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.
Epigenetics Chromatin ; 11(1): 70, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30445998

ABSTRACT

BACKGROUND: Epithelial mesenchymal transition (EMT) is tightly regulated by a network of transcription factors (EMT-TFs). Among them is the nuclear factor ZEB2, a member of the zinc-finger E-box binding homeobox family. ZEB2 nuclear localization has been identified in several cancer types, and its overexpression is correlated with the malignant progression. ZEB2 transcriptionally represses epithelial genes, such as E-cadherin (CDH1), by directly binding to the promoter of the genes it regulates and activating mesenchymal genes by a mechanism in which there is no full agreement. Recent studies showed that EMT-TFs interact with epigenetic regulatory enzymes that alter the epigenome, thereby providing another level of control. The role of epigenetic regulation on ZEB2 function is not well understood. In this study, we aimed to characterize the epigenetic effect of ZEB2 repressive function on the regulation of a small Rab GTPase RAB25. RESULTS: Using cellular models with conditional ZEB2 expression, we show a clear transcriptional repression of RAB25 and CDH1. RAB25 contributes to the partial suppression of ZEB2-mediated cell migration. Furthermore, a highly significant reverse correlation between RAB25 and ZEB2 expression in several human cancer types could be identified. Mechanistically, ZEB2 binds specifically to E-box sequences on the RAB25 promoter. ZEB2 binding is associated with the local increase in DNA methylation requiring DNA methyltransferases as well as histone deacetylation (H3K9Ac) depending on the activity of SIRT1. Surprisingly, SIRT1 and DNMTs did not interact directly with ZEB2, and while SIRT1 inhibition decreased the stability of long-term repression, it did not prevent down-regulation of RAB25 and CDH1 by ZEB2. CONCLUSIONS: ZEB2 expression is resulting in drastic changes at the chromatin level with both clear DNA hypermethylation and histone modifications. Here, we revealed that SIRT1-mediated H3K9 deacetylation helps to maintain gene repression but is not required for the direct ZEB2 repressive function. Targeting epigenetic enzymes to prevent EMT is an appealing approach to limit cancer dissemination, but inhibiting SIRT1 activity alone might have limited effect and will require drug combination to efficiently prevent EMT.


Subject(s)
Epigenesis, Genetic , Epithelial-Mesenchymal Transition/physiology , Sirtuin 1/metabolism , Zinc Finger E-box Binding Homeobox 2/metabolism , rab GTP-Binding Proteins/genetics , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , DNA Methylation , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/metabolism , Gene Expression Regulation, Neoplastic , Humans , Promoter Regions, Genetic , Sirtuin 1/genetics , Zinc Finger E-box Binding Homeobox 2/genetics , rab GTP-Binding Proteins/metabolism
2.
Mol Biol Cell ; 18(11): 4615-24, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17855508

ABSTRACT

Zinc finger transcription factors of the Snail/Slug and ZEB-1/SIP1 families control epithelial-mesenchymal transitions in development in cancer. Here, we studied SIP1-regulated mesenchymal conversion of epidermoid A431 cells. We found that concomitant with inducing invasive phenotype, SIP1 inhibited expression of cyclin D1 and induced hypophosphorylation of the Rb tumor suppressor protein. Repression of cyclin D1 was caused by direct binding of SIP1 to three sequence elements in the cyclin D1 gene promoter. By expressing exogenous cyclin D1 in A431/SIP1 cells and using RNA interference, we demonstrated that the repression of cyclin D1 gene by SIP1 was necessary and sufficient for Rb hypophosphorylation and accumulation of cells in G1 phase. A431 cells expressing SIP1 along with exogenous cyclin D1 were highly invasive, indicating that SIP1-regulated invasion is independent of attenuation of G1/S progression. However, in another epithelial-mesenchymal transition model, gradual mesenchymal conversion of A431 cells induced by a dominant negative mutant of E-cadherin produced no effect on the cell cycle. We suggest that impaired G1/S phase progression is a general feature of cells that have undergone EMT induced by transcription factors of the Snail/Slug and ZEB-1/SIP1 families.


Subject(s)
Cell Cycle , Cyclin D1/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cyclin D1/genetics , Down-Regulation , Humans , Mutation/genetics , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic/genetics , RNA-Binding Proteins/genetics , Transcription, Genetic/genetics
3.
Mol Cell Biol ; 27(1): 157-69, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17060462

ABSTRACT

The product of the von Hippel-Lindau gene (VHL) acts as the substrate-recognition component of an E3 ubiquitin ligase complex that ubiquitylates the catalytic alpha subunit of hypoxia-inducible factor (HIF) for oxygen-dependent destruction. Although emerging evidence supports the notion that deregulated accumulation of HIF upon the loss of VHL is crucial for the development of clear-cell renal cell carcinoma (CC-RCC), the molecular events downstream of HIF governing renal oncogenesis remain unclear. Here, we show that the expression of a homophilic adhesion molecule, E-cadherin, a major constituent of epithelial cell junctions whose loss is associated with the progression of epithelial cancers, is significantly down-regulated in primary CC-RCC and CC-RCC cell lines devoid of VHL. Reintroduction of wild-type VHL in CC-RCC (VHL(-/-)) cells markedly reduced the expression of E2 box-dependent E-cadherin-specific transcriptional repressors Snail and SIP1 and concomitantly restored E-cadherin expression. RNA interference-mediated knockdown of HIFalpha in CC-RCC (VHL(-/-)) cells likewise increased E-cadherin expression, while functional hypoxia or expression of VHL mutants incapable of promoting HIFalpha degradation attenuated E-cadherin expression, correlating with the disengagement of RNA polymerase II from the endogenous E-cadherin promoter/gene. These findings reveal a critical HIF-dependent molecular pathway connecting VHL, an established "gatekeeper" of the renal epithelium, with a major epithelial tumor suppressor, E-cadherin.


Subject(s)
Cadherins/biosynthesis , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Nerve Tissue Proteins/physiology , RNA-Binding Proteins/physiology , Transcription Factors/physiology , Von Hippel-Lindau Tumor Suppressor Protein/physiology , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Epithelial Cells/metabolism , Humans , Kidney/metabolism , Promoter Regions, Genetic , RNA Interference , RNA Polymerase II/metabolism , RNA, Small Interfering/metabolism , Snail Family Transcription Factors , Subcellular Fractions/metabolism
4.
Nucleic Acids Res ; 33(20): 6566-78, 2005.
Article in English | MEDLINE | ID: mdl-16314317

ABSTRACT

SIP1/ZEB2 is a member of the deltaEF-1 family of two-handed zinc finger nuclear factors. The expression of these transcription factors is associated with epithelial mesenchymal transitions (EMT) during development. SIP1 is also expressed in some breast cancer cell lines and was detected in intestinal gastric carcinomas, where its expression is inversely correlated with that of E-cadherin. Here, we show that expression of SIP1 in human epithelial cells results in a clear morphological change from an epithelial to a mesenchymal phenotype. Induction of this epithelial dedifferentiation was accompanied by repression of several cell junctional proteins, with concomitant repression of their mRNA levels. Besides E-cadherin, other genes coding for crucial proteins of tight junctions, desmosomes and gap junctions were found to be transcriptionally regulated by the transcriptional repressor SIP1. Moreover, study of the promoter regions of selected genes by luciferase reporter assays and chromatin immunoprecipitation shows that repression is directly mediated by SIP1. These data indicate that, during epithelial dedifferentiation, SIP1 represses in a coordinated manner the transcription of genes coding for junctional proteins contributing to the dedifferentiated state; this repression occurs by a general mechanism mediated by Smad Interacting Protein 1 (SIP1)-binding sites.


Subject(s)
Epithelial Cells/metabolism , Homeodomain Proteins/physiology , Intercellular Junctions/metabolism , Membrane Proteins/genetics , Repressor Proteins/physiology , Binding Sites , Cadherins/genetics , Cadherins/metabolism , Cell Line , Cell Line, Tumor , Chromatin/metabolism , Connexins/genetics , Connexins/metabolism , Down-Regulation , Epithelial Cells/cytology , Humans , Membrane Proteins/metabolism , Mesoderm/cytology , Mutation , Promoter Regions, Genetic , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...