Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Clin Exp Ophthalmol ; 42(1): 33-52, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24433356

RESUMO

Retinoblastoma is a paediatric ocular tumour that continues to reveal much about the genetic basis of cancer development. Study of genomic aberrations in retinoblastoma tumours has exposed important mechanisms of cancer development and identified oncogenes and tumour suppressors that offer potential points of therapeutic intervention. The recent development of next-generation genomic technologies has allowed further refinement of the genomic landscape of retinoblastoma at high resolution. In a relatively short period of time, a wealth of genetic and epigenetic data has emerged on a small number of tumour samples. These data highlight the inherent molecular complexity of this cancer despite the fact that most retinoblastomas are initiated by the inactivation of a single tumour suppressor gene. This review outlines the current understanding of the genomic, genetic and epigenetic changes in retinoblastoma, highlighting recent genome-wide analyses that have identified exciting candidate genes worthy of further validation as potential prognostic and therapeutic targets.


Assuntos
Genômica , Neoplasias da Retina/genética , Retinoblastoma/genética , Genes Supressores de Tumor , Genoma Humano/genética , Humanos , MicroRNAs/genética , Polimorfismo de Nucleotídeo Único
2.
Lancet Oncol ; 14(4): 327-34, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23498719

RESUMO

BACKGROUND: Retinoblastoma is the childhood retinal cancer that defined tumour-suppressor genes. Previous work shows that mutation of both alleles of the RB1 retinoblastoma suppressor gene initiates disease. We aimed to characterise non-familial retinoblastoma tumours with no detectable RB1 mutations. METHODS: Of 1068 unilateral non-familial retinoblastoma tumours, we compared those with no evidence of RB1 mutations (RB1(+/+)) with tumours carrying a mutation in both alleles (RB1(-/-)). We analysed genomic copy number, RB1 gene expression and protein function, retinal gene expression, histological features, and clinical data. FINDINGS: No RB1 mutations (RB1(+/+)) were reported in 29 (2·7%) of 1068 unilateral retinoblastoma tumours. 15 of the 29 RB1(+/+) tumours had high-level MYCN oncogene amplification (28-121 copies; RB1(+/+)MYCN(A)), whereas none of 93 RB1(-/-) primary tumours tested showed MYCN amplification (p<0·0001). RB1(+/+)MYCN(A) tumours expressed functional RB1 protein, had fewer overall genomic copy-number changes in genes characteristic of retinoblastoma than did RB1(-/-) tumours, and showed distinct aggressive histological features. MYCN amplification was the sole copy-number change in one RB1(+/+)MYCN(A) retinoblastoma. One additional MYCN(A) tumour was discovered after the initial frequencies were determined, and this is included in further analyses. Median age at diagnosis of the 17 children with RB1(+/+)MYCN(A) tumours was 4·5 months (IQR 3·5-10), compared with 24 months (15-37) for 79 children with non-familial unilateral RB1(-/-) retinoblastoma. INTERPRETATION: Amplification of the MYCN oncogene might initiate retinoblastoma in the presence of non-mutated RB1 genes. These unilateral RB1(+/+)MYCN(A) retinoblastomas are characterised by distinct histological features, only a few of the genomic copy-number changes that are characteristic of retinoblastoma, and very early age of diagnosis. FUNDING: National Cancer Institute-National Institutes of Health, Canadian Institutes of Health Research, German Research Foundation, Canadian Retinoblastoma Society, Hyland Foundation, Toronto Netralaya and Doctors Lions Clubs, Ontario Ministry of Health and Long Term Care, UK-Essen, and Foundations Avanti-STR and KiKa.


Assuntos
Dosagem de Genes , Proteínas Nucleares , Proteínas Oncogênicas , Proteína do Retinoblastoma , Retinoblastoma , Alelos , Linhagem Celular Tumoral , Criança , Pré-Escolar , Feminino , Amplificação de Genes , Regulação Neoplásica da Expressão Gênica , Genoma Humano , Humanos , Lactente , Mutação , Proteína Proto-Oncogênica N-Myc , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Polimorfismo de Nucleotídeo Único , Retinoblastoma/genética , Retinoblastoma/metabolismo , Retinoblastoma/patologia , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo
3.
Int J Cancer ; 130(8): 1844-54, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21618518

RESUMO

The novel oncogene KIF14 (kinesin family member 14) shows genomic gain and overexpression in many cancers including OvCa (ovarian cancer). We discovered that expression of the mitotic kinesin KIF14 is predictive of poor outcome in breast and lung cancers. We now determine the prognostic significance of KIF14 expression in primary OvCa tumors, and evaluate KIF14 action on OvCa cell tumorigenicity in vitro. Gene-specific multiplex PCR and real-time QPCR were used to measure KIF14 genomic (109 samples) and mRNA levels (122 samples) in OvCa tumors. Association of KIF14 with clinical variables was studied using Kaplan-Meier survival and Cox regression analyses. Cellular effects of KIF14 overexpression (stable transfection) and inhibition (stable shRNA knockdown) were studied by proliferation (cell counts), survival (Annexin V immunocytochemistry) and colony formation (soft-agar growth). KIF14 genomic gain (>2.6 copies) was present in 30% of serous OvCas, and KIF14 mRNA was elevated in 91% of tumors versus normal epithelium. High KIF14 in tumors independently predicted for worse outcome (p = 0.03) with loss of correlation with proliferation marker expression and increased rates of recurrence. Overexpression of KIF14 in OvCa cell lines increased proliferation and colony formation (p < 0.01), whereas KIF14 knockdown induced apoptosis and dramatically reduced colony formation (p < 0.05). Our findings indicate that KIF14 mRNA is an independent prognostic marker in serous OvCa. Dependence of OvCa cells on KIF14 for maintenance of in vitro colony formation suggests a role of KIF14 in promoting a tumorigenic phenotype, beyond its known role in proliferation.


Assuntos
Biomarcadores Tumorais/genética , Cinesinas/genética , Proteínas Oncogênicas/genética , Neoplasias Ovarianas/genética , Adenocarcinoma de Células Claras/genética , Adenocarcinoma de Células Claras/patologia , Adenocarcinoma de Células Claras/terapia , Biomarcadores Tumorais/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patologia , Cistadenocarcinoma Seroso/terapia , Variações do Número de Cópias de DNA , Feminino , Imunofluorescência , Regulação Neoplásica da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Cinesinas/metabolismo , Proteínas Oncogênicas/metabolismo , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Prognóstico , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Modelos de Riscos Proporcionais , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa/estatística & dados numéricos
4.
BMC Cancer ; 12: 69, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22336108

RESUMO

BACKGROUND: Most human cancers show inactivation of both pRB- and p53-pathways. While retinoblastomas are initiated by loss of the RB1 tumor suppressor gene, TP53 mutations have not been found. High expression of the p53-antagonist MDM2 in human retinoblastomas may compromise p53 tumor surveillance so that TP53 mutations are not selected for in retinoblastoma tumorigenesis. We previously showed that p14ARF protein, which activates p53 by inhibiting MDM2, is low in retinoblastomas despite high mRNA expression. METHODS: In human fetal retinas, adult retinas, and retinoblastoma cells, we determined endogenous p14ARF mRNA, ARF protein, and miR-24 expression, while integrity of p53 signalling in WERI-Rb1 cells was tested using an adenovirus vector expressing p14ARF. To study p14ARF biogenesis, retinoblastoma cells were treated with the proteasome inhibitor, MG132, and siRNA against miR-24. RESULTS: In human retinoblastoma cell lines, p14ARF mRNA was disproportionally high relative to the level of p14ARF protein expression, suggesting a perturbation of p14ARF regulation. When p14ARF was over-expressed by an adenovirus vector, expression of p53 and downstream targets increased and cell growth was inhibited indicating an intact p14ARF-p53 axis. To investigate the discrepancy between p14ARF mRNA and protein in retinoblastoma, we examined p14ARF biogenesis. The proteasome inhibitor, MG132, did not cause p14ARF accumulation, although p14ARF normally is degraded by proteasomes. miR-24, a microRNA that represses p14ARF expression, is expressed in retinoblastoma cell lines and correlates with lower protein expression when compared to other cell lines with high p14ARF mRNA. Transient over-expression of siRNA against miR-24 led to elevated p14ARF protein in retinoblastoma cells. CONCLUSIONS: In retinoblastoma cells where high levels of p14ARF mRNA are not accompanied by high p14ARF protein, we found a correlation between miR-24 expression and low p14ARF protein. p14ARF protein levels were restored without change in mRNA abundance upon miR-24 inhibition suggesting that miR-24 could functionally repress expression, effectively blocking p53 tumor surveillance. During retinal tumorigenesis, miR-24 may intrinsically compromise the p53 response to RB1 loss.


Assuntos
Regulação Neoplásica da Expressão Gênica , MicroRNAs/fisiologia , Neoplasias da Retina/metabolismo , Retinoblastoma/genética , Proteína Supressora de Tumor p14ARF/metabolismo , Proteína Supressora de Tumor p53/fisiologia , Linhagem Celular Tumoral , Inibidores de Cisteína Proteinase/farmacologia , Humanos , Leupeptinas/farmacologia , RNA/metabolismo , RNA Mitocondrial , RNA Interferente Pequeno/metabolismo , Retina/embriologia , Retina/metabolismo , Neoplasias da Retina/genética , Proteína Supressora de Tumor p14ARF/efeitos dos fármacos , Proteína Supressora de Tumor p14ARF/genética
5.
Gene ; 414(1-2): 95-105, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18372118

RESUMO

Bone morphogenetic protein (BMP)-4 signalling leads to the direct upregulation of ID3 proto-oncogene expression in human ovarian cancer cells. An upstream BMP4-responsive enhancer element consisting of a palindromic BMP response element (BRE) site and CAGA box was identified ~3.0 kb upstream of the human ID3 gene, and a nearly-identical element exists in the second intron of the ID3 gene. BMP4 stimulation leads to the direct binding of Smads 1/5 and Smad4 to the upstream and intronic enhancers, and together both enhancers cooperate to yield heightened BMP4-mediated ID3 promoter activity. We further demonstrate that ID3 is overexpressed in human ovarian cancer cells when compared to normal ovarian surface epithelial cells, and treatment of ovarian cancer cells with the BMP4 antagonist Noggin abrogates endogenous ID3 gene expression. Our findings define the mechanism of BMP4-mediated ID3 gene expression, and support the notion that ovarian cancer cells possess autocrine BMP4 signalling required to sustain ID3 overexpression which may contribute to human ovarian cancer pathogenesis.


Assuntos
Comunicação Autócrina , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Inibidoras de Diferenciação/genética , Proteínas de Neoplasias/genética , Neoplasias Ovarianas/genética , Transdução de Sinais , Animais , Sequência de Bases , Northern Blotting , Western Blotting , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/genética , Células COS , Células Cultivadas , Chlorocebus aethiops , Imunoprecipitação da Cromatina , Ensaio de Desvio de Mobilidade Eletroforética , Elementos Facilitadores Genéticos , Feminino , Humanos , Proteínas Inibidoras de Diferenciação/metabolismo , Luciferases , Dados de Sequência Molecular , Proteínas de Neoplasias/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Ovário/metabolismo , Ovário/patologia , Regiões Promotoras Genéticas/genética , Proto-Oncogene Mas , Elementos de Resposta , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Transfecção
6.
Am J Chin Med ; 32(5): 737-53, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15633809

RESUMO

We investigated the effects of Gu-Sui-Bu using in vitro bone cell cultures. Primary rabbit and mouse marrow cells were cultured with or without five different concentrations of Gu-Sui-Bu extract. Osteoclast numbers were assessed using tartrate-resistant acid phosphatase (TRAP) positive cell counts and for function, osteoclast resorption pits on bovine bone slices were performed. Alkaline phosphatase (AP) positive cell counts and mineralized nodule formation were examined to assess osteoblast function with Gu-Sui-Bu. TRAP+ osteoclast numbers increased, as did the number and size of resorption pits with 0.001 mg/ml of extract. Low doses of extract did not alter AP+ colony number or mineralized nodule formation, but both were inhibited by doses of 0.1 mg/ml or higher. The highest dose of extract (10 mg/ml) inhibited proliferation of all cell types. At 0.01 and 0.001 mg/ml doses, RANKL increased over time; however, osteoprotegerin levels only increased at doses > or = 0.1 mg/ml. Resorption pit formation was decreased without alteration in mature multinucleated (TRAP+) cell counts only at the highest dose of the putative active ingredient of Gu-Sui-Bu. In summary, lower concentrations of Gu-Sui-Bu extract had positive effects on osteoclast proliferation, survival and resorptive activity that may be mediated through enhanced prostaglandin secretion. However, high doses of extract proved detrimental to osteoclast and osteoblast survival. No effect of low doses of Gu-Sui-Bu extract was seen in osteoblast cultures. High doses of the putative active ingredient of Gu-Sui-Bu showed mild inhibition of mouse osteoclast function.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Osteoclastos/efeitos dos fármacos , Animais , Osso e Ossos/citologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Células Cultivadas , Citocinas/efeitos dos fármacos , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Medicamentos de Ervas Chinesas/química , Ensaio de Imunoadsorção Enzimática , Técnicas Imunoenzimáticas , Camundongos , Osteoclastos/metabolismo , Coelhos
7.
PLoS One ; 9(3): e91540, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24626475

RESUMO

KIF14 (kinesin family member 14) is a mitotic kinesin and an important oncogene in several cancers. Tumor KIF14 expression levels are independently predictive of poor outcome, and in cancer cells KIF14 can modulate metastatic behavior by maintaining appropriate levels of cell adhesion and migration proteins at the cell membrane. Thus KIF14 is an exciting potential therapeutic target. Understanding KIF14's regulation in cancer cells is crucial to the development of effective and selective therapies to block its tumorigenic function(s). We previously determined that close to 30% of serous ovarian cancers (OvCa tumors) exhibit low-level genomic gain, indicating one mechanism of KIF14 overexpression in tumors. We now report on transcriptional and epigenetic regulation of KIF14. Through promoter deletion analyses, we identified one cis-regulatory region containing binding sites for Sp1, HSF1 and YY1. siRNA-mediated knockdown of these transcription factors demonstrated endogenous regulation of KIF14 overexpression by Sp1 and YY1, but not HSF1. ChIP experiments confirmed an enrichment of both Sp1 and YY1 binding to the endogenous KIF14 promoter in OvCa cell lines with high KIF14 expression. A strong correlation was seen in primary serous OvCa tumors between Sp1, YY1 and KIF14 expression, further evidence that these transcription factors are important players in KIF14 overexpression. Hypomethylation patterns were observed in primary serous OvCa tumors, suggesting a minor role for promoter methylation in the control of KIF14 gene expression. miRNA expression analysis determined that miR-93, miR-144 and miR-382 had significantly lower levels of expression in primary serous OvCa tumors than normal tissues; treatment of an OvCa cell line with miRNA mimics and inhibitors specifically modulated KIF14 mRNA levels, pointing to potential novel mechanisms of KIF14 overexpression in primary tumors. Our findings reveal multiple mechanisms of KIF14 upregulation in cancer cells, offering new targets for therapeutic interventions to reduce KIF14 in tumors, aiming at improved prognosis.


Assuntos
Epigênese Genética , Regulação da Expressão Gênica , Cinesinas/metabolismo , Proteínas Oncogênicas/metabolismo , Neoplasias Ovarianas/metabolismo , Transcrição Gênica , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Metilação de DNA , Feminino , Deleção de Genes , Células HeLa , Humanos , Cinesinas/genética , Mitose , Metástase Neoplásica , Proteínas Oncogênicas/genética , Neoplasias Ovarianas/genética , Prognóstico , Regiões Promotoras Genéticas , RNA Interferente Pequeno/metabolismo , Fator de Transcrição Sp1/metabolismo , Regulação para Cima , Fator de Transcrição YY1/metabolismo
8.
J Ovarian Res ; 7: 123, 2014 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-25528264

RESUMO

OBJECTIVE: Previously, it has been shown that KIF14 mRNA is overexpressed in ovarian cancer (OvCa), regardless of histological subtype. KIF14 levels are independently predictive of poor outcome and increased rates of recurrence in serous OvCa patients. Furthermore, it has been shown that KIF14 also controls the in vivo tumorigenicity of OvCa cell lines. In this study, we evaluate the potential of KIF14 as a therapeutic target through selective inhibition of KIF14 in primary high-grade serous patient-derived OvCa cells. METHODS: To assess the dependence of primary serous OvCa cultures on KIF14, protein levels in 11 prospective high grade serous ovarian cancer samples were increased (KIF14 overexpression by transfection) or decreased (anti-KIF14 shRNA) in vitro, and proliferative capacity, anchorage independence and xenograft growth were assessed. RESULTS: Seven of eleven samples demonstrated increased/decreased in vitro proliferation in response to KIF14 overexpression/knockdown, respectively. When examining in vitro tumorigenicity (colony formation) and in vivo growth (subcutaneous xenografts) in response to KIF14 manipulation, none of the samples demonstrated growth in soft agar (11 samples), or xenograft growth (4 samples). CONCLUSIONS: Although primary high-grade serous OvCa cells may depend on KIF14 for in vitro proliferation we were unable to demonstrate a role for KIF14 on tumorigenicity or develop an in vivo model for assessment. We have, however developed an effective in vitro method to evaluate the effect of target gene manipulation on the proliferative capacity of primary OvCa cultures.


Assuntos
Cinesinas/fisiologia , Neoplasias Císticas, Mucinosas e Serosas/metabolismo , Proteínas Oncogênicas/fisiologia , Neoplasias Ovarianas/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Feminino , Expressão Gênica , Humanos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante de Neoplasias , Neoplasias Císticas, Mucinosas e Serosas/patologia , Neoplasias Ovarianas/patologia , Cultura Primária de Células
9.
Methods Mol Biol ; 1049: 323-36, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23913227

RESUMO

We have refined the technique for isolating and propagating cultures of primary epithelial ovarian cancer (EOC) cells derived from solid tumors and ascites. Both protocols involve a simple yet rapid method for the growth and propagation of EOC tumor and ascites cells in a basal culture medium without the addition of growth factors. Isolation of tumor EOC cells involves the mechanical disruption of the tumor tissue with the help of a cell scraper, while ascites-derived EOC cells are mixed with growth medium and placed directly into culture with very little manipulation. We further describe a partial trypsinization method to eliminate fibroblast contamination from primary EOC cells derived from solid tumors. These methods allow for the direct application of many molecular, cellular, and functional analyses within a few weeks of initial isolation, with the added potential of retrospective analyses of archived cells and tissues. Thus, we have included steps for long-term cryopreservation of early-passage EOC cells. Initial isolation of EOC cells can be completed within 1 h, and primary cells are further expanded in culture for several weeks.


Assuntos
Líquido Ascítico/patologia , Técnicas de Cultura de Células/métodos , Neoplasias Ovarianas/patologia , Cultura Primária de Células/métodos , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Ovarianas/genética
10.
J Cell Biol ; 199(6): 951-67, 2012 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-23209302

RESUMO

The small GTPase Rap1 regulates inside-out integrin activation and thereby influences cell adhesion, migration, and polarity. Several Rap1 effectors have been described to mediate the cellular effects of Rap1 in a context-dependent manner. Radil is emerging as an important Rap effector implicated in cell spreading and migration, but the molecular mechanisms underlying its functions are unclear. We report here that the kinesin KIF14 associates with the PDZ domain of Radil and negatively regulates Rap1-mediated inside-out integrin activation by tethering Radil on microtubules. The depletion of KIF14 led to increased cell spreading, altered focal adhesion dynamics, and inhibition of cell migration and invasion. We also show that Radil is important for breast cancer cell proliferation and for metastasis in mice. Our findings provide evidence that the concurrent up-regulation of Rap1 activity and increased KIF14 levels in several cancers is needed to reach optimal levels of Rap1-Radil signaling, integrin activation, and cell-matrix adhesiveness required for tumor progression.


Assuntos
Adenocarcinoma/metabolismo , Neoplasias da Mama/metabolismo , Proteínas de Transporte/metabolismo , Cinesinas/metabolismo , Proteínas Oncogênicas/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/secundário , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Proliferação de Células , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transdução de Sinais , Transplante Heterólogo
11.
J Ovarian Res ; 4: 10, 2011 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-21699706

RESUMO

Ovarian cancer continues to be the most lethal of the gynaecologic malignancies due to the lack of early detection, screening strategies and ineffective therapeutics for late-stage metastatic disease, particularly in the recurrent setting. The gathering of researchers investigating fundamental pathobiology of ovarian cancer and the clinicians who treat patients with this insidious disease is paramount to meeting the challenges we face. Since 2002, the Canadian Conference on Ovarian Cancer Research, held every two years, has served this essential purpose. The objectives of this conference have been to disseminate new information arising from the most recent ovarian cancer research and identify the most pressing challenges we still face as scientists and clinicians. This is best accomplished through direct encounters and exchanges of innovative ideas among colleagues and trainees from the realms of basic science and clinical disciplines. This meeting has and continues to successfully facilitate rapid networking and establish new collaborations from across Canada. This year, more guest speakers and participants from other countries have extended the breadth of the research on ovarian cancer that was discussed at the meeting. This report summarizes the key findings presented at the fifth biennial Canadian Conference on Ovarian Cancer Research held in Toronto, Ontario, and includes the important issues and challenges we still face in the years ahead to make a significant impact on this devastating disease.

13.
Carcinogenesis ; 28(6): 1153-62, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17272306

RESUMO

We identified previously an autocrine bone morphogenetic protein-4 (BMP4) signalling pathway in primary human normal ovarian surface epithelial (OSE) and epithelial ovarian cancer (OvCa) cells. Herein we show that treatment of OvCa cells with BMP4 produced morphological alterations and increased cellular adhesion, motility and invasion. The BMP4 inhibitor noggin blocked the BMP4-induced phenotype, and decreased autocrine BMP4-mediated OvCa cell motility and adherence. In response to exogenous BMP4, the epithelial-mesenchymal transition (EMT) markers Snail and Slug mRNA and protein were up-regulated, E-cadherin mRNA and protein were down-regulated and the network of alpha smooth muscle actin changed to resemble a mesenchymal cell. We also observed changes in the level of activated Rho GTPases in OvCa cells treated with BMP4, strongly suggesting that the changes in morphology, adhesion, motility and invasion are probably mediated through the activation of these molecules. Strikingly, treatment of normal OSE cells with BMP4 or noggin failed to alter cell motility, providing evidence that OSE and OvCa cells possess a distinct capability to respond to BMP4. Overall, our studies suggest a link between autocrine BMP signalling mediated through the Rho GTPase family and Snail- and Slug-induced EMT that may collectively contribute to aggressive OvCa behaviour.


Assuntos
Biomarcadores Tumorais/metabolismo , Proteínas Morfogenéticas Ósseas/fisiologia , Mesoderma/enzimologia , Neoplasias Ovarianas/enzimologia , Proteínas rho de Ligação ao GTP/metabolismo , Proteína Morfogenética Óssea 4 , Progressão da Doença , Ativação Enzimática/fisiologia , Epitélio/enzimologia , Epitélio/patologia , Feminino , Humanos , Mesoderma/patologia , Neoplasias Ovarianas/patologia , Células Tumorais Cultivadas
15.
Nat Protoc ; 1(6): 2643-9, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17406520

RESUMO

Our laboratory has refined the technique for isolating primary cultures of normal human ovarian surface epithelial (OSE) cells by combining two different protocols involving the enzymatic and mechanical removal of OSE cells from ovarian biopsies. A simple protocol of obtaining primary epithelial ovarian cancer (EOC) cells from the ascites fluid removed from patients with high-grade ovarian cancer is also described. These methods allow for the direct application of many molecular and cellular analyses of normal versus cancer cells isolated freshly from patients, with the added potential for retrospective analyses of archived cells and tissues. Thus, we have included optional steps for the immediate preparation of ascites-derived EOC cells to be used for subsequent cytological analyses. Initial isolation of OSE or EOC cells can be completed in 1 h, and primary cells are further expanded in culture for several weeks.


Assuntos
Técnicas de Cultura de Células , Células Epiteliais , Neoplasias Ovarianas , Células Tumorais Cultivadas , Ascite , Feminino , Humanos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa