Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Breast Cancer Res ; 20(1): 9, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29382358

RESUMEN

BACKGROUND: The Fos-related antigen 1 (FRA-1) transcription factor promotes tumor cell growth, invasion and metastasis. Phosphorylation of FRA-1 increases protein stability and function. We identify a novel signaling axis that leads to increased phosphorylation of FRA-1, increased extracellular matrix (ECM)-induced breast cancer cell invasion and is prognostic of poor outcome in patients with breast cancer. METHODS: While characterizing five breast cancer cell lines derived from primary human breast tumors, we identified BRC-31 as a novel basal-like cell model that expresses elevated FRA-1 levels. We interrogated the functional contribution of FRA-1 and an upstream signaling axis in breast cancer cell invasion. We extended this analysis to determine the prognostic significance of this signaling axis in samples derived from patients with breast cancer. RESULTS: BRC-31 cells display elevated focal adhesion kinase (FAK), SRC and extracellular signal-regulated (ERK2) phosphorylation relative to luminal breast cancer models. Inhibition of this signaling axis, with pharmacological inhibitors, reduces the phosphorylation and stabilization of FRA-1. Elevated integrin αVß3 and uPAR expression in these cells suggested that integrin receptors might activate this FAK-SRC-ERK2 signaling. Transient knockdown of urokinase/plasminogen activator urokinase receptor (uPAR) in basal-like breast cancer cells grown on vitronectin reduces FRA-1 phosphorylation and stabilization; and uPAR and FRA-1 are required for vitronectin-induced cell invasion. In clinical samples, a molecular component signature consisting of vitronectin-uPAR-uPA-FRA-1 predicts poor overall survival in patients with breast cancer and correlates with an FRA-1 transcriptional signature. CONCLUSIONS: We have identified a novel signaling axis that leads to phosphorylation and enhanced activity of FRA-1, a transcription factor that is emerging as an important modulator of breast cancer progression and metastasis.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas Proto-Oncogénicas c-fos/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/genética , Neoplasias de la Mama/patología , Matriz Extracelular/genética , Femenino , Humanos , Integrina alfaVbeta3/administración & dosificación , Integrina alfaVbeta3/genética , Células MCF-7 , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Fosforilación , Transducción de Señal/efectos de los fármacos , Vitronectina/administración & dosificación
2.
Mol Cancer ; 15(1): 58, 2016 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-27613122

RESUMEN

BACKGROUND: Ovarian cancer (OC) ascites consist in a proinflammatory tumor environment that is characterized by the presence of various cytokines, chemokines and growth factors. The presence of these inflammatory-related factors in ascites is associated with a more aggressive tumor phenotype. CCL18 is a member of CCL chemokines and its expression has been associated with poor prognosis in some cancers. However, its role in OC progression has not been established. Therefore, the aim of the current study was to elucidate the role of ascites CCL18 in OC progression. METHODS: ELISA and tissue microarrays were used to assess CCL18 in ascites and phospho-Pyk2 expression in cancer tissues respectively. Cell migration was assessed using Boyden chambers. CCL18 and ascites signaling was examined in ovarian cancer cells utilizing siRNA and exogenous gene expression. RESULTS: Here, we show that CCL18 levels are markedly increased in advanced serous OC ascites relative to peritoneal effusions from women with benign conditions. Ascites and CCL18 dose-dependently enhanced the migration of OC cell lines CaOV3 and OVCAR3. CCL18 levels in ascites positively correlated with the ability of ascites to promote cell migration. CCL18 blocking antibodies significantly attenuated ascites-induced cell migration. Ascites and CCL18 stimulated the phosphorylation of proline-rich tyrosine kinase 2 (Pyk2) in CaOV3 and OVCAR3 cells. Most importantly, the expression of phosphorylated Pyk2 in serous OC tumors was associated with shorter progression-free survival. Furthermore, enforced expression of Pyk2 promoted tumor cell migration while siRNA-mediated downregulation of Pyk2 attenuated cell migration. Downregulation of Pyk2 markedly inhibited ascites and CCL18-induced cell migration. CONCLUSIONS: Taken together, our findings establish an important role for CCL18, as a component of ascites, in the migration of tumor cells and identify Pyk2 as prognostic factor and a critical downstream signaling pathway for ascites-induced OC cell migration.


Asunto(s)
Ascitis/metabolismo , Quimiocinas CC/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Neoplasias Ováricas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Humanos , Fosforilación , Pronóstico , Transducción de Señal
3.
Int J Cancer ; 137(2): 289-98, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25482018

RESUMEN

Ovarian cancer ascites consist of a proinflammatory environment that is characterized by the presence of abundant human peritoneal mesothelial cells (HPMCs). Cytokines and growth factors in ascites modulate cell activities of tumor cells. The expression of proinflammatory cytokines in ascites is associated with a more aggressive tumor phenotype. The effect of ascites on HPMCs is for the most part unknown but this interplay is thought to be important for epithelial ovarian cancer (EOC) progression. Here, we examine the components of ascites, which stimulate patient-derived HPMC migration, from women with advanced EOC. We show that ovarian cancer ascites enhanced the migration of HPMCs. This effect was inhibited by heat treatment, hepatocyte growth factor (HGF) blocking antibodies and a HGF receptor (cMet) inhibitor. In ovarian cancer ascites, HGF is present at high concentration compared to benign fluids. Ascites-mediated activation of cMet was associated with Akt and EKR1/2 phosphorylation. This response was partly inhibited by heat treatment and cMet inhibitor. Ascites-induced migration and a cMet phosphorylation were strongly inhibited by epidermal growth factor receptor (EGFR) inhibitor PD153035, suggesting the transactivation of cMet by EGFR. Our study suggests that HGF and ligands of EGFR are factors that mediate ovarian cancer ascites-mediated migration of HPMCs by activating cMet and possibly downstream ERK1/2 and Akt pathways. The study provides evidence for the first time that ascites not only support tumor growth but also enhance the migratory potential of cancer-associated mesothelial cells, which in turn may support cancer progression.


Asunto(s)
Ascitis/metabolismo , Movimiento Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Factor de Crecimiento de Hepatocito/farmacología , Proteínas Proto-Oncogénicas c-met/metabolismo , Células Cultivadas , Citocinas/metabolismo , Citocinas/farmacología , Activación Enzimática/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Receptores ErbB/metabolismo , Femenino , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Immunoblotting , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Cavidad Peritoneal/patología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Pirazinas/farmacología , Transducción de Señal/efectos de los fármacos , Triazoles/farmacología
4.
BMC Cancer ; 15: 492, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26122176

RESUMEN

BACKGROUND: Platinum-based combination therapy is the standard first-line treatment for women with advanced serous epithelial ovarian carcinoma (EOC). However, about 20 % will not respond and are considered clinically resistant. The availability of biomarkers to predict responses to the initial therapy would provide a practical approach to identify women who would benefit from a more appropriate first-line treatment. Ascites is an attractive inflammatory fluid for biomarker discovery as it is easy and minimally invasive to obtain. The aim of this study was to evaluate whether six selected inflammation-regulating factors in ascites could serve as diagnostic or drug resistance biomarkers in patients with advanced serous EOC. METHODS: A total of 53 women with stage III/IV serous EOC and 10 women with benign conditions were enrolled in this study. Eleven of the 53 women with serous EOC were considered clinically resistant to treatment with progression-free survival<6 months. Ascites were collected at the time of the debulking surgery and the levels of cytokines were measured by ELISA. The six selected cytokines were evaluated for their ability to discriminate serous EOC from benign controls, and to discriminate platinum resistant from platinum sensitive patients. RESULTS: Median ascites levels of IL-6, IL-10 and osteoprotegerin (OPG) were significantly higher in women with advanced serous EOC than in controls (P≤0.012). There were no significant difference in the median ascites levels of leptin, soluble urokinase plasminogen activator receptor (suPAR) and CCL18 among serous EOC women and controls. In Receiver Operator curve (ROC) analysis, IL-6, IL-10 and OPG had a high area under the curve value of 0.905, 0.832 and 0.825 respectively for distinguishing EOC from benign controls. ROC analysis of individual cytokines revealed low discriminating potential to stratify patients according to their sensitivity to first-line treatment. The combination of biomarkers with the highest discriminating potential was with CA125 and leptin (AUC=0.936, 95% CI: 0.894-0.978). CONCLUSION: IL-6 was found to be strongly associated with advanced serous EOC and could be used in combination with serum CA125 to discriminate benign and EOC. Furthermore, the combination of serum CA125 and ascites leptin was a strong predictor of clinical resistance to first-line therapy.


Asunto(s)
Biomarcadores de Tumor/genética , Antígeno Ca-125/genética , Cistadenocarcinoma Seroso/genética , Interleucina-6/genética , Proteínas de la Membrana/genética , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Ováricas/genética , Adulto , Anciano , Anciano de 80 o más Años , Ascitis/metabolismo , Ascitis/patología , Líquido Ascítico/metabolismo , Carcinoma Epitelial de Ovario , Cistadenocarcinoma Seroso/tratamiento farmacológico , Cistadenocarcinoma Seroso/patología , Diagnóstico Diferencial , Supervivencia sin Enfermedad , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Persona de Mediana Edad , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Microambiente Tumoral/genética
5.
BMC Cancer ; 14: 288, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24761768

RESUMEN

BACKGROUND: Malignant ascites is often present at diagnostic in women with advanced ovarian cancer (OC) and its presence is associated with a worse outcome. Human peritoneal mesothelial cells (HPMCs) are key components of malignant ascites. Although the interplay between HPMCs and OC cells is believed to be critical for tumor progression, it has not been well characterized. The purpose of this study was to assess the effect of ascites on HPMCs and clarify the role of HPMCs in OC progression. METHODS: Human OC ascites and benign peritoneal fluids were assessed for their ability to stimulate HPMC proliferation. Conditioned medium from ascites- and benign fluid-stimulated HPMCs were compared for their ability to attenuate apoptosis induced by TNF-related apoptosis-inducing ligand (TRAIL). We conducted a comparative analysis of global expression changes in ascites-stimulated HPMCs using Agilent oligonucleotide microarrays. RESULTS: As compared to benign peritoneal fluids, malignant ascites stimulated the proliferation of HPMCs. TRAIL-induced apoptosis was attenuated in OC cells exposed to conditioned medium from ascites-stimulated HPMCs as compared to OC cells exposed to conditioned medium from benign fluid-stimulated HPMCs. A total of 649 genes were differentially expressed in ascites-stimulated HPMCs. Based on a ratio of more than 1.5-fold and a P < 0.05, 484 genes were up-regulated and 165 genes were down-regulated in ascites-exposed HPMCs. Stimulation of HPMCs with OC ascites resulted in differential expression of genes mainly associated with the regulation of cell growth and proliferation, cell death, cell cycle and cell assembly and organization, compared to benign peritoneal fluids. Top networks up-regulated by OC ascites included Akt and NF-κB survival pathways whereas vascular endothelial growth factor (VEGF) pathway was down-regulated. CONCLUSIONS: The results of this study not only provide evidence supporting the importance of the interplay between cancer cells and HPMCs but also define the role that the tumor environment plays in these interactions.


Asunto(s)
Proliferación Celular/genética , Células Epiteliales/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Mesoteliales/genética , Neoplasias Ováricas/genética , Apoptosis/genética , Ascitis/metabolismo , Ascitis/patología , Células Epiteliales/patología , Femenino , Humanos , Neoplasias Mesoteliales/complicaciones , Neoplasias Mesoteliales/patología , Neoplasias Ováricas/complicaciones , Neoplasias Ováricas/patología , Cavidad Peritoneal/patología , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Microambiente Tumoral/genética , Factor A de Crecimiento Endotelial Vascular
6.
BMC Cancer ; 14: 234, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24690311

RESUMEN

BACKGROUND: MUC16 (CA125) is a large transmembrane mucin protein (> 200 kDa) aberrantly expressed in approximately 80% of human epithelial ovarian cancers (EOC). MUC16 expression in EOC cells is associated with increased tumorigenesis and inhibiton of genotoxic drug-induced apoptosis. However, the mechanism by which MUC16 mediates these effects is unknown. In the present study, we investigated the mechanisms by which MUC16 attenuates TRAIL-induced apoptosis. METHODS: MUC16 expression was down-regulated by stably expressing an anti-MUC16 single-chain antibody (scFv) targeted to the endoplasmic reticulum (ER), which prevents cell surface localization of MUC16 in OVCAR3 cells. We also generated a MUC16 C-terminal domain (MUC16CTD) construct that was stably expressed in MUC16 negative SKOV3 cells. RESULTS: We show that MUC16 attenuates apoptosis, activation of caspase-8 and mitochondria activation in EOC cells in response to TRAIL. MUC16 decreases TRAIL receptor R2 (DR5) expression and inhibits pro-caspase-8 activation at the death-inducing signaling complex (DISC). MUC16CTD expression is sufficient to attenuate the TRAIL signaling cascade. MUC16 knockdown decreases caspase-8 inhibitor cFLIP mRNA levels, increases cFLIP degradation, and consequently increases TRAIL-induced apoptosis. Down-regulation of cFLIP following treatment of MUC16-expressing OVCAR3 cells with cFLIP siRNA also increases TRAIL-induced apoptosis. CONCLUSIONS: These findings indicate that MUC16 protects EOC cells against TRAIL-induced apoptosis through multiple mechanisms including the blockade of TRAIL R2 expression and the regulation of cFLIP expression at both the transcriptional and the protein level.


Asunto(s)
Antígeno Ca-125/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/biosíntesis , Proteínas de la Membrana/genética , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Ováricas/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Apoptosis/efectos de los fármacos , Apoptosis/genética , Antígeno Ca-125/metabolismo , Carcinogénesis/genética , Carcinoma Epitelial de Ovario , Caspasa 8/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de la Membrana/metabolismo , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Proteolisis , ARN Interferente Pequeño , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Transcripción Genética
7.
Mol Cancer ; 11: 84, 2012 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-23158473

RESUMEN

BACKGROUND: Ascites may affect the progression of ovarian cancer (OC). In particular, soluble factors present in OC ascites can create a protective environment for tumor cells that promote de novo resistance to drug- and death receptor-induced apoptosis. However, the underlying molecular mechanisms responsible for ascites-induced drug resistance are not well characterized. METHODS: Using human OC cell lines and tissues microarrays of human OC biopsies, we assessed the mechanism by which OC ascites increase Mcl-1 expression using Western blots, chemical inhibitors of ERK and small-inhibitory RNA treatments. RESULTS: In the present study, we found that both Mcl-1 mRNA and protein levels were upregulated within 2 h upon treatment of OC cells with ascites obtained from women with advanced OC. In contrast, the expression of other Bcl-2 family antiapoptotic members such as Bcl-2 and Bcl-XL was not affected by ascites. An increase of Mcl-1 expression was consistently observed across different ascites from women with advanced serous OC. The knockdown of Mcl-1 significantly blocked ascites-induced Mcl-1 upregulation and ascites-mediated inhibition of TRAIL-induced apoptosis. Ascites induced a rapid phosphorylation of ERK1/2 and Elk-1 transcription factor. Furthermore, we found that ERK1/2 inhibition or Elk-1 knockdown was sufficient to block ascites-induced Mcl-1 expression. In high grade serous OC, we found a positive correlation between phosphorylated ERK1/2 and Mcl-1 expression. CONCLUSIONS: These results indicate that ascites-induced ERK1/2/Elk-1 signaling is critical for Mcl-1 expression and for the ascites-mediated attenuation of TRAIL-induced apoptosis. The ERK1/2/Elk-1/Mcl-1 pathway represents a novel mechanism by which ascites induce de novo TRAIL resistance in OC cells.


Asunto(s)
Ascitis/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteína Elk-1 con Dominio ets/metabolismo , Apoptosis/genética , Ascitis/genética , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Clasificación del Tumor , Neoplasias Ováricas/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Transducción de Señal
8.
BMC Cancer ; 11: 210, 2011 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-21619709

RESUMEN

BACKGROUND: The acellular fraction of epithelial ovarian cancer (EOC) ascites promotes de novo resistance of tumor cells and thus supports the idea that tumor cells may survive in the surrounding protective microenvironment contributing to disease recurrence. Levels of the pro-inflammatory cytokines IL-6 and IL-8 are elevated in EOC ascites suggesting that they could play a role in tumor progression. METHODS: We measured IL-6 and IL-8 levels in the ascites of 39 patients with newly diagnosed EOC. Commercially available enzyme-linked immunosorbent assay (ELISA) was used to determine IL-6 and IL-8 ascites levels. Ascites cytokine levels were correlated with clinicopathological parameters and progression-free survival. RESULTS: Mean ascites levels for IL-6 and IL-8 were 6419 pg/ml (SEM: 1409 pg/ml) and 1408 pg/ml (SEM: 437 pg/ml) respectively. The levels of IL-6 and IL-8 in ascites were significantly lower in patients that have received prior chemotherapy before the surgery (Mann-Whitney U test, P = 0.037 for IL-6 and P = 0.008 for IL-8). Univariate analysis revealed that high IL-6 ascites levels (P = 0.021), serum CA125 levels (P = 0.04) and stage IV (P = 0.009) were significantly correlated with shorter progression-free survival. Including these variables in a multivariate analysis revealed that elevated IL-6 levels (P = 0.033) was an independent predictor of shorter progression-free survival. CONCLUSION: Elevated IL-6, but not IL-8, ascites level is an independent predictor of shorter progression-free survival.


Asunto(s)
Ascitis/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Neoplasias Glandulares y Epiteliales/diagnóstico , Neoplasias Glandulares y Epiteliales/fisiopatología , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/fisiopatología , Carcinoma Epitelial de Ovario , Femenino , Humanos , Estadificación de Neoplasias , Neoplasias Glandulares y Epiteliales/mortalidad , Neoplasias Ováricas/mortalidad , Pronóstico , Análisis de Supervivencia
9.
Gynecol Oncol ; 121(3): 434-43, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21421261

RESUMEN

OBJECTIVES: MUC16 (CA125) protein is a high molecular weight mucin overexpressed in the majority of epithelial ovarian cancers (EOC) but not in the epithelium of normal ovaries suggesting that it might play a role in EOC pathogenesis. Here, we explored the phenotypic consequences of MUC16 knockdown and expression of its C-terminal domain with the aim of establishing a role for MUC16 in tumorigenesis. METHODS: MUC16 was down-regulated by stably expressing an anti-MUC16 endoplasmic reticulum-targeted single-chain antibody which prevented MUC16 cell surface localization in NIH:OVCAR3 cells. In addition, we generated epitope tagged, N-terminal region-deleted MUC16 constructs with (MUC16TMU) and without (MUC16CTD) cytoplasmic tail deletions and stably expressed them in SKOV3 cells. RESULTS: Although MUC16 knockdown did not affect the cell growth rate, knockdown cells reached a stationary growth phase after 4 days whereas control cells continued to grow for up to 7 days. Colony formation assays in soft agar demonstrated that MUC16 knockdown cells had >8-fold reduction in their ability to form colonies. Importantly, MUC16 knockdown completely prevents the formation of subcutaneous tumors in nude mice. Conversely, we show that ectopic expression of the MUC16CTD enhances SKOV3 tumor cell growth, colony formation in soft agar and enhances tumor growth and metastases in SCID mice. In addition, MUC16CTD expression increases cell motility, invasiveness, and metastatic property. Deletion of the cytoplasmic tail from the MUC16CTD completely abolished its ability to enhance tumor cell growth, cell motility and invasiveness. Furthermore, the increased invasive properties of MUC16CTD-expressing cells correlated with decreased expression of E-cadherin and increased expression of N-cadherin and vimentin. CONCLUSION: These findings provide the first evidence for a critical role of MUC16 in tumor cell growth, tumorigenesis and metastases.


Asunto(s)
Antígeno Ca-125/biosíntesis , Proteínas de la Membrana/biosíntesis , Animales , Antígeno Ca-125/genética , Cadherinas/biosíntesis , Cadherinas/metabolismo , Carcinoma Epitelial de Ovario , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Regulación hacia Abajo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Desnudos , Ratones SCID , Metástasis de la Neoplasia , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Transfección , Trasplante Heterólogo , Vimentina/biosíntesis
10.
Mol Cancer ; 9: 118, 2010 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-20497550

RESUMEN

BACKGROUND: MUC16 is a cell surface mucin expressed at high levels by epithelial ovarian tumors. Following proteolytic cleavage, cell surface MUC16 (csMUC16) is shed in the extracellular milieu and is detected in the serum of cancer patients as the tumor marker CA125. csMUC16 acts as an adhesion molecule and facilitates peritoneal metastasis of ovarian tumors. Both sMUC16 and csMUC16 also protect cancer cells from cytotoxic responses of natural killer (NK) cells. In a previous study we demonstrated that sMUC16 binds to specific subset of NK cells. Here, we identify the csMUC16/sMUC16 binding partner expressed on immune cells. RESULTS: Analysis of immune cells from the peripheral blood and peritoneal fluid of ovarian cancer patients indicates that in addition to NK cells, sMUC16 also binds to B cells and monocytes isolated from the peripheral blood and peritoneal fluid. I-type lectin, Siglec-9, is identified as the sMUC16 receptor on these immune cells. Siglec-9 is expressed on approximately 30-40% of CD16pos/CD56dim NK cells, 20-30% of B cells and >95% of monocytes. sMUC16 binds to the majority of the Siglec-9pos NK cells, B cells and monocytes. sMUC16 is released from the immune cells following neuraminidase treatment. Siglec-9 transfected Jurkat cells and monocytes isolated from healthy donors bind to ovarian tumor cells via Siglec-9-csMUC16 interaction. CONCLUSIONS: Recent studies indicate that csMUC16 can act as an anti-adhesive agent that blocks tumor-immune cell interactions. Our results demonstrate that similar to other mucins, csMUC16 can also facilitate cell adhesion by interacting with a suitable binding partner such as mesothelin or Siglec-9. Siglec-9 is an inhibitory receptor that attenuates T cell and NK cell function. sMUC16/csMUC16-Siglec-9 binding likely mediates inhibition of anti-tumor immune responses.


