Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Br J Cancer ; 117(1): 89-101, 2017 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-28571043

RESUMO

BACKGROUND: MicroRNA-7 (miR-7) has been observed as a potent tumour suppressor in multiple cancer types including breast cancer. The aim of this study was to investigate the response sensitivities of metastatic breast cancer cells to miR-7 and the roles of miR-7 in the interaction of endothelial cells and metastatic cancer cells. METHODS: Expression profile of miRNAs in a breast cancer specimen cohort and breast cancer cells were determined using real-time quantitative miRNA assays. Effect of the altering expression of miR-7 on migration, invasion, proliferation, interaction and underlying molecular mechanism of breast cancer cells and endothelial cells was investigated after treatment with the synthesised mimic of miR-7. Luciferase activity analysis was performed to validate Wave-3 as a novel target of miR-7. RESULTS: miR-7 expression was negatively correlated with the stage, grade and survival of the breast cancer patients. There was also differential expression of miRNAs including miR-7 in the breast cancer cells. The synthesised mimic of miR-7 inhibits the motility and wound healing potential of breast cancer cells. The highly metastatic MDA-MB-231 cells are more sensitive to the miR-7 treatment than the poorly invasive MCF-7 cells. Treatment with miR-7 downregulated the expression of EGFR, IGF1R and Wave3 in MDA-MB-231 cells but not in MCF-7 cells. In addition, we further demonstrated that miR-7 inhibited the proliferation, migration and invasion of endothelial cells. And more importantly, miR-7 suppressed the homing and migration of endothelial cells to more aggressive tumour cell conditions. CONCLUSIONS: Given the dual inhibitory effect of miR-7 on metastatic breast cancer cells alone and the interaction of endothelial cells with the tumour-conditioned microenvironment, we suggest miR-7 may be a new therapeutic candidate for its capacity not only to prevent breast cancer cell spreading but also to inhibit tumour-associated angiogenesis in the metastatic breast cancer.


Assuntos
Neoplasias da Mama/genética , Células Endoteliais/metabolismo , MicroRNAs/genética , Apoptose , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/genética , Células Endoteliais/efeitos dos fármacos , Receptores ErbB/efeitos dos fármacos , Receptores ErbB/metabolismo , Feminino , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Células MCF-7 , MicroRNAs/metabolismo , MicroRNAs/farmacologia , Metástase Neoplásica , Estadiamento de Neoplasias , Prognóstico , Reação em Cadeia da Polimerase em Tempo Real , Receptor IGF Tipo 1 , Receptores CXCR/genética , Receptores CXCR4/genética , Receptores de Somatomedina/efeitos dos fármacos , Receptores de Somatomedina/metabolismo , Família de Proteínas da Síndrome de Wiskott-Aldrich/efeitos dos fármacos , Família de Proteínas da Síndrome de Wiskott-Aldrich/metabolismo
2.
Int J Oncol ; 48(6): 2488-96, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27035883

RESUMO

Epithelial protein lost in neoplasm-α (EPLIN-α) is a cytoskeletal protein whose expression is often lost or is aberrant in cancerous cells and tissues and whose loss is believed to be involved in aggressive phenotypes. This study examined this molecule in human epithelial ovarian tissues and investigated the cellular impact of EPLIN-α on ovarian cancer cells (EOC), SKOV3 and COV504. The expression of EPLIN-α in human ovarian tissues and EOC was assessed at both the mRNA and protein levels using reverse transcription-PCR (RT-PCR) and immunohistochemistry, respectively. In vitro assays for cellular matrix adhesion and migration (confirmed by an electrical cell substrate impedance sensing (ECIS) based method), invasion and cell growth were employed in order to assess the biological influence of EPLIN-α expression on EOC cells. Immunohistochemical analysis of ovarian cancer samples demonstrated that only a small number expressed EPLIN-α protein. Downregulation of EPLIN-α protein in EOC cell lines increased the growth, invasion, adhesion and migration in vitro. This EPLIN-α downregulation may have a prognostic value. From these data, we conclude that downregulation of EPLIN-α may be associated with poorer patient prognosis, and that this molecule appears to play a tumour suppressor role by inhibition of EOC growth and migration.


Assuntos
Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Regulação para Baixo , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/patologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma Epitelial do Ovário , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Prognóstico
3.
Anticancer Res ; 36(3): 1287-93, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26977027

RESUMO

BACKGROUND: Occludin is an integral membrane protein localised at tight junctions (TJ). There is no consensus regarding its paramount role in TJ. In previous work we showed that occludin is aberrantly expressed in both human breast tissues and cancer cell lines. This study demonstrates a link to bone metastasis in human cancer. MATERIALS AND METHODS: Primary breast tumours (n=124) and matched normal tissues (n=30) were processed for quantitative polymerase chain reaction (QPCR) analysis. A hammerhead ribozyme was constructed to create occludin knockdown cell lines, MDA-MB-231(ΔOcc) and PC-3(ΔOcc). The effect of human bone matrix extract (BME) was investigated using cell growth and electric cell impedance sensing (ECIS) technology to measure changes in attachment/migration. Trans-epithelial resistance (TER) was measured for assessing changes in TJ function. Cells used were MDA-MB-231, PC-3, CORL-23, SKMES-1 and A-549 human cancer cell lines. RESULTS: Tumours from patients with bone metastasis had significantly lower occludin expression compared to those remaining alive/well (60.7±21 vs. 331±98, respectively, p=0.008). This was striking in ductal carcinomas, where patients alive/well had significantly higher occludin expression compared to those with bone metastasis (391±12.5 vs. 67.9±28, respectively, p=0.0014). ECIS demonstrated that MDA-MB-231(ΔOcc) showed reduced attachment to 5% BME compared to controls (84% vs. 100%) that prevented closure of wounded cell layers. Moreover, these cells had reduced growth on BME. In addition, BME changed the TER of a number of human cell lines and was able to effect changes in the growth of MDA-MB-241 and PC-3 cells, with greater effect on knockdown cells. CONCLUSION: This is the first study to demonstrate that occludin expression has a clear relationship with bone metastasis in human cancer. The discrepancy between this and the in vitro data indicating a reduction in migration/growth rate of occludin knockdown indicates that loss of occludin leads to complex changes in human cancer cell phenotype.


Assuntos
Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Movimento Celular , Ocludina/metabolismo , Matriz Óssea/metabolismo , Neoplasias Ósseas/genética , Neoplasias Ósseas/mortalidade , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo , Impedância Elétrica , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Masculino , Ocludina/genética , Fenótipo , RNA Catalítico/genética , RNA Catalítico/metabolismo , Transdução de Sinais , Análise de Sobrevida , Junções Íntimas/metabolismo , Fatores de Tempo , Transfecção
4.
Int J Oncol ; 47(4): 1429-39, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26316204

RESUMO

Metastasis suppressor 1 (MTSS1) is a potential metastasis suppressor gene involved in the regulation of cytoskeleton dynamics and subsequently in cell motility. MTSS1 expression is frequently reduced in a variety of cancer cells and tissues and this loss may account for increased invasive traits in cancer cells. The present study aimed to assess the role of MTSS1 in epithelial ovarian cancer (EOC) cells. Expression of MTSS1 in human ovarian cells was assessed at both the mRNA and protein levels using reverse transcription-PCR (RT-PCR) and immunohistochemistry, respectively. Full-length MTSS1 cDNA expression vector was used to generate MTSS1 overexpressing cells. The effect of MTSS1 overexpression on cellular functions was examined in EOC cells using a variety of in vitro assays. MTSS1 expression was observed both in ovarian cancer tissues and EOC cells. Over-expression of MTSS1 protein reduced the growth, invasion, adhesion and migration of EOC cell lines in vitro. The present study revealed that MTSS1 plays an essential inhibitory role in the development and progression of ovarian cancers. MTSS1 overexpression is intimately related to migration and metastasis, suggesting that MTSS1 is a potential prognostic marker and therapeutic molecular target in human ovarian cancer.


Assuntos
Proteínas dos Microfilamentos/metabolismo , Invasividade Neoplásica/patologia , Proteínas de Neoplasias/metabolismo , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/patologia , Western Blotting , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Feminino , Imunofluorescência , Humanos , Imuno-Histoquímica , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
5.
Breast Cancer Res ; 16(1): R12, 2014 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-24457069

RESUMO

INTRODUCTION: Upregulation of PI3K/Akt/mTOR signalling in endocrine-resistant breast cancer (BC) has identified mTOR as an attractive target alongside anti-hormones to control resistance. RAD001 (everolimus/Afinitor®), an allosteric mTOR inhibitor, is proving valuable in this setting; however, some patients are inherently refractory or relapse during treatment requiring alternative strategies. Here we evaluate the potential for novel dual mTORC1/2 mTOR kinase inhibitors, exemplified by AZD8055, by comparison with RAD001 in ER + endocrine resistant BC cells. METHODS: In vitro models of tamoxifen (TamR) or oestrogen deprivation resistance (MCF7-X) were treated with RAD001 or AZD8055 alone or combined with anti-hormone fulvestrant. Endpoints included growth, cell proliferation (Ki67), viability and migration, with PI3K/AKT/mTOR signalling impact monitored by Western blotting. Potential ER cross-talk was investigated by immunocytochemistry and RT-PCR. RESULTS: RAD001 was a poor growth inhibitor of MCF7-derived TamR and MCF7-X cells (IC50 ≥1 µM), rapidly inhibiting mTORC1 but not mTORC2/AKT signalling. In contrast AZD8055, which rapidly inhibited both mTORC1 and mTORC2/AKT activity, was a highly effective (P <0.001) growth inhibitor of TamR (IC50 18 nM) and MCF7-X (IC50 24 nM), and of a further T47D-derived tamoxifen resistant model T47D-tamR (IC50 19 nM). AZD8055 significantly (P <0.05) inhibited resistant cell proliferation, increased cell death and reduced migration. Furthermore, dual treatment of TamR or MCF7-X cells with AZD8055 plus fulvestrant provided superior control of resistant growth versus either agent alone (P <0.05). Co-treating with AZD8055 alongside tamoxifen (P <0.01) or oestrogen deprivation (P <0.05) also effectively inhibited endocrine responsive MCF-7 cells. Although AZD8055 inhibited oestrogen receptor (ER) ser167 phosphorylation in TamR and MCF7-X, it had no effect on ER ser118 activity or expression of several ER-regulated genes, suggesting the mTOR kinase inhibitor impact was largely ER-independent. The capacity of AZD8055 for ER-independent activity was further evidenced by growth inhibition (IC5018 and 20 nM) of two acquired fulvestrant resistant models lacking ER. CONCLUSIONS: This is the first report demonstrating dual mTORC1/2 mTOR kinase inhibitors have potential to control acquired endocrine resistant BC, even under conditions where everolimus fails. Such inhibitors may prove of particular benefit when used alongside anti-hormonal treatment as second-line therapy in endocrine resistant disease, and also potentially alongside anti-hormones during the earlier endocrine responsive phase to hinder development of resistance.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Antagonistas do Receptor de Estrogênio/farmacologia , Morfolinas/farmacologia , Sirolimo/análogos & derivados , Serina-Treonina Quinases TOR/antagonistas & inibidores , Antineoplásicos Hormonais/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Everolimo , Feminino , Fulvestranto , Humanos , Imunossupressores/farmacologia , Células MCF-7 , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Complexos Multiproteicos/antagonistas & inibidores , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Estrogênio/antagonistas & inibidores , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Tamoxifeno/farmacologia
6.
Endocr Relat Cancer ; 20(5): 691-704, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23900794

RESUMO

The HER2 transmembrane receptor is a well-characterised predictive marker for trastuzumab benefit and may be associated with decreased benefit from endocrine therapy use. Despite the clinical effectiveness of anti-HER2 agents in such cases, resistance represents a significant limiting factor. Focal adhesion kinase (FAK) plays an important role in HER2 signalling, mediating downstream Akt activation in addition to HER2 cross talk with other growth factor receptors. In this study, we investigated the therapeutic potential of FAK in oestrogen receptor-positive (ER+)/HER2+ breast cancer using the novel FAK-specific inhibitor PF4554878 ('PF878'). The activation of the FAK/HER2 signalling pathway was assessed in ER+/HER2- (MCF7 and T47D) and ER+/HER2+ (BT-474 and MDAMB361) breast cancer cells in the presence or absence of PF878 and PF878±trastuzumab. The effects of PF878 on cell growth as a monotherapy and in combination with trastuzumab were assessed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and Coulter counting with isobologram analysis to determine synergy/additive effects. FAK activation (at Y861 but not at Y397) was highest in ER+/HER2+ cells, which also demonstrated the greatest sensitivity to PF878. As a monotherapy, PF878 prevented heregulin-induced MDA361 cell migration, but had no significant effect on cell growth. The treatment of ER+/HER2+ cells with PF878 and trastuzumab in combination resulted in the synergistic inhibition of cell proliferation. Underlying this was an abrogation of Akt activity and increased poly(ADP-ribose) polymerase cleavage, effects that were greatest in trastuzumab-refractory MDA361 cells. Collectively, these data support a role for FAK in ER+/HER2+ breast cancer, where its targeting has the potential to improve trastuzumab response. This is particularly important in the context of ER+/HER2+, trastuzumab-refractory disease, where FAK inhibition may present an important strategy to restore trastuzumab sensitivity.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Quinase 1 de Adesão Focal/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Neoplasias da Mama/metabolismo , Movimento Celular/efeitos dos fármacos , Feminino , Quinase 1 de Adesão Focal/metabolismo , Humanos , Receptor ErbB-2/metabolismo , Receptores de Estrogênio/metabolismo , Trastuzumab
7.
Front Psychol ; 4: 108, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23471214

RESUMO

The Uncanny Valley Hypothesis (Mori, 1970) predicts that perceptual difficulty distinguishing between a humanlike object (e.g., lifelike prosthetic hand, mannequin) and its human counterpart evokes negative affect. Research has focused on affect, with inconsistent results, but little is known about how objects along the hypothesis' dimension of human likeness (DHL) are actually perceived. This study used morph continua based on human and highly realistic computer-generated (avatar) faces to represent the DHL. Total number and dwell time of fixations to facial features were recorded while participants (N = 60) judged avatar versus human category membership of the faces in a forced choice categorization task. Fixation and dwell data confirmed the face feature hierarchy (eyes, nose, and mouth in this order of importance) across the DHL. There were no further findings for fixation. A change in the relative importance of these features was found for dwell time, with greater preferential processing of eyes and mouth of categorically ambiguous faces compared with unambiguous avatar faces. There were no significant differences between ambiguous and human faces. These findings applied for men and women, though women generally dwelled more on the eyes to the disadvantage of the nose. The mouth was unaffected by gender. In summary, the relative importance of facial features changed on the DHL's non-human side as a function of categorization ambiguity. This change was indicated by dwell time only, suggesting greater depth of perceptual processing of the eyes and mouth of ambiguous faces compared with these features in unambiguous avatar faces.

8.
BMC Cancer ; 12: 458, 2012 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-23039365

RESUMO

BACKGROUND: Acquired resistance to endocrine therapy in breast cancer is a significant problem with relapse being associated with local and/or regional recurrence and frequent distant metastases. Breast cancer cell models reveal that endocrine resistance is accompanied by a gain in aggressive behaviour driven in part through altered growth factor receptor signalling, particularly involving erbB family receptors. Recently we identified that CD44, a transmembrane cell adhesion receptor known to interact with growth factor receptors, is upregulated in tamoxifen-resistant (TamR) MCF7 breast cancer cells. The purpose of this study was to explore the consequences of CD44 upregulation in an MCF7 cell model of acquired tamoxifen resistance, specifically with respect to the hypothesis that CD44 may influence erbB activity to promote an adverse phenotype. METHODS: CD44 expression in MCF7 and TamR cells was assessed by RT-PCR, Western blotting and immunocytochemistry. Immunofluorescence and immunoprecipitation studies revealed CD44-erbB associations. TamR cells (± siRNA-mediated CD44 suppression) or MCF7 cells (± transfection with the CD44 gene) were treated with the CD44 ligand, hyaluronon (HA), or heregulin and their in vitro growth (MTT), migration (Boyden chamber and wound healing) and invasion (Matrigel transwell migration) determined. erbB signalling was assessed using Western blotting. The effect of HA on erbB family dimerisation in TamR cells was determined by immunoprecipitation in the presence or absence of CD44 siRNA. RESULTS: TamR cells overexpressed CD44 where it was seen to associate with erbB2 at the cell surface. siRNA-mediated suppression of CD44 in TamR cells significantly attenuated their response to heregulin, inhibiting heregulin-induced cell migration and invasion. Furthermore, TamR cells exhibited enhanced sensitivity to HA, with HA treatment resulting in modulation of erbB dimerisation, ligand-independent activation of erbB2 and EGFR and induction of cell migration. Overexpression of CD44 in MCF7 cells, which lack endogenous CD44, generated an HA-sensitive phenotype, with HA-stimulation promoting erbB/EGFR activation and migration. CONCLUSIONS: These data suggest an important role for CD44 in the context of tamoxifen-resistance where it may augment cellular response to erbB ligands and HA, factors that are reported to be present within the tumour microenvironment in vivo. Thus CD44 may present an important determinant of breast cancer progression in the setting of endocrine resistance.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica , Receptores de Hialuronatos/genética , Ácido Hialurônico/farmacologia , Neuregulina-1/farmacologia , Tamoxifeno/farmacologia , Antineoplásicos Hormonais/farmacologia , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Receptores ErbB/metabolismo , Feminino , Humanos , Receptores de Hialuronatos/metabolismo , Células MCF-7 , Microscopia de Fluorescência , Multimerização Proteica/efeitos dos fármacos , Interferência de RNA , Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Horm Mol Biol Clin Investig ; 5(2): 67-77, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25961242

RESUMO

Using MCF7 breast cancer cells, it has been shown that antihormones promote expression/activity of oestrogen-repressed tyrosine kinases, notably EGFR, HER2 and Src. These inductive events confer responsiveness to targeted inhibitors (e.g., gefitinib, trastuzumab, saracatinib). We observed that these antihormone-induced phenomena are common to ER+HER2- and ER+HER2+ breast cancer models in vitro, where targeting of EGFR, HER2 or Src alongside antihormone improves antitumour response and delays/prevents endocrine resistance. Such targeted inhibitors also subvert acquired endocrine resistant cells which retain increased EGFR, HER2 and Src (e.g., TAMR and FASR models derived after 6-12 months of tamoxifen or Faslodex treatment). Thus, antihormone-induced tyrosine kinases comprise "compensatory signalling" crucial in limiting maximal initial antihormone response and subsequently driving acquired resistance in vitro. However, despite such convincing preclinical findings from our group and others, clinical trials examining equivalent antigrowth factor strategies have proved relatively disappointing. Our new studies deciphering underlying causes reveal that further antihormone-promoted events could be pivotal in vivo. Firstly, Faslodex induces HER3 and HER4 which sensitise ER+ cells to heregulin, a paracrine growth factor that overcomes endocrine response and diminishes antitumour effect of agents targeting EGFR, HER2 or Src alongside antihormone. Secondly, extended antihormone exposure (experienced by ER+ cells prior to adjuvant clinical relapse) can "reprogramme" the compensatory kinase profile in vitro, hindering candidate antigrowth factor targeting of endocrine resistance. Faslodex resistant cells maintained with this antihormone for 3 years in vitro lose EGFR/HER2 dependency, gaining alternative mitogenic/invasion kinases. Deciphering these previously unrecognised antihormone-induced events could provide superior treatments to control endocrine relapse in the clinic.

10.
Endocrinology ; 149(10): 4912-20, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18583420

RESUMO

Antiestrogens such as tamoxifen are the mainstay of treatment for estrogen receptor-positive breast cancer. However, their effectiveness is limited by the development of endocrine resistance, allowing tumor regrowth and progression. Importantly, in vitro MCF7 cell models of acquired tamoxifen resistance (TamR cells) display an aggressive, invasive phenotype in which activation of epithelial growth factor receptor/IGF-I receptor/Src signaling plays a critical role. In this study, we report that TamR cells have increased levels of zinc and zinc transporter, ZIP7 [solute carrier family 39 (zinc transporter) member 7, also known as SLC39A7], resulting in an enhanced response to exogenous zinc, which is manifested as a greatly increased growth factor receptor activation, leading to increased growth and invasion. Removal of ZIP7, using small interfering RNA, destroys this activation of epithelial growth factor receptor/IGF-I receptor/Src signaling by reducing intracellular zinc levels. Similarly, it also blocks the activation of HER2, -3, and -4. These data suggest that intracellular zinc levels may be a critical factor in determining growth factor responses and that the targeting of zinc transporters may have novel therapeutic implications. We show that ZIP7 is a critical component in the redistribution of zinc from intracellular stores to the cytoplasm and, as such, is essential for the zinc-induced inhibition of phosphatases, which leads to activation of growth factor receptors. Removal of ZIP7 therefore offers a means through which zinc-induced activation of growth factor receptors may be effectively suppressed and provides a mechanism of targeting multiple growth factor pathways, increasing tumor kill, and preventing further development of resistance in breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Transdução de Sinais/fisiologia , Zinco/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Proteína Tirosina Quinase CSK , Proteínas de Transporte de Cátions/genética , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/fisiologia , Receptores ErbB/metabolismo , Moduladores de Receptor Estrogênico/farmacologia , Humanos , Proteínas Tirosina Quinases/metabolismo , RNA Interferente Pequeno , Receptor ErbB-2/metabolismo , Receptor ErbB-4 , Receptor IGF Tipo 1/metabolismo , Zinco/farmacologia , Quinases da Família src
11.
Clin Exp Metastasis ; 24(3): 157-67, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17394086

RESUMO

Src kinase is intimately involved in the control of matrix adhesion and cell migration through its ability to modulate the activity of focal adhesion kinase (FAK). In light of our previous observations that acquisition of tamoxifen resistance in breast cancer cells is accompanied by elevated Src kinase activity, we wish to investigate whether FAK function is also altered in these cells and if this leads to an enhanced migratory phenotype. In in vitro adhesion assays, tamoxifen-resistant (TamR) MCF7 cells had a greater affinity for the matrix proteins fibronectin, laminin, vitronectin and collagen and subsequently demonstrated a much greater migratory capacity across these substrates compared to their weakly-migratory, endocrine-sensitive counterparts. Additionally, elevated levels of activated Src in TamR cells promoted an increase in FAK phosphorylation at Y861 and Y925 and uncoupled FAK activation from an adhesion-dependent process. Inhibition of Src activity using the Src/Abl inhibitor AZD0530 reduced FAK activity, suppressed cell spreading on matrix-coated surfaces and significantly inhibited cell migration. Our data thus suggest that Src kinase plays a central role in the enhanced migratory phenotype that accompanies endocrine resistance through its modulation of FAK signalling and demonstrates the potential use of Src inhibitors as potent suppressors of tumour cell migration.


Assuntos
Neoplasias da Mama/patologia , Adesão Celular , Quinase 1 de Adesão Focal/metabolismo , Tamoxifeno/uso terapêutico , Quinases da Família src/fisiologia , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Movimento Celular , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Fosforilação
12.
Endocr Relat Cancer ; 13(4): 1085-99, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17158755

RESUMO

Our previous investigations using cell models of tamoxifen resistance have shown that the acquisition of an endocrine-insensitive state is accompanied by an invasive in vitro phenotype. In this study, we wished to determine whether this was specifically related to partial oestrogen receptor agonists or whether similar phenomena arise with the newer 'pure' anti-oestrogens, exemplified by fulvestrant. Our data demonstrate that the development of fulvestrant resistance in two breast cancer cell lines, MCF7 and T47D, is accompanied by an augmented migratory and invasive phenotype in vitro and overexpression of the HGF/SF receptor, c-Met. Importantly, upregulated c-Met expression in these cells facilitates their stimulation by HGF/SF-secreting stromal fibroblasts, leading to the activation of Src, Akt and ERK1/2 and a profound enhancement of their aggressive phenotype in vitro. These effects could be specifically attributable to activation of the c-Met receptor since the inclusion of neutralising antibodies to c-Met, or siRNA-mediated knockdown of c-Met expression, suppressed both invasion and migration stimulated by either exogenous HGF/SF, fibroblast-conditioned medium or following co-culture with fibroblast cells. Together, these in vitro data suggest that the development of fulvestrant resistance in vivo may confer a metastatic advantage to the cells by allowing their migratory and invasive behaviour to be augmented by surrounding stromal cells.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/metabolismo , Estradiol/análogos & derivados , Proteínas Proto-Oncogênicas c-met/metabolismo , Células Estromais/metabolismo , Western Blotting , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Estradiol/farmacologia , Moduladores de Receptor Estrogênico/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fulvestranto , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Fator de Crescimento de Hepatócito/farmacologia , Humanos , Invasividade Neoplásica/patologia , Proteínas de Neoplasias/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Células Estromais/efeitos dos fármacos , Células Tumorais Cultivadas , Regulação para Cima , Cicatrização/efeitos dos fármacos
13.
Breast Cancer Res Treat ; 87(2): 167-80, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15377841

RESUMO

The tamoxifen-resistant (TAM-R) MCF-7 breast cancer cell line has been used as a model to identify the signalling pathways that enable resistant cancer cells to grow independently of steroid hormones. In TAM-R cells, peptide growth factor signalling pathways appear to be important in modified cell behaviour, growth and survival. The PI3 kinase signalling components Akt1 and Akt2 are expressed at similar levels by both parental wild-type MCF-7 and TAM-R cells, but Akt1 phosphorylation is significantly increased in TAM-R cells grown under basal conditions. High levels of basal Akt, GSK3 alpha / beta and p70S6 kinase phosphorylation are all inhibited by the PI3 kinase inhibitor, LY 294002. The ligands for the EGFR/erbB1 receptor, EGF (epidermal growth factor) and TGF alpha (transforming growth factor- alpha ) demonstrate an increased ability to activate Akt in TAM-R compared with parental MCF-7 cells and it is proposed that the preferred autocrine or paracrine activation of Akt occurs via the erbB heterodimer EGFR/erbB2 in TAM-R cells. Akt phosphorylation is reduced by gefitinib ("Iressa"/ZD1839). The results suggest that the PI3 kinase pathway plays a role in proliferation of TAM-R cells and is important in the increased EGF induced membrane ruffling detected in the resistant cells. Increased Akt1 activation may contribute to the aggressive phenotype of tamoxifen resistant ER (oestrogen receptor) positive breast cancers.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/patologia , Fator de Crescimento Epidérmico/biossíntese , Proteínas Serina-Treonina Quinases/farmacologia , Proteínas Proto-Oncogênicas/farmacologia , Tamoxifeno/farmacologia , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Fenótipo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Receptores de Estrogênio/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Células Tumorais Cultivadas
14.
Hum Reprod Update ; 10(2): 95-105, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15073140

RESUMO

Pregnancy induces physiological alterations in thyroid function which may make difficult the interpretation of results of thyroid hormone measurement. A state of hyperstimulation of the thyroid gland is common in early pregnancy. In a few cases, thyroid hormone values will deviate from the normal range, which corresponds to the gestational transient thyrotoxicosis. This syndrome is closely associated with hyperemesis gravidarum. The relationship between the two syndromes, demonstrated by epidemiological studies, has been illustrated by an exceptional case of familial recurrent gestational thyrotoxicosis presenting as hyperemesis gravidarum due to hypersensitivity of the thyrotrophin receptor to hCG. However, the exact mechanisms of hyperemesis gravidarum have not yet been identified. Gestational transient thyrotoxicosis has to be distinguished from Graves' disease, because the latter is associated with potential maternal and fetal complications when thyrotoxicosis is not controlled, whereas the former has usually a favourable outcome. The existence of other cases of thyroid hypersensitivity or hCG endowed with abnormal thyrotrophic activity is suspected. They may be identified only by assessment of the thyroid function in cases of hyperemesis gravidarum. The identification of these cases would be helpful to understand the mechanisms of specificity of glycoprotein hormone receptors.


Assuntos
Complicações na Gravidez/etiologia , Gravidez/metabolismo , Receptores da Tireotropina/fisiologia , Sequência de Aminoácidos , Gonadotropina Coriônica/fisiologia , Feminino , Idade Gestacional , Doença de Graves/terapia , Humanos , Hiperêmese Gravídica/etiologia , Hiperêmese Gravídica/genética , Hipertireoidismo/etiologia , Hipertireoidismo/terapia , Dados de Sequência Molecular , Mutação , Complicações na Gravidez/terapia , Valores de Referência , Testes de Função Tireóidea , Glândula Tireoide/fisiologia , Tireotoxicose/metabolismo
15.
Clin Cancer Res ; 10(1 Pt 2): 346S-54S, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14734490

RESUMO

An increasing body of evidence demonstrates that growth factor networks are highly interactive with estrogen receptor signaling in the control of breast cancer growth. As such, tumor responses to antiestrogens are likely to be a composite of the estrogen receptor and growth factor-inhibitory activity of these agents, with alterations/aberrations in growth factor signaling providing a mechanism for the development of antiestrogen resistance. In this light, the current article focuses on illustrating the relationship between growth factor signaling and antiestrogen failure in our in-house tumor models of breast cancer and describing how we are now beginning to successfully target growth factor activity to improve the effects of antiestrogen drugs and to block aggressive disease progression.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/terapia , Moduladores de Receptor Estrogênico/farmacologia , Substâncias de Crescimento/metabolismo , Divisão Celular , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Glândulas Endócrinas/metabolismo , Receptores ErbB/metabolismo , Estrogênios/metabolismo , Humanos , Receptor IGF Tipo 1/metabolismo , Receptores de Estrogênio/metabolismo , Transdução de Sinais , Tamoxifeno/metabolismo , Tamoxifeno/farmacologia
16.
J Clin Endocrinol Metab ; 87(4): 1754-61, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11932312

RESUMO

Human TSH receptor (TSHR) autoantibodies with biological activity result in thyroid dysfunction, but antibodies that simply bind do not. We have applied flow cytometry to the measurements of IgG, IgA, and IgE immunoreactivity to the TSHR in patients with Graves' disease (GD) and thyroid eye disease (TED) and in normal controls. CHO cells stably expressing the extracellular domain of the TSHR with a glycophosphatidylinositol anchor were produced and found to express approximately 4 times as many receptors, but of similar affinity, as JP09 in TSH binding studies. Substantial increases in median fluorescence and peak channel fluorescence were obtained by flow cytometry using TSHR monoclonal antibodies on the glycophosphatidylinositol cells. IgG autoantibodies were demonstrated in 55 of 65 untreated GD patients, 3 of 25 normal subjects, and 4 of 8 atypical TED sera (negative for TSHR autoantibodies with biological activity) by flow cytometry and correlated poorly with thyroid-stimulating antibodies. IgA antibodies were present in 1 of 12 normal, 1 of 7 treated GD with TED, and 3 of 8 atypical TED sera. IgE binding was observed in 1 of 12 normal, 2 of 8 treated GD without TED, 1 of 6 treated GD with TED, and 0 of 8 atypical TED sera. In conclusion, we have demonstrated autoantibodies that bind directly to the TSHR in the majority of GD patients and in 50% of patients with atypical TED and a small number of normal controls lacking TSHR antibodies that affect function. Although predominantly IgG lambda, TSHR autoantibodies of the IgA and IgE isotypes are also detectable.


Assuntos
Autoanticorpos/análise , Doença de Graves/imunologia , Imunoglobulina A/análise , Imunoglobulina E/análise , Imunoglobulina G/análise , Receptores da Tireotropina/imunologia , Adulto , Idoso , Animais , Células CHO , Cricetinae , Feminino , Citometria de Fluxo , Humanos , Masculino , Pessoa de Meia-Idade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...