Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Reproduction ; 137(4): 721-6, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19176311

RESUMEN

Opioid binding protein/cell adhesion molecule-like gene (OPCML) is frequently inactivated in epithelial ovarian cancer, but the role of this membrane protein in normal reproductive function is unclear. The ovarian surface epithelium (OSE) is thought to be the cell of origin of most epithelial ovarian cancers, some of which arise after transformation of OSE cells lining ovarian inclusion cysts, formed during ovulation. We used immunohistochemistry, immunoblotting and quantitative RT-PCR (qRT-PCR) to investigate OPCML expression in the uteri and ovaries of cycling 3-month CD-1 mice, as well as in ovaries from older mice containing inclusion cysts derived from rete ovarii tubules. Immunoblotting showed OPCML bands in uterine, but not whole ovarian or muscle extracts. Strong OPCML immunoreactivity was observed in oviduct, rete ovarii and uterus, whereas in ovary more immunoreactivity was seen in granulosa cells than OSE. No staining was observed in OSE around ovulation sites, where OSE cells divide to cover the site. OPCML immunoreactivity was also weaker in more dysplastic cells lining large ovarian inclusion cysts, compared with normal rete ovarii. No significant changes in Opcml mRNA expression were observed in whole ovarian and uterine extracts at different stages of the cycle. We conclude that murine OPCML is more consistently expressed in cells lining the uterus, oviduct and rete ovarii than in ovary and is not expressed in OSE associated with ovulation sites. This observation supports the hypothesis that a proportion of epithelial ovarian cancers arise from ductal cells and other epithelia of the secondary Mullerian system, rather than the OSE.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Genitales Femeninos/metabolismo , Animales , Femenino , Proteínas Ligadas a GPI/metabolismo , Immunoblotting , Inmunohistoquímica , Ratones , Ovulación , Reacción en Cadena en Tiempo Real de la Polimerasa
2.
PLoS One ; 3(8): e2990, 2008 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-18714356

RESUMEN

BACKGROUND: Identification of tumor suppressor genes (TSGs) silenced by CpG methylation uncovers the molecular mechanism of tumorigenesis and potential tumor biomarkers. Loss of heterozygosity at 11q25 is common in multiple tumors including nasopharyngeal carcinoma (NPC). OPCML, located at 11q25, is one of the downregulated genes we identified through digital expression subtraction. METHODOLOGY/PRINCIPAL FINDINGS: Semi-quantitative RT-PCR showed frequent OPCML silencing in NPC and other common tumors, with no homozygous deletion detected by multiplex differential DNA-PCR. Instead, promoter methylation of OPCML was frequently detected in multiple carcinoma cell lines (nasopharyngeal, esophageal, lung, gastric, colon, liver, breast, cervix, prostate), lymphoma cell lines (non-Hodgkin and Hodgkin lymphoma, nasal NK/T-cell lymphoma) and primary tumors, but not in any non-tumor cell line and seldom weakly methylated in normal epithelial tissues. Pharmacological and genetic demethylation restored OPCML expression, indicating a direct epigenetic silencing. We further found that OPCML is stress-responsive, but this response is epigenetically impaired when its promoter becomes methylated. Ecotopic expression of OPCML led to significant inhibition of both anchorage-dependent and -independent growth of carcinoma cells with endogenous silencing. CONCLUSIONS/SIGNIFICANCE: Thus, through functional epigenetics, we identified OPCML as a broad tumor suppressor, which is frequently inactivated by methylation in multiple malignancies.


Asunto(s)
Carcinoma/genética , Moléculas de Adhesión Celular/genética , Metilación de ADN , Epigénesis Genética/genética , Silenciador del Gen , Genes Supresores de Tumor , Linfoma/genética , Proteínas del Tejido Nervioso/genética , Empalme Alternativo , Cromosomas Humanos Par 11 , Femenino , Proteínas Ligadas a GPI , Variación Genética , Humanos , Pérdida de Heterocigocidad , Masculino , Neoplasias Nasofaríngeas/genética , Valores de Referencia , Transcripción Genética
3.
Clin Cancer Res ; 11(16): 5764-8, 2005 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16115914

RESUMEN

PURPOSE: The IgLON family of cell adhesion molecules, comprising OPCML, HNT, LSAMP, and NEGR1, has recently been linked to cancer, through two of its members being proposed as tumor suppressors. We examined the expression profile of the family in human sporadic epithelial ovarian cancer and the normal ovary. EXPERIMENTAL DESIGN: We determined the expression level of each IgLON in a panel comprising 57 tumor and 11 normal ovarian samples by quantitative real-time reverse transcription-PCR. The results were statistically tested for associations with clinicopathologic variables. RESULTS: OPCML, LSAMP and NEGR1 exhibited reduced expression in the tumor samples relative to the normal samples, whereas HNT expression was elevated. Statistically significant changes were specific to histologic type. The expression levels of individual IgLONs were correlated, the most significant finding being a positive correlation between LSAMP and NEGR1. LSAMP expression was also negatively correlated with overall survival and was found to be a negative predictor of outcome. CONCLUSIONS: The expression of the IgLON family is altered in sporadic epithelial ovarian tumors in comparison to the normal ovary. In our small but representative cohort of patients, we have found significant correlations and associations in expression and clinicopathology that suggest a wider role of the family in ovarian cancer.


Asunto(s)
Moléculas de Adhesión Celular/genética , Perfilación de la Expresión Génica , Neoplasias Ováricas/patología , Adulto , Moléculas de Adhesión Celular Neuronal/genética , Femenino , Proteínas Ligadas a GPI , Humanos , Persona de Mediana Edad , Análisis Multivariante , Estadificación de Neoplasias , Moléculas de Adhesión de Célula Nerviosa/genética , Oportunidad Relativa , Neoplasias Ováricas/genética , Ovario/metabolismo , ARN Neoplásico/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Supervivencia
4.
Development ; 132(1): 177-87, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15576400

RESUMEN

During vertebrate eye development, the cells of the optic vesicle (OV) become either neuroretinal progenitors expressing the transcription factor Chx10, or retinal pigment epithelium (RPE) progenitors expressing the transcription factor Mitf. Chx10 mutations lead to microphthalmia and impaired neuroretinal proliferation. Mitf mutants have a dorsal RPE-to-neuroretinal phenotypic transformation, indicating that Mitf is a determinant of RPE identity. We report here that Mitf is expressed ectopically in the Chx10(or-J/or-J) neuroretina (NR), demonstrating that Chx10 normally represses the neuroretinal expression of Mitf. The ectopic expression of Mitf in the Chx10(or-J/or-J) NR deflects it towards an RPE-like identity; this phenotype results not from a failure of neuroretinal specification, but from a partial loss of neuroretinal maintenance. Using Chx10 and Mitf transgenic and mutant mice, we have identified an antagonistic interaction between Chx10 and Mitf in regulating retinal cell identity. FGF (fibroblast growth factor) exposure in a developing OV has also been shown to repress Mitf expression. We demonstrate that the repression of Mitf by FGF is Chx10 dependent, indicating that FGF, Chx10 and Mitf are components of a pathway that determines and maintains the identity of the NR.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/fisiología , Neuronas/metabolismo , Retina/embriología , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Animales , Linaje de la Célula , Ojo/embriología , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor de Transcripción Asociado a Microftalmía , Microscopía Fluorescente , Modelos Biológicos , Mutación , Fenotipo , Plásmidos/metabolismo , Retina/citología , Retina/metabolismo , Factores de Tiempo , Transgenes
5.
Cancer Res ; 63(24): 8648-55, 2003 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-14695176

RESUMEN

Microcell-mediated transfer of normal chromosome 11 (chr 11) to a clonal derivative of the ovarian cancer cell line, OVCAR3, was performed and generated independent hybrids with a common set of phenotypes: inhibition of cell growth and of cellular migration in vitro; and inhibition of tumor growth in vivo. Differential display reverse transcriptase-PCR (RT-PCR), cDNA-representational difference analysis, and hybridization of cDNA high-density filter arrays identified altered mRNAs associated with these phenotypic alterations. Quantitative RT-PCR-based validation of each altered mRNA eliminated false positives to identify a reduced set of expression differences. Twelve products were confirmed as up-regulated and 4 as down-regulated upon introduction of chr 11. Strikingly, 4 of the 12 up-regulated genes were located on chr 11. Expression analysis of selected products by quantitative RT-PCR in a series of 18 human primary ovarian tumors revealed several associations with clinicopathological features. Importantly, low expression of two products, the lysosomal protease CTSD and the lens crystallin CRYAB, was significantly associated with adverse patient survival. Immunohistochemical analysis of CTSD in a larger independent panel of 58 primary ovarian tumors confirmed that low CTSD was associated with poor survival. Furthermore, low CTSD was significantly associated with serous histology and advanced tumor stage. The combined approach of microcell-mediated chromosome transfer and expression difference analysis has identified several altered mRNAs in a model of chr 11-mediated ovarian tumor suppression. The detailed contextual characterization of these genes will determine the extent of their involvement in neoplastic development.


Asunto(s)
Cromosomas Humanos Par 11/genética , Genes Supresores de Tumor , Neoplasias Ováricas/genética , División Celular/genética , Línea Celular Tumoral , Femenino , Expresión Génica , Técnicas de Transferencia de Gen , Humanos , Neoplasias Ováricas/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Nat Genet ; 34(3): 337-43, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12819783

RESUMEN

Epithelial ovarian cancer (EOC), the leading cause of death from gynecological malignancy, is a poorly understood disease. The typically advanced presentation of EOC with loco-regional dissemination in the peritoneal cavity and the rare incidence of visceral metastases are hallmarks of the disease. These features relate to the biology of the disease, which is a principal determinant of outcome. EOC arises as a result of genetic alterations sustained by the ovarian surface epithelium (OSE; ref. 3). The causes of these changes are unknown but are manifest by activation of oncogenes and inactivation of tumor-suppressor genes (TSGs). Our analysis of loss of heterozygosity at 11q25 identified OPCML (also called OBCAM), a member of the IgLON family of immunoglobulin (Ig) domain-containing glycosylphosphatidylinositol (GPI)-anchored cell adhesion molecules, as a candidate TSG in EOC. OPCML is frequently somatically inactivated in EOC by allele loss and by CpG island methylation. OPCML has functional characteristics consistent with TSG properties both in vitro and in vivo. A somatic missense mutation from an individual with EOC shows clear evidence of loss of function. These findings suggest that OPCML is an excellent candidate for the 11q25 ovarian cancer TSG. This is the first description to our knowledge of the involvement of the IgLON family in cancer.


Asunto(s)
Proteínas Portadoras/genética , Moléculas de Adhesión Celular/genética , Cromosomas Humanos Par 11/genética , Genes Supresores de Tumor/fisiología , Pérdida de Heterocigocidad , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Ováricas/genética , Animales , Azacitidina/farmacología , Neoplasias de la Mama/genética , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/metabolismo , Estudios de Casos y Controles , Moléculas de Adhesión Celular/antagonistas & inhibidores , Moléculas de Adhesión Celular/metabolismo , Islas de CpG , ADN/genética , ADN/metabolismo , Metilación de ADN , Inhibidores Enzimáticos/farmacología , Femenino , Proteínas Ligadas a GPI , Humanos , Ratones , Ratones Desnudos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación/genética , Proteínas del Tejido Nervioso/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Células Tumorales Cultivadas/trasplante
7.
Int J Oncol ; 21(5): 929-33, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12370737

RESUMEN

Epithelial ovarian cancer (EOC) is the most common cause of death from gynaecological malignancy. Resistance to platinum chemotherapy is a major reason for treatment failure and poor prognosis. The human homeobox gene BARX2 is located within a minimal region at 11q25 that is associated with frequent loss of heterozygosity (LOH) and adverse survival in EOC. BARX2 is a transcription factor known to regulate transcription of specific cell adhesion molecules in the mouse. We have previously shown that BARX2 expression is low in clear cell/endometrioid and high in serous adenocarcinomas of the ovary, histologic variants that are less and more sensitive, respectively to platinum chemotherapy. The aim of this study was to define whether BARX2 could modulate sensitivity to cisplatin in human epithelial ovarian cancer. In two cell line series sequentially derived from ovarian cancer patients pre- and post-cisplatin chemotherapy, BARX2 expression was downregulated in the cell lines established upon tumor recurrence after platinum therapy. Transfection of BARX2 into a platinum resistant cell line significantly reversed cisplatin resistance compared with its isogenic platinum sensitive parent, in both growth inhibition and clonogenic assays. Taken together, our data demonstrate that the homeobox gene BARX2 may be a biological factor involved in determining sensitivity or resistance to the cytotoxic effects of cisplatin.


Asunto(s)
Cisplatino/farmacología , Genes Homeobox/fisiología , Proteínas de Homeodominio/genética , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Northern Blotting , Resistencia a Antineoplásicos , Femenino , Humanos , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/patología , Transfección , Células Tumorales Cultivadas
8.
Clin Cancer Res ; 8(4): 1101-7, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11948120

RESUMEN

PURPOSE: Matriptase is a type II transmembrane serine protease expressed by cells of surface epithelial origin, including epithelial ovarian tumor cells. Matriptase cleaves and activates proteins implicated in the progression of ovarian cancer and represents a potential prognostic and therapeutic target. The aim of this study was to examine the expression of matriptase, and its inhibitor, hepatocyte growth factor activator inhibitor-1 (HAI-1), in epithelial ovarian cancer and to assign clinicopathological correlations. EXPERIMENTAL DESIGN: We have determined by immunohistochemistry the expression of matriptase and HAI-1 in 54 epithelial ovarian cancers. Statistical analyses of immunohistochemistry expression data with clinical outcome and clinicopathological parameters were then performed. RESULTS: Of 54 tumors tested, 39 (72%) and 11 (20%) were positive for matriptase and for HAI-1, respectively. All HAI-1-positive tumors were also matriptase positive. Analysis of clinicopathological parameters demonstrated a loss of matriptase associated with stage III/IV tumors as compared with stage I/II tumors (P = 0.030). There was also a loss of HAI-1 expression associated with stage III/IV tumors (P = 0.039). Of 34 stage I/II tumors, 28 (82%) stained positive for matriptase, and 10 (29%) stained positive for HAI-1; 10 (29%) tumors showed coexpression. Of 20 stage III/IV tumors, however, 11 stained positive for matriptase (55%), only 1 of which coexpressed HAI-1 (P = 0.039). CONCLUSIONS: Advanced-stage ovarian tumors that express matriptase are more likely to do so in the absence of its inhibitor, HAI-1, indicating that an imbalance in the matriptase:HAI-1 ratio could be important in the development of advanced disease. Such an imbalance could promote the proteolytic activity of matriptase and, consequently, a more invasive phenotype.


Asunto(s)
Glicoproteínas de Membrana/biosíntesis , Neoplasias Ováricas/patología , Serina Endopeptidasas/biosíntesis , Tripsina/biosíntesis , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Hibridación in Situ , Glicoproteínas de Membrana/genética , Estadificación de Neoplasias , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Modelos de Riesgos Proporcionales , Proteínas Inhibidoras de Proteinasas Secretoras , ARN Mensajero/genética , ARN Mensajero/metabolismo , Serina Endopeptidasas/genética , Análisis de Supervivencia , Tripsina/genética , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA