Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
2.
PLoS One ; 10(7): e0131232, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26196517

RESUMEN

Androgen receptor (AR) variants are associated with resistance to anti androgen therapy both in human prostate cancer cell lines and clinical samples. These observations support the hypothesis that AR isoform accumulation is a consequence of selective therapeutic pressure on the full length AR. The Pten deficient prostate cancer model proceeds with well-defined kinetics including progression to castration resistant prostate cancer (CRPC). While surgical castration and enzalutamide treatments yield an initial therapeutic response, Pten-/-epithelia continue to proliferate yielding locally invasive primary tumor pathology. That most epithelium remains AR positive, but ligand independent, suggests the presence of oncogenic AR variants. To address this hypothesis, we have used a panel of recently described Pten-/- tumor cell lines derived from both from hormone intact (E4, E8) and castrated Pten mutants (cE1, cE2) followed by RACE PCR to identify and characterize three novel truncated, amino terminus containing AR variants (mAR-Va, b, c). Variants appear not only conserved throughout progression but are correlated with nearly complete loss of full length AR (AR-FL) at castrate androgen levels. The overexpression of variants leads to enhanced transcriptional activity of AR while knock down studies show reduced transcriptional output. Collectively, the identification of truncated AR variants in the conditional PTEN deletion model supports a role for maintaining the CRPC phenotype and provides further therapeutic applications of this preclinical model.


Asunto(s)
Variación Genética , Fosfohidrolasa PTEN/deficiencia , Neoplasias de la Próstata Resistentes a la Castración/genética , Receptores Androgénicos/genética , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Sitios de Empalme de ARN , Regulación hacia Arriba
3.
PLoS One ; 10(2): e0117758, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25693195

RESUMEN

Prostate cancer (PCa) is the second-leading cause of cancer-related mortality, after lung cancer, in men from developed countries. In its early stages, primary tumor growth is dependent on androgens, thus generally can be controlled by androgen deprivation therapy (ADT). Eventually however, the disease progresses to castration-resistant prostate cancer (CRPC), a lethal form in need of more effective treatments. G-protein coupled receptors (GPCRs) comprise a large clan of cell surface proteins that have been implicated as therapeutic targets in PCa growth and progression. The findings reported here provide intriguing evidence of a role for the newly characterized glutamate family member GPR158 in PCa growth and progression. We found that GPR158 promotes PCa cell proliferation independent of androgen receptor (AR) functionality and that this requires its localization in the nucleus of the cell. This suggests that GPR158 acts by mechanisms different from other GPCRs. GPR158 expression is stimulated by androgens and GPR158 stimulates AR expression, implying a potential to sensitize tumors to low androgen conditions during ADT via a positive feedback loop. Further, we found GPR158 expression correlates with a neuroendocrine (NE) differentiation phenotype and promotes anchorage-independent colony formation implying a role for GPR158 in therapeutic progression and tumor formation. GPR158 expression was increased at the invading front of prostate tumors that formed in the genetically defined conditional Pten knockout mouse model, and co-localized with elevated AR expression in the cell nucleus. Kaplan-Meier analysis on a dataset from the Memorial Sloan Kettering cancer genome portal showed that increased GPR158 expression in tumors is associated with lower disease-free survival. Our findings strongly suggest that pharmaceuticals targeting GPR158 activities could represent a novel and innovative approach to the prevention and management of CRPC.


Asunto(s)
Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/patología , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Andrógenos/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Supervivencia sin Enfermedad , Relación Dosis-Respuesta a Droga , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Humanos , Masculino , Ratones , Células Neuroendocrinas/efectos de los fármacos , Células Neuroendocrinas/patología , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/genética , Receptores Androgénicos/metabolismo
4.
J Cell Physiol ; 229(9): 1170-6, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24752784

RESUMEN

We previously established a role for cancer-associated fibroblasts (CAF) in enhancing the self-renewal and differentiation potentials of putative prostate cancer stem cells (CSC). Our published work focused on androgen-dependent prostate cancer (ADPC) using the conditional Pten deletion mouse model. Employing the same model, we now describe the interaction of CAF and CSC in castration-resistant prostate cancer (CRPC). CAF isolated from ADPC (ADPCAF) and from CRPC (CRPCAF) were compared in terms of their ability to support organoid formation and tumor initiation by CSC from CRPC (CRPCSC) in vitro and in vivo. CRPCSC formed spheroids in vitro and well-differentiated glandular structures under the renal capsules of recipient mice in vivo more effectively in the presence of CRPCAF compared to ADPCAF. Furthermore, whereas CSC with CAF from ADPC formed mostly well-differentiated tumors in our previous study, we now show that CRPCSC, when combined with CRPCAF (but not ADPCAF), can form aggressive, poorly-differentiated tumors. The potential of CRPCAF to support organoid/tumor formation by CRPCSC remained greater even when compared to 10-fold more ADPCAF, suggesting that paracrine factors produced specifically by CRPCAF preferentially potentiate the stemness and tumorigenic properties of the corresponding CSC. This apparently unique property of CRPCAF was notable when the CAF and CSC were grafted in either intact or castrated recipient mice. In both environments, CRPCAF induced in the epithelial compartment higher proliferative activity compared to ADPCAF, indicated by a higher Ki67 index. Factors released by CRPCAF to regulate CRPCSC may be targeted to develop novel therapeutic approaches to manage advanced prostate cancer.


Asunto(s)
Fibroblastos/patología , Células Madre Neoplásicas/patología , Comunicación Paracrina , Neoplasias de la Próstata Resistentes a la Castración/patología , Animales , Biomarcadores de Tumor/metabolismo , Castración , Diferenciación Celular , Proliferación Celular , Fibroblastos/metabolismo , Fibroblastos/trasplante , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/trasplante , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/genética , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Transducción de Señal , Esferoides Celulares , Carga Tumoral , Células Tumorales Cultivadas , Microambiente Tumoral
5.
Mol Cancer Res ; 12(4): 607-21, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24464914

RESUMEN

UNLABELLED: Annexin A1 (AnxA1), a phospholipid-binding protein and regulator of glucocorticoid-induced inflammatory signaling, has implications in cancer. Here, a role for AnxA1 in prostate adenocarcinoma was determined using primary cultures and a tumor cell line (cE1), all derived from the conditional Pten deletion mouse model of prostate cancer. AnxA1 secretion by prostate-derived cancer-associated fibroblasts (CAF) was significantly higher than by normal prostate fibroblasts (NPF). Prostate tumor cells were sorted to enrich for epithelial subpopulations based on nonhematopoietic lineage, high SCA-1, and high or medium levels of CD49f. Compared with controls, AnxA1 enhanced stem cell-like properties in high- and medium-expression subpopulations of sorted cE1 and primary cells, in vitro, through formation of greater number of spheroids with increased complexity, and in vivo, through generation of more, larger, and histologically complex glandular structures, along with increased expression of p63, a basal/progenitor marker. The differentiated medium-expression subpopulations from cE1 and primary cells were most susceptible to gain stem cell-like properties as shown by increased spheroid and glandular formation. Further supporting this increased plasticity, AnxA1 was shown to regulate epithelial-to-mesenchymal transition in cE1 cells. These results suggest that CAF-secreted AnxA1 contributes to tumor stem cell dynamics via two separate but complementary pathways: induction of a dedifferentiation process leading to generation of stem-like cells from a subpopulation of cancer epithelial cells and stimulation of proliferation and differentiation of the cancer stem-like cells. IMPLICATIONS: AnxA1 participates in a paradigm in which malignant prostate epithelial cells that are not cancer stem cells are induced to gain cancer stem cell-like properties.


Asunto(s)
Anexina A1/metabolismo , Células Madre Neoplásicas/patología , Neoplasias de la Próstata Resistentes a la Castración/patología , Animales , Diferenciación Celular/fisiología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Transducción de Señal
6.
Endocr Relat Cancer ; 20(6): 861-74, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24042462

RESUMEN

Several studies have focused on the effect of bone morphogenetic protein (BMP) on prostate cancer homing and growth at distant metastatic sites, but very little effect at the primary site. Here, we used two cell lines, one (E8) isolated from a primary tumor and the other (cE1) from a recurrent tumor arising at the primary site, both from the conditional Pten deletion mouse model of prostatic adenocarcinoma. Over-expression of the BMP antagonist noggin inhibited proliferation of cE1 cells in vitro while enhancing their ability to migrate. On the other hand, cE1/noggin grafts grown in vivo showed a greater mass and a higher proliferation index than the cE1/control grafts. For suppression of BMP activity in the context of cancer-associated fibroblasts (CAFs), we used noggin-transduced CAFs from the same mouse model to determine their effect on E8- or cE1-induced tumor growth. CAF/noggin led to increased tumor mass and greater de-differentiation of the E8 cell when compared with tumors formed in the presence of CAF/control cells. A trend of increase in the size of the tumor was also noted for cE1 cells when inoculated with CAF/noggin. Together, the results may point to a potential inhibitory role of BMP in the growth or re-growth of prostate tumor at the primary site. Additionally, results for cE1/noggin, and cE1 mixed with CAF/noggin, suggested that suppression of BMP activity in the cancer cells may have a stronger growth-enhancing effect on the tumor than its suppression in the fibroblastic compartment of the tumor microenvironment.


Asunto(s)
Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Recurrencia Local de Neoplasia/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata/metabolismo , Animales , Western Blotting , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Movimiento Celular , Proliferación Celular , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Fosfohidrolasa PTEN/fisiología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/metabolismo , Células del Estroma/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
PLoS One ; 8(7): e69484, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936028

RESUMEN

The inhibitor of apoptosis protein survivin is expressed in most cancers. Using the conditional PTEN deletion mouse model, we previously reported that survivin levels increase with prostate tumor growth. Here we evaluated the functional role of survivin in prostate tumor growth. First, we demonstrated that mice lacking the survivin gene in prostate epithelium were fertile and had normal prostate growth and development. We then serially, from about 10-56 weeks of age, evaluated histopathologic changes in the prostate of mice with PTEN deletion combined with survivin mono- or bi-allelic gene deletion. While within this time period most of the animals with wild-type or monoallelic survivin deletion developed adenocarcinomas, the most severe lesions in the biallelic survivin deleted mice were high-grade prostatic intra-epithelial neoplasia with distinct histopathology. Many atypical cells contained large hypertrophic cytoplasm and desmoplastic reaction in the prostatic intra-epithelial neoplasia lesions of this group was minimal until the late ages. A reduced proliferation index as well as apoptotic and senescent cells were detected in the lesions of mice with compound PTEN/survivin deficiency throughout the time points examined. Survivin deletion was also associated with reduced tumor expression of another inhibitor of apoptosis member, the X-linked inhibitor of apoptosis. Our findings suggest that survivin participates in the progression of prostatic intraepithelial neoplasia to adenocarcinoma, and that survivin interference at the prostatic intraepithelial neoplasia stages may be a potential therapeutic strategy to halt or delay further progression.


Asunto(s)
Adenocarcinoma/genética , Epitelio/metabolismo , Proteínas Inhibidoras de la Apoptosis/genética , Próstata/metabolismo , Neoplasias de la Próstata/genética , Proteínas Represoras/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Carcinogénesis/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Epitelio/patología , Femenino , Histonas/metabolismo , Humanos , Inmunohistoquímica , Proteínas Inhibidoras de la Apoptosis/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Ratones Transgénicos , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Próstata/patología , Neoplasia Intraepitelial Prostática/genética , Neoplasia Intraepitelial Prostática/metabolismo , Neoplasia Intraepitelial Prostática/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Represoras/metabolismo , Survivin
8.
Endocr Relat Cancer ; 20(4): R155-70, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23580590

RESUMEN

Mouse models of prostate cancer (PCa) are critical for understanding the biology of PCa initiation, progression, and treatment modalities. Here, we summarize recent advances in PCa mouse models that led to new insights into specific gene functions in PCa. For example, the study of transgenic mice with TMPRSS2/ERG, an androgen-regulated fusion protein, revealed its role in developing PCa precursor lesions, prostate intraepithelial neoplasia; however, it is not sufficient for PCa development. Double deficiency of Pten and Smad4 leads to a high incidence of metastatic PCa. Targeted deletion of Pten in castration-resistant Nkx3-1-expressing cells results in rapid carcinoma formation after androgen-mediated regeneration, indicating that progenitor cells with luminal characteristics can play a role in initiation of PCa. Transgenic mice with activated oncogenes, growth factors, and steroid hormone receptors or inactivated tumor suppressors continue to provide insights into disease progression from initiation to metastasis. Further development of new PCa models with spatial and temporal regulation of candidate gene expression will probably enhance our understanding of the complex events that lead to PCa initiation and progression, thereby invoking novel strategies to combat this common disease in men.


Asunto(s)
Modelos Animales de Enfermedad , Neoplasias de la Próstata , Animales , Investigación Biomédica , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones Transgénicos , Células Madre , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Cancer Discov ; 2(10): 906-21, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22777769

RESUMEN

We show that the VEGF receptor neuropilin-2 (NRP2) is associated with high-grade, PTEN-null prostate cancer and that its expression in tumor cells is induced by PTEN loss as a consequence of c-Jun activation. VEGF/NRP2 signaling represses insulin-like growth factor-1 receptor (IGF-IR) expression and signaling, and the mechanism involves Bmi-1-mediated transcriptional repression of the IGF-IR. This mechanism has significant functional and therapeutic implications that were evaluated. IGF-IR expression positively correlates with PTEN and inversely correlates with NRP2 in prostate tumors. NRP2 is a robust biomarker for predicting response to IGF-IR therapy because prostate carcinomas that express NRP2 exhibit low levels of IGF-IR. Conversely, targeting NRP2 is only modestly effective because NRP2 inhibition induces compensatory IGF-IR signaling. Inhibition of both NRP2 and IGF-IR, however, completely blocks tumor growth in vivo.


Asunto(s)
Neuropilina-2/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Neoplasias de la Próstata/metabolismo , Receptor IGF Tipo 1/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/genética , Complejo Represivo Polycomb 1/genética , Neoplasias de la Próstata/genética , Interferencia de ARN , ARN Interferente Pequeño , Receptor IGF Tipo 1/genética , Transducción de Señal/genética , Transcripción Genética , Activación Transcripcional , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular/genética
10.
Am J Transl Res ; 4(2): 165-74, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22611469

RESUMEN

Chronic inflammation is proposed to prime the development of prostate cancer. However, the mechanisms of prostate cancer initiation and development are not completely understood. The α(v)ß(6) integrin has been shown to play a role in epithelial development, wound healing and some epithelial cancers [1, 2]. Here, we investigate the expression of α(v)ß(6) in mouse models of prostatic inflammation and prostate cancer to establish a possible relationship between inflammation of the prostate, α(v)ß(6) expression and the progression of prostate cancer. Using immunohistochemical techniques, we show expression of α(v)ß(6) in two in vivo mouse models; the Pten(pc)-/- model containing a prostate- specific Pten tumor suppressor deletion that causes cancer, and the prostate ovalbumin-expressing transgenic (POET) inflammation mouse model. We show that the α(v)ß(6) integrin is induced in prostate cancer and inflammation in vivo in these two mouse models. α(v)ß(6) is expressed in all the mice with cancer in the Pten(pc-/-) model but not in age-matched wild-type mice. In the POET inflammation model, α(v)ß(6) is expressed in mice injected with activated T-cells, but in none of the control mice. In the POET model, we also used real time PCR to assess the expression of Transforming Growth Factor Beta 1 (TGFß1), a factor in inflammation that is activated by α(v)ß(6). In conclusion, through in vivo evidence, we conclude that α(v)ß(6) integrin may be a crucial link between prostatic inflammation and prostatic adenocarcinoma.

11.
Prostate ; 72(10): 1052-9, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22314496

RESUMEN

BACKGROUND: The "New Western-style Diet" (NWD) characterized by high in fat and low in fiber, vitamin D, calcium, and methyl donors--are considered as a risk factor for prostate cancer. Previous studies have shown that premalignant lesions of human prostate have decreased expression of the Retinoid X Receptor alpha (RXRα). This study was to determine the effect of diet in RXRα knockout mice in developing high-grade prostate intraepithelial neoplasia (mPIN). METHODS: Male mice (n = 54) with or without the RXRα prostate null mutation were fed either NWD or AIN-76A control diet for 10 months; prostates were harvested at 11 months of age and examined for prostate mPIN. RESULTS: mPIN was seen in 79% of RXRα prostate null mice fed NWD (n = 19), 30.8% RXRα prostate null mice fed AIN-76A (n = 13), 42.9% RXRα wild-type mice fed NWD (n = 14), and 12.5% RXRα wild-type mice fed AIN-76A (n = 8). Unconditional Logistic analysis showed a significant joint effect of NWD and RXRα status in developing mPIN 26.3 (95% CI: 2.5-280), but interaction was not significant owing to the small sample size 1.6 (0.09-27.7, P = 0.7441). CONCLUSION: This study provides preliminary data to support a joint RXRα-diet effect in prostate carcinogenesis.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Neoplasia Intraepitelial Prostática/metabolismo , Neoplasias de la Próstata/metabolismo , Receptor alfa X Retinoide/deficiencia , Animales , Masculino , Ratones , Ratones Noqueados , Neoplasia Intraepitelial Prostática/etiología , Neoplasia Intraepitelial Prostática/genética , Neoplasias de la Próstata/etiología , Neoplasias de la Próstata/genética , Distribución Aleatoria , Receptor alfa X Retinoide/genética
12.
Dis Model Mech ; 5(3): 366-74, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22279083

RESUMEN

Regions in the 8q24 gene desert contribute significantly to the risk of prostate cancer and other adult cancers. This region contains several DNA regions with enhancer activity in cultured cells. One such segment, histone acetylation peak 10 (AcP10), contains a risk single nucleotide polymorphism (SNP) that is significantly associated with the pathogenesis of colorectal, prostate and other cancers. The mechanism by which AcP10 influences cancer risk remains unknown. Here we show that AcP10 contains a sequence that is highly conserved across terrestrial vertebrates and is capable in transgenic mice of directing reporter gene expression to a subset of prostate lumenal epithelial cells. These cells include a small population of Nkx3.1-positive cells that persist even after androgen ablation. Castration-resistant Nkx3.1-positive (CARN) cells were shown by others to function both as stem cells and cells of origin of prostate cancer. Our results thus provide a mechanism by which AcP10 could influence prostate cancer risk.


Asunto(s)
Cromosomas Humanos Par 8/genética , Elementos de Facilitación Genéticos , Células Epiteliales/metabolismo , Predisposición Genética a la Enfermedad/genética , Próstata/citología , Neoplasias de la Próstata/genética , Células Madre/metabolismo , Animales , Secuencia de Bases , Secuencia Conservada/genética , Femenino , Expresión Génica , Marcación de Gen , Genes Reporteros/genética , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Especificidad de Órganos/genética , Fosfohidrolasa PTEN/genética , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo , Factores de Transcripción/metabolismo , Transgenes/genética , Vertebrados/genética , beta-Galactosidasa/metabolismo
13.
Horm Cancer ; 2(5): 298-309, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21948155

RESUMEN

We reported previously that bone morphogenetic protein 7 (BMP7) could induce epithelial-mesenchymal transition (EMT) in PC-3 prostate cancer cells grown in tissue culture plates. In this study, we examined BMP7-induced morphological and molecular expression changes that are characteristic of EMT using these cells under both two- (2D) and three-dimensional (3D) culture conditions. Filamentous outgrowths from spheroid structures that were formed from PC-3 cells in 3D cultures were strikingly evident when the spheroids were exposed to extracellular BMP7. This morphological change in 3D was accompanied by down-regulation of E-cadherin, which is an essential adhesion molecule for the integrity of epithelial phenotype. Invasiveness of the cancer cells was significantly enhanced with BMP7 treatment along with activation and up-regulation of proteases such as MMP1, MMP13, and urokinase plasminogen activator. Signal transduction of EMT conversion was examined by the use of certain pathway-specific inhibitors. Of the chemical inhibitors tested, inhibitors of PI3 kinase and Erk were found to suppress BMP-induced morphological changes both in 2D and 3D conditions. These results suggest that, besides the Smad signaling pathways, BMP-induced activation of PI3K and Erk contribute to EMT morphologic conversion of the PC-3 prostate cancer cells. Together, the results support the notion that the complexity of EMT may be better evaluated in terms of both spatial and temporal processes in 3D cell culture models that are physiologically more relevant than the cell growth in tissue culture plates.


Asunto(s)
Proteína Morfogenética Ósea 7/farmacología , Carcinoma/patología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias de la Próstata/patología , Cadherinas/genética , Cadherinas/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Transición Epitelial-Mesenquimal/fisiología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/fisiología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo
14.
PLoS One ; 6(1): e16205, 2011 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-21267413

RESUMEN

BACKGROUND: Despite its initial positive response to hormone ablation therapy, prostate cancers invariably recur in more aggressive, treatment resistant forms. The lack of our understanding of underlying genetic alterations for the transition from androgen-dependent (AD) to ADI prostate cancer growth hampers our ability to develop target-driven therapeutic strategies for the efficient treatment of ADI prostate cancer. METHODOLOGY/PRINCIPAL FINDINGS: By screening a library of activated human kinases, we have identified TPL2, encoding a serine/threonine kinase, as driving ADI prostate cancer growth. TPL2 activation by over-expressing either wild-type or a constitutively activated form of TPL2 induced ADI growth, whereas the suppression of TPL2 expression and its kinase activity in ADI prostate cancer cells inhibited cell proliferation under androgen-depleted conditions. Most importantly, TPL2 is upregulated in ADI prostate cancers of both the Pten deletion mouse model and the clinical prostate cancer specimens. CONCLUSIONS/SIGNIFICANCE: Together these data suggest that TPL2 kinase plays a critical role in the promotion of ADI prostate cancer progression. Furthermore, the suppression of TPL2 diminishes ADI prostate cancer growth and a high frequency of TPL2 overexpression in human ADI prostate cancer samples validates TPL2 as a target for the treatment of this deadly disease.


Asunto(s)
Quinasas Quinasa Quinasa PAM/fisiología , Neoplasias de la Próstata/enzimología , Proteínas Proto-Oncogénicas/fisiología , Andrógenos , Animales , Proliferación Celular , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Masculino , Ratones , Neoplasias de la Próstata/etiología , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Regulación hacia Arriba/genética
15.
Cancer Res ; 70(18): 7294-303, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20807814

RESUMEN

Signals originating from cancer-associated fibroblasts (CAF) may positively regulate proliferation and tumorigenicity in prostate cancer. In this study, we investigated whether CAFs may regulate the biology of prostate cancer stem cells (CSC) by using a conditional Pten deletion mouse model of prostate adenocarcinoma to isolate both CAF cultures and CSC-enriched cell fractions from the tumors. CSCs that were isolated possessed self-renewal, spheroid-forming, and multipotential differentiation activities in tissue culture, segregating with a cell fraction exhibiting a signature expression phenotype, including SCA-1 (high), CD49f (high), CK5 (high), p63 (high), Survivin (high), RUNX2 (high), CD44 (low), CD133 (low), CK18 (low), and Androgen Receptor (low). CSC spheroid-forming efficiency was differentially influenced by the nature of fibroblasts in a coculture system: Compared with mouse urogenital sinus mesenchyme or normal prostate fibroblasts, CAFs enhanced spheroid formation, with the spheroids displaying generally larger sizes and more complex histology. Graft experiments showed that CSCs admixed with CAFs produced prostatic glandular structures with more numerous lesions, high proliferative index, and tumor-like histopathologies, compared with those formed in the presence of normal prostate fibroblasts. Together, our findings underscore a significant role of CAFs in CSC biology.


Asunto(s)
Adenocarcinoma/patología , Células Madre Neoplásicas/patología , Neoplasias de la Próstata/patología , Adenocarcinoma/genética , Animales , Células Epiteliales/patología , Fibroblastos/patología , Eliminación de Gen , Expresión Génica , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Fosfohidrolasa PTEN/genética , Neoplasias de la Próstata/genética , Células del Estroma/patología
16.
Horm Cancer ; 1(1): 44-54, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20631921

RESUMEN

The clinical course of prostate cancer is grouped into two broad phases. The first phase, which is the growth of the androgen-dependent cancer (AD-Ca) responds well to androgen depletion treatment while the second phase, that could be termed as androgen depletion-independent cancer (ADI-Ca) does not. We used two separate prostate tumors, one AD-Ca and one ADI-Ca from the conditional Pten deletion mouse model to generate from each a pair of cell lines. The AD-Ca cell lines (E2 and E4) and the ADI-Ca cell lines (cE1 and cE2) display bi-allelic deletion at the Pten gene locus, an event which is specific for the prostate epithelium for this mouse model, and a fairly similar level of expression of the androgen receptor (AR). The ADI-Ca cell lines (cE series) grow well in the absence of androgen, display increased AR transcription under androgen-deprived environment, and retain the sensitivity to increased proliferation when androgen is supplemented. The AD-Ca cell lines (E series) grow slowly in the absence of androgen, and, unlike cE cells, do not show increased AR expression when maintained in the absence of androgen. The detection of epithelial cell markers, such as CK8, CK14, CK18 and E-cadherin in the cE series is conforming with the polygonal epithelial morphology of these cells in culture. The E cells also present mostly polygonal-shaped morphology with a small percent of cells with fibroblastoid morphology, and produce little or very low levels of cytokeratins, but increased levels of vimentin, Twist and Slug, the markers known to be associated with epithelial-mesenchymal transition. Each of the cell lines, when inoculated subcutaneously into male or female NOD.SCID mice induced tumors within eight weeks with 100% incidence. Histopathological examinations of the tumor sections, however, led to noticeable biological differences. The cE series engenders adenocarcinomas, particularly in male hosts, and the E series induces sarcomatoid carcinomas (positively stained for CK8 and AR as well as vimentin expression) in either male or female hosts. These new cell lines are promising models for the elucidation of the androgen metabolism and their role in prostate cancer.


Asunto(s)
Línea Celular Tumoral/patología , Recurrencia Local de Neoplasia/patología , Neoplasias Experimentales/patología , Neoplasias Hormono-Dependientes/patología , Neoplasias de la Próstata/patología , Animales , Western Blotting , Castración , Línea Celular Tumoral/metabolismo , Femenino , Masculino , Ratones , Ratones Noqueados , Ratones SCID , Recurrencia Local de Neoplasia/metabolismo , Trasplante de Neoplasias , Neoplasias Hormono-Dependientes/metabolismo , Fosfohidrolasa PTEN/genética , Neoplasias de la Próstata/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Horm Cancer ; 1(6): 297-305, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21761361

RESUMEN

For a study of interactions between the cancer-associated fibroblasts (CAFs) and the putative prostate cancer stem cells (CSCs), we used a conditional Pten deletion mouse model of prostatic adenocarcinoma to isolate both CAF cultures and CSC-enriched cell fractions from the primary tumors. The CSC subpopulation exhibited a collective phenotype of Lin(-)/SCA-1(hi)/CD49f(hi)/p63(hi)/CK5(hi)/AR(lo)/CK18(lo)/Survivin(hi)/Runx2(hi) and contained cells with the ability to both self-renew and differentiate into basal and luminal cells in vitro. The spheroids generated from the CSC-enriched subpopulation mimicked the glandular structures that could be produced from a similarly isolated cell fraction from the normal mouse prostate. The efficiency of spheroid formation was found to be influenced differentially by the nature of the fibroblasts that were co-cultured in the 3-D system. The growth and differentiation properties of the CSCs were significantly more enhanced by factors released from CAFs relative to normal prostate fibroblasts (NPFs). Additionally, increased commitment to differentiation to the luminal cell lineage was noted when CAFs were present. When CSCs admixed with either CAFs or NPFs were examined for formation of prostatic glandular structures in renal grafts in vivo, the lesions formed were generally more in numbers in the presence of CAFs than NPFs. Furthermore, lesions formed with CAFs often displayed tumor-like complex histopathology and contained increased numbers of proliferating cells. Taken together, the results suggested that the CAFs in the prostate tumor microenvironment can contribute to the biologic properties of the CSCs and by this account may play a major role in prostate tumorigenesis and progression. Thus, it would be important now to identify the paracrine and/or juxtacrine factors that are responsible for the stimulation of the cancer stem cells.


Asunto(s)
Adenocarcinoma/patología , Comunicación Celular/fisiología , Células Madre Neoplásicas/patología , Neoplasias de la Próstata/patología , Microambiente Tumoral , Animales , Diferenciación Celular/fisiología , Linaje de la Célula , Técnicas de Cocultivo , Progresión de la Enfermedad , Fibroblastos/patología , Masculino , Ratones , Ratones Transgénicos
18.
Lab Invest ; 90(2): 222-33, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19949374

RESUMEN

Previously we described that bone morphogenetic protein-7 (BMP7) could protect prostate cancer C4-2B cells from serum starvation-induced apoptosis via survivin induction. Here, for the first time, we identify Runx2 as a key regulator of survivin transcription. In C4-2B cells grown normally, suppression of Runx2 reduced survivin expression. Using ChIP assays, two regions of the survivin promoter, -1953 to -1812 (I) and -1485 to -1119 (II) encompassing consensus Runx-binding sites were examined. Runx2 was found to be associated with both regions, with a stronger affinity to region-I. In serum-starved cells neither region was occupied, but BMP7 restored association to region-II and not region-I. In reporter assays, transcription activity by BMP7 was significantly reduced when sequences including binding sites of region-II were deleted. Additionally, Runx2 expression was enhanced by BMP7 in these cells. Along with a strong survivin expression, a trend in increased Runx2 expression in human prostate cancer cells and tissues was noted. In the conditional Pten-knockout mouse, Runx2 level increased with growth of prostate tumor. The data define a novel role of Runx2 in regulating survivin expression in malignant epithelial cells and identify it as a critical factor in BMP signaling that protects cancer cells against apoptosis.


Asunto(s)
Adenocarcinoma/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Neoplasias de la Próstata/metabolismo , Animales , Apoptosis , Proteína Morfogenética Ósea 7/fisiología , Línea Celular Tumoral , Humanos , Proteínas Inhibidoras de la Apoptosis , Masculino , Ratones , Análisis por Matrices de Proteínas , Survivin , Transfección , Regulación hacia Arriba
19.
Virology ; 386(1): 16-22, 2009 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-19203775

RESUMEN

A new proviral integration site for feline leukemia virus (FeLV), termed flit-1, was identified from feline thymic lymphoma. Among 35 FeLV-related tumors examined, 5 of 25 thymic lymphomas demonstrated proviral insertion within flit-1 locus whereas none of four alimentary and five multicentric lymphomas and one T-lymphoid leukemia examined had rearrangement in this region. Extensive sequence analysis has shown that flit-1, which is noncoding, is conserved on human chromosome 12 and mouse chromosome 15. The human and murine homologs of flit-1 are positioned approximately 30-kb upstream to activin-A receptor type II-like 1 (ACVRL1/ALK1) gene. Expression of ACVRL1 mRNA was examined in two of five lymphomas with flit-1 rearrangement and detected in both of the two whereas normal thymuses and seven lymphoid tumors without flit-1 rearrangement had no detectable expression. Therefore, flit-1 appears to represent a novel FeLV proviral common integration domain that may influence lymphomagenesis as insertional mutagenesis.


Asunto(s)
Virus de la Leucemia Felina/fisiología , Linfoma/virología , Provirus/fisiología , Timo/virología , Integración Viral , Animales , Enfermedades de los Gatos/virología , Gatos , Orden Génico , Reordenamiento Génico , Linfoma/patología , Timo/patología
20.
J Urol ; 181(1): 354-62, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19010487

RESUMEN

PURPOSE: Testing immunotherapeutic strategies for prostate cancer has been impeded by the lack of relevant tumor models in immunocompetent animals. This opportunity is now provided by the recent development of prostate specific PTEN knockout mice, which show spontaneous development of true adenocarcinoma arising from prostate epithelium and more faithfully recapitulate the human disease than any previous model. We investigated the feasibility of using tumor cells derived from this model to test tumor vaccination and adoptive immunotherapeutic strategies for prostate cancer. MATERIALS AND METHODS: PTEN-CaP8 adenocarcinoma cells derived from the biallelic PTEN knockout prostate cancer model were used to vaccinate nontumor bearing litter mates. Tumor specific effector cells were generated from splenocytes of vaccinated mice by mixed lymphocyte-tumor reactions, and antiproliferative effects and cytokine generation were examined in vitro. The effect of vaccination or adoptive immunotherapy on luciferase marked PTEN-CaP8 subcutaneous tumors was monitored by tumor volumetric measurements and noninvasive bioluminescence imaging. RESULTS: Vaccination of litter mate mice with irradiated PTEN-CaP8 cells showed a significant prophylactic effect against the subsequent tumor challenge. Effector cells harvested from vaccinated litter mates showed significant interferon-gamma secretion upon co-incubation with PTEN-CaP8 target cells and they were capable of efficient target cell growth inhibition in vitro. Intratumor adoptive transfer of effector cells resulted in significant growth inhibition of preestablished prostate tumors in vivo. CONCLUSIONS: The PTEN knockout model serves as a highly useful model in which to investigate tumor cell vaccination and adoptive immunotherapeutic strategies in the context of true adenocarcinoma of the prostate. This model should accelerate efforts to develop effective immunotherapies for human prostate cancer.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Vacunas contra el Cáncer/uso terapéutico , Inmunización , Neoplasias de la Próstata/tratamiento farmacológico , Adenocarcinoma/inmunología , Animales , Masculino , Ratones , Ratones Noqueados , Fosfohidrolasa PTEN/genética , Neoplasias de la Próstata/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA