Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Bases de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Endocrinology ; 160(11): 2692-2708, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31433456

RESUMEN

Human prostate stem and progenitor cells express estrogen receptor (ER)α and ERß and exhibit proliferative responses to estrogens. In this study, membrane-initiated estrogen signaling was interrogated in human prostate stem/progenitor cells enriched from primary epithelial cultures and stem-like cell lines from benign and cancerous prostates. Subcellular fractionation and proximity ligation assays localized ERα and ERß to the cell membrane with caveolin-1 interactions. Exposure to 17ß-estradiol (E2) for 15 to 60 minutes led to sequential phosphorylation of signaling molecules in MAPK and AKT pathways, IGF1 receptor, epidermal growth factor receptor, and ERα, thus documenting an intact membrane signalosome that activates diverse downstream cascades. Treatment with an E2-dendrimer conjugate or ICI 182,870 validated E2-mediated actions through membrane ERs. Overexpression and knockdown of ERα or ERß in stem/progenitor cells identified pathway selectivity; ERα preferentially activated AKT, whereas ERß selectively activated MAPK cascades. Furthermore, prostate cancer stem-like cells expressed only ERß, and brief E2 exposure activated MAPK but not AKT cascades. A gene subset selectively regulated by nongenomic E2 signaling was identified in normal prostate progenitor cells that includes BGN, FOSB, FOXQ1, and MAF. Membrane-initiated E2 signaling rapidly modified histone methyltransferases, with MLL1 cleavage observed downstream of phosphorylated AKT and EZH2 phosphorylation downstream of MAPK signaling, which may jointly modify histones to permit rapid gene transcription. Taken together, the present findings document ERα and ERß membrane-initiated signaling in normal and cancerous human prostate stem/progenitor cells with differential engagement of downstream effectors. These signaling pathways influence normal prostate stem/progenitor cell homeostasis and provide novel therapeutic sites to target the elusive prostate cancer stem cell population.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Células Madre Neoplásicas/metabolismo , Próstata/metabolismo , Caveolina 1/metabolismo , Células Cultivadas , Histona Metiltransferasas/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Fosforilación , Fosfotransferasas/metabolismo , Próstata/citología
2.
Stem Cell Res ; 23: 1-12, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28651114

RESUMEN

Using primary cultures of normal human prostate epithelial cells, we developed a novel prostasphere-based, label-retention assay that permits identification and isolation of stem cells at a single cell level. Their bona fide stem cell nature was corroborated using in vitro and in vivo regenerative assays and documentation of symmetric/asymmetric division. Robust WNT10B and KRT13 levels without E-cadherin or KRT14 staining distinguished individual stem cells from daughter progenitors in spheroids. Following FACS to isolate label-retaining stem cells from label-free progenitors, RNA-seq identified unique gene signatures for the separate populations which may serve as useful biomarkers. Knockdown of KRT13 or PRAC1 reduced sphere formation and symmetric self-renewal highlighting their role in stem cell maintenance. Pathways analysis identified ribosome biogenesis and membrane estrogen-receptor signaling enriched in stem cells with NF-ĸB signaling enriched in progenitors; activities that were biologically confirmed. Further, bioassays identified heightened autophagy flux and reduced metabolism in stem cells relative to progenitors. These approaches similarly identified stem-like cells from prostate cancer specimens and prostate, breast and colon cancer cell lines suggesting wide applicability. Together, the present studies isolate and identify unique characteristics of normal human prostate stem cells and uncover processes that maintain stem cell homeostasis in the prostate gland.


Asunto(s)
Separación Celular/métodos , Células Epiteliales/citología , Próstata/citología , Análisis de la Célula Individual/métodos , Adulto , División Celular Asimétrica , Autofagia , Autorrenovación de las Células , Células Cultivadas , Fluoresceínas/metabolismo , Técnicas de Silenciamiento del Gen , Ontología de Genes , Humanos , Queratinas/metabolismo , Masculino , Mitocondrias/metabolismo , Células Madre Neoplásicas/metabolismo , Regeneración , Análisis de Secuencia de ARN , Esferoides Celulares/citología , Coloración y Etiquetado , Succinimidas/metabolismo , Transcriptoma/genética , Adulto Joven
3.
Endocrinology ; 155(3): 805-17, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24424067

RESUMEN

Previous studies in rodent models have shown that early-life exposure to bisphenol A (BPA) reprograms the prostate and enhances its susceptibility to hormonal carcinogenesis with aging. To determine whether the human prostate is similarly sensitive to BPA, the current study used human prostate epithelial stem-like cells cultured from prostates of young, disease-free donors. Similar to estradiol-17ß (E2), BPA increased stem-progenitor cell self-renewal and expression of stem-related genes in a dose-dependent manner. Further, 10 nM BPA and E2 possessed equimolar membrane-initiated signaling with robust induction of p-Akt and p-Erk at 15 minutes. To assess in vivo carcinogenicity, human prostate stem-progenitor cells combined with rat mesenchyme were grown as renal grafts in nude mice, forming normal human prostate epithelium at 1 month. Developmental BPA exposure was achieved through oral administration of 100 or 250 µg BPA/kg body weight to hosts for 2 weeks after grafting, producing free BPA levels of 0.39 and 1.35 ng/mL serum, respectively. Carcinogenesis was driven by testosterone plus E2 treatment for 2 to 4 months to model rising E2 levels in aging men. The incidence of high-grade prostate intraepithelial neoplasia and adenocarcinoma markedly increased from 13% in oil-fed controls to 33% to 36% in grafts exposed in vivo to BPA (P < .05). Continuous developmental BPA exposure through in vitro (200 nM) plus in vivo (250 µg/kg body weight) treatments increased high-grade prostate intraepithelial neoplasia/cancer incidence to 45% (P < .01). Together, the present findings demonstrate that human prostate stem-progenitor cells are direct BPA targets and that developmental exposure to BPA at low doses increases hormone-dependent cancer risk in the human prostate epithelium.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Epitelio/patología , Estrógenos no Esteroides/toxicidad , Fenoles/toxicidad , Próstata/citología , Neoplasias de la Próstata/patología , Células Madre/efectos de los fármacos , Animales , Carcinogénesis , Células Cultivadas , Cromatografía Líquida de Alta Presión , Epitelio/efectos de los fármacos , Estrógenos/metabolismo , Citometría de Flujo , Humanos , Masculino , Ratones , Ratones Desnudos , Próstata/efectos de los fármacos , Neoplasias de la Próstata/inducido químicamente , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Células Madre/citología , Espectrometría de Masas en Tándem , Factores de Tiempo , Adulto Joven
4.
Neoplasia ; 15(7): 749-60, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23814487

RESUMEN

Urothelial carcinoma (UC) causes substantial morbidity and mortality worldwide. However, the molecular mechanisms underlying urothelial cancer development and tumor progression are still largely unknown. Using informatics analysis, we identified Sh3gl2 (endophilin A1) as a bladder urothelium-enriched transcript. The gene encoding Sh3gl2 is located on chromosome 9p, a region frequently altered in UC. Sh3gl2 is known to regulate endocytosis of receptor tyrosine kinases implicated in oncogenesis, such as the epidermal growth factor receptor (EGFR) and c-Met. However, its role in UC pathogenesis is unknown. Informatics analysis of expression profiles as well as immunohistochemical staining of tissue microarrays revealed Sh3gl2 expression to be decreased in UC specimens compared to nontumor tissues. Loss of Sh3gl2 was associated with increasing tumor grade and with muscle invasion, which is a reliable predictor of metastatic disease and cancer-derived mortality. Sh3gl2 expression was undetectable in 19 of 20 human UC cell lines but preserved in the low-grade cell line RT4. Stable silencing of Sh3gl2 in RT4 cells by RNA interference 1) enhanced proliferation and colony formation in vitro, 2) inhibited EGF-induced EGFR internalization and increased EGFR activation, 3) stimulated phosphorylation of Src family kinases and STAT3, and 4) promoted growth of RT4 xenografts in subrenal capsule tissue recombination experiments. Conversely, forced re-expression of Sh3gl2 in T24 cells and silenced RT4 clones attenuated oncogenic behaviors, including growth and migration. Together, these findings identify loss of Sh3gl2 as a frequent event in UC development that promotes disease progression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Carcinoma/genética , Transformación Celular Neoplásica/genética , Neoplasias de la Vejiga Urinaria/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Carcinoma/metabolismo , Carcinoma/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Progresión de la Enfermedad , Receptores ErbB/metabolismo , Perfilación de la Expresión Génica , Silenciador del Gen , Humanos , Ratones , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Carga Tumoral/genética , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Familia-src Quinasas/metabolismo
5.
Cell Mol Bioeng ; 2(1): 133-143, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20161062

RESUMEN

Mechanical loads are essential towards maintaining bone mass and skeletal integrity. Such loads generate various stimuli at the cellular level, including cyclic hydraulic pressure (CHP) and fluid shear stress (FSS). To gain insight into the anabolic responses of osteoblasts to CHP and FSS, we subjected MC3T3-E1 preosteoblasts to either FSS (12 dynes/cm(2)) or CHP varying from 0 to 68 kPa at 0.5 Hz. As with FSS, CHP produced a significant increase in ATP release over static controls within 5 min of onset. Cell stiffness examined by atomic force microscopy increased after 15 min of either CHP or FSS stimulation, which was attenuated when extracellular ATP was hydrolyzed with apyrase. As previously shown FSS induced polymerization of actins into stress fibers. However, the microtubule network was completely disrupted under FSS. In contrast, CHP appeared to maintain strong microtubule and f-actin networks. The purinergic signaling was found to be involved in the remodeling of f-actin, but not microtubule. Both CHP and FSS applied for 1 hour increased expression of COX-2. These data indicate that, while CHP and FSS produce similar anabolic responses, these stimuli have very different effects on the cytoskeleton remodeling and could contribute to loss of mechanosensitivity with extended loading.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA