Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 286(26): 23467-75, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21518759

RESUMO

Reversible modulation of integrin-regulated cell-matrix adhesion and epithelial (E)-cadherin-mediated cell-cell adhesion plays a critical role in the establishment of ovarian cancer metastases. In contrast to most epithelial cell-derived tumors that down-regulate E-cadherin expression during progression, acquisition of E-cadherin expression accompanies malignant transformation of the ovarian surface epithelium and is maintained in peritoneal metastases. Metastatic epithelial ovarian cancer cells are disseminated intraperitoneally and preferentially adhere via integrins to interstitial collagens in the peritoneal cavity. This study was undertaken to determine whether integrin engagement influences E-cadherin and ß-catenin localization and function. The data demonstrate that multivalent integrin engagement results in increased internalization of E-cadherin, inhibition of GSK-3ß, elevated levels of nuclear ß-catenin, increased ß-catenin-regulated promoter activation, and transcriptional activation of Wnt/ß-catenin target genes. Blocking ß-catenin transcriptional control with inhibitor of ß-catenin and Tcf-4 reduces cellular invasion, suggesting a key role for ß-catenin nuclear signaling in EOC invasion and metastasis. These studies support a model wherein cell-matrix engagement regulates the functional integrity of cell-cell contacts, leading to increased ß-catenin nuclear signaling and enhanced cellular invasive activity. Furthermore, these results provide a mechanism for activation of Wnt/ß-catenin signaling in the absence of activating mutations in this pathway.


Assuntos
Regulação Neoplásica da Expressão Gênica , Integrinas/metabolismo , Modelos Biológicos , Proteínas de Neoplasias/metabolismo , Neoplasias Ovarianas/metabolismo , Transdução de Sinais , beta Catenina/metabolismo , Caderinas/metabolismo , Comunicação Celular , Linhagem Celular Tumoral , Feminino , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias Ovarianas/patologia , Transcrição Gênica
2.
Cancer Res ; 67(5): 2030-9, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17332331

RESUMO

Reversible modulation of cell-cell adhesion, cell-matrix adhesion, and proteolytic activity plays a critical role in remodeling of the neoplastic ovarian epithelium during metastasis, implicating cadherins, integrins, and proteinases in i.p. metastatic dissemination of epithelial ovarian carcinoma (EOC). Aberrant epithelial differentiation is an early event in ovarian carcinogenesis; thus, in contrast to most carcinomas that lose E-cadherin expression with progression, E-cadherin is abundant in primary EOC. Metastasizing EOCs engage in integrin-mediated adhesion to submesothelial interstitial collagens and express matrix metalloproteinases (MMP) that facilitate collagen invasion, thereby anchoring secondary lesions in the submesothelial matrix. As metalloproteinases have also been implicated in E-cadherin ectodomain shedding, the current study was undertaken to model the effects of matrix-induced integrin clustering on proteinase-catalyzed E-cadherin ectodomain shedding. Aggregation of collagen-binding integrins induced shedding of an 80-kDa E-cadherin ectodomain [soluble E-cadherin (sE-cad)] in a MMP- and Src kinase-dependent manner, and sE-cad was prevalent in ascites from ovarian cancer patients. Expression of MMP-9 was elevated by integrin aggregation, integrin-mediated ectodomain shedding was inhibited by a MMP-9 function blocking antibody, and incubation of cells with exogenous MMP-9 catalyzed E-cadherin ectodomain shedding. In contrast to other tumors wherein sE-cad is released into the circulation, EOC tumors maintain direct contact with sE-cad-rich ascites at high concentration, and incubation of EOC cells with physiologically relevant concentrations of recombinant sE-cad disrupted adherens junctions. These data support a novel mechanism for posttranslational modification of E-cadherin function via MMP-9 induction initiated by cell-matrix contact and suggest a mechanism for promotion of EOC metastatic dissemination.


Assuntos
Caderinas/metabolismo , Carcinoma/patologia , Colágeno/metabolismo , Integrinas/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Neoplasias Ovarianas/patologia , Junções Aderentes/metabolismo , Junções Aderentes/fisiologia , Carcinoma/metabolismo , Junções Célula-Matriz/fisiologia , Feminino , Humanos , Invasividade Neoplásica , Neoplasias Ovarianas/metabolismo , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Células Tumorais Cultivadas
3.
Gynecol Oncol ; 111(1): 125-31, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18678400

RESUMO

OBJECTIVE: Serous borderline ovarian tumors (SBOT) are slow growing, noninvasive ovarian epithelial neoplasms, which tend to recur as low-grade invasive carcinomas (LGC) with a much worse prognosis. We investigated the molecular basis of this progression. METHODS: We established cultures of three SBOTs and one LGC from tumor biopsies, and inactivated p53, Rb and PP2A in the cells with SV40 large T (LT) and small T (ST) antigen. They were examined for cadherins by immunofluorescence and immunoblotting, invasiveness in Boyden chambers, motility by scratch-wound healing assay, anchorage independence by growth in agarose, and protease activity by gelatin zymography, immunoassay and colorimetry. Cells were overexpressed with N-cadherin using an adenovirus. RESULTS: Inactivation of p53, Rb and PP2A by SV40 LT/ST antigen resulted in greatly enhanced growth potential, invasiveness, motility and anchorage independence, and in epithelio-mesenchymal transition, as indicated by morphology and substitution of N-cadherin for E-cadherin. Overexpressed N-cadherin did not induce invasiveness of SBOT cells and there was no consistent change in protease activities, suggesting that these were not primary effectors of the enhanced neoplastic characteristics. Low passage LGC cells were more invasive than SBOT cells, but this difference disappeared with the introduction of LT/ST into the two cell types. CONCLUSION: Downregulation or inactivation of p53, Rb and/or PP2A plays a role in the progression from SBOT to invasive ovarian carcinomas.


Assuntos
Antígenos Transformantes de Poliomavirus/genética , Transformação Celular Neoplásica/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Adenoviridae/genética , Adulto , Caderinas/genética , Movimento Celular/genética , Transformação Celular Neoplásica/patologia , Progressão da Doença , Regulação para Baixo , Células Epiteliais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Genes do Retinoblastoma , Genes p53 , Vetores Genéticos/genética , Humanos , Mesoderma/patologia , Pessoa de Meia-Idade , Peptídeo Hidrolases/metabolismo , Proteína Fosfatase 2/genética , Transfecção , Células Tumorais Cultivadas
4.
Cancer Res ; 65(6): 2234-42, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15781636

RESUMO

Elevated levels of the bioactive lipid lysophosphatidic acid (LPA) are detectable in the majority of patients with both early- and late-stage ovarian cancer, suggesting that LPA promotes early events in ovarian carcinoma dissemination. LPA contributes to the development, progression, and metastasis of ovarian cancer in part by inducing the expression of genes that contribute to proliferation, survival, or invasion, including cyclooxgenase-2 (COX-2) and matrix metalloproteinase-2 (MMP-2). We have previously shown that LPA promotes proMMP-2 activation and MMP-2-dependent migration and invasion in ovarian cancer cells. The purpose of the current study was to determine whether the effect of LPA on acquisition of the metastatic phenotype in ovarian cancer cells is mediated via a COX-2-dependent mechanism. Immunohistochemical analysis of 173 ovarian tumors showed strong COX-2 immunoreactivity in 63% of tumor specimens, including 50% of borderline tumors. LPA increased COX-2 protein expression in a time- and concentration-dependent manner in two of three immortalized borderline ovarian epithelial cells as well as in four of six ovarian cancer cell lines. This was accomplished by both activation of the Edg/LPA receptor and LPA-mediated transactivation of the epidermal growth factor receptor, which increased COX-2 expression via the Ras/mitogen-activated protein kinase pathway. COX-2 also played a role in LPA-induced invasion and migration, as treatment with the COX-2 specific inhibitor NS-398 reduced LPA-induced proMMP-2 protein expression and activation and blocked MMP-dependent motility and invasive activity. These data show that COX-2 functions as a downstream mediator of LPA to potentiate aggressive cellular behavior.


Assuntos
Lisofosfolipídeos/farmacologia , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/patologia , Prostaglandina-Endoperóxido Sintases/fisiologia , Movimento Celular/genética , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/farmacologia , Indução Enzimática/efeitos dos fármacos , Precursores Enzimáticos/metabolismo , Receptores ErbB/biossíntese , Receptores ErbB/genética , Feminino , Gelatinases/metabolismo , Humanos , Imuno-Histoquímica , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas de Membrana , Metaloendopeptidases/metabolismo , Prostaglandina-Endoperóxido Sintases/biossíntese , Receptores de Lisoesfingolipídeo/fisiologia , Ativação Transcricional/efeitos dos fármacos
5.
Mol Endocrinol ; 16(1): 45-57, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11773438

RESUMO

PRL is essential for normal lobulo-alveolar growth of the mammary gland and may contribute to mammary cancer development or progression. However, analysis of the mechanism of action of PRL in these processes is complicated by the production of PRL within mammary epithelia. To examine PRL actions in a mammary cell-specific context, we selected MCF-7 cells that lacked endogenous PRL synthesis, using PRL stimulation of interferon-gamma-activated sequence-related PRL response elements. Derived clones exhibited a greater proliferative response to PRL than control cells. To understand the mechanism, we examined, by Western analysis, levels of proteins essential for cell cycle progression as well as phosphorylation of retinoblastoma protein. The expression of cyclin D1, a critical regulator of the G1/S transition, was significantly increased by PRL and was associated with hyperphosphorylation of retinoblastoma protein at Ser(780). Cyclin B1 was also increased by PRL. In contrast, PRL decreased the Cip/Kip family inhibitor, p21, but not p16 or p27. These studies demonstrate that PRL can stimulate the cell cycle in mammary epithelia and identify specific targets in this process. This model system will enable further molecular dissection of the pathways involved in PRL-induced proliferation, increasing our understanding of this hormone and its interactions with other factors in normal and pathogenic processes.


Assuntos
Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteínas de Ciclo Celular/metabolismo , Prolactina/farmacologia , Sequência de Bases , Proteínas de Ciclo Celular/genética , Ciclina B/efeitos dos fármacos , Ciclina B/metabolismo , Ciclina B1 , Ciclina D1/efeitos dos fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/efeitos dos fármacos , Ciclinas/metabolismo , Inibidores Enzimáticos/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Humanos , Interferon gama/genética , Interferon gama/metabolismo , Sistema de Sinalização das MAP Quinases , Dados de Sequência Molecular , Oligonucleotídeos Antissenso/farmacologia , Fosforilação , Prolactina/metabolismo , Proteína do Retinoblastoma/efeitos dos fármacos , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais
6.
J Oncol ; 2012: 501492, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22593767

RESUMO

Ovarian cancer metastasizes via exfoliation of free-floating cells and multicellular aggregates from the primary tumor to the peritoneal cavity. A key event in EOC metastasis is disruption of cell-cell contacts via modulation of intercellular junctional components including cadherins. Ascites is rich in lysophosphatidic acid (LPA), a bioactive lipid that may promote early events in ovarian cancer dissemination. The objective of this paper was to assess the effect of LPA on E-cadherin junctional integrity. We report a loss of junctional E-cadherin in OVCAR3, OVCA429, and OVCA433 cells exposed to LPA. LPA-induced loss of E-cadherin was concentration and time dependent. LPA increased MMP-9 expression and promoted MMP-9-catalyzed E-cadherin ectodomain shedding. Blocking LPA receptor signaling inhibited MMP-9 expression and restored junctional E-cadherin staining. LPA-treated cells demonstrated a significant decrease in epithelial cohesion. Together these data support a model wherein LPA induces MMP-9 expression and MMP-9-catalyzed E-cadherin ectodomain shedding, resulting in loss of E-cadherin junctional integrity and epithelial cohesion, facilitating metastatic dissemination of ovarian cancer cells.

7.
Cancer Res ; 68(12): 4606-13, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18559505

RESUMO

Epidermal growth factor (EGF) receptor (EGFR) is frequently elevated in epithelial ovarian cancer, and E-cadherin expression is often reduced in advanced disease. In this study, we investigated a mechanism by which EGFR activation promotes disruption of adherens junctions through induction of matrix metalloproteinase 9 (MMP-9). We show that EGFR activation down-modulates E-cadherin, and broad spectrum MMP inhibition ameliorates EGF-stimulated junctional disruption and loss of E-cadherin protein. MMP-9 involvement in EGF-dependent down-regulation of E-cadherin was determined by siRNA specifically directed against MMP-9. Furthermore, treatment with recombinant MMP-9 or transient expression of MMP-9 is sufficient to reduce E-cadherin levels in differentiated ovarian tumor cells. Stable overexpression of MMP-9 led to a loss of E-cadherin and junctional integrity, and promoted a migratory and invasive phenotype. Thus, elevated MMP-9 protein expression is sufficient for junctional disruption and loss of E-cadherin in these cells. The associations between EGFR activation, MMP-9 expression, and E-cadherin were investigated in human ovarian tumors and paired peritoneal metastases wherein immunohistochemical staining for activated (phospho) EGFR and MMP-9 colocalized with regions of reduced E-cadherin. These data suggest that regulation of MMP-9 by EGFR may represent a novel mechanism for down-modulation of E-cadherin in ovarian cancer.


Assuntos
Caderinas/metabolismo , Receptores ErbB/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Junções Aderentes/fisiologia , Líquido Ascítico/enzimologia , Western Blotting , Caderinas/antagonistas & inibidores , Caderinas/genética , Linhagem Celular Tumoral , Movimento Celular , Fator de Crescimento Epidérmico/farmacologia , Feminino , Citometria de Fluxo , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Metaloproteinase 9 da Matriz/genética , Microscopia de Fluorescência , Invasividade Neoplásica , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Proteínas Recombinantes/metabolismo , Análise Serial de Tecidos , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA