Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Int J Mol Sci ; 22(3)2021 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-33503805

RESUMEN

Castration-resistant prostate cancer (CRPC) is an advanced and androgen-independent form of prostate cancer. Recent studies of rapid actions mediated by estrogen in the prostate and its relationship with CRPC are emerging. We have previously shown that estrogen receptor (ER) promotes migration and invasion of the androgen-independent prostate cancer cells PC-3, but the signaling pathways involved in these events remain to be elucidated. Therefore, this study aimed to analyze the role of ERα and ERß in the activation of SRC, and the involvement of SRC and PI3K/AKT on invasion and colony formation of the PC-3 cells. Our results showed that the activation of ERα (using ERα-selective agonist PPT) and ERß (using ERß-selective agonist DPN) increased phosphorylation of SRC in PC-3 cells. In the presence of the selective inhibitor for SRC-family kinases PP2, the effects of DPN and PPT on transmigration and soft agar colony formation assays were decreased. Furthermore, SRC is involved in the expression of the non-phosphorylated ß-catenin. Finally, using PI3K specific inhibitor Wortmannin and AKT inhibitor MK2206, we showed that PI3K/AKT are also required for invasion and colony formation of PC-3 cells simulated by ER. This study provides novel insights into molecular mechanisms of ER in PC-3 cells by demonstrating that ER, located outside the cell nucleus, activates rapid responses molecules, including SRC and PI3K/AKT, which enhance the tumorigenic potential of prostate cancer cells, increasing cell proliferation, migration, invasion, and tumor formation.


Asunto(s)
Andrógenos/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores de Estrógenos/metabolismo , Transducción de Señal , Línea Celular Tumoral , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Humanos , Inmunohistoquímica , Masculino , Células PC-3 , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , beta Catenina/metabolismo , Familia-src Quinasas/metabolismo
2.
Oncol Rep ; 49(5)2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36960864

RESUMEN

The aim of the present study was to investigate the role of estrogen receptor (ER)α and ERß, and galectin­3 (GAL­3) in migration and invasion of androgen­independent DU­145 prostate cancer cells, and to examine the regulation of the expression of GAL­3 by the activation of these receptors. Wound healing and cell invasion assays were performed using the control (basal level of cellular function) and treated DU­145 cells. At 24 h of treatment, 17ß­estradiol (E2), the ERα­selective agonist, 4,4',4"­(4­propyl­(1H)­pyrazole­1,3,5­triyl)trisphenol (PPT), or the ERß­selective agonist, 2,3­bis(4­hydroxyphenyl)­propionitrile (diarylprepionitrile; DPN), increased the migration and invasion of the DU­145 cells. Pre­treatment with the ERα­ and ERß­selective antagonists blocked these effects, indicating that ERα and ERß are upstream receptors regulating these processes. Western blot analysis and immunofluorescence staining for the detection of the GAL­3 were performed using the control and treated DU­145 cells. Treatment of the DU­145 cells with E2, PPT or DPN for 24 h increased the expression of the GAL­3 compared to the control. Furthermore, a specific inhibitor of GAL­3 (VA03) inhibited the migration and invasion of DU­145 cells, indicating the involvement of the complex ERα/GAL­3 and ERß/GAL­3 in the regulation of these processes. On the whole, the present study demonstrates that the activation of both ERs increases the expression and signaling of GAL­3, and promotes the migration and invasion of DU­145 cells. The findings of the present study provide novel insight into the signatures and molecular mechanisms of ERα and ERß in DU­145 cells.


Asunto(s)
Neoplasias de la Próstata , Receptores de Estrógenos , Masculino , Humanos , Receptor alfa de Estrógeno/metabolismo , Galectina 3 , Andrógenos , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Estradiol/farmacología
3.
Exp Cell Res ; 316(19): 3207-26, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20727350

RESUMEN

Growth and survival of tumors at a site of metastasis involve interactions with stromal cells in the surrounding environment. Stromal cells aid tumor cell growth by producing cytokines as well as by modifying the environment surrounding the tumor through modulation of the extracellular matrix (ECM). Small leucine-rich proteoglycans (SLRPs) are biologically active components of the ECM which can be altered in the stroma surrounding tumors. The influence tumor cells have on stromal cells has been well elucidated. However, little is understood about the effect metastatic cancer cells have on the cell biology and behavior of the local stromal cells. Our data reveal a significant down-regulation in the expression of ECM components such as collagens I, II, III, and IV, and the SLRPs, decorin, biglycan, lumican, and fibromodulin in stromal cells when grown in the presence of two metastatic prostate cancer cell lines PC3 and DU145. Interestingly, TGF-ß down-regulation was observed in stromal cells, as well as actin depolymerization and increased vimentin and α5ß1 integrin expression. MT1-MMP expression was upregulated and localized in stromal cell protrusions which extended into the ECM. Moreover, enhanced stromal cell migration was observed after cross-talk with metastatic prostate tumor cells. Xenografting metastatic prostate cancer cells together with "activated" stromal cells led to increased tumorigenicity of the prostate cancer cells. Our findings suggest that metastatic prostate cancer cells create a metastatic niche by altering the phenotype of local stromal cells, leading to changes in the ECM.


Asunto(s)
Diferenciación Celular/genética , Regulación hacia Abajo/genética , Matriz Extracelular/genética , Fibroblastos/patología , Neoplasias de la Próstata/genética , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Comunicación Celular/efectos de los fármacos , Comunicación Celular/genética , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Técnicas de Cocultivo , Citoesqueleto/efectos de los fármacos , Citoesqueleto/genética , Regulación hacia Abajo/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Metaloproteinasa 1 de la Matriz/genética , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Metástasis de la Neoplasia , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Transporte de Proteínas/efectos de los fármacos , Proteoglicanos/genética , Proteoglicanos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Receptores de Vitronectina/genética , Receptores de Vitronectina/metabolismo , Transducción de Señal/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Células del Estroma/patología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Artículo en Inglés | MEDLINE | ID: mdl-32328032

RESUMEN

Prostate cancer is initially dependent on the androgen, gradually evolves into an androgen-independent form of the disease, also known as castration-resistant prostate cancer (CRPC). At this stage, current therapies scantily improve survival of the patient. Androgens and estrogens are involved in normal prostate and prostate cancer development. The mechanisms by which estrogens/estrogen receptors (ERs) induce prostate cancer and promote prostate cancer progression have not yet been fully identified. Our laboratory has shown that androgen-independent prostate cancer cells PC-3 express both ERα and ERß. The activation of ERß increases the expression of ß-catenin and proliferation of PC-3 cells. We now report that the activation of ERß promotes the increase of migration, invasion and anchorage-independent growth of PC-3 cells. Furthermore, the activation of ERα also plays a role in invasion and anchorage-independent growth of PC-3 cells. These effects are blocked by pretreatment with PKF 118-310, compound that disrupts the complex ß-catenin/TCF/LEF, suggesting that ERs/ß-catenin are involved in all cellular characteristics of tumor development in vitro. Furthermore, PKF 118-310 also inhibited the upregulation of vascular endothelial growth factor A (VEGFA) induced by activation of ERs. VEGF also is involved on invasion of PC-3 cells. In conclusion, this study provides novel insights into the signatures and molecular mechanisms of ERß in androgen-independent prostate cancer cells PC-3. ERα also plays a role on invasion and colony formation of PC-3 cells.


Asunto(s)
Adenocarcinoma/patología , Movimiento Celular , Proliferación Celular , Neoplasias de la Próstata/patología , Receptores de Estrógenos/fisiología , Andrógenos/farmacología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Estradiol/farmacología , Humanos , Masculino , Invasividad Neoplásica , Células PC-3 , Transducción de Señal/efectos de los fármacos , Ensayo de Tumor de Célula Madre , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo
5.
Cancer Lett ; 491: 108-120, 2020 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-32841713

RESUMEN

Breast cancer is the most common malignant tumor among women worldwide, and triple-negative breast cancer is the most aggressive type of breast cancer, which does not respond to hormonal therapies. The protease inhibitor, EcTI, extracted from seeds of Enterolobium contortisiliquum, acts on the main signaling pathways of the MDA-MB-231 triple-negative breast cancer cells. This inhibitor, when bound to collagen I of the extracellular matrix, triggers a series of pathways capable of decreasing the viability, adhesion, migration, and invasion of these cells. This inhibitor can interfere in the cell cycle process through the main signaling pathways such as the adhesion, Integrin/FAK/SRC, Akt, ERK, and the cell death pathway BAX and BCL-2. It also acts by reducing the main inflammatory cytokines such as TGF-α, IL-6, IL-8, and MCP-1, besides NFκB, a transcription factor, responsible for the aggressive and metastatic characteristics of this type of tumor. Thus, the inhibitor was able to reduce the main processes of carcinogenesis of this type of cancer.


Asunto(s)
Citocinas/antagonistas & inhibidores , Fabaceae/química , Glicosaminoglicanos/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Inhibidores de Tripsina/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colágeno Tipo I/metabolismo , Citocinas/biosíntesis , Femenino , Humanos , Metaloproteinasas de la Matriz/metabolismo , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Inhibidores de Tripsina/uso terapéutico
6.
Mol Cell Endocrinol ; 483: 11-23, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30660702

RESUMEN

The aim of the present study was to investigate the subcellular localization of estrogen receptors ERα and ERß in androgen-independent prostate cancer cell line DU-145, and the possible role of exportin CRM1 on ERs distribution. In addition, we evaluated the ERs contribution to activation of ERK1/2 and AKT. Immunostaining of ERα and ERß was predominantly found in the extranuclear regions of DU-145 cells. CRM1 inhibitor Leptomycin B reduced drastically the presence of ERα and ERß in the extranuclear regions and increased in the nuclei, indicating the possible involvement of CRM1 on ERs nuclear-cytoplasmic shuttling. 17ß-estradiol (E2), ERα-selective agonist PPT and ERß-selective agonist DPN induced a rapid increase on ERK1/2 phosphorylation. E2-induced ERK1/2 activation was partially inhibited when cells were pretreated with ERα- or ERß-selective antagonists, and blocked by simultaneous pretreatment with both antagonists, suggesting ERα/ß heterodimers formation. Furthermore, E2 treatment did not activate AKT pathway. Therefore, we highlighted a possible crosstalk between extranuclear and nuclear ERs and their upstream and downstream signaling molecules as an important mechanism to control ER function as a potential therapeutic target in prostate cancer cells.


Asunto(s)
Citoplasma/metabolismo , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Carioferinas/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Estradiol/farmacología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Fosforilación/efectos de los fármacos , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Exportina 1
7.
Int J Biochem Cell Biol ; 96: 40-50, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29341930

RESUMEN

The aim of the present study was to investigate the impact of the activation of estrogen receptors on expression and localization of N-cadherin, E-cadherin and non-phosphorylated ß-catenin in androgen-independent prostate cancer cells (PC-3 and DU-145) and in human post pubertal prostate epithelial cells (PNT1A). Expression of N-cadherin was detected in PNT1A and PC-3 cells, but not in DU-145 cells. E-cadherin was detected only in DU-145 cells and ß-catenin was detected in all cells studied. N-cadherin and ß-catenin were located preferentially in the cellular membrane of PNT1A cells and in the cytoplasm of PC-3 cells. E-cadherin and ß-catenin were located preferentially in the cellular membrane of DU-145 cells. 17ß-estradiol (E2) or the ERα-selective agonist PPT did not affect the content and localization of N-cadherin in PC-3 and PNT1A cells or E-cadherin in DU-145 cells. In PC-3 cells, ERß-selective agonist DPN decreased the expression of N-cadherin. DPN-induced downregulation of N-cadherin was blocked by pretreatment with the ERß-selective antagonist (PHTPP), indicating that ERß1 is the upstream receptor regulating the expression of N-cadherin. In DU-145 cells, the activation of ERß1 by DPN increased the expression of E-cadherin. Taken together, these results suggest that activation of ERß1 is required to maintain an epithelial phenotype in PC-3 and DU-145 cells. The activation of ERß1 also increased the expression of ß-catenin in cytoplasm of PC-3 and in the cellular membrane of DU-145 cells. In conclusion, our results indicate differential expression and localization of N-cadherin, E-cadherin and ß-catenin in androgen-independent prostate cancer cells. The reduction of N-cadherin content by activation of ERß, exclusively observed in androgen-independent prostate cancer cells (PC-3), may be related to the activation of signaling pathways, such as the release of ß-catenin into the cytoplasm, translocation of ß-catenin to the nucleus and activation of gene transcription.


Asunto(s)
Antígenos CD/biosíntesis , Cadherinas/biosíntesis , Receptor beta de Estrógeno/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/biosíntesis , Neoplasias de la Próstata/metabolismo , beta Catenina/biosíntesis , Antígenos CD/genética , Cadherinas/genética , Línea Celular Tumoral , Receptor beta de Estrógeno/genética , Humanos , Masculino , Proteínas de Neoplasias/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Transducción de Señal , beta Catenina/genética
8.
Mol Cell Endocrinol ; 430: 12-24, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27107935

RESUMEN

The aim of the present study was to characterize the mechanism underlying estrogen effects on the androgen-independent prostate cancer cell line PC-3. 17ß-estradiol and the ERß-selective agonist DPN, but not the ERα-selective agonist PPT, increased the incorporation of [methyl-(3)H]thymidine and the expression of Cyclin D2, suggesting that ERß mediates the proliferative effect of estrogen on PC-3 cells. In addition, upregulation of Cyclin D2 and incorporation of [methyl-(3)H]thymidine induced by 17ß-estradiol and DPN were blocked by the ERß-selective antagonist PHTPP in PC-3 cells. Upregulation of Cyclin D2 and incorporation of [methyl-(3)H]thymidine induced by DPN were also blocked by PKF118-310, a compound that disrupts ß-catenin-TCF (T-cell-specific transcription factor) complex, suggesting the involvement of ß-catenin in the estradiol effects in PC-3 cells. A diffuse immunostaining for non-phosphorylated ß-catenin was detected in the cytoplasm of PC-3 cells. Low levels of non-phosphorylated ß-catenin immunostaining were also detected near the plasma membrane and in nuclei. Treatment of PC-3 cells with 17ß-estradiol or DPN markedly increased non-phosphorylated ß-catenin expression. These effects were blocked by pretreatment with the ERß-selective antagonist PHTPP, PI3K inhibitor Wortmannin or AKT inhibitor MK-2206, indicating that ERß-PI3K/AKT mediates non-phosphorylated ß-catenin expression. Cycloheximide blocked the DPN-induced upregulation of non-phosphorylated ß-catenin, suggesting de novo synthesis of this protein. In conclusion, these results suggest that estrogen may play a role in androgen-independent prostate cancer cell proliferation through a novel pathway, involving ERß-mediated activation of ß-catenin.


Asunto(s)
Receptor beta de Estrógeno/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , beta Catenina/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D1/metabolismo , Ciclina D2/metabolismo , Cicloheximida/farmacología , Estradiol/farmacología , Receptor beta de Estrógeno/agonistas , Humanos , Masculino , Nitrilos/farmacología , Fenoles/farmacología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Timidina/metabolismo
9.
J Exp Clin Cancer Res ; 34: 25, 2015 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-25887999

RESUMEN

BACKGROUND: SULF2 is a 6-O-endosulfatase which removes 6-O sulfate residues from N-glucosamine present on heparan sulfate (HS). The sulfation pattern of HS influences signaling events mediated by heparan sulfate proteoglycans (HSPGs) located on cell surface, which are critical for the interactions with growth factors and their receptors. Alterations in SULF2 expression have been identified in the context of several cancer types but its function in cancer is still unclear where the precise molecular mechanism involved has not been fully deciphered. To further investigate SULF2 role in tumorigenesis, we overexpressed such gene in prostate cancer cell lines. METHODS: The normal prostate epithelial cell line RWPE-1 and the prostate cancer cells DU-145, and PC3 were transfected with SULF2-expressing plasmid pcDNA3.1/Myc-His(-)-Hsulf-2. Transfected cells were then submitted to viability, migration and colony formation assays. RESULTS: Transfection of DU-145 and PC3 prostate cancer cells with SULF2 resulted in increased viability, which did not occur with normal prostate cells. The effect was reverted by the knockdown of SULF2 using specific siRNAs. Furthermore, forced expression of SULF2 augmented cell migration and colony formation in both prostate cell lines. Detailed structural analysis of HS from cells overexpressing SULF2 showed a reduction of the trisulfated disaccharide UA(2S)-GlcNS(6S). There was an increase in epithelial-mesenchymal transition markers and an increase in WNT signaling pathway. CONCLUSIONS: These results indicate that SULF2 have a pro-tumorigenic effect in DU-145 and PC3 cancer cells, suggesting an important role of this enzyme in prostatic cancer metastasis.


Asunto(s)
Transformación Celular Neoplásica/genética , Expresión Génica , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Sulfotransferasas/genética , Biomarcadores , Línea Celular Tumoral , Movimiento Celular/genética , Supervivencia Celular/genética , Técnicas de Cocultivo , Activación Enzimática , Transición Epitelial-Mesenquimal/genética , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Neoplasias de la Próstata/metabolismo , ARN Interferente Pequeño , Células del Estroma/metabolismo , Sulfatasas , Sulfotransferasas/metabolismo , Ensayo de Tumor de Célula Madre
10.
Mol Cancer Res ; 13(3): 510-23, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25477293

RESUMEN

UNLABELLED: Heparan sulfate endosulfatase-1 and -2 (SULF1 and SULF2) are two important extracellular 6-O-endosulfatases that remove 6-O sulfate groups of N-glucosamine along heparan sulfate (HS) proteoglycan chains often found in the extracellular matrix. The HS sulfation pattern influences signaling events at the cell surface, which are critical for interactions with growth factors and their receptors. SULFs are overexpressed in several types of human tumors, but their role in cancer is still unclear because their molecular mechanism has not been fully explored and understood. To further investigate the functions of these sulfatases in tumorigenesis, stable overexpression models of these genes were generated in the colorectal cancer cells, Caco-2 and HCT-116. Importantly, mimicking overexpression of these sulfatases resulted in increased viability and proliferation, and augmented cell migration. These effects were reverted by shRNA-mediated knockdown of SULF1 or SULF2 and by the addition of unfractionated heparin. Detailed structural analysis of HS from cells overexpressing SULFs showed reduction in the trisulfated disaccharide UA(2S)-GlcNS(6S) and corresponding increase in UA(2S)-GlcNS disaccharide, as well as an unexpected rise in less common disaccharides containing GlcNAc(6S) residues. Moreover, cancer cells transfected with SULFs demonstrated increased Wnt signaling. In summary, SULF1 or SULF2 overexpression contributes to colorectal cancer cell proliferation, migration, and invasion. IMPLICATIONS: This study reveals that sulfatases have oncogenic effects in colon cancer cells, suggesting an important role for these enzymes in cancer progression.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Sulfotransferasas/metabolismo , Células CACO-2 , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Neoplasias Colorrectales/genética , Células HCT116 , Heparitina Sulfato/química , Heparitina Sulfato/metabolismo , Humanos , Sulfatasas , Sulfotransferasas/genética , Vía de Señalización Wnt
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA