Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
1.
Oncogene ; 30(37): 3930-42, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21478913

RESUMO

The mechanisms underlying the progression of noninvasive serous borderline ovarian tumors (SBOT) to low-grade invasive carcinomas are poorly understood. We recently showed that inhibition of p53 induces SBOT invasion by activating the PI3K/Akt pathway and transcriptionally repressing E-cadherin. In human cancers, aberrant DNA methylation is a common phenomenon, and it is thought to be involved in the progression from noninvasive to invasive ovarian carcinomas. In this study, we tested the hypothesis that inhibition of p53 downregulates E-cadherin by regulating the methylation of its promoter in SBOT cells. Here, we show that DNA methyltransferase-1 (DNMT1), but not DNMT3a or DNMT3b, was increased in SV40 LT-infected SBOT4 cells, SBOT4-LT and the low-grade invasive serous ovarian carcinoma-derived cell line MPSC1. Treatment with 5-Aza-dC, a DNMT1 inhibitor, restored E-cadherin promoter methylation and expression, and inhibited cell invasion in both invasive SBOT4-LT and MPSC1 cells. Moreover, knockdown of endogenous p53 using siRNA in SBOT3.1 cells induced DNMT1 expression and led to an increase in E-cadherin promoter methylation. Additionally, activation of the PI3K/Akt pathway is required for p53 inhibition-induced DNMT1 expression. The increase in DNMT1 was associated with the inhibition of p53-induced downregulation of E-cadherin and cell invasion. Our findings show an important role for p53 in the progression of SBOT to an invasive carcinoma, and suggest that downregulation of E-cadherin by DNMT1-mediated promoter methylation contributes to this process.


Assuntos
Caderinas/genética , Carcinoma/patologia , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Neoplasias Ovarianas/patologia , Proteína Supressora de Tumor p53/metabolismo , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Caderinas/antagonistas & inibidores , Carcinoma/metabolismo , Linhagem Celular Tumoral , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Decitabina , Feminino , Técnicas de Silenciamento de Genes , Humanos , Invasividade Neoplásica , Neoplasias Ovarianas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína Supressora de Tumor p53/antagonistas & inibidores , DNA Metiltransferase 3B
2.
Oncogene ; 30(9): 1020-31, 2011 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-20972462

RESUMO

Serous borderline ovarian tumors (SBOTs) are slow-growing, non-invasive ovarian epithelial neoplasms. SBOTs are considered to be distinct entities that give rise to invasive low-grade serous carcinomas (LGCs), which have a relatively poor prognosis and are unrelated to high-grade serous carcinomas (HGCs). The mechanisms underlying the progression of non-invasive SBOTs to invasive epithelial ovarian carcinomas are not understood. We recently established short-term cultures of SBOT cells from tumor biopsies and showed that inactivation of p53, retinoblastoma (Rb) and/or PP2A by the simian virus 40 (SV40) large (LT) and small T antigens extends the life span of the cells and endows them with the ability to invade Matrigel-coated transwells. In this study, we show that concurrent inhibition of p53 and Rb by the SV40 LT produces cells (referred to as SBOT4-LT) with increased life span and cell invasion. To distinguish the roles of p53 and Rb in the progression from SBOTs to invasive ovarian carcinomas, we performed small interfering RNA-mediated knockdown of endogenous p53 in a spontaneously immortalized SBOT cell line, SBOT3.1, which increased cell invasion. This increased invasive activity was associated with the transcriptional downregulation of E-cadherin, correlated with an increase in PIK3CA levels and the increased activation of Akt. Conversely, in invasive LGC-derived MPSC1 cells, enhancing the levels of p53 decreased cell invasion and diminished the phosphatidylinositol 3-kinase (PI3K)/Akt-mediated downregulation of E-cadherin. Inhibition of Rb also enhanced invasiveness, but did not affect the levels of PIK3CA and E-cadherin in SBOT3.1 cells, suggesting that it functions by a different pathway. To our knowledge, this study is the first to show that p53 has an important role in the progression from SBOTs to invasive carcinomas. In addition, our findings suggest that downregulation of E-cadherin by the PI3K/Akt pathway contributes to this progression.


Assuntos
Caderinas/antagonistas & inibidores , Cistadenocarcinoma Seroso/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína Supressora de Tumor p53/antagonistas & inibidores , Antígenos Virais de Tumores/metabolismo , Western Blotting , Carcinoma Epitelial do Ovário , Sobrevivência Celular/efeitos dos fármacos , Classe I de Fosfatidilinositol 3-Quinases , Cistadenocarcinoma Seroso/genética , Feminino , Imunofluorescência , Técnicas de Silenciamento de Genes , Humanos , Hibridização In Situ , Invasividade Neoplásica , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , RNA Interferente Pequeno/genética , Proteína do Retinoblastoma/antagonistas & inibidores , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vírus 40 dos Símios/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
3.
Int J Gynecol Cancer ; 18(6): 1234-47, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18217967

RESUMO

Serous borderline ovarian tumors (SBOTs) are differentiated, slow growing, noninvasive, and have a better prognosis than their invasive counterparts, but recurrence and progression to invasive carcinomas are common, and unlike high-grade serous carcinomas, they tend to be nonresponsive to chemotherapy. However, due to a lack of culture systems and animal models, information about the properties of SBOT and their changes with neoplastic progression is extremely limited. Our objective was to establish a cell culture model for SBOTs and to characterize their phenotype and genotype. We compared cultures derived from two SBOTs, one of which was a short-term culture containing a BRAF mutation but few other cytogenetic changes while the other culture developed into a spontaneously immortalized permanent cell line and had numerical and structural chromosomal abnormalities but lacked RAS/BRAF mutations. Both cultures formed whorl-like epithelial colonies and resembled low-grade invasive carcinomas by their secretion of CA125 and oviduct-specific glycoprotein, production of matrix metalloproteinases, E-cadherin expression, and telomerase activity. Other characteristics associated with neoplastic transformation, including invasiveness, anchorage-independent growth, and tumorigenicity, were not observed. Importantly, cell motility was reduced in both lines, likely contributing to the lack of invasiveness. The results reveal a striking phenotypic similarity between the two cell lines, regardless of their cytogenetic diversity, which suggests that their characteristic phenotype is regulated to a large degree by epigenetic and environmental factors. In conclusion, we have established the first permanent SBOT cell line, which provides a new model to elucidate the undefined relationship of SBOTs to invasive ovarian carcinomas.


Assuntos
Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Adulto , Animais , Sequência de Bases , Biomarcadores , Diferenciação Celular , Movimento Celular , Feminino , Dosagem de Genes/genética , Genótipo , Saúde , Humanos , Camundongos , Mutação/genética , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Fatores de Tempo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Oncogene ; 26(34): 4961-8, 2007 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-17310993

RESUMO

Polypyrimidine tract-binding protein (PTB) is an RNA-binding protein with multiple functions in the regulation of RNA processing and IRES-mediated translation. We report here overexpression of PTB in a majority of epithelial ovarian tumors revealed by immunoblotting and tissue microarray (TMA) staining. By western blotting, we found that PTB was overexpressed in 17 out of 19 ovarian tumor specimens compared to their matched-normal tissues. By TMA staining, we found PTB expression in 38 out of 44 ovarian cancer cases but only in two out of nine normal adjacent tissues. PTB is also overexpressed in SV40 large T-antigen immortalized ovarian epithelial cells compared to normal human ovarian epithelial cells. Using doxycycline-inducible small interfering RNA technology, we found that knockdown of PTB expression in the ovarian tumor cell line A2780 substantially impaired tumor cell proliferation, anchorage-independent growth and in vitro invasiveness. These results suggest that overexpression of PTB is an important component of the multistep process of tumorigenesis, and might be required for the development and maintenance of epithelial ovarian tumors. Moreover, because of its novel role in tumor cell growth and invasiveness, shown here for the first time, PTB may be a novel therapeutic target in the treatment of ovarian cancer.


Assuntos
Neoplasias Ovarianas/patologia , Proteína de Ligação a Regiões Ricas em Polipirimidinas/antagonistas & inibidores , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Imuno-Histoquímica , Invasividade Neoplásica , Neoplasias Ovarianas/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Interferência de RNA , Análise Serial de Tecidos
5.
Minerva Ginecol ; 55(4): 297-314, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-14581855

RESUMO

The ovarian surface epithelium (OSE) is thought to be the origin of most ovarian cancers in humans. OSE is a single cell layer of squamous or cuboidal cells on the surface of ovaries. In contrast to its relatively simple structure, it gives rise to a highly complex group of neoplasms, the epithelial ovarian carcinomas. Thus, in this review, we summarize the development and the main characteristics of normal adult OSE with emphasis on those properties that might contribute to its propensity to undergo neoplastic progression. Secondly, a brief comparison of the biology and endocrinology of normal and neoplastic OSE is being described to understand the mechanisms in the transformation of normal OSE to its neoplastic counterparts. Lastly, recent advances in experimental approaches including in vitro and in vivo models that have contributed to our understanding of the nature of epithelial ovarian cancers are being briefly explored.


Assuntos
Neoplasias Ovarianas/etiologia , Ovário/citologia , Animais , Diferenciação Celular , Transformação Celular Neoplásica , Células Cultivadas , Células Epiteliais , Feminino , Substâncias de Crescimento/fisiologia , Hormônios/fisiologia , Humanos , Ovário/embriologia , Ovário/fisiologia
6.
Int J Gynecol Cancer ; 12(6): 691-703, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12445245

RESUMO

The etiology and early events in the progression of epithelial ovarian carcinomas are among the least understood of all major human malignancies. There are no adequate means for early detection of these neoplasms and, as a result, they are usually diagnosed in late stages. The purpose of this review is to point out some of the peculiar problems and limitations that have hampered progress in ovarian carcinogenesis research and to summarize new approaches and recent advances in our understanding of this process. The review first presents an overview of the properties of the ovarian surface epithelium (OSE) which is thought to be the source of epithelial ovarian carcinomas, followed by a discussion of recent research based on human OSE. This includes sections on methodology for the attainment and study of OSE, investigations of OSE from women with predisposing mutations, and attempts to convert normal OSE to malignancy. This overview is followed by a discussion of the contributions, potential, and limitations of animal models. The knowledge gained by these approaches will likely lead to improvements in our ability to prevent, diagnose, and treat ovarian cancer.


Assuntos
Carcinoma/patologia , Neoplasias Ovarianas/patologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Projetos de Pesquisa
7.
J Clin Endocrinol Metab ; 86(10): 5075-8, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11600588

RESUMO

Epithelial ovarian carcinomas are the most common cause of death from gynecological malignancies and appear to arise from ovarian surface epithelium (OSE), but the exact mechanism of ovarian tumorigenesis has not been elucidated. Recent cloning of a second form of gonadotropin-releasing hormone (GnRH-II) has been reported in various human tissues including the ovary. However, the expression and role of GnRH-II in human OSE and ovarian carcinomas is not known. In the present study, we demonstrated that in addition to the GnRH receptor (GnRH-R), GnRH-II mRNA is expressed in normal OSE, immortalized OSE (IOSE) cells, primary cultures of ovarian tumors and ovarian cancer cell lines. Treatments with increasing doses (10(-9)-10(-7) M) of GnRH-I and -II resulted in a growth-inhibition in both non-tumorigenic IOSE-29 and tumorigenic IOSE-29EC cells. These results indicate for the first time the expression and potential anti-proliferative effect of GnRH-II, suggesting that GnRH-II, similar to GnRH-I, may have a growth-regulatory effect in normal and neoplastic OSE cells.


Assuntos
Hormônio Liberador de Gonadotropina/genética , Inibidores do Crescimento/farmacologia , Neoplasias Ovarianas/metabolismo , Ovário/metabolismo , RNA Mensageiro/análise , Divisão Celular/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/farmacologia , Humanos , Receptores LHRH/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
In Vitro Cell Dev Biol Anim ; 37(8): 515-21, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11669285

RESUMO

The epithelial ovarian carcinomas arise in the ovarian surface epithelium (OSE) which is the mesothelial covering of the ovary. Studies of human USE have been hampered by the small amounts and limited lifespan of this epithelium in culture. OSE cells expressing SV40 large T antigen (Tag) or the HPV genes E6 and E7 have increased growth potentials but lack some of the normal characteristics of OSE. In this study, we used conditional SV40 Tag expression to produce OSE cells with increased proliferative potentials but relatively normal phenotypes. Primary OSE cultures from three women, one of whom had a BRCA1 mutation, were infected with a temperature-sensitive Tag construct (tsTag), and from these, 28 monoclonal and four polyclonal lines were isolated. The effects of temperature changes were examined in two monoclonal and two polyclonal lines. At the permissive temperature (34 degrees C), these cell lines underwent 52-71 population doublings (PD) compared to 15-20 PD for normal OSE. Nuclear SV40-Tag and p53 expression, demonstrated by immunofluorescence, showed that tsTag was uniformly present and biologically active in all lines. At 34 degrees C, culture morphologies ranged from epithelial to mesenchymal. The mean percentage of cells expressing the epithelial differentiation marker, keratin. varied between lines from 20 to 97%. Collagen type III, a mesenchymal marker expressed by OSE in response to explantation into culture, was present in 24-43% of cells. At 39 degrees C, tsTag was inactivated by 2 d while nuclear p53 staining diminished to control levels over 2 wk. Over 3 d. the cells assumed more epithelial morphologies, keratin expression reached 85-100% in all lines and collagen expression increased significantly in two lines. The cultures with the BRCA1 mutation expressed the most keratin and the least collage n III at both temperatures. As indicated by beta-galactosidase staining at pH 6.0, changes leading to senescence were initiated at 39 degrees C by 6 h and were present in all cells after 24 h. However, the cells underwent 1-3 population doublings over up to 1 wk before growth arrest and widespread cell death, thus providing an experimental system where large numbers of OSE cells with different genetic backgrounds and growth potentials can be studied without the concurrent influence of Tag.


Assuntos
Antígenos Transformantes de Poliomavirus/genética , Diferenciação Celular , Divisão Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Expressão Gênica , Ovário/citologia , Colágeno Tipo III/análise , Células Epiteliais/química , Feminino , Imunofluorescência , Genes BRCA1 , Humanos , Queratinas/análise , Mutação , Temperatura , Proteína Supressora de Tumor p53/análise
9.
Gynecol Oncol ; 82(2): 305-11, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11531284

RESUMO

OBJECTIVE: The aim of this study was to study the pattern of protein kinase expression in a culture model of epithelial ovarian carcinogenesis. METHODS: Cultures of normal human ovarian surface epithelium (OSE), OSE from women with BRCA1 mutations, a cell culture model of preneoplastic (SV40 T-antigen-immortalized, nontumorigenic) and neoplastic (SV40-E-cadherin transfected, tumorigenic) OSE, and three ovarian cancer cell lines were used to represent OSE phenotypes of different genetic backgrounds and at different, progressive stages of neoplastic transformation. The protein kinase network signaling was studied by Western blotting, simultaneously using multiple antibodies for specific protein kinases. RESULTS: High levels of cGMP-dependent protein kinase were found in normal and preneoplastic OSE, but were absent in neoplastic OSE. In contrast, expression of MEK6 was detected exclusively in neoplastic OSE. The expressions of casein kinase II (CK2), p38 mitogen-activated protein kinase (MAPK), cyclin-dependent kinase, and the phosphatidylinositol 3-kinase (PI3K) effectors Akt2 and p70 S6 kinase (S6K) were several-fold higher in neoplastic OSE than in normal OSE, whereas the expressions of the MAPKs extracellular signal-regulated kinases ERK1 and -2 were unchanged. Importantly, constitutive phosphorylation of Akt2 and p70 S6K, as found in neoplastic OSE, was also observed in overtly normal OSE from women with predisposing BRCA1 gene mutations. CONCLUSIONS: These data demonstrate that different repertoires of downstream signaling proteins, particularly those of the MEK6-p38 MAPK-CK2 pathway and the PI3K pathway, are correlated with phenotypic manifestations of a cell culture model of OSE at progressive stages in the development of ovarian cancer. Changes in PI3K effectors are already found in overtly normal OSE from women with BRCA1 mutations.


Assuntos
Transformação Celular Neoplásica/metabolismo , Neoplasias Ovarianas/enzimologia , Lesões Pré-Cancerosas/enzimologia , Proteínas Quinases/biossíntese , Animais , Antígenos Transformantes de Poliomavirus/genética , Antígenos Transformantes de Poliomavirus/fisiologia , Western Blotting , Caderinas/genética , Progressão da Doença , Eletroforese em Gel de Poliacrilamida , Epitélio/enzimologia , Epitélio/patologia , Feminino , Genes BRCA1/genética , Fator de Crescimento de Hepatócito/farmacologia , Humanos , Camundongos , Neoplasias Ovarianas/patologia , Ovário/enzimologia , Ovário/patologia , Fosforilação , Lesões Pré-Cancerosas/patologia , Proteínas Quinases/metabolismo , Transdução de Sinais , Propriedades de Superfície , Transfecção , Células Tumorais Cultivadas
10.
J Clin Endocrinol Metab ; 86(5): 2125-35, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11344217

RESUMO

Most ovarian neoplasms arise from the ovarian surface epithelium (OSE), and multiple growth factors have been implicated to influence the transformation from OSE. The present study was performed to investigate the role of activin and transforming growth factor-beta (TGFbeta) in normal and neoplastic OSE cells. An immortalized OSE cell line (IOSE-29) was generated from normal OSE by transfecting simian virus 40 large T antigen and was rendered tumorigenic after subsequent transfection with the E-cadherin gene (IOSE-29EC). The activin/inhibin subunits and activin receptors were expressed at both messenger ribonucleic acids and protein levels in these cells, suggesting that activin may have an autocrine role in neoplastic OSE cells. Treatments with activin (1-100 ng/mL) resulted in a significant decrease in cell proliferation in both IOSE-29 and IOSE-29EC cells, although we have shown that it stimulated the growth of ovarian cancer cells and had no effect on normal OSE. This inhibitory effect was attenuated with cotreatment with follistatin. Treatment with TGFbeta (0.1-10 ng/mL) also significantly decreased the proliferation of normal, IOSE-29, and IOSE-29EC cells in a dose-dependent manner. In addition, treatments with both activin and TGFbeta resulted in an increase in DNA fragmentation in IOSE-29EC cells in a dose-dependent manner. This apoptotic effect of activin was attenuated by cotreatment with follistatin. Treatment with TGFbeta (1 and 10 ng/mL) resulted in a significant decrease in Bcl-2 protein (up to 50%) in IOSE-29EC, whereas no difference was observed in Bax protein levels. Therefore, down-regulated Bcl-2 by TGFbeta may eventually induce apoptosis in IOSE-29EC cells. In contrast, no difference was observed in Bax and Bcl-2 protein expression after treatment with activin. In conclusion, the present study indicates that activin and TGFbeta inhibited growth and induced apoptosis in early neoplastic (IOSE-29) and tumorigenic OSE (IOSE-29EC) cells. Furthermore, antiapoptotic Bcl-2 protein was down-regulated by TGFbeta, whereas no difference was produced in Bax protein by activin or TGFbeta treatment or in Bcl-2 protein by activin. These results suggest that activin and TGFbeta may play a role in growth inhibition and induction of apoptosis in early neoplastic and tumorigenic stage of ovarian cancer.


Assuntos
Apoptose/efeitos dos fármacos , Inibinas/farmacologia , Neoplasias Ovarianas/patologia , Fator de Crescimento Transformador beta/farmacologia , Receptores de Ativinas , Ativinas , Linhagem Celular , Feminino , Humanos , Inibinas/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , RNA Mensageiro/análise , Receptores de Fatores de Crescimento/análise , Receptores de Fatores de Crescimento/genética , Fator de Crescimento Transformador beta/genética , Proteína X Associada a bcl-2
11.
J Natl Cancer Inst ; 93(10): 762-8, 2001 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-11353786

RESUMO

BACKGROUND: Lysophosphatidic acid (LPA) stimulates ovarian tumor growth at concentrations present in ascitic fluid. Vascular endothelial growth factor (VEGF) stimulates angiogenesis and plays a pivotal role in the formation of ovarian cancer-associated ascites. We examined whether LPA promotes ovarian tumor growth by increasing angiogenesis via VEGF. METHODS: VEGF expression was examined in a simian virus 40 T-antigen-immortalized ovarian surface epithelial cell line (IOSE-29) and in ovarian cancer cell lines (OVCAR-3, SKOV-3, and CAOV-3) treated with LPA. VEGF promoter activity was measured in OVCAR-3 cells after transfection or cotransfection with c-Fos and c-Jun, components of AP1 transcription factor, potential binding sites for which are present in the VEGF promoter. The expression of the LPA receptors Edg2 and Edg4 was also assessed. All statistical tests were two-sided. RESULTS: LPA treatment increased steady-state VEGF messenger RNA (mRNA) levels in OVCAR-3 cells in a time- and dose-dependent fashion and stimulated VEGF promoter activity without prolonging mRNA half-life in these cells, but LPA had little effect on IOSE-29 cells. Forced overexpression of c-Jun and c-Fos in OVCAR-3 cells stimulated VEGF promoter activity fourfold. LPA also elevated VEGF protein levels by 1.5-fold in SKOV-3 cells (P =.0148), 1.9-fold in CAOV-3 cells (P<.001), and threefold in OVCAR-3 cells (P<.0001). Both Edg2 and Edg4 were detected in ovarian cancer cells; however, only Edg2 was present in normal ovarian surface epithelial cells and IOSE-29 cells. CONCLUSIONS: LPA stimulates ovarian tumor growth, at least in part, via induction of VEGF expression through transcriptional activation. However, this LPA response is not evident in normal ovarian surface epithelial cells. Our data suggest that Edg4, but not Edg2, plays a role in LPA stimulation of ovarian tumor growth.


Assuntos
Fatores de Crescimento Endotelial/biossíntese , Linfocinas/biossíntese , Lisofosfolipídeos/farmacologia , Neoplasias Ovarianas/metabolismo , Receptores Acoplados a Proteínas G , Antibióticos Antineoplásicos/farmacologia , Sítios de Ligação , Northern Blotting , Linhagem Celular , Linhagem Celular Transformada , Meios de Cultivo Condicionados/metabolismo , Dactinomicina/farmacologia , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Feminino , Deleção de Genes , Humanos , Luciferases/metabolismo , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/biossíntese , Receptores de Ácidos Lisofosfatídicos , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo , Fator de Transcrição AP-1/metabolismo , Transfecção , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
12.
Endocrinology ; 142(6): 2351-60, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11356682

RESUMO

Most epithelial ovarian tumors appear to arise from the ovarian surface epithelium (OSE). Even though it has been suggested that estrogen may be associated with ovarian tumorigenesis, the exact role of estrogen in the regulation of apoptosis in neoplastic OSE cells remains uncertain. Immortalized OSE (IOSE) cell lines were generated from human normal OSE. These cell lines represent early neoplastic (IOSE-29), tumorigenic (IOSE-29EC), and late neoplastic (IOSE-29EC/T4 and IOSE-29EC/T5) transformation stages from human normal OSE. The present studies demonstrated that both mRNAs and proteins of estrogen receptor (ER) alpha and beta were expressed in IOSE cell lines. No difference was observed in normal OSE and IOSE-29 cells, whereas treatment with 17beta-estradiol (E(2); 10(-8)-10(-6) M) resulted in an increased thymidine incorporation and DNA content per culture in IOSE-29EC cells. This effect of E(2) was attenuated with tamoxifen treatment (10(-6) M), the estrogen antagonist, suggesting that the effect of E(2) is mediated through specific ERs. There was no stimulatory effect on thymidine incorporation before day 6, but after 6 days of E(2) treatment, thymidine incorporation was significantly increased. Because the ratio of thymidine incorporation to DNA content per culture did not change, this E(2) effect does not appear to indicate stimulation of proliferation but, rather, inhibition of apoptosis. In addition, treatment with tamoxifen (10(-6) M) induced apoptosis up to 3-fold in IOSE-29EC cells, whereas cotreatment with E(2) (10(-8)-10(-6) M) plus tamoxifen attenuated tamoxifen-induced apoptosis in a dose-dependent manner. Both proapoptotic bax and antiapoptotic bcl-2 at messenger RNA (mRNA) and protein levels were expressed in IOSE cell lines. Interestingly, treatments with E(2) resulted in a significant increase of bcl-2 mRNA and protein levels (2- and 1.7-fold, respectively), whereas no difference was observed in bax mRNA level. Thus, E(2) may enhance survival of IOSE-29EC by up-regulating bcl-2, and antiapoptotic bcl-2 may be a dominant regulator of apoptotic pathway in these cells. In conclusion, the present study indicates that early neoplastic (IOSE-29), tumorigenic (IOSE-29EC), and late neoplastic (IOSE-29EC/T4 and T5) OSE cells expressed both ERalpha and ERbeta at the mRNA and protein levels. In addition, E(2) prevented tamoxifen induced-apoptosis through ERs. The mechanism of E(2) action may be associated with up-regulation of bcl-2 gene at mRNA and protein levels. These results suggest that estrogen may play a role in ovarian tumorigenesis by preventing apoptosis in tumorigenic OSE cells.


Assuntos
Apoptose/genética , Estradiol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Neoplasias Ovarianas/química , Proteínas Proto-Oncogênicas c-bcl-2/análise , Proteínas Proto-Oncogênicas c-bcl-2/genética , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , DNA de Neoplasias/biossíntese , Células Epiteliais/química , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Feminino , Humanos , Neoplasias Ovarianas/patologia , RNA Mensageiro/análise , Receptores de Estrogênio/análise , Receptores de Estrogênio/genética , Células Tumorais Cultivadas
13.
Oncogene ; 20(11): 1318-28, 2001 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11313876

RESUMO

Since autocrine regulation of HGF-Met is implicated in many forms of human cancer, we investigated whether the predisposition to develop ovarian cancer in women with hereditary ovarian cancer syndromes involves changes in the expression of HGF-Met by the tissue of origin of epithelial ovarian cancers, the ovarian surface epithelium (OSE). We compared cultures of normal OSE from women with (FH-OSE) (n=20) and with no (NFH-OSE) (n=48) family histories of ovarian cancer, SV40 Tag immortalized OSE lines (IOSE, n=5) and ovarian cancer cell lines (n=3). Cultures derived from 21/22 women with NFH-OSE and 13/13 women with FH-OSE expressed Met mRNA initially. After two to three passages, Met was downregulated in 37% of NFH-OSE cultures but persisted in 100% of FH-OSE cultures and ovarian cancer lines, like other epithelial differentiation markers that are stabilized in FH-OSE and neoplasia. HGF and Met mRNA were concomitantly expressed by NFH-OSE from only three of 32 women but in FH-OSE from eight of 13 women, and also in five of five IOSE and two of three ovarian cancer lines. Conditioned media from FH-OSE, but not NFH-OSE, contained immunoreactive HGF and induced cohort migration which was inhibited by neutralizing HGF antibody. Several signaling molecules of the PI3K pathway, including Akt2 and p70 S6K, were constitutively activated in FH-OSE from six of six women but in NFH-OSE from only four of eight women. Exogenous HGF was mitogenic in OSE, and that effect was regulated through the MAP kinase (ERK1/ERK2) and FRAP/p70 S6K pathways. The proliferative response to HGF was greater in NFH-OSE than in FH-OSE cultures. The results show that FH-OSE cultures differ from NFH-OSE by increased stability of Met expression and by HGF secretion. Constitutive phosphorylation of kinases and a diminished growth response to HGF suggest the presence of autocrine regulation in FH-OSE. In analogy with other cell types where an autocrine HGF-Met loop has been implicated in tumorigenic transformation, this change in FH-OSE may play a role in the enhanced susceptibility to ovarian carcinogenesis in women with hereditary ovarian cancer syndromes.


Assuntos
Células Epiteliais/citologia , Fator de Crescimento de Hepatócito/biossíntese , Neoplasias Ovarianas/etiologia , Neoplasias Ovarianas/genética , Proteínas Proto-Oncogênicas c-met/biossíntese , Comunicação Autócrina , Movimento Celular , Transformação Celular Neoplásica , Feminino , Predisposição Genética para Doença , Humanos , Morfogênese
14.
Endocr Rev ; 22(2): 255-88, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11294827

RESUMO

The epithelial ovarian carcinomas, which make up more than 85% of human ovarian cancer, arise in the ovarian surface epithelium (OSE). The etiology and early events in the progression of these carcinomas are among the least understood of all major human malignancies because there are no appropriate animal models, and because methods to culture OSE have become available only recently. The objective of this article is to review the cellular and molecular mechanisms that underlie the control of normal and neoplastic OSE cell growth, differentiation, and expression of indicators of neoplastic progression. We begin with a brief discussion of the development of OSE, from embryonic to the adult. The pathological and genetic changes of OSE during neoplastic progression are next summarized. The histological characteristics of OSE cells in culture are also described. Finally, the potential involvement of hormones, growth factors, and cytokines is discussed in terms of their contribution to our understanding of the physiology of normal OSE and ovarian cancer development.


Assuntos
Epitélio/fisiologia , Neoplasias Ovarianas/patologia , Ovário/fisiologia , Adulto , Células Cultivadas , Epitélio/patologia , Feminino , Humanos , Neoplasias Ovarianas/metabolismo , Ovário/patologia
15.
Mol Cell Endocrinol ; 174(1-2): 99-110, 2001 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-11306176

RESUMO

Ovarian surface epithelium (OSE) is the tissue of origin for the majority of ovarian cancers. The mechanism underlying the neoplastic transformation of OSE to ovarian cancer is poorly understood. Activin, a member of the transforming growth factor-beta superfamily, has been shown to increase cell proliferation in ovarian cancer cells. The present study was carried out to investigate the expression and regulation of activin/inhibin subunits and activin receptors in normal and neoplastic OSE. Using reverse transcriptase-polymerase chain reaction and Southern blot analysis, the mRNA levels of alpha, betaA and betaB subunits and activin receptor type IIA and IIB were analyzed in normal OSE and the ovarian cancer cell line, OVCAR-3 cells. The alpha and betaA subunits were highly expressed in normal OSE when compared to OVCAR-3 cells. By contrast, betaB subunit was highly expressed in OVCAR-3 cells, when compared to normal OSE cells. Interestingly, activin receptor IIB mRNA levels were significantly higher in OVCAR-3 when compared to normal OSE cells, whereas activin receptor IIA mRNA levels were the same in both cell types. To characterize the growth modulatory role of activin during neoplastic progression, normal OSE and OVCAR-3 cells were treated with recombinant human activin A (rh-activin A). At concentrations of 1,10 and 100 ng/ml, rh-activin A stimulated the growth of OVCAR-3 cells, but not of normal OSE. Treatment with follistatin, binding protein of activin, attenuates the stimulatory effect of activin. To determine whether the growth stimulatory action of activin in the neoplastic OSE is mediated via an autocrine regulatory mechanism, OVCAR-3 cells were treated with rh-activin A in a dose- and time-dependent manner and the expression levels of activin/inhibin subunits and activin receptors were investigated. Treatments with activin increased the alpha and betaA subunit mRNA levels in a dose- and time-dependent manner. However, no difference was observed in levels of betaB subunit, or in activin receptor type IIA and IIB mRNAs following activin treatments in OVCAR-3 cells. Taken together, these results suggest that different levels of activin/inhibin and activin receptor isoforms are expressed in normal and neoplastic OSE cells. In addition, the altered expression of the activin/inhibin subunits, as well as the cell proliferative effect of activin observed in OVCAR-3 but not in normal OSE cells, indicate that activin may act as an autocrine regulator of neoplastic OSE progression.


Assuntos
Células Epiteliais/efeitos dos fármacos , Inibinas/metabolismo , Inibinas/farmacologia , Neoplasias Ovarianas/patologia , RNA Mensageiro/efeitos dos fármacos , Receptores de Fatores de Crescimento/genética , Receptores de Ativinas , Ativinas , Comunicação Autócrina , Divisão Celular/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Substâncias de Crescimento/metabolismo , Substâncias de Crescimento/farmacologia , Humanos , Inibinas/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/farmacologia , Subunidades Proteicas , RNA Mensageiro/metabolismo , Receptores de Fatores de Crescimento/efeitos dos fármacos , Receptores de Fatores de Crescimento/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
16.
Endocrinology ; 142(2): 580-8, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11159828

RESUMO

In the present study, we investigated the expression of estrogen receptors (ERalpha and ERbeta) in human ovarian surface epithelial (hOSE) cells and the ovarian cancer cell line, OVCAR-3, and provided novel evidence that estrogen may have a growth regulatory effect in these cells. Expression levels of ERalpha messenger RNA (mRNA) were 1.5-fold higher in OVCAR-3 cells than in hOSE cells, as revealed by semiquantitative RT-PCR and Southern blot analysis. A significant increase (3.3-fold) in ERss mRNA levels was observed in OVCAR-3 cells compared with hOSE cells. In parallel with mRNA levels, expression levels of ERalpha and ERbeta proteins were also higher in OVCAR-3 cells compared with hOSE cells. We recently proposed that GnRH and its receptor may have an autocrine role in hOSE and ovarian cancer cells. To determine whether estrogen regulates GnRH and GnRH receptor (GnRHR), hOSE and OVCAR-3 cells were treated with various concentrations of 17beta-estradiol for 24 h. Expression levels of GnRH and GnRHR mRNA were examined using quantitative and competitive RT-PCR, respectively. Treatment with 17beta-estradiol induced a significant down-regulation of GnRH mRNA in OVCAR-3 cells, but not in hOSE cells and of GnRHR mRNA in both hOSE and OVCAR-3 cells. Tamoxifen, an estrogen antagonist, prevented the effects of 17ssestradiol, suggesting that estradiol action is mediated via the ER. Finally, the effect of estrogen on the growth of hOSE and OVCAR-3 cells was investigated. The cells were treated with various concentrations of 17ss-estradiol, and the proliferative index of cells was measured using [(3)H]thymidine incorporation and DNA fluorometric assays. 17beta-Estradiol stimulated the growth of OVCAR-3 cells in a dose- and time-dependent manner. In contrast, 17beta-estradiol failed to stimulate the growth of hOSE cells. As estrogen down-regulated GnRH and GnRHR mRNA, we investigated whether estrogen treatment blocks the growth inhibitory effect of a GnRH agonist in OVCAR-3 and hOSE cells. Cells were treated with 17beta-estradiol (10(-7) M) together with (D-Ala(6))-GnRH (10(-7) M), and the proliferative index of cells was measured. Pre- or cotreatment of cells with 17beta-estradiol significantly attenuated the growth inhibitory effect of the GnRH agonist in OVCAR-3 cells, whereas no effect of 17ss-estradiol treatment was observed in hOSE cells. To our knowledge, these results provide the first demonstration of a potential interaction between the estradiol/ER and GnRH/GnRHR systems, which may be important in the growth regulation of normal and neoplastic hOSE cells.


Assuntos
Estradiol/farmacologia , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Inibidores do Crescimento/farmacologia , Neoplasias Ovarianas/patologia , Ovário/citologia , Receptores LHRH/metabolismo , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/agonistas , Hormônio Liberador de Gonadotropina/genética , Inibidores do Crescimento/antagonistas & inibidores , Humanos , Neoplasias Ovarianas/genética , Ovário/efeitos dos fármacos , RNA Mensageiro/metabolismo , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Receptores LHRH/genética
17.
Methods Mol Med ; 39: 169-74, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-21340768

RESUMO

The ovarian surface epithelium (OSE) is the part of the pelvic mesothelium that covers the ovary. It comprises only a minute fraction of the ovary-however, it is the source of the epithelial ovarian carcinomas which are the prime cause of death from gynecological malignancies in North American and European women. The implication that OSE is the source of the epithelial ovarian cancers is based mainly on the demonstration of histopathological changes (1). The early stages of epithelial ovarian car-cinogenesis are poorly understood because these carcinomas are usually diagnosed in late stages and there are no adequate animal models for their study.

18.
Gynecol Oncol ; 78(2): 158-65, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10926796

RESUMO

OBJECTIVE: Hepatocyte growth factor (HGF) enhances cell dissociation and morphogenesis in many forms of carcinomas including some, but not all, cervical carcinomas. In this study, we examined the effects of HGF on two cervical cancer cell lines, derived from the same tumor, with different growth patterns in vivo and in vitro. METHODS: Two cell lines, derived from the same cervical carcinoma, express spinous (C-4I) and basal (C4-II) squamous cell differentiation, respectively. A cell scattering assay was used to determine whether HGF would stimulate cell dissociation and motility. The morphogenetic capacity of HGF was assessed in collagen gel cultures, expression of the HGF receptor c-Met by Western blot analysis, and cadherin expression by immunofluorescence microscopy. RESULTS: HGF-induced cell scattering was intense in C-4II, but limited in C-4I cultures. In collagen gels, C-4I cells formed large, spherical compact colonies with necrotic centers while C-4II cells formed small, irregular colonies with no necrosis. HGF induced proliferation and branching morphogenesis in both lines, but more prominently in C-4II cultures. There was no difference in c-Met or E- and P-cadherin expression between C-4I and C-4II cultures, but the lines differed in their signal transduction responses to HGF. The scatter response was mediated primarily by phosphatidylinositol 3-kinase in line C-4I, but by mitogen-activated protein kinase in line C-4II. HGF induced collagen gel contraction by C-4 cells, demonstrating for the first time that HGF has the capacity to induce this function. CONCLUSIONS: The HGF-induced cell dispersion, morphogenesis, and collagen gel contraction in two cervical carcinoma cell lines were greatly influenced by differences between the lines in differentiation-associated properties. These properties, which include variations in extracellular matrix, junctional proteins, and signal transduction, may also modulate HGF action in vivo and thus determine patterns of invasiveness and growth of cervical carcinomas.


Assuntos
Movimento Celular/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Neoplasias do Colo do Útero/patologia , Animais , Caderinas/biossíntese , Técnicas de Cultura de Células , Linhagem Celular , Colágeno , Feminino , Humanos , Rim/citologia , Rim/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Microscopia de Fluorescência , Morfogênese , Proteínas Proto-Oncogênicas c-met/biossíntese , Células Tumorais Cultivadas/efeitos dos fármacos , Neoplasias do Colo do Útero/metabolismo
19.
Br J Cancer ; 82(8): 1415-20, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10780520

RESUMO

In this study, we have investigated the expression of inhibin subunits and activin receptors (ActRs) in normal and malignant ovarian cells. Each product of the inhibin subunits (alpha, betaa, betab) and activin receptors (ActRs) amplified by reverse transcription polymerase chain reaction were detected as a single band in human granulosa cells, surface epithelial cells (OSE), and the ovarian cancer cell lines OVCAR 3 and SKOV 3. Western blot analysis was performed using polyclonal antibodies against ActR IIa or IIb peptides based on 13 COOH-terminal amino acids; cultured human granulosa cells were used as a positive control. Using ActR IIa antibody, one major band corresponding to approximately 80 kDa and one minor band corresponding to 105 kDa were observed in the samples. One single band at approximately 60 kDa was detected in OVCAR 3 and a 50 kDa band was detected with ActR IIb antibody in cultured granulosa cell, OSE and SKOV 3. Although no detectable change was induced in Smad 4 mRNA in OVCAR 3, Smad 2 mRNA levels were increased during 48 h treatment with activin A (50 ng ml(-1)). These data provide a better understanding as the first step in the mechanism of action of the activin in the epithelial ovarian carcinoma.


Assuntos
Adenocarcinoma/fisiopatologia , Células Epiteliais/fisiologia , Inibinas/fisiologia , Neoplasias Ovarianas/fisiopatologia , Ovário/fisiologia , Transdução de Sinais , Receptores de Ativinas , Ativinas , Adenocarcinoma/patologia , Proteínas de Ligação a DNA/genética , Células Epiteliais/citologia , Células Epiteliais/patologia , Feminino , Células da Granulosa/citologia , Células da Granulosa/fisiologia , Substâncias de Crescimento/fisiologia , Humanos , Inibinas/genética , Neoplasias Ovarianas/patologia , Ovário/citologia , Ovário/patologia , Receptores de Fatores de Crescimento/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Smad2 , Proteína Smad4 , Transativadores/genética , Transcrição Gênica , Células Tumorais Cultivadas
20.
Int J Cancer ; 85(3): 430-7, 2000 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-10652437

RESUMO

Epithelial ovarian carcinomas are thought to originate in the ovarian surface epithelium (OSE), i.e., the mesothelium covering the ovary, but experimental evidence for this origin has been lacking. Contrary to most epithelia, where neoplastic progression is associated with a reduction of E-cadherin, this cell-cell adhesion molecule is sparse in normal human OSE but its expression increases with the development of ovarian epithelial metaplasia and neoplasia. Concurrently, the tumors tend to acquire characteristics of the complex epithelia of the oviduct and uterus. The high proportion of ovarian cancers where such aberrant Mullerian differentiation occurs suggests that this change may confer a selective advantage on the transforming cells. We previously demonstrated that increased E-cadherin expression may be a cause, rather than a consequence, of such Mullerian differentiation. E-cadherin was transfected into SV40 large T antigen-immortalized, E-cadherin-negative cells derived from normal OSE. Constitutive expression of E-cadherin re-established normal epithelial markers that had been lost in culture, such as keratin, and induced markers of metaplasia and neoplasia, such as CA125. In the present study, SV40-immortalized, E-cadherin-transfected cells, but not the E-cadherin-negative controls, were found to be anchorage-independent and to form transplantable, invasive s.c. and i.p. adenocarcinomas in 100% of injected SCID mice. Tumor cells injected i.p. seeded the mesenteries and omentum, invaded the liver and thigh musculature and produced ascites. The presence of SV40 large T antigen in the tumor cell nuclei confirmed their origin as transfected OSE cells. Our results demonstrate that ovarian adenocarcinomas can be derived by genetic manipulation of normal human OSE.


Assuntos
Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Antígenos Transformantes de Poliomavirus/metabolismo , Caderinas/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Adenocarcinoma/imunologia , Animais , Western Blotting , Transformação Celular Neoplásica , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Imunofluorescência , Humanos , Imuno-Histoquímica , Camundongos , Camundongos SCID , Tumor Mulleriano Misto/metabolismo , Tumor Mulleriano Misto/patologia , Neoplasias Ovarianas/imunologia , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...