Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Oncogene ; 34(18): 2325-36, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-24954505

ABSTRACT

Disease progression and recurrence are major barriers to survival for breast cancer patients. Understanding the etiology of recurrent or metastatic breast cancer and underlying mechanisms is critical for the development of new treatments and improved survival. Here, we report that two commonly overexpressed breast cancer oncogenes, Ron (Recepteur d'Origine Nantaise) and DEK, cooperate to promote advanced disease through multipronged effects on ß-catenin signaling. The Ron receptor is commonly activated in breast cancers, and Ron overexpression in human disease stimulates ß-catenin nuclear translocation and is an independent predictor of metastatic dissemination. Dek is a chromatin-associated oncogene whose expression has been linked to cancer through multiple mechanisms, including ß-catenin activity. We demonstrate here that Dek is a downstream target of Ron receptor activation in murine and human models. The absence of Dek in the MMTV-Ron mouse model led to a significant delay in tumor development, characterized by decreased cell proliferation, diminished metastasis and fewer cells expressing mammary cancer stem cell markers. Dek complementation of cell lines established from this model was sufficient to promote cellular growth and invasion. Mechanistically, Dek expression stimulated the production and secretion of Wnt ligands to sustain an autocrine/paracrine canonical ß-catenin signaling loop. Finally, we show that Dek overexpression promotes tumorigenic phenotypes in immortalized human mammary epithelial MCF10A cells and, in the context of Ron receptor activation, correlates with disease recurrence and metastasis in patients. Overall, our studies demonstrate that DEK overexpression, due in part to Ron receptor activation, drives breast cancer progression through the induction of Wnt/ß-catenin signaling.


Subject(s)
Breast Neoplasms/pathology , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Wnt Signaling Pathway , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasms, Experimental , Poly-ADP-Ribose Binding Proteins
2.
Oncogene ; 30(50): 4990-8, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-21625214

ABSTRACT

The Ron receptor tyrosine kinase (TK) is overexpressed in many cancers, including prostate cancer. To examine the significance of Ron in prostate cancer in vivo, we utilized a genetically engineered mouse model, referred to as TRAMP mice, that is predisposed to develop prostate tumors. In this model, we show that prostate tumors from 30-week-old TRAMP mice have increased Ron expression compared with age-matched wild-type prostates. Based on the upregulation of Ron in human prostate cancers and in this murine model of prostate tumorigenesis, we hypothesized that this receptor has a functional role in the development of prostate tumors. To test this hypothesis, we crossed TRAMP mice with mice that are deficient in Ron signaling (TK-/-). Interestingly, TK-/- TRAMP+ mice show a significant decrease in prostate tumor mass relative to TRAMP mice containing functional Ron. Moreover, TK-/- TRAMP+ prostate tumors exhibited decreased tumor vascularization relative to TK+/+ TRAMP+ prostate tumors, which correlated with reduced levels of the angiogenic molecules vascular endothelial growth factor and CXCL2. Although Ron loss did not alter tumor cell proliferation, a significant decrease in cell survival was observed. Similarly, murine prostate cancer cell lines containing a Ron deficiency exhibited decreased levels of active nuclear factor-κB, suggesting that Ron may be important in regulating prostate cell survival at least partly through this pathway. In total, our data show for the first time that Ron promotes prostate tumor growth, prostate tumor angiogenesis and prostate cancer cell survival in vivo.


Subject(s)
Cell Proliferation , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/metabolism , Prostatic Neoplasms/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Animals , Cell Survival/genetics , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , Crosses, Genetic , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Proteins/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Tumor Necrosis Factor, Member 25/genetics
3.
Oncogene ; 30(34): 3694-704, 2011 Aug 25.
Article in English | MEDLINE | ID: mdl-21423209

ABSTRACT

Our previous studies demonstrated that selective overexpression of the Ron receptor tyrosine kinase in the murine mammary epithelium leads to mammary tumor formation. Biochemical analysis of mammary tumor lysates showed that Ron overexpression was associated with increases in ß-catenin expression and tyrosine phosphorylation. ß-Catenin has also been shown to be regulated through tyrosine phosphorylation by the receptor tyrosine kinases Met, Fer and Fyn. However, the molecular and physiological roles of ß-catenin and ß-catenin tyrosine phosphorylation downstream of Ron are not known. To investigate this association, we show that Ron and ß-catenin are coordinately elevated in human breast cancers. Our data also demonstrate that activation of Ron, through ligand binding by hepatocyte growth factor-like protein (HGFL), induces the tyrosine phosphorylation of ß-catenin, primarily on tyrosine residues Tyr 654 and Tyr 670. In addition, HGFL-mediated Ron activation induces both ß-catenin nuclear localization and transcriptional activity, with Tyr 654 and Tyr 670 residues of ß-catenin being critical for these processes. We also demonstrate that a knockdown of Ron in breast cancer cell lines leads to a loss of HGFL-induced ß-catenin-dependent transcriptional activation and cell growth, which can be rescued by activation of canonical Wnt/ß-catenin signaling. Moreover, we show that HGFL-dependent Ron activation mediates upregulation of the ß-catenin target genes cyclin D1 and c-myc, and that expression of these target genes in breast cancer cells is decreased following inhibition of Ron and/or ß-catenin. Finally, we show that genetic ablation of ß-catenin in Ron-expressing breast cancer cells decreases cellular proliferation in vitro, as well as mammary tumor growth and metastasis, following orthotopic transplantation into the mammary fat pad. Together, our data suggest that ß-catenin is a crucial downstream regulator of Ron receptor activation and is an important mediator of mammary tumorigenesis.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , Receptor Protein-Tyrosine Kinases/physiology , beta Catenin/metabolism , Animals , Base Sequence , Cell Proliferation , DNA Primers , Immunohistochemistry , Mammary Neoplasms, Experimental/pathology , Mice , Phosphorylation , Polymerase Chain Reaction , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/metabolism , Tyrosine/metabolism
4.
Oncogene ; 30(24): 2741-52, 2011 Jun 16.
Article in English | MEDLINE | ID: mdl-21317931

ABSTRACT

Breast cancer is a major cause of cancer-related deaths in American women; therefore, the identification of novel breast cancer-related molecules for the discovery of new markers and drug targets remains essential. The human DEK gene, which encodes a chromatin-binding protein and DNA topology regulator, is upregulated in many types of cancer. DEK has been implicated as an oncogene in breast cancer based on mRNA expression studies, but its functional significance in breast cancer growth and progression has not yet been tested directly. We demonstrate that DEK is highly expressed in breast cancer cells compared with normal tissue, and functionally important for cellular growth, invasion and mammosphere formation. DEK overexpression in non-tumorigenic MCF10A cells resulted in increased growth and motility, with a concomitant downregulation of E-cadherin. Conversely, DEK knockdown in MCF7 and MDA-MB-468 breast cancer cells resulted in decreased growth and motility with upregulation of E-cadherin. The use of DEK-proficient and -deficient breast cancer cells in orthotopic xenografts provided further in vivo evidence that DEK contributes to tumor growth. Activation of the ß-catenin signaling pathway is important for normal and cancer stem cell character, growth and metastasis. We show that DEK expression stimulated, and DEK knockdown repressed ß-catenin nuclear translocation and activity. Importantly, the expression of constitutively active ß-catenin rescued breast cancer invasion defects of DEK knockdown cells. Together, our data indicate that DEK expression stimulates the growth, stem cell character and motility of breast cancer cells, and that DEK-dependent cellular invasion occurs at least in part via ß-catenin activation.


Subject(s)
Breast Neoplasms/pathology , Chromosomal Proteins, Non-Histone/genetics , Neoplastic Stem Cells/pathology , Oncogene Proteins/genetics , Proto-Oncogenes , Signal Transduction/physiology , beta Catenin/physiology , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Chromosomal Proteins, Non-Histone/physiology , Female , Humans , Mice , Neoplasm Invasiveness , Oncogene Proteins/physiology , Poly-ADP-Ribose Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...