Asunto(s)
Antígenos CD/metabolismo , Linfocitos B/metabolismo , Antígeno Ca-125/metabolismo , Células Asesinas Naturales/metabolismo , Lectinas/metabolismo , Proteínas de la Membrana/metabolismo , Monocitos/metabolismo , Neoplasias Ováricas/metabolismo , Antígenos CD/inmunología , Linfocitos B/inmunología , Western Blotting , Antígeno Ca-125/inmunología , Separación Celular , Femenino , Citometría de Flujo , Humanos , Células Jurkat , Células Asesinas Naturales/inmunología , Lectinas/inmunología , Proteínas de la Membrana/inmunología , Monocitos/inmunología , Neoplasias Ováricas/inmunología , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico
11.
Mol Cancer ; 9: 11, 2010 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-20089172

RESUMEN

BACKGROUND: Cancer cells utilize a variety of mechanisms to evade immune detection and attack. Effective immune detection largely relies on the formation of an immune synapse which requires close contact between immune cells and their targets. Here, we show that MUC16, a heavily glycosylated 3-5 million Da mucin expressed on the surface of ovarian tumor cells, inhibits the formation of immune synapses between NK cells and ovarian tumor targets. Our results indicate that MUC16-mediated inhibition of immune synapse formation is an effective mechanism employed by ovarian tumors to evade immune recognition. RESULTS: Expression of low levels of MUC16 strongly correlated with an increased number of conjugates and activating immune synapses between ovarian tumor cells and primary naïve NK cells. MUC16-knockdown ovarian tumor cells were more susceptible to lysis by primary NK cells than MUC16 expressing controls. This increased lysis was not due to differences in the expression levels of the ligands for the activating receptors DNAM-1 and NKG2D. The NK cell leukemia cell line (NKL), which does not express KIRs but are positive for DNAM-1 and NKG2D, also conjugated and lysed MUC16-knockdown cells more efficiently than MUC16 expressing controls. Tumor cells that survived the NKL challenge expressed higher levels of MUC16 indicating selective lysis of MUC16(low) targets. The higher csMUC16 levels on the NKL resistant tumor cells correlated with more protection from lysis as compared to target cells that were never exposed to the effectors. CONCLUSION: MUC16, a carrier of the tumor marker CA125, has previously been shown to facilitate ovarian tumor metastasis and inhibits NK cell mediated lysis of tumor targets. Our data now demonstrates that MUC16 expressing ovarian cancer cells are protected from recognition by NK cells. The immune protection provided by MUC16 may lead to selective survival of ovarian cancer cells that are more efficient in metastasizing within the peritoneal cavity and also at overcoming anti-tumor innate immune responses.


Asunto(s)
Antígeno Ca-125/inmunología , Citoprotección/inmunología , Sinapsis Inmunológicas/inmunología , Células Asesinas Naturales/inmunología , Proteínas de la Membrana/inmunología , Neoplasias Ováricas/inmunología , Antígenos de Diferenciación de Linfocitos T/metabolismo , Adhesión Celular , Recuento de Células , Línea Celular Tumoral , Membrana Celular/metabolismo , Supervivencia Celular , Citotoxicidad Inmunológica , Femenino , Técnicas de Silenciamiento del Gen , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Células Asesinas Naturales/patología , Ligandos , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Neoplasias Ováricas/patología
12.
Mol Cell Endocrinol ; 301(1-2): 146-53, 2009 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-18812208

RESUMEN

We investigated the relative involvement of three reductive 17beta-hydroxysteroid dehydrogenase (17beta-HSD) isoforms, namely types 1, 7 and 12, in the formation of potent estrogen estradiol (E2) in 10 human breast cancer cell lines (T-47D, MCF-7, ZR-75-1, CAMA-1, BT-20, BRC-17, BRC-31, BRC-32, BRC-36 and BRN-196) and also in 1 choriocarcinoma cell line (JEG-3) using selective inhibitors. In T-47D, BT-20 and JEG-3 cells, a 17beta-HSD1 inhibitor almost completely inhibited the formation of E2 at 1microM from 60nM of estrone (E1) (98%, 91% and 90%, respectively), whereas no significant inhibition of E2 formation was obtained using inhibitors of types 7 and 12. However, we obtained lower levels of inhibition (32%, 36% and 35% respectively using inhibitors of types 1, 7 and 12 at 10microM) in MCF-7 cells and even lower and variable levels of inhibition (15%, 23% and 18% respectively using inhibitors of types 1, 7 and 12 at 10microM) in ZR-75-1 cells. No inhibition of E2 formation was observed in CAMA-1 cells with a 17beta-HSD1 inhibitor at 1microM whereas inhibitors of types 7 and 12 inhibited 40% and 30% of E2 formation, respectively. In BRC and BRN cell lines, types 1, 7 and 12 17beta-HSDs were all involved in the formation of E2, but type 12 seemed to predominate. At 10microM, each inhibitor inhibited 10-50% of the formation of E2. Using MCF-7 and BRC-32 cell lines, a combination of the three inhibitors (3x10microM) does not fully inhibit the 17beta-HSD activity (65% and 75%). In addition to identify the relative importance of types 1, 7 and 12 17beta-HSDs in the formation of E2 in human breast cancer cell lines, our results show also a great variability between each cell line. In some cases the formation of E2 was completely inhibited, but this was not the result observed in other cell lines, suggesting the presence of another enzyme involved in the biosynthesis of E2.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , 17-Hidroxiesteroide Deshidrogenasas/metabolismo , Neoplasias de la Mama/enzimología , Inhibidores Enzimáticos/farmacología , Estradiol/biosíntesis , 17-Hidroxiesteroide Deshidrogenasas/clasificación , Neoplasias de la Mama/genética , Línea Celular Tumoral , Estradiol/química , Estrona/química , Estrona/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especificidad por Sustrato/efectos de los fármacos
13.
Gynecol Oncol ; 115(3): 407-13, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19747716

RESUMEN

OBJECTIVE: Little is known about the biological functions of CA125/MUC16 tumor antigen. Here, we examined the role of CA125/MUC16 in regulating the sensitivity of epithelial ovarian carcinoma (EOC) cells to different drugs. METHODS: An endoplasmic reticulum targeted single-chain antibody (scFv) was used to down-regulate cell surface expression of CA125/MUC16 in NIH:OVCAR3 cells and the C-terminal domain (CTD) of MUC16 was ectopically expressed in CA125-negative SKOV3 cells. Sensitivity to genotoxic agents and to inhibitors of microtubule depolymerization was examined in NIH:OVCAR3 and SKOV3 cell sublines. Cell viability was determined by XTT assay, apoptosis by propidium iodide staining and caspase activation by Western blot and fluorogenic assay. RESULTS: Down-regulation of cell surface MUC16 decreases cisplatin IC(50) by 5-fold in NIH:OVCAR3 cells but does not affect paclitaxel IC(50). We found that the sensitivity to other genotoxic agents such as cyclophosphamide, doxorubicine and etoposide was also increased by down-regulation of MUC16. Caspase-9 and caspase-3 activation also significantly augmented in cisplatin-treated NIH:OVCAR3 cells expressing the anti-MUC16 scFv. Ectopic expression of MUC16 CTD has the opposite effect. Cisplatin sensitivity and caspases activation are decreased by the ectopic expression of MUC16 CTD in SKOV3 cells. CONCLUSIONS: CA125/MUC16 selectively modulates the sensitivity of EOC cells to genotoxic agents. The MUC16 CTD appears to be sufficient to promote cisplatin resistance.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Apoptosis/efectos de los fármacos , Antígeno Ca-125/biosíntesis , Neoplasias Ováricas/tratamiento farmacológico , Antígeno Ca-125/genética , Antígeno Ca-125/inmunología , Caspasas/metabolismo , Línea Celular Tumoral , Cisplatino/farmacología , Ciclofosfamida/farmacología , Regulación hacia Abajo/efectos de los fármacos , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Activación Enzimática/efectos de los fármacos , Etopósido/farmacología , Femenino , Humanos , Fragmentos de Inmunoglobulinas/inmunología , Fragmentos de Inmunoglobulinas/farmacología , Concentración 50 Inhibidora , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo
14.
BMC Cancer ; 8: 346, 2008 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-19032793

RESUMEN

BACKGROUND: Serous epithelial ovarian tumors can be subdivided into benign (BOV), low malignant potential (LMP) or borderline and invasive (TOV) tumors. Although the molecular characteristics of serous BOV, LMP and low grade (LG) TOV tumors has been initiated, definitive immunohistochemical markers to distinguish between these tumor types have not been defined. METHODS: In the present study, we used a tissue array composed of 27 BOVs, 78 LMPs and 23 LG TOVs to evaluate the protein expression of a subset of selected candidates identified in our previous studies (Ape1, Set, Ran, Ccne1 and Trail) or known to be implicated in epithelial ovarian cancer disease (p21, Ccnb1, Ckd1). RESULTS: Statistically significant difference in protein expression was observed for Ccnb1 when BOV tumors were compared to LMP tumors (p = 0.003). When BOV were compared to LG TOV tumors, Trail was significantly expressed at a higher level in malignant tumors (p = 0.01). Expression of p21 was significantly lower in LG tumors when compared with either BOVs (p = 0.03) or LMPs (p = 0.001). We also observed that expression of p21 was higher in LMP tumors with no (p = 0.02) or non-invasive (p = 0.01) implants compared to the LMP associated with invasive implants. CONCLUSION: This study represents an extensive analyse of the benign and highly differentiated ovarian disease from an immunohistochemical perspective.


Asunto(s)
Biomarcadores de Tumor/análisis , Cistadenoma Seroso/metabolismo , Cistadenoma Seroso/patología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Femenino , Humanos , Inmunohistoquímica , Análisis de Matrices Tisulares
15.
Mol Cancer ; 5(1): 50, 2006 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-17067392

RESUMEN

BACKGROUND: The mucin MUC16 and the glycosylphosphatidylinositol anchored glycoprotein mesothelin likely facilitate the peritoneal metastasis of ovarian tumors. The biochemical basis and the kinetics of the binding between these two glycoproteins are not clearly understood. Here we have addressed this deficit and provide further evidence supporting the role of the MUC16-mesothelin interaction in facilitating cell-cell binding under conditions that mimic the peritoneal environment. RESULTS: In this study we utilize recombinant-Fc tagged human mesothelin to measure the binding kinetics of this glycoprotein to MUC16 expressed on the ovarian tumor cell line OVCAR-3. OVCAR-3 derived sublines that did not express MUC16 showed no affinity for mesothelin. In a flow cytometry-based assay mesothelin binds with very high affinity to the MUC16 on the OVCAR-3 cells with an apparent Kd of 5-10 nM. Maximum interaction occurs within 5 mins of incubation of the recombinant mesothelin with the OVCAR-3 cells and significant binding is observed even after 10 sec. A five-fold molar excess of soluble MUC16 was unable to completely inhibit the binding of mesothelin to the OVCAR-3 cells. Oxidation of the MUC16 glycans, removal of its N-linked oligosaccharides, and treatment of the mucin with wheat germ agglutinin and erythroagglutinating phytohemagglutinin abrogates its binding to mesothelin. These observations suggest that at least a subset of the MUC16-asscociated N-glycans is required for binding to mesothelin. We also demonstrate that MUC16 positive ovarian tumor cells exhibit increased adherence to A431 cells transfected with mesothelin (A431-Meso+). Only minimal adhesion is observed between MUC16 knockdown cells and A431-Meso+ cells. The binding between the MUC16 expressing ovarian tumor cells and the A431-Meso+ cells occurs even in the presence of ascites from patients with ovarian cancer. CONCLUSION: The strong binding kinetics of the mesothelin-MUC16 interaction and the cell adhesion between ovarian tumor cells and A431-Meso+ even in the presence of peritoneal fluid strongly support the importance of these two glycoproteins in the peritoneal metastasis of ovarian tumors. The demonstration that N-linked glycans are essential for mediating mesothlein-MUC16 binding may lead to novel therapeutic targets to control the spread of ovarian carcinoma.


Asunto(s)
Antígeno Ca-125/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Polisacáridos/metabolismo , Unión Competitiva/fisiología , Antígeno Ca-125/genética , Adhesión Celular/fisiología , Línea Celular Tumoral , Femenino , Citometría de Flujo , Proteínas Ligadas a GPI , Humanos , Cinética , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/genética , Mesotelina , Modelos Biológicos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Neoplasias Peritoneales/metabolismo , Neoplasias Peritoneales/secundario
16.
Clin Exp Metastasis ; 33(8): 839-852, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27612856

RESUMEN

Epithelial ovarian cancer (EOC) dissemination is primarily mediated by the shedding of tumor cells from the primary site into ascites where they form multicellular spheroids that rapidly lead to peritoneal carcinomatosis. While the clinical importance and fundamental role of multicellular spheroids in EOC is increasingly appreciated, the mechanisms that regulate their formation and dictate their cellular composition remain poorly characterized. To investigate these important questions, we characterized spheroids isolated from ascites of women with EOC. We found that in these spheroids, a core of mesothelial cells was encased in a shell of tumor cells. Analysis further revealed that EOC spheroids are dynamic structures of proliferating, non-proliferating and hypoxic regions. To recapitulate these in vivo findings, we developed a three-dimensional co-culture model of primary EOC and mesothelial cells. Our analysis indicated that, compared to the OVCAR3 cell line, primary EOC cells isolated from ascites as well as mesothelial cells formed compact spheroids. Analysis of heterotypic spheroid microarchitecture revealed a structure that grossly resembles the structure of spheroids isolated from ascites. Cells that formed compact spheroids had elevated expression of ß1 integrin and low expression of E-cadherin. Addition of ß1 integrin blocking antibody or siRNA-mediated downregulation of ß1 integrin resulted in reduced tightness of the spheroids. Interestingly, the loss of MUC16 and E-cadherin expression resulted in the formation of more compact spheroids. Therefore, our findings support the heterotypic nature of spheroids from malignant EOC ascites. In addition, our data describe an unusual link between E-cadherin expression and less compact spheroids. Our data also emphasize the role of MUC16 and ß1 integrin in EOC spheroid formation.


Asunto(s)
Antígeno Ca-125/biosíntesis , Cadherinas/biosíntesis , Integrina beta1/biosíntesis , Proteínas de la Membrana/biosíntesis , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Ováricas/genética , Neoplasias Peritoneales/patología , Ascitis/genética , Ascitis/patología , Líquido Ascítico/patología , Antígeno Ca-125/genética , Cadherinas/genética , Carcinoma/genética , Carcinoma/patología , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Epitelio/metabolismo , Epitelio/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Integrina beta1/genética , Proteínas de la Membrana/genética , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/patología , Neoplasias Peritoneales/genética , Neoplasias Peritoneales/secundario , Esferoides Celulares/metabolismo , Esferoides Celulares/patología
17.
Int J Oncol ; 46(1): 91-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25338620

RESUMEN

MUC16 (CA125) is a transmembrane mucin that contributes to the progression of epithelial ovarian cancer (EOC). Expression of MUC16 is not detectable in the epithelial surface of normal ovaries. MUC16 expression is, however, common in serous EOC as well as in metastatic and recurrent tumors. Despite these observations, its contribution to the development of EOC is unknown. We stably expressed either empty vector, MUC16 C-terminal domain (MUC16 CTD) or MUC16 TMU (a construct that lacks the cytoplasmic tail) in NIH3T3 mouse fibroblast cells. In this study, we provide evidence for the role of MUC16 CTD in oncogenic transformation. We show that ectopic expression of MUC16 CTD enhances the growth of NIH3T3 cells under normal and low serum conditions, and promotes anchorage-dependent colony formation. The deletion of the cytoplasmic tail abrogated these effects. MUC16 CTD expression in NIH3T3 cells also enhances the formation of colony in soft agar as compared to MUC16 TMU. MUC16 CTD expression enhances tumor formation in nude mice. Our findings provide the first evidence that MUC16 induces the transformation of NIH3T3 cells and indicate that MUC16 functions as an oncogene. Furthermore, our data suggest that the cytoplasmic tail is critical for MUC16 oncogenic properties.


Asunto(s)
Antígeno Ca-125/química , Antígeno Ca-125/genética , Transformación Celular Neoplásica/genética , Fibroblastos/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Animales , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Transformación Celular Neoplásica/metabolismo , Medio de Cultivo Libre de Suero/farmacología , Citoplasma , Fibroblastos/patología , Ratones , Ratones Desnudos , Células 3T3 NIH , Oncogenes , Estructura Terciaria de Proteína/genética
18.
Am J Cancer Res ; 5(1): 219-30, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25628932

RESUMEN

After shedding from the primary tumor site, ovarian cancer cells form three-dimensional multicellular aggregates that serve as vehicle for cancer cell dissemination in the peritoneal cavity. MUC16 mucin (CA125) is aberrantly expressed by most advanced serous ovarian cancers and can promote proliferation, migration and metastasis. MUC16 associates with E-cadherin and ß-catenin, two proteins involved in regulation of cell adhesion and the formation of multicellular aggregates. However, the role of MUC16 in the formation of multicellular aggregates remains to be defined. Here, we show that MUC16 alters E-cadherin cellular localization and expression. Consistent with this, MUC16 knockdown inhibited the formation of multicellular aggregates and, conversely, forced expression of MUC16 C-terminal domain (CTD) enhanced the formation of multicellular aggregates. MUC16 knockdown induces ß-catenin relocation from the cell membrane to the cytoplasm, decreases its expression by increasing degradation and decreases ß-catenin target gene expression. MUC16 CTD inhibits GSK-3ß-mediated phosphorylation and degradation of ß-catenin, leading to increased ß-catenin levels. Importantly, knockdown of ß-catenin inhibited multicellular aggregation. These findings indicate that MUC16 promotes the formation of multicellular aggregates by inhibiting ß-catenin degradation.

19.
J Ovarian Res ; 6(1): 82, 2013 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-24267510

RESUMEN

BACKGROUND: Resistance to apoptosis is a major problem in ovarian cancer (OC) and correlates with poor prognosis. Osteoprotegerin (OPG) is a soluble secreted factor that acts as a decoy receptor for receptor activator of NF-κB ligand (RANKL) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). OPG has been reported to attenuate TRAIL-induced apoptosis in a variety of cancer cells, including OC cells. OPG-mediated protection against TRAIL has been attributed to its decoy receptor function. However, OPG activates integrin/focal adhesion kinase (FAK) signaling in endothelial cells. In OC cells, activation of integrin/FAK signaling inhibits TRAIL-induced apoptosis. Based on these observations, we hypothesized that OPG could attenuate TRAIL-induced apoptosis in OC cells through integrin/FAK signaling. METHODS: In vitro experiments including immunoblots, colony formation assays, and apoptosis measurements were used to assess the effect of OPG on TRAIL-induced apoptosis. RESULTS: Exogenous OPG protected from TRAIL-induced apoptosis in a TRAIL binding-independent manner and OPG protection was αvß3 and αvß5 integrin/FAK signaling-dependent. Moreover, OPG-mediated activation of integrin/FAK signaling resulted in the activation of Akt. Inhibition of both integrin/FAK and Akt signaling significantly inhibited OPG-mediated attenuation of TRAIL-induced apoptosis. Although OPG also stimulated ERK1/2 phosphorylation, inhibition of ERK1/2 signaling did not significantly altered OPG protection. CONCLUSIONS: Our studies provide evidence, for the first time, that OPG can attenuate TRAIL-induced apoptosis in a TRAIL binding-independent manner through the activation of integrin/FAK/Akt signaling in OC cells.

20.
J Ovarian Res ; 5(1): 34, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23153223

RESUMEN

BACKGROUND: Resistance to apoptosis is a major problem in ovarian cancer and correlates with poor prognosis. Osteoprotegerin (OPG) is a secreted factor in malignant ascites and acts as a decoy receptor for receptor activator of NF-κB ligand (RANKL) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). TRAIL promotes apoptosis in ovarian cancer cells. Ovarian cancer ascites attenuate TRAIL-induced apoptosis raising the possibility that OPG contained in ascites may abrogate the anti-tumor activity of TRAIL. METHODS: Determination of OPG levels in ascites was measured by ELISA. Effect of OPG on TRAIL-induced cell death was determined by XTT and colony forming assays in ovarian cancer cell lines and primary tumor cells. Apoptosis was assessed by ELISA. RESULTS: We found that recombinant OPG and malignant ascites attenuates TRAIL-induced cell death and apoptosis in a dose-dependent manner in ovarian cancer cell lines and primary ovarian tumor cells. OPG is present at high levels in the ascites of patients with ovarian cancer. We found a positive correlation between the levels of OPG in ascites and the ability of the ascites to attenuate TRAIL-induced cell death. The anti-apoptotic effect of ascites was not reversed by co-incubation with an OPG blocking antibody. CONCLUSIONS: OPG and malignant ascites protect ovarian cancer cells from TRAIL-induced apoptosis. Although malignant ascites contain high levels of OPG, OPG is not a critical component that contributes to ascites-mediated attenuation of TRAIL-induced apoptosis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA