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(46): 5760-70, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26387539

ABSTRACT

The canonical Wnt pathway (TCF4/ß-catenin) has important roles during normal differentiation and in disease. Some Wnt functions depend on signaling gradients requiring the pathway to be tightly regulated. A key Wnt target is the transcription factor ZEB1 whose expression by cancer cells promotes tumor invasiveness by repressing the expression of epithelial specification markers and activating mesenchymal genes, including a number of Wnt targets such as LAMC2 and uPA. The ability of ZEB1 to activate/repress its target genes depends on its recruitment of corepressors (CtBP, BRG1) or coactivators (p300) although conditions under which ZEB1 binds these cofactors are not elucidated. Here, we show that TCF4 and ZEB1 reciprocally modulate each other's transcriptional activity: ZEB1 enhances TCF4/ß-catenin-mediated transcription and, in turn, Wnt signaling switches ZEB1 from a repressor into an activator. In colorectal cancer (CRC) cells with active Wnt signaling, ZEB1 enhances transcriptional activation of LAMC2 and uPA by TCF4/ß-catenin. However, in CRC cells with inactive Wnt, ZEB1 represses both genes. Reciprocal modulation of ZEB1 and TCF4 activities involves their binding to DNA and mutual interaction. Wnt signaling turns ZEB1 into an activator by replacing binding of CtBP/BRG1 in favor of p300. Using a mouse model of Wnt-induced intestinal tumorigenesis, we found that downregulation of ZEB1 reduces the expression of LAMC2 in vivo. These results identify a mechanism through which Wnt and ZEB1 transcriptional activities are modulated, offering new approaches in cancer therapy.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Wnt Signaling Pathway , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Line, Tumor , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Homeodomain Proteins/genetics , Humans , Mice , Mice, Transgenic , Promoter Regions, Genetic , Transcription Factor 4 , Transcription Factors/genetics , Transcription, Genetic , Zinc Finger E-box-Binding Homeobox 1 , beta Catenin/metabolism
2.
Cell Death Differ ; 21(2): 247-57, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24013721

ABSTRACT

Mantle cell lymphoma (MCL) is a B-cell malignancy characterized by a poor response to treatment and prognosis. Constitutive activation of different signaling pathways in subsets of MCLs, through genetic and/or nongenetic alterations, endows tumor cells with enhanced proliferation and reduced apoptosis. The canonical Wnt pathway (ß-catenin/TCF-LEF), implicated in the pathogenesis of numerous cancers, is constitutively active in half of MCLs. Here, we show that ZEB1, a transcription factor better known for promoting metastasis in carcinomas, is expressed in primary MCLs with active Wnt signaling. ZEB1 expression in MCL cells depends on Wnt, being downregulated by ß-catenin knockdown or blocking of Wnt signaling by salinomycin. Knockdown of ZEB1 reduces in vitro cell viability and proliferation in MCL cells, and, importantly, tumor growth in mouse xenograft models. ZEB1 activates proliferation-associated (HMGB2, UHRF1, CENPF, MYC, MKI67, and CCND1) and anti-apoptotic (MCL1, BCL2, and BIRC5) genes and inhibits pro-apoptotic ones (TP53, BBC3, PMAIP1, and BAX). We show that ZEB1 expression in MCL cells determines differential resistance to chemotherapy drugs and regulates transporters involved in drug influx/efflux. Downregulation of ZEB1 by salinomycin increases the sensitivity of MCL cells to the cytotoxic effect of doxorubicin, cytarabine and gemcitabine. Lastly, salinomycin and doxorubicin display a synergistic effect in established and primary MCL cells. These results identify ZEB1 in MCL where it promotes cell proliferation, enhanced tumor growth and a differential response to chemotherapy drugs. ZEB1 could thus potentially become a predictive biomarker and therapeutic target in this lymphoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Epithelial-Mesenchymal Transition/drug effects , Homeodomain Proteins/metabolism , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/pathology , Transcription Factors/metabolism , Animals , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , HEK293 Cells , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , Lymphoma, Mantle-Cell/metabolism , Mice , Pyrans/pharmacology , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Zinc Finger E-box-Binding Homeobox 1
3.
Oncogene ; 29(41): 5579-90, 2010 Oct 14.
Article in English | MEDLINE | ID: mdl-20676136

ABSTRACT

E2F1-3 proteins appear to have distinct roles in progenitor cells and in differentiating cells undergoing cell cycle exit. However, the function of these proteins in paradigms of terminal differentiation that involve continued cell division has not been examined. Using compound E2F1/E2F2-deficient mice, we have examined the effects of E2F1 and E2F2 loss on the differentiation and simultaneous proliferation of bone-marrow-derived cells toward the macrophage lineage. We show that E2F1/E2F2 deficiency results in accelerated DNA replication and cellular division during the initial cell division cycles of bone-marrow-derived cells, arguing that E2F1/E2F2 are required to restrain proliferation of pro-monocyte progenitors during their differentiation into macrophages, without promoting their cell cycle exit. Accelerated proliferation is accompanied by early expression of DNA replication and cell cycle regulators. Remarkably, rapid proliferation of E2F1/E2F2 compound mutant cultures is temporally followed by induction of a DNA damage response and the implementation of a p21(CIP1)-dependent senescence. We further show that differentiating E2F1/E2F2-knockout macrophages do not trigger a DNA damage response pathway in the absence of DNA replication. These findings underscore the relevance of E2F1 and E2F2 as suppressors of hematopoietic progenitor expansion. Our data indicate that their absence in differentiating macrophages initiates a senescence program that results from enforcement of a DNA damage response triggered by DNA hyper-replication.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , DNA Replication , E2F1 Transcription Factor/metabolism , E2F2 Transcription Factor/metabolism , Macrophages/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Cycle , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p21/genetics , E2F1 Transcription Factor/genetics , E2F2 Transcription Factor/genetics , Flow Cytometry , Immunoblotting , Macrophages/cytology , Mice , Mice, Knockout , Time Factors
4.
Oncogene ; 29(24): 3490-500, 2010 Jun 17.
Article in English | MEDLINE | ID: mdl-20418909

ABSTRACT

Loss of E-cadherin is a key initial step in the transdifferentiation of epithelial cells to a mesenchymal phenotype, which occurs when tumor epithelial cells invade into surrounding tissues. Expression of the nuclear factor ZEB1 induces an epithelial-to-mesenchymal transition and confers a metastatic phenotype on carcinomas by repressing the E-cadherin gene at the transcriptional level. In this study, we show that ZEB1 interacts with the SWI/SNF chromatin-remodeling protein BRG1 to regulate E-cadherin independently of CtBP, its traditional co-repressor. Blocking the interaction between ZEB1 and BRG1 induces expression of E-cadherin and downregulation of the mesenchymal marker vimentin. ZEB1 and BRG1 colocalize in E-cadherin-negative cells from cancer lines and in the stroma of normal colon. Colocalization of ZEB1 and BRG1 in epithelial cells is only found in those de-differentiated cells characterized by nuclear beta-catenin staining at the invasive edge of the tumor. Our results identify ZEB1/BRG1 as a new transcriptional mechanism regulating E-cadherin expression and epithelial-to-mesenchymal transdifferentiation that may be involved during the initial stages of tumor invasion.


Subject(s)
Cadherins/antagonists & inhibitors , DNA Helicases/metabolism , Epithelial Cells/cytology , Homeodomain Proteins/metabolism , Mesoderm/cytology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Dedifferentiation , Cell Line, Tumor , Colon/cytology , Colon/metabolism , Colon/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Organ Specificity , Promoter Regions, Genetic/genetics , Protein Transport , Transcription, Genetic , Zinc Finger E-box-Binding Homeobox 1
SELECTION OF CITATIONS
SEARCH DETAIL
...