymmetry at 6-12 weeks post-grafting. In addition, cell migration and long-term survival in vivo were not associated with significant cell proliferation. These data indicate that CTX0E03 has the appropriate biological and manufacturing characteristics necessary for development as a therapeutic cell line.


Asunto(s)
Corteza Cerebral/citología , Infarto de la Arteria Cerebral Media/cirugía , Células Neuroepiteliales/fisiología , Trasplante de Células Madre , Células Madre/fisiología , Análisis de Varianza , Animales , Conducta Animal , Diferenciación Celular/fisiología , Movimiento Celular , Corteza Cerebral/embriología , Células Clonales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Feto , Humanos , Hidroxitestosteronas/farmacología , Masculino , Actividad Motora/fisiología , Células Neuroepiteliales/citología , ARN Mensajero/biosíntesis , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Trasplante de Células Madre/métodos , Telomerasa/efectos de los fármacos , Telomerasa/metabolismo , Factores de Tiempo , Transducción Genética/métodos
20.
Biochem Biophys Res Commun ; 327(1): 70-5, 2005 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-15629431

RESUMEN

A chicken B lymphoma line, DT40, hypermutates immunoglobulin (Ig) genes spontaneously during culture. Thus, cultured DT 40 cells constitute a useful Ig library for screening antibodies (Abs) in vitro. To fix desirable Ig mutants by stopping hypermutation or to resume mutation for further improvement of Ab affinity, activation-induced cytidine deaminase (AID), a key enzyme responsible for the Ig mutation machinery, must be switched on or off. To this end, we generated a DT40 line whose one AID allele was disrupted, and the other allele was replaced by the loxP-flanked AID construct. In this engineered cell line designated as DT40-SW, AID expression could be switched reversibly by tamoxifen-regulated Cre recombinase. Devices were also introduced to discriminate between the "AID-ON" and the "AID-OFF" cells by GFP expression and puromycin resistance, respectively. Starting from a single DT40-SW cell, Ig gene repertoire was efficiently diversified during culture only when AID expression was on.


Asunto(s)
Linfocitos B/metabolismo , Pollos/genética , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Inmunoglobulinas/genética , Hipermutación Somática de Inmunoglobulina/genética , Tamoxifeno/análogos & derivados , Animales , Sitios de Ligazón Microbiológica/genética , Linfocitos B/patología , Línea Celular Tumoral , Pollos/inmunología , Resistencia a Medicamentos/genética , Proteínas Fluorescentes Verdes/genética , Hidroxitestosteronas/farmacología , Puromicina/farmacología , Tamoxifeno/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA