Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 51(5): 559-72, 2013 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-23973329

RESUMO

TGF-ß signaling is a therapeutic target in advanced cancers. We identified tumor necrosis factor receptor-associated factor 4 (TRAF4) as a key component mediating pro-oncogenic TGF-ß-induced SMAD and non-SMAD signaling. Upon TGF-ß stimulation, TRAF4 is recruited to the active TGF-ß receptor complex, where it antagonizes E3 ligase SMURF2 and facilitates the recruitment of deubiquitinase USP15 to the TGF-ß type I receptor (TßRI). Both processes contribute to TßRI stabilization on the plasma membrane and thereby enhance TGF-ß signaling. In addition, the TGF-ß receptor-TRAF4 interaction triggers Lys 63-linked TRAF4 polyubiquitylation and subsequent activation of the TGF-ß-activated kinase (TAK)1. TRAF4 is required for efficient TGF-ß-induced migration, epithelial-to-mesenchymal transition, and breast cancer metastasis. Elevated TRAF4 expression correlated with increased levels of phosphorylated SMAD2 and phosphorylated TAK1 as well as poor prognosis among breast cancer patients. Our results demonstrate that TRAF4 can regulate the TGF-ß pathway and is a key determinant in breast cancer pathogenesis.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator 4 Associado a Receptor de TNF/genética , Fator 4 Associado a Receptor de TNF/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Fosforilação , Poliubiquitina/metabolismo , Prognóstico , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Transdução de Sinais , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Ubiquitina-Proteína Ligases/metabolismo
2.
Breast Cancer Res ; 17: 28, 2015 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-25849225

RESUMO

INTRODUCTION: Increased expression of αv integrins is frequently associated with tumor cell adhesion, migration, invasion and metastasis, and correlates with poor prognosis in breast cancer. However, the mechanism by which αv integrins can enhance breast cancer progression is still largely unclear. The effects of therapeutic targeting of αv integrins in breast cancer also have yet to be investigated. METHODS: We knocked down αv integrin in MDA-MB-231 and MCF10A-M4 breast cancer cells, or treated these cells with the αv antagonist GLPG0187. The effects of αv integrin depletion on mesenchymal markers, transforming growth factor-ß (TGF-ß)/Smad signaling and TGF-ß-induced target gene expression were analyzed in MDA-MB-231 cells by RNA analysis or Western blotting. The function of αv integrin on breast cancer cell migration was investigated by transwell assay in vitro, and its effect on breast cancer progression was assessed by both zebrafish and mouse xenografts in vivo. In the mouse model, GLPG0187 was administered separately, or in combination with the standard-of-care anti-resorptive agent zoledronate and the chemotherapeutic drug paclitaxel, to study the effects of combinational treatments on breast cancer metastasis. RESULTS: Genetic interference and pharmacological targeting of αv integrin with GLPG0187 in different breast cancer cell lines inhibited invasion and metastasis in the zebrafish or mouse xenograft model. Depletion of αv integrin in MDA-MB-231 cells inhibited the expression of mesenchymal markers and the TGF-ß/Smad response. TGF-ß induced αv integrin mRNA expression and αv integrin was required for TGF-ß-induced breast cancer cell migration. Moreover, treatment of MDA-MB-231 cells with non-peptide RGD antagonist GLPG0187 decreased TGF-ß signaling. In the mouse xenografts GLPG0187 inhibited the progression of bone metastasis. Maximum efficacy of inhibition of bone metastasis was achieved when GLPG0187 was combined with the standard-of-care metastatic breast cancer treatments. CONCLUSION: These findings show that αv integrin is required for efficient TGF-ß/Smad signaling and TGF-ß-induced breast cancer cell migration, and for maintaining a mesenchymal phenotype of the breast cancer cells. Our results also provide evidence that targeting αv integrin could be an effective therapeutic approach for treatment of breast cancer tumors and/or metastases that overexpress αv integrin.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Deleção de Genes , Integrina alfaV/genética , Animais , Animais Geneticamente Modificados , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Progressão da Doença , Transição Epitelial-Mesenquimal/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Integrina alfaV/metabolismo , Camundongos , Metástase Neoplásica , Fator de Crescimento Transformador beta/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Peixe-Zebra
3.
Arterioscler Thromb Vasc Biol ; 34(1): 177-86, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24158517

RESUMO

OBJECTIVE: Endothelial cells (ECs), pericytes, and vascular smooth muscle cells (vSMCs) are essential for vascular development, and their dysfunction causes multiple cardiovascular diseases. Primary vascular cells for research are, however, difficult to obtain. Human-induced pluripotent stem cells (hiPSCs) derived from somatic tissue are a renewable source of ECs and vSMCs; however, their use as disease models has been limited by low and inconsistent efficiencies of differentiation and the lack of phenotypic bioassays. APPROACH AND RESULTS: Here, we developed defined conditions for simultaneous derivation of ECs and pericytes with high efficiency from hiPSCs of different tissue origin. The protocol was equally efficient for all lines and human embryonic stem cells (hESCs). The ECs could undergo sequential passage and were phenotypically indistinguishable, exhibiting features of arterial-like embryonic ECs. Moreover, hiPSC-derived ECs formed an authentic vascular plexus when cocultured with hiPSC-derived pericytes. The coculture system recapitulated (1) major steps of vascular development including EC proliferation and primary plexus remodeling, and (2) EC-mediated maturation and acquisition of contractile vSMC phenotype by pericytes. In addition, hiPSC-derived ECs integrated into developing vasculature as xenografts in zebrafish. This contrasts with more widely used ECs from human umbilical vein, which form only unstable vasculature and were completely unable to integrate into zebrafish blood vessels. CONCLUSIONS: We demonstrate that vascular derivatives of hiPSC, such as ECs and pericytes, are fully functional and can be used to study defective endothelia-pericyte interactions in vitro for disease modeling and studies on tumor angiogenesis.


Assuntos
Diferenciação Celular , Células Endoteliais/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Neovascularização Fisiológica , Pericitos/fisiologia , Animais , Biomarcadores/metabolismo , Comunicação Celular , Linhagem Celular , Linhagem da Célula , Proliferação de Células , Técnicas de Cocultura , Células Endoteliais/metabolismo , Células Endoteliais/transplante , Regulação da Expressão Gênica , Xenoenxertos , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/fisiologia , Pericitos/metabolismo , Pericitos/transplante , Fenótipo , Vasoconstrição , Peixe-Zebra
4.
Nucleic Acids Res ; 40(13): 5988-6000, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22492511

RESUMO

The MYB proto-oncogene is expressed in most estrogen receptor-positive (ERα(+)) breast tumors and cell lines. Expression of MYB is controlled, in breast cancer and other cell types, by a transcriptional pausing mechanism involving an attenuation site located ∼1.7 kb downstream from the transcription start site. In breast cancer cells, ligand-bound ERα binds close to, and drives transcription beyond this attenuation site, allowing synthesis of complete transcripts. However, little is known, in general, about the factors involved in relieving transcriptional attenuation, or specifically how ERα coordinates such factors to promote transcriptional elongation. Using cyclin dependent kinase 9 (CDK9) inhibitors, reporter gene assays and measurements of total and intronic MYB transcription, we show that functionally active CDK9 is required for estrogen-dependent transcriptional elongation. We further show by ChIP and co-immunoprecipitation studies that the P-TEFb complex (CDK9/CyclinT1) is recruited to the attenuation region by ligand-bound ERα, resulting in increased RNA polymerase II Ser-2 phosphorylation. These data provide new insights into MYB regulation, and given the critical roles of MYB in tumorigenesis, suggest targeting MYB elongation as potential therapeutic strategy.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Genes myb , Fator B de Elongação Transcricional Positiva/metabolismo , Transcrição Gênica , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Estradiol/farmacologia , Estrogênios/farmacologia , Feminino , Células HEK293 , Humanos , Íntrons , Fosforilação , Fator B de Elongação Transcricional Positiva/antagonistas & inibidores , Proto-Oncogene Mas , RNA Polimerase II/metabolismo , Elementos Reguladores de Transcrição
5.
J Biol Chem ; 287(36): 30701-10, 2012 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-22730322

RESUMO

FAS-associated factor 1 (FAF1) antagonizes Wnt signaling by stimulating ß-catenin degradation. However, the molecular mechanism underlying this effect is unknown. Here, we demonstrate that the E3 ubiquitin ligase ß-transducin repeat-containing protein (ß-TrCP) is required for FAF1 to suppress Wnt signaling and that FAF1 specifically associates with the SCF (Skp1-Cul1-F-box protein)-ß-TrCP complex. Depletion of ß-TrCP reduced FAF1-mediated ß-catenin polyubiquitination and impaired FAF1 in antagonizing Wnt/ß-catenin signaling. FAF1 was shown to act as a scaffold for ß-catenin and ß-TrCP and thereby to potentiate ß-TrCP-mediated ß-catenin ubiquitination and degradation. Data mining revealed that FAF1 expression is statistically down-regulated in human breast carcinoma compared with normal breast tissue. Consistent with this, FAF1 expression is higher in epithelial-like MCF7 than mesenchymal-like MDA-MB-231 human breast cancer cells. Depletion of FAF1 in MCF7 cells resulted in increased ß-catenin accumulation and signaling. Importantly, FAF1 knockdown promoted a decrease in epithelial E-cadherin and an increase in mesenchymal vimentin expression, indicative for an epithelial to mesenchymal transition. Moreover, ectopic FAF1 expression reduces breast cancer cell migration in vitro and invasion/metastasis in vivo. Thus, our studies strengthen a tumor-suppressive function for FAF1.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Neoplasias da Mama/metabolismo , Proteólise , Proteínas Supressoras de Tumor/metabolismo , Ubiquitinação , Via de Sinalização Wnt , Proteínas Contendo Repetições de beta-Transducina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Reguladoras de Apoptose , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Caderinas/genética , Caderinas/metabolismo , Movimento Celular/genética , Proteínas Culina/genética , Proteínas Culina/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Células HEK293 , Células HeLa , Humanos , Proteínas Quinases Associadas a Fase S/genética , Proteínas Quinases Associadas a Fase S/metabolismo , Proteínas Supressoras de Tumor/genética , beta Catenina/genética , Proteínas Contendo Repetições de beta-Transducina/genética
6.
Breast Cancer Res ; 15(6): R106, 2013 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-24196484

RESUMO

INTRODUCTION: The transforming growth factor beta (TGF-ß) signalling pathway is known to control human breast cancer invasion and metastasis. We demonstrate that the zebrafish xenograft assay is a robust and dependable animal model for examining the role of pharmacological modulators and genetic perturbation of TGF-ß signalling in human breast tumour cells. METHODS: We injected cancer cells into the embryonic circulation (duct of cuvier) and examined their invasion and metastasis into the avascular collagenous tail. Various aspects of the TGF-ß signalling pathway were blocked by chemical inhibition, small interfering RNA (siRNA), or small hairpin RNA (shRNA). Analysis was conducted using fluorescent microscopy. RESULTS: Breast cancer cells with different levels of malignancy, according to in vitro and in vivo mouse studies, demonstrated invasive and metastatic properties within the embryonic zebrafish model that nicely correlated with their differential tumourigenicity in mouse models. Interestingly, MCF10A M2 and M4 cells invaded into the caudal hematopoietic tissue and were visible as a cluster of cells, whereas MDA MB 231 cells invaded into the tail fin and were visible as individual cells. Pharmacological inhibition with TGF-ß receptor kinase inhibitors or tumour specific Smad4 knockdown disturbed invasion and metastasis in the zebrafish xenograft model and closely mimicked the results we obtained with these cells in a mouse metastasis model. Inhibition of matrix metallo proteinases, which are induced by TGF-ß in breast cancer cells, blocked invasion and metastasis of breast cancer cells. CONCLUSIONS: The zebrafish-embryonic breast cancer xenograft model is applicable for the mechanistic understanding, screening and development of anti-TGF-ß drugs for the treatment of metastatic breast cancer in a timely and cost-effective manner.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Fator de Crescimento Transformador beta/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Peixe-Zebra/metabolismo , Animais , Benzamidas/farmacologia , Neoplasias da Mama/tratamento farmacológico , Cromonas/farmacologia , Dioxóis/farmacologia , Dipeptídeos/farmacologia , Modelos Animais de Doenças , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Embrião não Mamífero , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Inibidores de Metaloproteinases de Matriz/farmacologia , Morfolinas/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , RNA Interferente Pequeno/farmacologia , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
7.
Breast Cancer Res ; 15(6): R113, 2013 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-24283570

RESUMO

INTRODUCTION: Epithelial-to-mesenchymal transition (EMT) promotes cell migration and is important in metastasis. Cellular proliferation is often downregulated during EMT, and the reverse transition (MET) in metastases appears to be required for restoration of proliferation in secondary tumors. We studied the interplay between EMT and proliferation control by MYB in breast cancer cells. METHODS: MYB, ZEB1, and CDH1 expression levels were manipulated by lentiviral small-hairpin RNA (shRNA)-mediated knockdown/overexpression, and verified with Western blotting, immunocytochemistry, and qRT-PCR. Proliferation was assessed with bromodeoxyuridine pulse labeling and flow cytometry, and sulforhodamine B assays. EMT was induced with epidermal growth factor for 9 days or by exposure to hypoxia (1% oxygen) for up to 5 days, and assessed with qRT-PCR, cell morphology, and colony morphology. Protein expression in human breast cancers was assessed with immunohistochemistry. ZEB1-MYB promoter binding and repression were determined with Chromatin Immunoprecipitation Assay and a luciferase reporter assay, respectively. Student paired t tests, Mann-Whitney, and repeated measures two-way ANOVA tests determined statistical significance (P < 0.05). RESULTS: Parental PMC42-ET cells displayed higher expression of ZEB1 and lower expression of MYB than did the PMC42-LA epithelial variant. Knockdown of ZEB1 in PMC42-ET and MDA-MB-231 cells caused increased expression of MYB and a transition to a more epithelial phenotype, which in PMC42-ET cells was coupled with increased proliferation. Indeed, we observed an inverse relation between MYB and ZEB1 expression in two in vitro EMT cell models, in matched human breast tumors and lymph node metastases, and in human breast cancer cell lines. Knockdown of MYB in PMC42-LA cells (MYBsh-LA) led to morphologic changes and protein expression consistent with an EMT. ZEB1 expression was raised in MYBsh-LA cells and significantly repressed in MYB-overexpressing MDA-MB-231 cells, which also showed reduced random migration and a shift from mesenchymal to epithelial colony morphology in two dimensional monolayer cultures. Finally, we detected binding of ZEB1 to MYB promoter in PMC42-ET cells, and ZEB1 overexpression repressed MYB promoter activity. CONCLUSIONS: This work identifies ZEB1 as a transcriptional repressor of MYB and suggests a reciprocal MYB-ZEB1 repressive relation, providing a mechanism through which proliferation and the epithelial phenotype may be coordinately modulated in breast cancer cells.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Transição Epitelial-Mesenquimal/genética , Proteínas de Homeodomínio/genética , Proteínas Proto-Oncogênicas c-myb/genética , Fatores de Transcrição/genética , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-myb/metabolismo , RNA Interferente Pequeno , Células Tumorais Cultivadas , Homeobox 1 de Ligação a E-box em Dedo de Zinco
8.
Cancer Metastasis Rev ; 31(3-4): 553-68, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22714591

RESUMO

The transforming growth factor-ß (TGF-ß) system signals via protein kinase receptors and SMAD mediators to regulate a large number of biological processes. Alterations of the TGF-ß signalling pathway are implicated in human cancer. Prior to tumour initiation and early during progression, TGF-ß acts as a tumour suppressor; however, at later stages, it is often a tumour promoter. Knowledge about the mechanisms involved in TGF-ß signal transduction has allowed a better understanding of cancer progression, invasion, metastasis and epithelial-to-mesenchymal transition. Furthermore, several molecular targets with great potential in therapeutic interventions have been identified. This review discusses the TGF-ß signalling pathway, its involvement in cancer and current therapeutic approaches.


Assuntos
Metástase Neoplásica , Neoplasias/etiologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Progressão da Doença , Transição Epitelial-Mesenquimal , Humanos , Mutação , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Proteínas Smad/fisiologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Microambiente Tumoral
9.
Biochem Biophys Res Commun ; 435(1): 58-63, 2013 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-23618854

RESUMO

TGF-ß plays a dual role in cancer; in early stages it inhibits tumor growth, whereas later it promotes invasion and metastasis. TGF-ß is thought to be pro-invasive by inducing epithelial-to-mesenchymal transition (EMT) via induction of transcriptional repressors, including Slug and Snail. In this study, we investigated the role of Snail and Slug in TGF-ß-induced invasion in an in vitro invasion assay and in an embryonic zebrafish xenograft model. Ectopic expression of Slug or Snail promoted invasion of single, rounded amoeboid cells in vitro. In an embryonic zebrafish xenograft model, forced expression of Slug and Snail promoted single cell invasion and metastasis. Slug and Snail are sufficient for the induction of single-cell invasion in an in vitro invasion assay and in an embryonic zebrafish xenograft model.


Assuntos
Movimento Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta3/farmacologia , Animais , Western Blotting , Linhagem Celular , Movimento Celular/genética , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Embrião não Mamífero/patologia , Transição Epitelial-Mesenquimal/genética , Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Invasividade Neoplásica/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Peixe-Zebra/embriologia , Peixe-Zebra/genética
10.
J Mammary Gland Biol Neoplasia ; 16(2): 97-108, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21494783

RESUMO

The contribution of transforming growth factor ß (TGF-ß) signaling to breast cancer has been studied for more than two decades. In an early phase TGF-ß may act as a tumour suppressor, while later, when cells have become resistant to its anti-mitogenic effects, the role of TGF-ß switches towards malignant conversion and progression. TGF-ß stimulates cell invasion and modifies the microenvironment to the advantage of cancer cells. Studies have shown that TGF-ß promotes bone and lung metastasis via different mechanisms. The therapeutic strategies to target the TGF-ß pathway in breast cancer are becoming increasingly clear. This review will focus on the role TGF-ß in breast cancer invasion and metastasis.


Assuntos
Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Feminino , Humanos , Invasividade Neoplásica
11.
Breast Cancer Res ; 12(4): R55, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20659323

RESUMO

INTRODUCTION: MYB is highly expressed in estrogen receptor positive (ER + ve) breast tumours and tumour cell lines. We recently demonstrated that MYB is essential for the proliferation of ER + ve breast cancer cells, and have now investigated its role in mammary epithelial differentiation. METHODS: MCF-7 breast cancer cells were treated with sodium butyrate, vitamin E succinate or 12-O-tetradecanoylphorbol-13-acetate to induce differentiation as measured by Nile Red staining of lipid droplets and ß-casein expression. The non-tumorigenic murine mammary epithelial cell (MEC) line, HC11, was induced to differentiate with lactogenic hormones. MYB levels were manipulated by inducible lentiviral shRNA-mediated knockdown and retroviral overexpression. RESULTS: We found that MYB expression decreases following chemically-induced differentiation of the human breast cancer cell line MCF-7, and hormonally-induced differentiation of a non-tumorigenic murine mammary epithelial cell (MEC) line, HC11. We also found that shRNA-mediated MYB knockdown initiated differentiation of breast cancer cells, and greatly sensitised them to the differentiative and pro-apoptotic effects of differentiation-inducing agents (DIAs). Sensitisation to the pro-apoptotic effects DIAs is mediated by decreased expression of BCL2, which we show here is a direct MYB target in breast cancer cells. Conversely, enforced expression of MYB resulted in the cells remaining in an undifferentiated state, with concomitant suppression of apoptosis, in the presence of DIAs. CONCLUSIONS: Taken together, these data imply that MYB function is critical in regulating the balance between proliferation, differentiation, and apoptosis in MECs. Moreover, our findings suggest MYB may be a viable therapeutic target in breast cancer and suggest specific approaches for exploiting this possibility.


Assuntos
Apoptose/fisiologia , Diferenciação Celular/fisiologia , Proteínas Proto-Oncogênicas c-myb/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Butiratos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Sinergismo Farmacológico , Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Feminino , Fulvestranto , Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos , Microscopia de Fluorescência , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-myb/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Acetato de Tetradecanoilforbol/farmacologia , Vitamina E/farmacologia , Vitaminas/farmacologia
12.
Histol Histopathol ; 32(7): 673-686, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27933602

RESUMO

Advances in scientific techniques have provided researchers with exceptional new opportunities to identify and monitor changes between different cancer types, during different stages of progression, between individual tumor cells and in the surrounding stroma. The wealth of information that can be obtained from new scientific techniques places additional requirements on the conventional cancer models. New models that could be used to rapidly access the (potential) functional importance of newly identified (epi)genetic and proteomic changes and test the efficacy on emerging (combinatorial) therapies are desperately required. The distinctive characteristics of zebrafish are progressively being applied to create more relevant models of human diseases. Zebrafish embryos provide a powerful tool to develop functional cancer models. This is a tool that can be used from drug discovery and development to assessment of drug toxicity. This review will summarise the use of zebrafish xenograft models to study human cancers, and discuss the benefits and limitations of these models.


Assuntos
Ensaios Antitumorais Modelo de Xenoenxerto , Peixe-Zebra , Animais , Linhagem Celular Tumoral , Embrião não Mamífero , Humanos , Larva , Transplante de Neoplasias
13.
Nat Commun ; 8(1): 1537, 2017 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-29146913

RESUMO

Macroautophagy can regulate cell signalling and tumorigenesis via elusive molecular mechanisms. We establish a RAS mutant cancer cell model where the autophagy gene ATG5 is dispensable in A549 cells in vitro, yet promotes tumorigenesis in mice. ATG5 represses transcriptional activation by the TGFß-SMAD gene regulatory pathway. However, autophagy does not terminate cytosolic signal transduction by TGFß. Instead, we use proteomics to identify selective degradation of the signalling scaffold TRAF3. TRAF3 autophagy is driven by RAS and results in activation of the NF-κB family member RELB. We show that RELB represses TGFß target promoters independently of DNA binding at NF-κB recognition sequences, instead binding with SMAD family member(s) at SMAD-response elements. Thus, autophagy antagonises TGFß gene expression. Finally, autophagy-deficient A549 cells regain tumorigenicity upon SMAD4 knockdown. Thus, at least in this setting, a physiologic function for autophagic regulation of gene expression is tumour growth.


Assuntos
Autofagia/genética , Regulação Neoplásica da Expressão Gênica , Proteínas Smad/genética , Fator 3 Associado a Receptor de TNF/genética , Fator de Transcrição RelB/genética , Células A549 , Animais , Células Cultivadas , Feminino , Células HEK293 , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Proteínas Smad/metabolismo , Fator 3 Associado a Receptor de TNF/metabolismo , Fator de Transcrição RelB/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Transplante Heterólogo
14.
Methods Mol Biol ; 1213: 107-19, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25173378

RESUMO

Human pluripotent stem cells (hPSCs) are proving to be a valuable source of endothelial cells (ECs), pericytes, and vascular smooth muscle cells (vSMCs). Although an increasing number of phenotypic markers are becoming available to determine the phenotypes of these cells in vitro, the ability to integrate and form functional vessels in the host organism, typically mouse, remains critical for the assessment of EC functional competence. However, current mouse models require relatively large numbers of cells that might be difficult to derive simultaneously from multiple hPSCs lines. Therefore, there is an urgent need for new functional assays that are robust and can be performed with small numbers of cells. Here we describe a novel zebrafish xenograft model to test functionality of hPSC-derived ECs. The assay can be performed in 10 days and requires only ~100-400 human cells per embryo. Thus, the zebrafish xenograft model can be useful for the accurate and rapid assessment of functionality of hPSC-derived ECs in a lower vertebrate model that is widely viewed by regulatory authorities as a more acceptable alternative to adult mice.


Assuntos
Diferenciação Celular , Embrião não Mamífero/metabolismo , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/transplante , Animais , Técnicas de Cultura de Células , Xenoenxertos , Humanos , Peixe-Zebra
15.
Nat Protoc ; 9(6): 1514-31, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24874816

RESUMO

Human endothelial cells (ECs) and pericytes are of great interest for research on vascular development and disease, as well as for future therapy. This protocol describes the efficient generation of ECs and pericytes from human pluripotent stem cells (hPSCs) under defined conditions. Essential steps for hPSC culture, differentiation, isolation and functional characterization of ECs and pericytes are described. Substantial numbers of both cell types can be derived in only 2-3 weeks: this involves differentiation (10 d), isolation (1 d) and 4 or 10 d of expansion of ECs and pericytes, respectively. We also describe two assays for functional evaluation of hPSC-derived ECs: (i) primary vascular plexus formation upon coculture with hPSC-derived pericytes and (ii) incorporation in the vasculature of zebrafish xenografts in vivo. These assays can be used to test the quality and drug sensitivity of hPSC-derived ECs and model vascular diseases with patient-derived hPSCs.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Células Endoteliais/citologia , Pericitos/citologia , Células-Tronco Pluripotentes/citologia , Animais , Proliferação de Células , Células Endoteliais/fisiologia , Xenoenxertos/irrigação sanguínea , Xenoenxertos/citologia , Humanos , Pericitos/fisiologia , Células-Tronco Pluripotentes/fisiologia , Peixe-Zebra
16.
Nat Commun ; 5: 3388, 2014 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-24584437

RESUMO

In advanced cancers, the TGF-ß pathway acts as an oncogenic factor and is considered to be a therapeutic target. Here using a genome-wide cDNA screen, we identify nuclear receptor NR4A1 as a strong activator of TGF-ß signalling. NR4A1 promotes TGF-ß/SMAD signalling by facilitating AXIN2-RNF12/ARKADIA-induced SMAD7 degradation. NR4A1 interacts with SMAD7 and AXIN2, and potently and directly induces AXIN2 expression. Whereas loss of NR4A1 inhibits TGF-ß-induced epithelial-to-mesenchymal transition and metastasis, slight NR4A1 ectopic expression stimulates metastasis in a TGF-ß-dependent manner. Importantly, inflammatory cytokines potently induce NR4A1 expression, and potentiate TGF-ß-mediated breast cancer cell migration, invasion and metastasis in vitro and in vivo. Notably, NR4A1 expression is elevated in breast cancer patients with high immune infiltration and its expression weakly correlates with phosphorylated SMAD2 levels, and is an indicator of poor prognosis. Our results uncover inflammation-induced NR4A1 as an important determinant for hyperactivation of pro-oncogenic TGF-ß signalling in breast cancer.


Assuntos
Neoplasias Mamárias Animais/metabolismo , Metástase Neoplásica/fisiopatologia , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Células Cultivadas , Imunoprecipitação da Cromatina , Feminino , Imuno-Histoquímica , Neoplasias Mamárias Animais/genética , Camundongos , Camundongos Knockout , Metástase Neoplásica/genética , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Smad3/genética , Proteína Smad3/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Fator de Crescimento Transformador beta/genética , Ubiquitinação/genética , Ubiquitinação/fisiologia , Peixe-Zebra
17.
Nat Cell Biol ; 14(7): 717-26, 2012 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-22706160

RESUMO

The stability and membrane localization of the transforming growth factor-ß (TGF-ß) type I receptor (TßRI) determines the levels of TGF-ß signalling. TßRI is targeted for ubiquitylation-mediated degradation by the SMAD7-SMURF2 complex. Here we performed a genome-wide gain-of-function screen and identified ubiquitin-specific protease (USP) 4 as a strong inducer of TGF-ß signalling. USP4 was found to directly interact with TßRI and act as a deubiquitylating enzyme, thereby controlling TßRI levels at the plasma membrane. Depletion of USP4 mitigates TGF-ß-induced epithelial to mesenchymal transition and metastasis. Importantly, AKT (also known as protein kinase B), which has been associated with poor prognosis in breast cancer, directly associates with and phosphorylates USP4. AKT-mediated phosphorylation relocates nuclear USP4 to the cytoplasm and membrane and is required for maintaining its protein stability. Moreover, AKT-induced breast cancer cell migration was inhibited by USP4 depletion and TßRI kinase inhibition. Our results uncover USP4 as an important determinant for crosstalk between TGF-ß and AKT signalling pathways.


Assuntos
Neoplasias da Mama/enzimologia , Membrana Celular/metabolismo , Proteínas Oncogênicas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Ubiquitina Tiolesterase/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Movimento Celular , Estabilidade Enzimática , Transição Epitelial-Mesenquimal , Feminino , Regulação da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Camundongos , Camundongos Knockout , Mutação , Invasividade Neoplásica , Proteínas Oncogênicas/deficiência , Proteínas Oncogênicas/genética , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Transporte Proteico , Proteínas Proto-Oncogênicas , Interferência de RNA , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Receptores de Fatores de Crescimento Transformadores beta/genética , Transdução de Sinais , Fatores de Tempo , Transfecção , Fator de Crescimento Transformador beta/metabolismo , Ubiquitina Tiolesterase/genética , Proteases Específicas de Ubiquitina , Ubiquitinação , Peixe-Zebra/embriologia
18.
Cancer Res ; 71(22): 7029-37, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21948968

RESUMO

MYB oncogene upregulation is associated with estrogen receptor (ER)-positive breast cancer, but disease requirements for MYB function in vivo have not been explored. In this study, we provide evidence of a critical requirement for MYB functions in models of human and murine breast cancer. In human breast cancer, we found that MYB expression was critical for tumor cell growth both in vitro and in vivo in xenograft settings. In transgenic knockout mice, tissue-specific deletion of the murine MYB gene caused a transient defect in mammary gland development that was reflected in delayed ductal branching and defective apical bud formation. In mouse mammary tumor virus (MMTV)-NEU mice where tumors are initiated by activation of HER2, MYB deletion was sufficient to abolish tumor formation. In the more aggressive MMTV-PyMT model system, MYB deletion delayed tumorigenesis significantly. Together, the findings in these transgenic knockout models implied that MYB was critical during an early window in mammary development when it was essential for tumor initiation, even though MYB loss did not exert a lasting impact upon normal mammary function. Two important MYB-target genes that promote cell survival, BCL2 and GRP78/BIP, were each elevated compared with nontransformed mammary epithelial cells, thereby promoting survival as confirmed in colony formation assays in vitro. Taken together, our findings establish a role for MYB at the hub of ER- and HER2-dependent pathways in mammary carcinogenesis.


Assuntos
Neoplasias Mamárias Experimentais/etiologia , Proteínas Oncogênicas v-myb/fisiologia , Animais , Antígenos Virais de Tumores/genética , Linhagem Celular Tumoral , Chaperona BiP do Retículo Endoplasmático , Receptor alfa de Estrogênio/análise , Etilnitrosoureia , Feminino , Neoplasias Mamárias Experimentais/química , Vírus do Tumor Mamário do Camundongo/genética , Camundongos , Receptor ErbB-2/análise
19.
Hum Gene Ther ; 21(8): 1005-17, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20615123

RESUMO

Manipulation of gene expression is an invaluable tool to study gene function in vitro and in vivo. The application of small inhibitory RNAs to knock down gene expression provides a relatively simple, elegant, but transient approach to study gene function in many cell types as well as in whole animals. Short hairpin structures (shRNAs) are a logical advance as they can be expressed continuously and are hence suitable for stable gene knockdown. Drug-inducible systems have now been developed; however, application of the technology has been hampered by persistent problems with low or transient expression, leakiness or poor inducibility of the short hairpin, and lack of reversibility. We have developed a robust, versatile, single lentiviral vector tool that delivers tightly regulated, fully reversible, doxycycline-responsive knockdown of target genes (FOXP3 and MYB), using single short hairpin RNAs. To demonstrate the capabilities of the vector we targeted FOXP3 because it plays a critical role in the development and function of regulatory T cells. We also targeted MYB because of its essential role in hematopoiesis and implication in breast cancer progression. The versatility of this vector is hence demonstrated by knockdown of distinct genes in two biologically separate systems.


Assuntos
Técnicas de Silenciamento de Genes/métodos , Vetores Genéticos , Lentivirus/genética , RNA Interferente Pequeno/metabolismo , Animais , Doxiciclina/metabolismo , Fatores de Transcrição Forkhead/genética , Expressão Gênica , Marcação de Genes , Células HEK293 , Humanos , Lentivirus/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-myb/genética , RNA Interferente Pequeno/genética , Transfecção
20.
Proc Natl Acad Sci U S A ; 104(34): 13762-7, 2007 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-17690249

RESUMO

MYB (the human ortholog of c-myb) is expressed in a high proportion of human breast tumors, and that expression correlates strongly with estrogen receptor (ER) positivity. This may reflect the fact that MYB is a target of estrogen/ER signaling. Because in many cases MYB expression appears to be regulated by transcriptional attenuation or pausing in the first intron, we first investigated whether this mechanism was involved in estrogen/ER modulation of MYB. We found that this was the case and that estrogen acted directly to relieve attenuation due to sequences within the first intron, specifically, a region potentially capable of forming a stem-loop structure in the transcript and an adjacent poly(dT) tract. Secondly, given the involvement of MYB in hematopoietic and colon tumors, we also asked whether MYB was required for the proliferation of breast cancer cells. We found that proliferation of ER(+) but not ER(-) breast cancer cell lines was inhibited when MYB expression was suppressed by using either antisense oligonucleotides or RNA interference. Our results show that MYB is an effector of estrogen/ER signaling and provide demonstration of a functional role of MYB in breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Estrogênios/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myb/metabolismo , Receptores de Estrogênio/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Genes Reporter/genética , Humanos , Proteínas Proto-Oncogênicas c-myb/genética , Interferência de RNA , RNA Mensageiro/genética , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA