801
|
Alves CC, Rosivatz E, Schott C, Hollweck R, Becker I, Sarbia M, Carneiro F, Becker KF. Slug is overexpressed in gastric carcinomas and may act synergistically with SIP1 and Snail in the down-regulation of E-cadherin. J Pathol 2007; 211:507-515. [PMID: 17299729 DOI: 10.1002/path.2138] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Epithelial-mesenchymal transition (EMT) involving down-regulation of E-cadherin is known to play an important role in tumour progression. The aim of our study was to investigate the mRNA expression of two EMT regulators-Slug and E12/E47-in primary human gastric carcinomas and to compare this with the expression of E-cadherin and other EMT regulators (Snail, Twist, and SIP1). We studied a series of 59 gastric carcinomas by real-time quantitative RT-PCR in formalin-fixed and paraffin-embedded tissues. Thirty-four cases (58%) showed Slug up-regulation in the tumour; reduced or negative expression of E-cadherin was present in 24 of these (71%, p<0.0001). Twenty-one cases (36%) showed E12/E47 up-regulation that was not significantly associated with E-cadherin down-regulation (p=0.5734). Slug up-regulation accompanied by E-cadherin down-regulation correlated with the presence of distant metastases (p=0.0029) and with advanced pTNM stages (p=0.0424). A statistically significant association was found between Slug up-regulation and the expression of SIP1 in intestinal (p=0.0014) and Snail in diffuse (p=0.0067) carcinomas. We present the first study integrating the analysis of several EMT regulators in primary gastric carcinomas and conclude that Slug up-regulation is associated with E-cadherin down-regulation in diffuse and intestinal-type gastric carcinoma, and that this effect could be complemented by the presence of other EMT regulators.
Collapse
Affiliation(s)
- C Castro Alves
- Institut für Pathologie, Technische Universität München, Trogerstrasse 18, D-81765 Munich, Germany
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Rua Dr Roberto Frias, S/N 4200-465 Porto, Portugal
| | - E Rosivatz
- Institut für Pathologie, Technische Universität München, Trogerstrasse 18, D-81765 Munich, Germany
- Division of Cell and Molecular Biology, Imperial College London, Exhibition Road, London SW7 2AZ, UK
| | - C Schott
- Institut für Pathologie, Technische Universität München, Trogerstrasse 18, D-81765 Munich, Germany
| | - R Hollweck
- Institut für Medizinische Statistik und Epidemiologie, Technische Universität München, Ismaningerstrasse 22, D-81675 Munich, Germany
| | - I Becker
- Institut für Pathologie, Technische Universität München, Trogerstrasse 18, D-81765 Munich, Germany
- Pathologie Rosenheim, Germany
| | - M Sarbia
- Institut für Pathologie, Technische Universität München, Trogerstrasse 18, D-81765 Munich, Germany
- Institut für Pathologie, Krankenhaus Lichtenberg, Berlin, Germany
| | - F Carneiro
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Rua Dr Roberto Frias, S/N 4200-465 Porto, Portugal
- Faculdade de Medicina da Universidade do Porto and Hospital de S João, Alameda Hernâni Monteiro, 4200-319 Porto, Portugal
| | - K-F Becker
- Institut für Pathologie, Technische Universität München, Trogerstrasse 18, D-81765 Munich, Germany
| |
Collapse
|
802
|
Pagan JK, Arnold J, Hanchard KJ, Kumar R, Bruno T, Jones MJK, Richard DJ, Forrest A, Spurdle A, Verdin E, Crossley M, Fanciulli M, Chenevix-Trench G, Young DB, Khanna KK. A novel corepressor, BCoR-L1, represses transcription through an interaction with CtBP. J Biol Chem 2007; 282:15248-57. [PMID: 17379597 DOI: 10.1074/jbc.m700246200] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Corepressors play a crucial role in negative gene regulation and are defective in several diseases. BCoR is a corepressor for the BCL6 repressor protein. Here we describe and functionally characterize BCoR-L1, a homolog of BCoR. When tethered to a heterologous promoter, BCoR-L1 is capable of strong repression. Like other corepressors, BCoR-L1 associates with histone deacetylase (HDAC) activity. Specifically, BCoR-L1 coprecipitates with the Class II HDACs, HDAC4, HDAC5, and HDAC7, suggesting that they are involved in its role as a transcriptional repressor. BCoR-L1 also interacts with the CtBP corepressor through a CtBP-interacting motif in its amino terminus. Abrogation of the CtBP binding site within BCoR-L1 partially relieves BCoR-L1-mediated transcriptional repression. Furthermore, BCoR-L1 is located on the E-cadherin promoter, a known CtBP-regulated promoter, and represses the E-cadherin promoter activity in a reporter assay. The inhibition of BCoR-L1 expression by RNA-mediated interference results in derepression of E-cadherin in cells that do not normally express E-cadherin, indicating that BCoR-L1 contributes to the repression of an authentic endogenous CtBP target.
Collapse
Affiliation(s)
- Julia K Pagan
- Queensland Institute of Medical Research, 300 Herston Road, Herston 4029, Queensland, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
803
|
Taneyhill LA, Coles EG, Bronner-Fraser M. Snail2 directly represses cadherin6B during epithelial-to-mesenchymal transitions of the neural crest. Development 2007; 134:1481-90. [PMID: 17344227 PMCID: PMC2595139 DOI: 10.1242/dev.02834] [Citation(s) in RCA: 154] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The neural crest, a transient population of migratory cells, forms the craniofacial skeleton and peripheral nervous system, among other derivatives in vertebrate embryos. The transcriptional repressor Snail2 is thought to be crucial for the epithelial-to-mesenchymal transition (EMT) that promotes neural crest delamination from the neural tube; however, little is known about its downstream targets. To this end, we depleted avian Snail2 in the premigratory neural crest using morpholino antisense oligonucleotides and examined effects on potential targets by quantitative PCR. Several dorsal neural tube genes were upregulated by alleviating Snail2 repression; moreover, the cell adhesion molecule cadherin6B was derepressed within 30 minutes of blocking Snail2 translation. Examination of the chick cadherin6B genomic sequence reveals that the regulatory region contains three pairs of clustered E boxes, representing putative Snail2 binding sites. Furthermore, in vivo and in vitro biochemical analyses demonstrate that Snail2 directly binds to these sites and regulates cadherin6B transcription. These results are the first to describe a direct target of Snail2 repression in vivo and in the context of the EMT that characterizes neural crest development.
Collapse
|
804
|
Leroy P, Mostov KE. Slug is required for cell survival during partial epithelial-mesenchymal transition of HGF-induced tubulogenesis. Mol Biol Cell 2007; 18:1943-52. [PMID: 17344479 PMCID: PMC1855025 DOI: 10.1091/mbc.e06-09-0823] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Transcription factors of the Snail family are key regulators of epithelial-mesenchymal transition (EMT). In many processes during development or disease, cells do not acquire all the characteristics associated with EMT, leading to what we refer to as partial EMT (p-EMT). However, little is known of the implications of the Snail transcription factors in processes that only involve a p-EMT. To assess this, we used the hepatocyte growth factor (HGF)-induced Madin-Darby canine kidney tubulogenesis system, which provides a three-dimensional culture model of a morphogenetic process including a p-EMT. We found that although Slug (Snail2) is highly and transitory up-regulated during the p-EMT phase of tubulogenesis, it is not a repressor of E-cadherin during this process. Using inducible knockdown of Slug, we demonstrate that Slug is not an inducer of cell movement and instead is required for survival during p-EMT. We conclude that in epithelial cells, promoting cell survival can be a primary function of Slug, rather than being acquired concomitantly with EMT.
Collapse
Affiliation(s)
- Pascale Leroy
- Department of Anatomy, University of California, San Francisco, CA 94158-2517, USA.
| | | |
Collapse
|
805
|
Yasmeen A, Bismar TA, Al Moustafa AE. ErbB receptors and epithelial-cadherin-catenin complex in human carcinomas. Future Oncol 2007; 2:765-81. [PMID: 17155902 DOI: 10.2217/14796694.2.6.765] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The ErbB family of receptor tyrosine kinases have important roles in maintaining normal epithelial cell function. The ErbBs are involved in the interaction between cells and cell-matrix adhesion molecules and have proven critical in maintaining the integrity of the epithelial cell environment. Deregulation of these tyrosine receptors has been associated with several human diseases. In particular, the expression or activation of epidermal growth factor receptor (EGFR) and ErbB2 is altered in many epithelial tumors. Epithelial (E)-cadherin is another major molecule expressed by epithelial cells. To create efficient cell-cell adhesion, E-cadherin couples its cytoplasmic domain to catenins and the actin cytoskeleton. The loss of intercellular adhesion appears to be a fundamental aspect of the neoplastic phenomena. In addition, EGFR and ErbB2 signaling associated with the E-cadherin-catenin complex has been demonstrated in normal and cancer cells. This signaling is involved in regulating cell adhesion and the invasive growth of cancers. This article provides an overview of the interaction between the ErbB tyrosine receptors and the E-cadherin-catenin complex in human carcinomas.
Collapse
Affiliation(s)
- Amber Yasmeen
- McGill University, Program in Cancer Genetics, Department of Oncology, Montreal, Quebec, Canada.
| | | | | |
Collapse
|
806
|
Morales AV, Acloque H, Ocaña OH, de Frutos CA, Gold V, Nieto MA. Snail genes at the crossroads of symmetric and asymmetric processes in the developing mesoderm. EMBO Rep 2007; 8:104-9. [PMID: 17124510 PMCID: PMC1796742 DOI: 10.1038/sj.embor.7400867] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2006] [Revised: 10/02/2006] [Accepted: 10/10/2006] [Indexed: 01/03/2023] Open
Abstract
Retinoic acid (RA) signalling ensures that vertebrate mesoderm segmentation is bilaterally synchronized, and corrects transient interferences from asymmetric left-right (L-R) signals involved in organ lateralization. Snail genes participate in both these processes and, although they are expressed symmetrically in the presomitic mesoderm (PSM), Snail1 transcripts are asymmetrically distributed in the L-R lateral mesoderm. We show that the alteration of the symmetric Snail expression in the PSM induces asynchronous somite formation. Furthermore, in the absence of RA signalling, normal asymmetric Snail1 expression in the lateral mesoderm is extended to the PSM, desynchronizing somitogenesis. Thus, Snail1 is the first cue corrected by RA in the PSM to ensure synchronized bilateral segmentation.
Collapse
Affiliation(s)
- Aixa V Morales
- Instituto Cajal, CSIC, Doctor Arce 37, 28002 Madrid, Spain
| | - Hervé Acloque
- Instituto Cajal, CSIC, Doctor Arce 37, 28002 Madrid, Spain
- Instituto de Neurociencias de Alicante, CSIC-UMH, Apartado 18, Sant Joan d'Alacant, 03550, Spain
| | - Oscar H Ocaña
- Instituto Cajal, CSIC, Doctor Arce 37, 28002 Madrid, Spain
- Instituto de Neurociencias de Alicante, CSIC-UMH, Apartado 18, Sant Joan d'Alacant, 03550, Spain
| | - Cristina A de Frutos
- Instituto Cajal, CSIC, Doctor Arce 37, 28002 Madrid, Spain
- Instituto de Neurociencias de Alicante, CSIC-UMH, Apartado 18, Sant Joan d'Alacant, 03550, Spain
| | - Veronica Gold
- Instituto Cajal, CSIC, Doctor Arce 37, 28002 Madrid, Spain
- Instituto de Neurociencias de Alicante, CSIC-UMH, Apartado 18, Sant Joan d'Alacant, 03550, Spain
| | - M Angela Nieto
- Instituto Cajal, CSIC, Doctor Arce 37, 28002 Madrid, Spain
- Instituto de Neurociencias de Alicante, CSIC-UMH, Apartado 18, Sant Joan d'Alacant, 03550, Spain
| |
Collapse
|
807
|
Mejlvang J, Kriajevska M, Berditchevski F, Bronstein I, Lukanidin EM, Pringle JH, Mellon JK, Tulchinsky EM. Characterization of E-cadherin-dependent and -independent events in a new model of c-Fos-mediated epithelial–mesenchymal transition. Exp Cell Res 2007; 313:380-93. [PMID: 17141758 DOI: 10.1016/j.yexcr.2006.10.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2006] [Revised: 10/13/2006] [Accepted: 10/19/2006] [Indexed: 01/05/2023]
Abstract
Fos proteins have been implicated in control of tumorigenesis-related genetic programs including invasion, angiogenesis, cell proliferation and apoptosis. In this study, we demonstrate that c-Fos is able to induce mesenchymal transition in murine tumorigenic epithelial cell lines. Expression of c-Fos in MT1TC1 cells led to prominent alterations in cell morphology, increased expression of mesenchymal markers, vimentin and S100A4, DNA methylation-dependent down-regulation of E-cadherin and abrogation of cell-cell adhesion. In addition, c-Fos induced a strong beta-catenin-independent proliferative response in MT1TC1 cells and stimulated cell motility, invasion and adhesion to different extracellular matrix proteins. To explore whether loss of E-cadherin plays a role in c-Fos-mediated mesenchymal transition, we expressed wild-type E-cadherin and two different E-cadherin mutants in MT1TC1/c-fos cells. Expression of wild-type E-cadherin restored epithelioid morphology and enhanced cellular levels of catenins. However, exogenous E-cadherin did not influence expression of c-Fos-dependent genes, only partly suppressed growth of MT1TC1/c-fos cells and produced no effect on c-Fos-stimulated cell motility and invasion in matrigel. On the other hand, re-expression of E-cadherin specifically negated c-Fos-induced adhesion to collagen type I, but not to laminin or fibronectin. Of interest, mutant E-cadherin which lacks the ability to form functional adhesive complexes had an opposite, potentiating effect on cell adhesion to collagen I. These data suggest that cell adhesion to collagen I is regulated by the functional state of E-cadherin. Overall, our data demonstrate that, with the exception of adhesion to collagen I, c-Fos is dominant over E-cadherin in relation to the aspects of mesenchymal transition assayed in this study.
Collapse
Affiliation(s)
- Jakob Mejlvang
- Department of Cancer Studies and Molecular Medicine, University of Leicester, Hodgkin Bldg., Lancaster Rd, LE1 9HN, Leicester, UK
| | | | | | | | | | | | | | | |
Collapse
|
808
|
Klein EA, Yung Y, Castagnino P, Kothapalli D, Assoian RK. Cell Adhesion, Cellular Tension, and Cell Cycle Control. Methods Enzymol 2007; 426:155-75. [PMID: 17697884 DOI: 10.1016/s0076-6879(07)26008-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Cooperative signaling between growth factor receptor tyrosine kinases, integrins, and the actin cytoskeleton is required for activation of the G1-phase cyclin-dependent kinases and progression through G1-phase. Increasing evidence suggests that there is cell type specificity in these cooperative interactions and that the compliance of the underlying substratum can strongly affect adhesion-dependent signaling to the cell cycle. This chapter reviews our current methods for studying how cell type specificity and changes in substratum compliance can contribute to G1-phase cell cycle control. We also describe several of our current analytical procedures.
Collapse
Affiliation(s)
- Eric A Klein
- Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | | | | | | | |
Collapse
|
809
|
Zhang C, Carl TF, Trudeau ED, Simmet T, Klymkowsky MW. An NF-kappaB and slug regulatory loop active in early vertebrate mesoderm. PLoS One 2006; 1:e106. [PMID: 17205110 PMCID: PMC1762408 DOI: 10.1371/journal.pone.0000106] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2006] [Accepted: 11/23/2006] [Indexed: 01/11/2023] Open
Abstract
Background In both Drosophila and the mouse, the zinc finger transcription factor Snail is required for mesoderm formation; its vertebrate paralog Slug (Snai2) appears to be required for neural crest formation in the chick and the clawed frog Xenopus laevis. Both Slug and Snail act to induce epithelial to mesenchymal transition (EMT) and to suppress apoptosis. Methodology & Principle Findings Morpholino-based loss of function studies indicate that Slug is required for the normal expression of both mesodermal and neural crest markers in X. laevis. Both phenotypes are rescued by injection of RNA encoding the anti-apoptotic protein Bcl-xL; Bcl-xL's effects are dependent upon IκB kinase-mediated activation of the bipartite transcription factor NF-κB. NF-κB, in turn, directly up-regulates levels of Slug and Snail RNAs. Slug indirectly up-regulates levels of RNAs encoding the NF-κB subunit proteins RelA, Rel2, and Rel3, and directly down-regulates levels of the pro-apopotic Caspase-9 RNA. Conclusions/Significance These studies reveal a Slug/Snail–NF-κB regulatory circuit, analogous to that present in the early Drosophila embryo, active during mesodermal formation in Xenopus. This is a regulatory interaction of significance both in development and in the course of inflammatory and metastatic disease.
Collapse
Affiliation(s)
- Chi Zhang
- Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Timothy F. Carl
- Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Evan D. Trudeau
- Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Thomas Simmet
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, University of Ulm, Ulm, Germany
| | - Michael W. Klymkowsky
- Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
810
|
Abstract
Metastasis is the deadly face of epithelial tumors. The studies performed in the last decade have shed considerable light on the processes involved in the metastatic cascade. In particular, much effort has focused on defining the molecular changes that govern the conversion from an epithelial to a mesenchymal cell, a process known as epithelial-mesenchymal transition (EMT). The process of EMT is considered a fundamental event in the metastatic cascade (i.e. during invasion and/or intravasation) and several molecules involved in EMT have been described, including epithelial markers, transcription factors, as well as extracellular proteins and growth factors. In this green series article, we will focus our attention on the new molecules described in the recent years that appear to influence EMT and that are therefore relevant to epithelial carcinogenesis. Furthermore, we will try to explain how these molecules collaborate with the tumor microenvironment to trigger metastasis. Recent advances in our understanding of this process is generating a wide range of molecules that could be potentially considered as new therapeutic targets for drug design to block metastatic spreading.
Collapse
Affiliation(s)
- H Peinado
- Departamento de Bioquímica, Facultad de Medicina, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM. Madrid. Spain.
| | | |
Collapse
|
811
|
Sivertsen S, Hadar R, Elloul S, Vintman L, Bedrossian C, Reich R, Davidson B. Expression of Snail, Slug and Sip1 in malignant mesothelioma effusions is associated with matrix metalloproteinase, but not with cadherin expression. Lung Cancer 2006; 54:309-17. [PMID: 16996643 DOI: 10.1016/j.lungcan.2006.08.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2006] [Revised: 06/26/2006] [Accepted: 08/22/2006] [Indexed: 11/28/2022]
Abstract
Snail, Slug and Sip1 regulate cadherin and protease expression and mediate epithelial-mesenchymal transition in cancer. We analyzed the expression of cadherins and matrix metalloproteinases (MMP) and their transcriptional regulators in malignant mesothelioma (MM). One hundred and ten MM specimens (86 solid, 24 effusions) and 10 non-malignant effusions with reactive mesothelial cells (RMC) were analyzed for E-cadherin, N-cadherin and P-cadherin protein expression using immunhistochemistry. MM effusions were further analyzed for expression of Snail, Slug, Sip1, E-cadherin, MMP-2, MMP-9, MT1-MMP (MMP-14) and the MMP inhibitor TIMP-2, and for MMP-2 and MMP-9 activity using RT-PCR, Western blotting, immunhistochemistry and zymography. Results were analyzed for relationship with specimen type (biopsy versus effusion) and anatomic site (pleural versus peritoneal). E-cadherin, N-cadherin and P-cadherin expression was found in 69/110 (63%), 87/110 (79%) and 84/110 (76%) MM cases, respectively. Pleural and peritoneal MM showed comparable expression, but all three cadherins were upregulated in effusions compared to solid tumors (p<0.001). RMC were uniformly negative for E-cadherin and N-cadherin, and showed P-cadherin expression in 7/10 specimens. Immunohistochemistry localized MMP-2, MMP-9 and TIMP-2 to MM cells in 11/15, 14/15 and 8/15 effusions, respectively. RT-PCR showed direct association between MMP-2 mRNA expression level and the levels of MT1-MMP (p=0.027) and TIMP-2 (p=0.011). Snail protein expression showed positive association with MT1-MMP (p=0.016) and TIMP-2 (p=0.02) mRNA expression, but its expression was unrelated to MMP-2 and MMP-9 expression or activity. Snail, Slug and Sip1 levels did not show inverse association with E-cadherin levels. Our data show that E-cadherin and N-cadherin are selectively expressed in malignant mesothelial cells, and that P-cadherin and N-cadherin are expressed with similar frequency in MM. In agreement with our earlier data for ovarian carcinoma, cadherin expression is upregulated in effusions compared to solid lesions. The increased E-cadherin expression in effusions may be related to lack of negative regulation at the epigenetic level. The relationship between Snail and MMP in MM is uncertain at present.
Collapse
Affiliation(s)
- Stine Sivertsen
- Department of Pathology, Radiumhospitalet-Rikshospitalet Medical Center, University of Oslo, Montebello, N-0310 Oslo, Norway
| | | | | | | | | | | | | |
Collapse
|
812
|
Rukstalis JM, Habener JF. Snail2, a mediator of epithelial-mesenchymal transitions, expressed in progenitor cells of the developing endocrine pancreas. Gene Expr Patterns 2006; 7:471-9. [PMID: 17185046 PMCID: PMC2698037 DOI: 10.1016/j.modgep.2006.11.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2006] [Revised: 11/02/2006] [Accepted: 11/03/2006] [Indexed: 01/13/2023]
Abstract
The mammalian pancreas develops by the expansion and morphogenesis of the epithelial cells of the foregut endoderm via the sequential activation of transcription factors that direct differentiation into the various pancreatic lineages. Implicit in this growth and differentiation are the temporal and spatial processes of cell migration and three-dimensional organization, which cooperate to form a properly functioning organ. In many organ systems, such as the kidney, heart, and neural crest derivatives, migration and tissue morphogenesis is accomplished by the transient conversion of stationary epithelial cells to migratory mesenchymal-like cells in a process known as epithelial-mesenchymal transition (EMT). We report the identification of the expression of the transcription factor Snail2/Slug, a known inducer of EMT and cell movement, in both the endocrine and exocrine cells of the developing mouse pancreas. Snail2 is expressed in Neurogenin3-positive endocrine progenitor cells, and expression is maintained during endocrine cell differentiation where it becomes increasingly restricted to the insulin-producing beta cells and somatostatin-producing delta cells. In the adult pancreas, the expression of Snail2 is maintained at low but detectable levels in all beta cells, indicating a latent role for Snail2 in the mature islet. These findings of Snail2 expression during endocrine pancreas development are relevant to the recent evidence demonstrating the involvement of EMT in the expansion of human islet tissue in vitro. EMT-like events appear to be involved in the development of the mammalian pancreas in vivo.
Collapse
Affiliation(s)
- J Michael Rukstalis
- Laboratory of Molecular Endocrinology, Massachusetts General Hospital, Harvard Medical School and Howard Hughes Medical Institute, Boston, MA 02114, USA
| | | |
Collapse
|
813
|
Boutet A, De Frutos CA, Maxwell PH, Mayol MJ, Romero J, Nieto MA. Snail activation disrupts tissue homeostasis and induces fibrosis in the adult kidney. EMBO J 2006; 25:5603-13. [PMID: 17093497 PMCID: PMC1679761 DOI: 10.1038/sj.emboj.7601421] [Citation(s) in RCA: 262] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2006] [Accepted: 10/10/2006] [Indexed: 02/06/2023] Open
Abstract
During embryonic development, the kidney epithelium originates from cells that undergo a mesenchymal to epithelial transition (MET). The reverse process, epithelium to mesenchyme transition (EMT), has been implicated in epithelial tumor progression and in the fibrosis that leads to end-stage kidney failure. Snail transcription factors induce both natural and pathological EMT, but their implication in renal development and disease is still unclear. We show that Snail genes are downregulated during the MET that occurs during renal development and that this is correlated with Cadherin-16 expression. Snail suppresses Cadherin-16 via the direct repression of the kidney differentiation factor HNF-1beta, a novel route by which Snail disrupts epithelial homeostasis. Indeed, Snail activation is sufficient to induce EMT and kidney fibrosis in adult transgenic mice. Significantly, Snail is also activated in patients with renal fibrosis. Thus, Snail expression is suppressed during renal development and it must remain silent in the mature kidney where its aberrant activation leads to fibrosis.
Collapse
Affiliation(s)
- Agnès Boutet
- Instituto de Neurociencias de Alicante, CSIC-UMH, Sant Joan d'Alacant, Alicante, Spain
| | - Cristina A De Frutos
- Instituto de Neurociencias de Alicante, CSIC-UMH, Sant Joan d'Alacant, Alicante, Spain
| | - Patrick H Maxwell
- Department of Nephrology, Imperial College London, Hammersmith Campus, London, UK
| | - M José Mayol
- Department of Anatomopathology and Urology, Sant Joan d'Alacant University Hospital, Sant Joan d'Alacant, Spain
| | - J Romero
- Department of Anatomopathology and Urology, Sant Joan d'Alacant University Hospital, Sant Joan d'Alacant, Spain
| | - M Angela Nieto
- Instituto de Neurociencias de Alicante, CSIC-UMH, Sant Joan d'Alacant, Alicante, Spain
- Instituto de Neurociencias de Alicante, CSIC-UMH, Apartado 18, Sant Joan d'Alacant, Alicante 03550, Spain. Tel.: +34 96 591 92 43; Fax: +34 96 591 95 61; E-mail:
| |
Collapse
|
814
|
Ding JX, Feng YJ, Yao LQ, Yu M, Jin HY, Yin LH. The reinforcement of invasion in epithelial ovarian cancer cells by 17β-Estradiol is associated with up-regulation of Snail. Gynecol Oncol 2006; 103:623-30. [PMID: 16806441 DOI: 10.1016/j.ygyno.2006.04.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2006] [Revised: 04/16/2006] [Accepted: 04/25/2006] [Indexed: 11/24/2022]
Abstract
OBJECTIVE The transcription factor Snail, which is implicated in the triggering of epithelial-mesenchymal transitions (EMT), plays an important role in adhesion, invasion and metastasis of tumor cells. In the present study, we assessed 17beta-Estradiol (E2)'s effect on Snail, E-cadherin and MMP-2 expression of epithelial ovarian cancer cell line ES-2 and SKOV3. Then we induced Snail gene silencing by RNA interference to explore the effect of E2 on E-cadherin and MMP-2 expression when Snail gene expression was blocked. METHODS Treated by 10(-8) M E2, Snail, E-cadherin and MMP-2 mRNA expression of the cells was measured by RT-PCR; Snail, MMP-2 protein expression was detected by IHC; and MMP-2 activity was determined by Zymography. E-cadherin protein level was measured by Western blot. We constructed the small interfering dsRNA expression vector (pRNAT-U6.1/Neo-Snail) targeting Snail gene, as well as a negative control vector (pRNAT-U6.1/Neo-Neg). Then the cells were transiently transfected with the vectors. Western blot and zymography were conducted to determine E-cadherin protein level and matrix metalloproteinase activity of the cells transfected with pRNAT-U6.1/Neo-Snail or pRNAT-U6.1/Neo-Neg after treated with E2 for 24 h. RESULTS The expression of ER alpha mRNA and protein was negative in ES-2 cells and positive in SKOV3 cells, and ER beta expression was positive in both cell lines. 10(-8) mol/l E2 elevated expression of Snail and MMP-2 mRNA and protein in both ES-2 and SKOV3 cells, and reduced expression of E-cadherin mRNA and protein in SKOV3 cells. While in the RNAi group transfected with the small interfering dsRNA expression vector (pRNAT-U6.1/Neo-Snail) targeting Snail gene, E2 treatment did not have a significant effect on MMP-2 activity or E-cadherin protein in ES-2 and SKOV3 cells. CONCLUSIONS 17beta-Estradiol increased Snail expression in both ER alpha-negative ES-2 cells and ER alpha-positive SKOV3 cells independent of the existence of ER alpha. The increase of MMP-2 expression in ES-2 and SKOV3 cells and decrease of E-cadherin expression in SKOV3 cells induced by E2 were associated with up-regulation of Snail.
Collapse
MESH Headings
- Adenocarcinoma, Clear Cell/genetics
- Adenocarcinoma, Clear Cell/metabolism
- Adenocarcinoma, Clear Cell/pathology
- Cadherins/biosynthesis
- Cadherins/genetics
- Cell Line, Tumor
- Cystadenocarcinoma, Serous/genetics
- Cystadenocarcinoma, Serous/metabolism
- Cystadenocarcinoma, Serous/pathology
- Epithelial Cells/pathology
- Estradiol/pharmacology
- Female
- Gene Silencing
- Humans
- Matrix Metalloproteinase 2/biosynthesis
- Matrix Metalloproteinase 2/genetics
- Matrix Metalloproteinase 2/metabolism
- Neoplasm Invasiveness
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Receptors, Estrogen/biosynthesis
- Receptors, Estrogen/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Snail Family Transcription Factors
- Transcription Factors/biosynthesis
- Transcription Factors/genetics
- Up-Regulation
Collapse
Affiliation(s)
- Jing-Xin Ding
- Gynecologic and Obstetric Hospital, Fudan University, Department of Gynecology and Obstetrics, Shanghai Medical College, Fudan University, Shanghai, PR China
| | | | | | | | | | | |
Collapse
|
815
|
Huang HC, Hu CH, Tang MC, Wang WS, Chen PM, Su Y. Thymosin β4 triggers an epithelial–mesenchymal transition in colorectal carcinoma by upregulating integrin-linked kinase. Oncogene 2006; 26:2781-90. [PMID: 17072345 DOI: 10.1038/sj.onc.1210078] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The epithelial-mesenchymal transition (EMT) is crucial for the invasion and metastasis of many epithelial tumors including colorectal carcinoma (CRC). In the present study, a scattering and fibroblastic morphology with reduced intercellular contacts was found in the SW480 colon cancer cells overexpressing the gene encoding thymosin beta4 (Tbeta4), which was accompanied by a loss of E-cadherin as well as a cytosolic accumulation of beta-catenin, two most prominent markers of EMT. Whereas E-cadherin downregulation was likely to be accounted by a ZEB1-mediated transcriptional repression, the accumulation of beta-catenin was a result of glycogen synthase kinase-3beta inactivation mediated by integrin-linked kinase (ILK) and/or its downstream effector, Akt. Intriguingly, ILK upregulation in Tbeta4-overexpressing SW480 cells seemed to be attributed mainly to a stabilization of this kinase by complexing with particularly interesting new Cys-His protein (PINCH) more efficiently. In the meantime, a strong correlation between the expression levels of Tbeta4, ILK and E-cadherin in CRC patients was also revealed by immunohistochemical analysis. Taken together, these data suggest a novel role of Tbeta4 in promoting CRC progression by inducing an EMT in tumor cells via upregulating ILK and consequentially its signal transduction.
Collapse
Affiliation(s)
- H-C Huang
- Institute of Bioscience and Biotechnology, College of Life Science, National Taiwan Ocean University, Keelung, and Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
| | | | | | | | | | | |
Collapse
|
816
|
Yang JY, Zong CS, Xia W, Wei Y, Ali-Seyed M, Li Z, Broglio K, Berry DA, Hung MC. MDM2 promotes cell motility and invasiveness by regulating E-cadherin degradation. Mol Cell Biol 2006; 26:7269-82. [PMID: 16980628 PMCID: PMC1592879 DOI: 10.1128/mcb.00172-06] [Citation(s) in RCA: 156] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Gene amplification and protein overexpression of MDM2, which is often found in certain types of cancers, indicate that MDM2 plays an important role in tumorigenesis. Interestingly, several clinical reports have demonstrated that amplification of the MDM2 gene correlates with the metastatic stage. Using an antibody array assay, we identified E-cadherin as an MDM2-binding protein and confirmed that E-cadherin is a substrate for the MDM2 E3 ubiquitin ligase. We demonstrate that MDM2 interacts in vivo with E-cadherin, resulting in its ubiquitination and degradation. This regulation appears to be clinically relevant, as we found a significant correlation between high MDM2 and low E-cadherin protein levels in resected tumor specimens recovered from breast cancer patients with lymph node metastases. Ectopic expression of MDM2 in breast cancer cells was found to disrupt cell-cell contacts and enhance cell motility and invasive potential. We found that E-cadherin and MDM2 colocalized on the plasma membrane and in the early endosome, where ubiquitin moieties were attached to E-cadherin. Blocking endocytosis with dominant-negative mutants of dynamin abolished the association of MDM2 with E-cadherin, prevented E-cadherin degradation, and attenuated cell motility as observed by fluorescence microscopy. Thus, we provide evidence to support a novel role for MDM2 in regulating cell adhesions by a mechanism that involves degrading and down-regulating the expression of E-cadherin via an endosome pathway. This novel MDM2-regulated pathway is likely to play a biologically relevant role in cancer metastasis.
Collapse
Affiliation(s)
- Jer-Yen Yang
- Department of Molecular and Cellular Oncology, University of Texas M. D. Anderson Cancer Center, Unit 108, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
817
|
Cicchini C, Filippini D, Coen S, Marchetti A, Cavallari C, Laudadio I, Spagnoli FM, Alonzi T, Tripodi M. Snail controls differentiation of hepatocytes by repressing HNF4alpha expression. J Cell Physiol 2006; 209:230-8. [PMID: 16826572 DOI: 10.1002/jcp.20730] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Epithelial-to-mesenchymal transition (EMT) is a coordinated process, occurring both during morphogenesis and tumor progression, that allows epithelial cells to dissociate from initial contacts and migrate to secondary sites. The transcriptional repressors of the Snail family induce EMT in different epithelial cell lines and their expression is strictly correlated with EMT during the development and progression of carcinomas. We have previously shown that EMT in hepatocytes correlates with the downregulation of hepatic differentiation key factors HNFs (hepatocyte nuclear factors), and in particular of HNF4alpha. Here, we demonstrate that Snail overexpression is sufficient (i) to induce EMT in hepatocytes with conversion of morphology, downregulation of several epithelial adhesion molecules, reduction of proliferation and induction of matrix metalloproteinase 2 expression and, (ii) most relevantly, to repress the transcription of the HNF4alpha gene through a direct binding to its promoter. These finding demonstrate that Snail is at the crossroads of the regulation of EMT in hepatocytes by a dual control of epithelial morphogenesis and differentiation.
Collapse
Affiliation(s)
- Carla Cicchini
- Dipartimento di Biotecnologie Cellulari ed Ematologia, Università La Sapienza, Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
818
|
Martín-Villar E, Megías D, Castel S, Yurrita MM, Vilaró S, Quintanilla M. Podoplanin binds ERM proteins to activate RhoA and promote epithelial-mesenchymal transition. J Cell Sci 2006; 119:4541-53. [PMID: 17046996 DOI: 10.1242/jcs.03218] [Citation(s) in RCA: 282] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Podoplanin is a small membrane mucin expressed in tumors associated with malignant progression. It is enriched at cell-surface protrusions where it colocalizes with members of the ERM (ezrin, radixin, moesin) protein family. Here, we found that human podoplanin directly interacts with ezrin (and moesin) in vitro and in vivo through a cluster of basic amino acids within its cytoplasmic tail, mainly through a juxtamembrane dipeptide RK. Podoplanin induced an epithelial-mesenchymal transition in MDCK cells linked to the activation of RhoA and increased cell migration and invasiveness. Fluorescence time-lapse video observations in migrating cells indicate that podoplanin might be involved in ruffling activity as well as in retractive processes. By using mutant podoplanin constructs fused to green fluorescent protein we show that association of the cytoplasmic tail with ERM proteins is required for upregulation of RhoA activity and epithelial-mesenchymal transition. Furthermore, expression of either a dominant-negative truncated variant of ezrin or a dominant-negative mutant form of RhoA blocked podoplanin-induced RhoA activation and epithelial-mesenchymal transition. These results provide a mechanistic basis to understand the role of podoplanin in cell migration or invasiveness.
Collapse
Affiliation(s)
- Ester Martín-Villar
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
| | | | | | | | | | | |
Collapse
|
819
|
Olmeda D, Jordá M, Peinado H, Fabra A, Cano A. Snail silencing effectively suppresses tumour growth and invasiveness. Oncogene 2006; 26:1862-74. [PMID: 17043660 DOI: 10.1038/sj.onc.1209997] [Citation(s) in RCA: 206] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The transcription factor Snail has been recently proposed as an important mediator of tumour invasion because of its role in downregulation of E-cadherin and induction of epithelial-mesenchymal transitions (EMT). This behaviour has led to the consideration of Snail as a potential therapeutic target to block tumour progression. In this report, we provide evidence for this hypothesis. We show that silencing of Snail by stable RNA interference in MDCK-Snail cells induces a complete mesenchymal to epithelial transition (MET), associated to the upregulation of E-cadherin, downregulation of mesenchymal markers and inhibition of invasion. More importantly, stable interference of endogenous Snail in two independent carcinoma cell lines leads to a dramatic reduction of in vivo tumour growth, accompanied by increased tumour differentiation and a significant decrease in the expression of MMP-9 and angiogenic markers and invasiveness. These results indicate that use of RNA interference can be an effective tool for blocking Snail function, opening the way for its application in new antiinvasive therapies.
Collapse
Affiliation(s)
- D Olmeda
- Departamento de Bioquímica, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, c/Arturo Duperier, Madrid, Spain
| | | | | | | | | |
Collapse
|
820
|
Elloul S, Silins I, Tropé CG, Benshushan A, Davidson B, Reich R. Expression of E-cadherin transcriptional regulators in ovarian carcinoma. Virchows Arch 2006; 449:520-8. [PMID: 17024425 DOI: 10.1007/s00428-006-0274-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2006] [Accepted: 07/12/2006] [Indexed: 11/29/2022]
Abstract
Unlike most epithelial cancers, E-cadherin expression is upregulated in ovarian carcinoma effusions compared with corresponding primary tumors. In the present study, we analyzed the anatomic site-specific expression of transcription factors that negatively regulate E-cadherin in ovarian carcinoma. Using reverse-transcription polymerase chain reaction, mRNA in situ hybridization, and Western blotting, we analyzed the expression and localization of the Snail, Slug, and SIP1 transcription factors and E-cadherin in 78 effusions, 41 primary carcinomas, and 15 solid metastases. Slug mRNA and protein expression was highest in metastases (p=0.042 and p<0.001, respectively). Snail mRNA was comparable at all anatomic sites, but higher protein expression was found in primary tumors and solid metastases compared with effusions (p<0.001). SIP1 mRNA expression was higher in effusions (p<0.001) compared to other sites. Confocal microscopy analysis of fresh and cultured cells from effusion specimens revealed cytoplasmic localization of the Snail protein in primary tumor cells, with a nuclear shift following culturing of these cells. In conclusion, E-cadherin and its negative regulators show site-dependent expression in ovarian carcinoma. In solid tumors, E-cadherin is negatively regulated by Snail and Slug. In effusions, SIP1 may be the main regulator of E-cadherin, but with a lesser level of suppression compared with primary tumors and solid metastases.
Collapse
Affiliation(s)
- Sivan Elloul
- Department of Pharmacology and Experimental Therapeutics, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | | | | | | | | | | |
Collapse
|
821
|
Hayashida Y, Urata Y, Muroi E, Kono T, Miyata Y, Nomata K, Kanetake H, Kondo T, Ihara Y. Calreticulin Represses E-cadherin Gene Expression in Madin-Darby Canine Kidney Cells via Slug. J Biol Chem 2006; 281:32469-84. [PMID: 16943193 DOI: 10.1074/jbc.m607240200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Calreticulin (CRT) is a multifunctional Ca(2+)-binding molecular chaperone in the endoplasmic reticulum. In mammals, the expression level of CRT differs markedly in a variety of organs and tissues, suggesting that CRT plays a specific role in each cell type. In the present study, we focused on CRT functions in the kidney, where overall expression of CRT is quite low, and established CRT-overexpressing kidney epithelial cell-derived Madin-Darby canine kidney cells by gene transfection. We demonstrated that, in CRT-overexpressing cells, the morphology was apparently changed, and the original polarized epithelial cell phenotype was destroyed. Furthermore, CRT-overexpressing cells showed enhanced migration through Matrigel-coated Boyden chamber wells, compared with controls. E-cadherin expression was significantly suppressed at the protein and transcriptional levels in CRT-overexpressing cells compared with controls. On the other hand, the expression of mesenchymal protein markers, such as N-cadherin and fibronectin, was up-regulated. We also found that the expression of Slug, a repressor of the E-cadherin promoter, was up-regulated by overexpression of CRT through altered Ca(2+) homeostasis, and this led to enhanced binding of Slug to the E-box element in the E-cadherin promoter. Thus, we conclude that CRT regulates the epithelial-mesenchymal transition-like change of cellular phenotype by modulating the Slug/E-cadherin pathway through altered Ca(2+) homeostasis in cells, suggesting a novel function of CRT in cell-cell interaction of epithelial cells.
Collapse
Affiliation(s)
- Yasushi Hayashida
- Department of Biochemistry and Molecular Biology in Disease, Atomic Bomb Disease Institute, and Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
822
|
Spaderna S, Schmalhofer O, Hlubek F, Berx G, Eger A, Merkel S, Jung A, Kirchner T, Brabletz T. A transient, EMT-linked loss of basement membranes indicates metastasis and poor survival in colorectal cancer. Gastroenterology 2006; 131:830-40. [PMID: 16952552 DOI: 10.1053/j.gastro.2006.06.016] [Citation(s) in RCA: 379] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2006] [Accepted: 05/25/2006] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Loss of the basement membrane (BM) is considered an important step toward tumor malignancy. However, the BM is still expressed in most typical colorectal adenocarcinomas; nevertheless, these tumors can invade and develop metastases. The aim of this study was to investigate the role, mechanisms, and clinical relevance of BM turnover in malignant colorectal cancer (CRC) progression. METHODS Expression of BM components and their transcriptional regulation and clinical relevance were investigated in human CRCs and cell lines. RESULTS Our data show new aspects in BM turnover in CRCs with impact on malignant tumor progression: (1) The BM is still expressed in the main tumor mass of most colorectal adenocarcinomas, but selectively lost at invasive regions of the tumor in many cases. (2) Selective loss of the BM at the invasive front has high clinical and tumor biologic relevance for distant metastasis and survival. (3) The BM is reexpressed in metastases, indicating that its loss is transient and regulated by environmental factors. (4) This transient loss is not only due to proteolytic breakdown but to a down-regulated synthesis and linked to an epithelial-mesenchymal transition (EMT) in tumor cells, and, thereby, zinc-finger-enhancer protein 1 (ZEB1) is the crucial transcriptional repressor of BM components in CRCs. CONCLUSIONS A transient BM loss at the invasive front is correlated with increased distant metastasis and poor patient survival, indicating its tumor biologic relevance and usefulness as a prognostic marker. Targeting ZEB1 might be a promising therapeutic option to prevent metastasis.
Collapse
Affiliation(s)
- Simone Spaderna
- Department of Pathology, University of Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
823
|
Whitbread AK, Veveris-Lowe TL, Lawrence MG, Nicol DL, Clements JA. The role of kallikrein-related peptidases in prostate cancer: potential involvement in an epithelial to mesenchymal transition. Biol Chem 2006; 387:707-14. [PMID: 16800731 DOI: 10.1515/bc.2006.089] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Several members of the kallikrein-related peptidase family of serine proteases have proteolytic activities that may affect cancer progression; however, the in vivo significance of these activities remains uncertain. We have demonstrated that expression of PSA or KLK4, but not KLK2, in PC-3 prostate cancer cells changed the cellular morphology from epithelial to spindle-shaped, markedly reduced E-cadherin expression, increased vimentin expression and increased cellular migration. These changes are indicative of an epithelial to mesenchymal transition (EMT), a process important in embryonic development and cancer progression. The potential novel role of kallikrein-related peptidases in this process is the focus of this brief review.
Collapse
Affiliation(s)
- Astrid K Whitbread
- Hormone-Dependent Cancer Program, School of Life Sciences and Institute of Health and Biomedical Innovation, Queensland University of Technology, and Department of Urology, Princess Alexandria Hospital, Brisbane 4000, QLD, Australia
| | | | | | | | | |
Collapse
|
824
|
Vernon AE, LaBonne C. Slug stability is dynamically regulated during neural crest development by the F-box protein Ppa. Development 2006; 133:3359-70. [PMID: 16887825 DOI: 10.1242/dev.02504] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The neural crest is a population of stem-cell-like precursors found only in vertebrates. Slug, a member of the Snail family of zincfinger transcriptional repressors, is a critical regulator of neural crest development and has also been implicated in the acquisition of invasive behavior during tumor progression. Despite its central role in these two important processes, little is known about the mechanisms that control the expression and/or activity of Slug. We demonstrate that Slug is a labile protein whose stability is positively reinforced through activation of the neural crest regulatory program. We identify Partner of paired (Ppa) as the F-box component of a modular E3 ligase, and show that it is expressed in neural crest-forming regions, and that it binds to and promotes ubiquitin-mediated proteasomal degradation of Slug. Misexpression of Ppa inhibits the formation of neural crest precursors, and Slug mutants in which Ppa binding has been abrogated rescue this inhibition. These results provide novel insight into the regulation of Slug, a protein that plays a central role in neural crest precursor formation, as well as in developmental and pathological epithelial to mesenchymal transitions.
Collapse
Affiliation(s)
- Ann E Vernon
- Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, Evanston, IL 60208, USA
| | | |
Collapse
|
825
|
Robert G, Gaggioli C, Bailet O, Chavey C, Abbe P, Aberdam E, Sabatié E, Cano A, Garcia de Herreros A, Ballotti R, Tartare-Deckert S. SPARC represses E-cadherin and induces mesenchymal transition during melanoma development. Cancer Res 2006; 66:7516-23. [PMID: 16885349 DOI: 10.1158/0008-5472.can-05-3189] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
During progression of melanoma, loss of the cell-cell adhesion molecule E-cadherin contributes to uncontrolled growth and invasive behavior of transformed melanocytes. Secreted protein acidic and rich in cysteine (SPARC) is a nonstructural matricellular protein that regulates cell-matrix interactions leading to alterations in cell adhesion and proliferation. Overexpression of SPARC has been associated with progression of various cancers, including melanoma; however, its role in primary tumor development is not well defined. We show that normal human melanocytes overexpressing SPARC adopt a fibroblast-like morphology, concomitant with loss of E-cadherin and P-cadherin expression, and increased expression of mesenchymal markers. Concurrent with these changes, SPARC expression stimulates melanocyte motility and melanoma cell invasion. Expression of SPARC results in transcriptional down-regulation of E-cadherin that correlates with induction of Snail, a repressor of E-cadherin. Conversely, SPARC depletion leads to up-regulation of E-cadherin and reduces Snail levels, and SPARC-null cells exhibit a marked change in their mesenchymal phenotype. Finally, analysis of SPARC, Snail, and E-cadherin levels in melanocytes and malignant melanoma cell lines further supports the functional relationship among these proteins during melanoma progression. Our findings provide evidence for the role of SPARC in early transformation of melanocytes and identify a novel mechanism, whereby tumor-derived SPARC promotes tumorigenesis by mediating Snail induction and E-cadherin suppression.
Collapse
Affiliation(s)
- Guillaume Robert
- INSERM Unité 597, Biologie et Pathologies des Cellules Mélanocytaires, Faculté de Médecine, Université de Nice Sophia-Antipolis, 28 avenue de Valombrose, 06107 Nice Cédex 2, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
826
|
Turner FE, Broad S, Khanim FL, Jeanes A, Talma S, Hughes S, Tselepis C, Hotchin NA. Slug regulates integrin expression and cell proliferation in human epidermal keratinocytes. J Biol Chem 2006; 281:21321-21331. [PMID: 16707493 DOI: 10.1074/jbc.m509731200] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The human epidermis is a self-renewing epithelial tissue composed of several layers of keratinocytes. Within the epidermis there exists a complex array of cell adhesion structures, and many of the cellular events within the epidermis (differentiation, proliferation, and migration) require that these adhesion structures be remodeled. The link between cell adhesion, proliferation, and differentiation within the epidermis is well established, and in particular, there is strong evidence to link the process of terminal differentiation to integrin adhesion molecule expression and function. In this paper, we have analyzed the role of a transcriptional repressor called Slug in the regulation of adhesion molecule expression and function in epidermal keratinocytes. We report that activation of Slug, which is expressed predominantly in the basal layer of the epidermis, results in down-regulation of a number of cell adhesion molecules, including E-cadherin, and several integrins, including alpha3, beta1, and beta4. We demonstrate that Slug binds to the alpha3 promoter and that repression of alpha3 transcription by Slug is dependent on an E-box sequence within the promoter. This reduction in integrin expression is reflected in decreased cell adhesion to fibronectin and laminin-5. Despite the reduction in integrin expression and function, we do not observe any increase in differentiation. We do, however, find that activation of Slug results in a significant reduction in keratinocyte proliferation.
Collapse
Affiliation(s)
- Frances E Turner
- School of Biosciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Simon Broad
- Cancer Research UK, 44 Lincoln's Inn Fields, London WC2A 3PX, United Kingdom
| | - Farhat L Khanim
- School of Biosciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Alexa Jeanes
- School of Biosciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Sonia Talma
- School of Biosciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Sharon Hughes
- School of Medicine, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Chris Tselepis
- School of Medicine, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Neil A Hotchin
- School of Biosciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom.
| |
Collapse
|
827
|
Dohadwala M, Yang SC, Luo J, Sharma S, Batra RK, Huang M, Lin Y, Goodglick L, Krysan K, Fishbein MC, Hong L, Lai C, Cameron RB, Gemmill RM, Drabkin HA, Dubinett SM. Cyclooxygenase-2-dependent regulation of E-cadherin: prostaglandin E(2) induces transcriptional repressors ZEB1 and snail in non-small cell lung cancer. Cancer Res 2006; 66:5338-45. [PMID: 16707460 DOI: 10.1158/0008-5472.can-05-3635] [Citation(s) in RCA: 225] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Elevated tumor cyclooxygenase-2 (COX-2) expression is associated with tumor invasion, metastasis, and poor prognosis in non-small cell lung cancer (NSCLC). Here, we report that COX-2-dependent pathways contribute to the modulation of E-cadherin expression in NSCLC. First, whereas genetically modified COX-2-sense (COX-2-S) NSCLC cells expressed low E-cadherin and showed diminished capacity for cellular aggregation, genetic or pharmacologic inhibition of tumor COX-2 led to increased E-cadherin expression and resulted in augmented homotypic cellular aggregation among NSCLC cells in vitro. An inverse relationship between COX-2 and E-cadherin was shown in situ by double immunohistochemical staining of human lung adenocarcinoma tissue sections. Second, treatment of NSCLC cells with exogenous prostaglandin E(2) (PGE(2)) significantly decreased the expression of E-cadherin, whereas treatment of COX-2-S cells with celecoxib (1 mumol/L) led to increased E-cadherin expression. Third, the transcriptional suppressors of E-cadherin, ZEB1 and Snail, were up-regulated in COX-2-S cells or PGE(2)-treated NSCLC cells but decreased in COX-2-antisense cells. PGE(2) exposure led to enhanced ZEB1 and Snail binding at the chromatin level as determined by chromatin immunoprecipitation assays. Small interfering RNA-mediated knockdown of ZEB1 or Snail interrupted the capacity of PGE(2) to down-regulate E-cadherin. Fourth, an inverse relationship between E-cadherin and ZEB1 and a direct relationship between COX-2 and ZEB1 were shown by immunohistochemical staining of human lung adenocarcinoma tissue sections. These findings indicate that PGE(2), in autocrine or paracrine fashion, modulates transcriptional repressors of E-cadherin and thereby regulates COX-2-dependent E-cadherin expression in NSCLC. Thus, blocking PGE(2) production or activity may contribute to both prevention and treatment of NSCLC.
Collapse
Affiliation(s)
- Mariam Dohadwala
- Lung Cancer Research Program of the University of California at Los Angeles Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
828
|
Saito T, Nagai M, Ladanyi M. SYT-SSX1 and SYT-SSX2 Interfere with Repression of E-Cadherin by Snail and Slug: A Potential Mechanism for Aberrant Mesenchymal to Epithelial Transition in Human Synovial Sarcoma. Cancer Res 2006; 66:6919-27. [PMID: 16849535 DOI: 10.1158/0008-5472.can-05-3697] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Synovial sarcoma is a primitive mesenchymal neoplasm characterized in almost all cases by a t(X;18) fusing the SYT transcriptional coactivator gene with either SSX1 or SSX2, with the resulting fusion gene encoding an aberrant transcriptional regulator. A subset of synovial sarcoma, predominantly cases with the SYT-SSX1 fusion, shows foci of morphologic epithelial differentiation in the form of nests of glandular epithelium. The striking spontaneous mesenchymal to epithelial differentiation in this cancer is reminiscent of a developmental switch, but the only clue to its mechanistic basis has been the observation that most cases of synovial sarcoma with glandular epithelial differentiation (GED) contain SYT-SSX1 instead of SYT-SSX2. We report here that SYT-SSX1 and SYT-SSX2 interact preferentially with Snail or Slug, respectively, and prevent these transcriptional repressors from binding to the proximal E-cadherin promoter as shown by coimmunoprecipitation and chromatin immunoprecipitation. Luciferase reporter assays reveal that SYT-SSX1 and SYT-SSX2 can respectively overcome the Snail- or Slug-mediated repression of E-cadherin transcription. This provides a mechanism by which E-cadherin expression, a prerequisite of epithelial differentiation, is aberrantly derepressed in synovial sarcoma and may also explain the association of GED with the SYT-SSX1 fusion because it interferes with Snail, the stronger repressor of the E-cadherin promoter. Thus, our data provide a mechanistic basis for the observed heterogeneity in the acquisition of epithelial characteristics in synovial sarcoma and highlight the potential role of differential interactions with Snail or Slug in modulating this phenotypic transition.
Collapse
Affiliation(s)
- Tsuyoshi Saito
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA
| | | | | |
Collapse
|
829
|
Slattery C, McMorrow T, Ryan MP. Overexpression of E2A proteins induces epithelial-mesenchymal transition in human renal proximal tubular epithelial cells suggesting a potential role in renal fibrosis. FEBS Lett 2006; 580:4021-30. [PMID: 16814783 DOI: 10.1016/j.febslet.2006.06.039] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2006] [Revised: 06/06/2006] [Accepted: 06/09/2006] [Indexed: 11/24/2022]
Abstract
Epithelial-mesenchymal transition (EMT), a process whereby renal tubular epithelial cells lose phenotype and gain fibroblast-like characteristics, has been demonstrated to contribute significantly to the development of renal fibrosis. The immunosuppressant cyclosporine A (CsA) has been shown to induce renal fibrosis, a major complication of CsA therapy. The mechanisms that drive CsA-induced fibrosis remain undefined, however, CsA has been demonstrated to induce EMT in human renal proximal tubular epithelial cells (RPTEC). E2A transcription factors were identified as being upregulated by CsA treatment. To further examine the role of E2A proteins in EMT, E12 and E47 were overexpressed, alone and in combination, in human RPTEC. Both E12 and E47 elicited EMT effects on tubular epithelial cells with E47 more potent in inducing the fibroblast-like phenotype. These results indicate the important role of the E2A gene products in the progression of CsA-induced EMT and provide novel insights into CsA-induced renal fibrosis.
Collapse
Affiliation(s)
- Craig Slattery
- UCD School of Biomolecular and Biomedical Sciences, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | |
Collapse
|
830
|
Chen M, Chen LM, Chai KX. Androgen regulation of prostasin gene expression is mediated by sterol-regulatory element-binding proteins and SLUG. Prostate 2006; 66:911-20. [PMID: 16541421 DOI: 10.1002/pros.20325] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Prostasin is downregulated in hormone-refractory prostate cancers (HRPC). The mechanisms by which androgens regulate prostasin expression are unclear. METHODS LNCaP cells were treated with dihydrotestosterone (DHT), and mRNA expression of prostasin, SREBPs, SNAIL, and SLUG was examined by real-time PCR following reverse transcription. A human prostasin promoter was evaluated in HEK-293 cells co-transfected with transcription factor cDNAs. Regulation of endogenous prostasin expression by transfected SREBP-2 or SLUG was evaluated. Expression of SNAIL and SLUG mRNA in DU-145 cells treated with epidermal growth factor (EGF) was examined. RESULTS Prostasin mRNA expression in LNCaP cells was not responsive to DHT treatment. DHT marginally upregulated mRNA expression of SREBP-1c, SREBP-2, and SNAIL, but not SREBP-1a, while dramatically increased SLUG mRNA expression, in a dose-dependent manner. Co-transfection of prostasin promoter and SREBP cDNA in HEK-293 cells resulted in stimulation of promoter activity at approximately twofold by SREBP-1c, and up to sixfold by SREBP-2; while co-transfection with SNAIL or SLUG cDNA resulted in repression of promoter activity to 43% or 59%, respectively. Co-transfection of the SLUG cDNA negated SREBP-2's stimulation of prostasin promoter in a dose-dependent manner. Transfection of an SREBP-2 cDNA in HEK-293 and DU-145 resulted in upregulation of prostasin while transfection of a SLUG cDNA in LNCaP repressed prostasin expression. EGF upregulated SNAIL and SLUG mRNA in DU-145. CONCLUSIONS DHT regulates prostasin expression in prostate cells via SREBP stimulation and SLUG repression of prostasin promoter. SLUG is upregulated by DHT and EGF, providing a molecular mechanism by which epithelial cell-specific genes are silenced during prostate cancer development and progression.
Collapse
Affiliation(s)
- Mengqian Chen
- Department of Molecular Biology and Microbiology, University of Central Florida, Orlando, Florida 32816-2364, USA
| | | | | |
Collapse
|
831
|
Rukstalis JM, Ubeda M, Johnson MV, Habener JF. Transcription factor snail modulates hormone expression in established endocrine pancreatic cell lines. Endocrinology 2006; 147:2997-3006. [PMID: 16556769 DOI: 10.1210/en.2005-1396] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The development of differentiated cells from undifferentiated progenitor cells is one of the central tenets of developmental biology. However, under conditions of tissue morphogenesis, regeneration, and cancer, this process of development is reversed and fully differentiated cells transition to an undifferentiated phenotype. Here we present evidence that the zinc-finger transcription factor Snail modulates this transition in differentiated pancreatic endocrine cell lines. During passage and growth of these cell lines, Snail expression is induced in a subset of cells within the culture, concomitant with a decrease in insulin and/or glucagon expression. As the cells cluster and exit the cell division cycle, nuclear levels of Snail are reduced and hormone expression is resumed. Snail represses proinsulin and proglucagon gene transcription, and reduction of Snail levels by small interfering RNA treatment increases proinsulin gene expression. We propose that Snail modulates the dynamic balance between differentiated and dedifferentiated cells allowing their migration and proliferation. These findings may be relevant to providing approaches for the enhancement of beta-cell growth in individuals with diabetes mellitus.
Collapse
Affiliation(s)
- J Michael Rukstalis
- Laboratory of Molecular Endocrinology, Massachusetts General Hospital, Harvard Medical School, Howard Hughes Medical Institute, Boston, Massachusetts 02114, USA
| | | | | | | |
Collapse
|
832
|
Alexander NR, Tran NL, Rekapally H, Summers CE, Glackin C, Heimark RL. N-cadherin gene expression in prostate carcinoma is modulated by integrin-dependent nuclear translocation of Twist1. Cancer Res 2006; 66:3365-9. [PMID: 16585154 DOI: 10.1158/0008-5472.can-05-3401] [Citation(s) in RCA: 190] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The gain of N-cadherin expression in carcinomas has been shown to be important in the regulation of cell migration, invasion, and survival. Here, we show that N-cadherin mRNA expression in PC-3 prostate carcinoma cells is dependent on beta(1) integrin-mediated cell adhesion to fibronectin and the basic helix-loop-helix transcription factor Twist1. Depletion of Twist1 mRNA by small interfering RNA resulted in decreased expression of both Twist1 and N-cadherin and the inhibition of cell migration. Whereas Twist1 gene expression was independent of beta(1) integrin-mediated adhesion, Twist1 protein failed to accumulate in the nuclei of cells cultured in anchorage-independent conditions. The increased nuclear accumulation of Twist1 following cell attachment was suppressed by treatment with an inhibitor of Rho kinase or a beta(1) integrin neutralizing antibody. The effect of Twist1 on induction of N-cadherin mRNA required an E-box cis-element located within the first intron (+2,627) of the N-cadherin gene. These data raise the possibility that integrin-mediated adhesion to interstitial matrix proteins during metastasis differentially regulates the nuclear/cytoplasmic translocation and DNA binding of Twist1, activating N-cadherin transcription.
Collapse
Affiliation(s)
- Nelson R Alexander
- Cancer Biology Graduate Interdisciplinary Program, and Department of Surgery, University of Arizona Health Sciences Center, 1501 North Campbell Avenue, Tucson, AZ 85724, USA
| | | | | | | | | | | |
Collapse
|
833
|
Battle MA, Konopka G, Parviz F, Gaggl AL, Yang C, Sladek FM, Duncan SA. Hepatocyte nuclear factor 4alpha orchestrates expression of cell adhesion proteins during the epithelial transformation of the developing liver. Proc Natl Acad Sci U S A 2006; 103:8419-24. [PMID: 16714383 PMCID: PMC1482507 DOI: 10.1073/pnas.0600246103] [Citation(s) in RCA: 195] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Epithelial formation is a central facet of organogenesis that relies on intercellular junction assembly to create functionally distinct apical and basal cell surfaces. How this process is regulated during embryonic development remains obscure. Previous studies using conditional knockout mice have shown that loss of hepatocyte nuclear factor 4alpha (HNF4alpha) blocks the epithelial transformation of the fetal liver, suggesting that HNF4alpha is a central regulator of epithelial morphogenesis. Although HNF4alpha-null hepatocytes do not express E-cadherin (also called CDH1), we show here that E-cadherin is dispensable for liver development, implying that HNF4alpha regulates additional aspects of epithelial formation. Microarray and molecular analyses reveal that HNF4alpha regulates the developmental expression of a myriad of proteins required for cell junction assembly and adhesion. Our findings define a fundamental mechanism through which generation of tissue epithelia during development is coordinated with the onset of organ function.
Collapse
Affiliation(s)
- Michele A. Battle
- *Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226-0509
| | - Genevieve Konopka
- *Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226-0509
| | - Fereshteh Parviz
- *Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226-0509
| | - Alexandra Lerch Gaggl
- *Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226-0509
| | - Chuhu Yang
- Department of Cell Biology and Neuroscience, University of California, Riverside, CA 92521; and
- WiCell Research Institute, Madison, WI 53707
| | - Frances M. Sladek
- Department of Cell Biology and Neuroscience, University of California, Riverside, CA 92521; and
| | - Stephen A. Duncan
- *Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226-0509
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
834
|
Abstract
Members of the Snail gene superfamily, which encode zinc finger transcriptional repressors, play critical roles in the establishment of the vertebrate body plan. The Snail1 (Snai1) gene promotes epithelial-mesenchymal transitions during development and disease progression, and Snai1 null mouse embryos exhibit defects in gastrulation. However, the early embryonic lethality of Snai1 null embryos precludes the study of Snai1 function in other developmental contexts or diseases. To overcome this restriction, we generated a Snai1 conditional null allele by flanking the promoter and first two exons of the Snai1 gene with loxP sites. Cre-mediated deletion of the Snai1(flox) allele generates the Snai1(del2) allele, which behaves genetically as a Snai1 null allele. This conditional null allele will enable investigation of Snai1 function in a variety of developmental and pathological contexts.
Collapse
|
835
|
Lui WY, Lee WM. Regulation of junction dynamics in the testis--transcriptional and post-translational regulations of cell junction proteins. Mol Cell Endocrinol 2006; 250:25-35. [PMID: 16431017 DOI: 10.1016/j.mce.2005.12.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Cell junctions are the sites at which cells attach to the neighboring cells. They do not only maintain tissue integrity, their turnover also plays a crucial role in cell development and morphogenesis. In the testis, tight junctions and adherens junctions are dynamically remodeled to allow the movement of post-meiotic germ cells across the seminiferous epithelium and the timely release of spermatids into the tubular lumen. There is growing evidence that this dynamic remodeling of cell junctions is mediated by several mechanisms at the transcriptional and post-translational levels. This review summarizes what is known about the transcriptional regulation, ubiquitination and endocytosis that are involved in modulating junction dynamics in epithelial cells. It also highlights the recent findings on the regulation of junction dynamics in the testis and the specific areas that require further research for a thorough understanding of the role of junction remodeling in spermatogenesis. Understanding the junction dynamics in the seminiferous epithelium may unfold new targets for non-hormonal male contraceptive development.
Collapse
Affiliation(s)
- Wing-Yee Lui
- Department of Zoology, The University of Hong Kong, Pokfulam, Hong Kong, China
| | | |
Collapse
|
836
|
Martínez-Estrada OM, Cullerés A, Soriano FX, Peinado H, Bolós V, Martínez FO, Reina M, Cano A, Fabre M, Vilaró S. The transcription factors Slug and Snail act as repressors of Claudin-1 expression in epithelial cells. Biochem J 2006; 394:449-57. [PMID: 16232121 PMCID: PMC1408675 DOI: 10.1042/bj20050591] [Citation(s) in RCA: 229] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Claudin-1 is an integral membrane protein component of tight junctions. The Snail family of transcription factors are repressors that play a central role in the epithelial-mesenchymal transition, a process that occurs during cancer progression. Snail and Slug members are direct repressors of E-cadherin and act by binding to the specific E-boxes of its proximal promoter. In the present study, we demonstrate that overexpression of Slug or Snail causes a decrease in transepithelial electrical resistance. Overexpression of Slug and Snail in MDCK (Madin-Darby canine kidney) cells down-regulated Claudin-1 at protein and mRNA levels. In addition, Snail and Slug are able to effectively repress human Claudin-1-driven reporter gene constructs containing the wild-type promoter sequence, but not those with mutations in two proximal E-box elements. We also demonstrate by band-shift assay that Snail and Slug bind to the E-box motifs present in the human Claudin-1 promoter. Moreover, an inverse correlation in the levels of Claudin-1 and Slug transcripts were observed in breast cancer cell lines. E-box elements in the Claudin-1 promoter were found to play a critical negative regulatory role in breast cancer cell lines that expressed low levels of Claudin-1 transcript. Significantly, in invasive human breast tumours, high levels of Snail and Slug correlated with low levels of Claudin-1 expression. Taken together, these results support the hypothesis that Claudin-1 is a direct downstream target gene of Snail family factors in epithelial cells.
Collapse
|
837
|
Bindels S, Mestdagt M, Vandewalle C, Jacobs N, Volders L, Noël A, van Roy F, Berx G, Foidart JM, Gilles C. Regulation of vimentin by SIP1 in human epithelial breast tumor cells. Oncogene 2006; 25:4975-85. [PMID: 16568083 DOI: 10.1038/sj.onc.1209511] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The expression of Smad interacting protein-1 (SIP1; ZEB2) and the de novo expression of vimentin are frequently involved in epithelial-to-mesenchymal transitions (EMTs) under both normal and pathological conditions. In the present study, we investigated the potential role of SIP1 in the regulation of vimentin during the EMT associated with breast tumor cell migration and invasion. Examining several breast tumor cell lines displaying various degrees of invasiveness, we found SIP1 and vimentin expression only in invasive cell lines. Also, using a model of cell migration with human mammary MCF10A cells, we showed that SIP1 is induced specifically in vimentin-positive migratory cells. Furthermore, transfection of SIP1 cDNA in MCF10A cells increased their vimentin expression both at the mRNA and protein levels and enhanced their migratory abilities in Boyden Chamber assays. Inversely, inhibition of SIP1 expression by RNAi strategies in BT-549 cells and MCF10A cells decreased vimentin expression. We also showed that SIP1 transfection did not activate the TOP-FLASH reporter system, suggesting that the beta-catenin/TCF pathway is not implicated in the regulation of vimentin by SIP1. Our results therefore implicate SIP1 in the regulation of vimentin observed in the EMT associated with breast tumor cell migration, a pathway that may contribute to the metastatic progression of breast cancer.
Collapse
Affiliation(s)
- S Bindels
- Laboratory of Tumor and Developmental Biology, Center for Biomedical Integrated Genoproteomics, University of Liège, CHU Sart-Tilman, Liège, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
838
|
Li X, Deng W, Nail CD, Bailey SK, Kraus MH, Ruppert JM, Lobo-Ruppert SM. Snail induction is an early response to Gli1 that determines the efficiency of epithelial transformation. Oncogene 2006; 25:609-21. [PMID: 16158046 PMCID: PMC1361531 DOI: 10.1038/sj.onc.1209077] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Gli family members mediate constitutive Hedgehog signaling in the common skin cancer, basal cell carcinoma (BCC). Snail/Snai1 is rapidly induced by Gli1 in vitro, and is coexpressed with Gli1 in human hair follicles and skin tumors. In the current study, we generated a dominant-negative allele of Snail, SnaZFD, composed of the zinc-finger domain and flanking sequence. In promoter-reporter assays, SnaZFD blocked the activity of wild-type Snail on the E-cadherin promoter. Snail loss-of-function mediated by SnaZFD or by one of several short hairpin RNAs inhibited transformation of RK3E epithelial cells by Gli1. Conversely, enforced expression of Snail promoted transformation in vitro by Gli1, but not by other genes that were tested, including Notch1, ErbB2, and N-Ras. As observed for Gli1, wild-type Snail repressed E-cadherin in RK3E cells and induced blebbing of the cytoplasmic membrane. Induction of a conditional Gli1 transgene in the basal keratinocytes of mouse skin led to rapid upregulation of Snail transcripts and to cell proliferation in the interfollicular epidermis. Established Gli1-induced skin lesions exhibited molecular similarities to BCC, including loss of E-cadherin. The results identify Snail as a Gli1-inducible effector of transformation in vitro, and an early Gli1-responsive gene in the skin.
Collapse
Affiliation(s)
| | - Wentao Deng
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Clinton D. Nail
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Sarah K. Bailey
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Matthias H. Kraus
- Department of Cell Biology
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - J. Michael Ruppert
- Department of Cell Biology
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Susan M. Lobo-Ruppert
- Department of Cell Biology
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
- *Correspondence: Department of Medicine, Room 570 WTI, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294-3300. Phone: (205) 975-0556; Fax: (205) 934-9511;
| |
Collapse
|
839
|
Abstract
AIM: To explore the expression of cadherin isoforms in cultured human gastric carcinoma cells and its regulation.
METHODS: The expressions of cell adhesion molecules (including E-cadherin, N-cadherin, α-catenin, β-catenin) and cadherin transcription factors including snail, slug and twist were determined by reverse transcriptase-polymerase chain reaction(RT-PCR), immunoblotting and immunofluorescence in SV40-immortalized human gastric cell line Ges-1 and human gastric cancer cell lines MGC-803, BGC-823 and SGC-7901.
RESULTS: All cell lines expressed N-cadherin, but not E-cadherin. N-cadherin immunofluorescence was detected at cell membranous adherents junctions where co-localization with immunofluorescent staining of inner surface adhesion proteins α- and β-catenins was observed. The transformed Ges-1 and gastric cancer cell lines all expressed transcription factors (snail, slug and twist) which inhibited the expression of E-cadherin and triggered epithelial-mesenchymal transformation.
CONCLUSION: Cadherin isoforms can change from E-cadherin to N-cadherin in transformed human gastric cancer cells, which is associated with intracellular events of stomach carcinogenesis and high expression of corresponding transcription factors.
Collapse
Affiliation(s)
- Bing-Jing Wang
- Beijing Institute for Cancer Research, Peking University, Beijing 100036, China
| | | | | |
Collapse
|
840
|
Shih JY, Tsai MF, Chang TH, Chang YL, Yuan A, Yu CJ, Lin SB, Liou GY, Lee ML, Chen JJW, Hong TM, Yang SC, Su JL, Lee YC, Yang PC. Transcription repressor slug promotes carcinoma invasion and predicts outcome of patients with lung adenocarcinoma. Clin Cancer Res 2006; 11:8070-8. [PMID: 16299238 DOI: 10.1158/1078-0432.ccr-05-0687] [Citation(s) in RCA: 170] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE In a previous genome-wide gene expression profiling analysis using an invasion cancer cell lines model, we have identified Slug as selectively overexpressed in the highly invasive cancer cells. Here, we investigated the clinical significance of Slug in lung adenocarcinoma and the role of Slug in the process of cancer cell invasion and metastasis. EXPERIMENTAL DESIGN Real-time quantitative reverse transcription-PCR was used to investigate Slug mRNA in surgically resected lung adenocarcinoma of 54 patients and its correlation with survival. We overexpressed Slug in a lung adenocarcinoma cell line with very low Slug levels and investigated the in vitro and in vivo effects of Slug expression. RESULTS High expression of Slug mRNA in lung cancer tissue was significantly associated with postoperative relapse (P = 0.03) and shorter patient survival (P < 0.001). The overexpression of Slug enhanced xenograft tumor growth and increased microvessel counts in angiogenesis assay. Both inducible and constitutive overexpression of Slug suppressed the expression of E-cadherin and increased the in vitro invasive ability. Zymography revealed increased matrix metalloproteinase-2 activity in Slug overexpressed cells. ELISA, reverse transcription-PCR, and immunohistochemistry confirmed the increase of matrix metalloproteinase-2 proteins and mRNA in Slug overexpressed cells and xenograft tumors. CONCLUSIONS Slug expression can predict the clinical outcome of lung adenocarcinoma patients. Slug is a novel invasion-promoting gene in lung adenocarcinoma.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/pathology
- Adenocarcinoma/surgery
- Aged
- Animals
- Blotting, Northern
- Cadherins/metabolism
- Cell Line, Tumor
- Cell Movement
- Cell Proliferation
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Lung Neoplasms/surgery
- Male
- Matrix Metalloproteinase 2/metabolism
- Mice
- Mice, SCID
- Middle Aged
- Neoplasm Invasiveness
- Neoplasm Recurrence, Local
- Neoplasm Transplantation
- Neoplasms, Experimental/blood supply
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/metabolism
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Prognosis
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Snail Family Transcription Factors
- Survival Analysis
- Transcription Factors/genetics
- Transcription Factors/physiology
- Transfection
- Transplantation, Heterologous
- Treatment Outcome
Collapse
Affiliation(s)
- Jin-Yuan Shih
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
841
|
Liu YN, Lee WW, Wang CY, Chao TH, Chen Y, Chen JH. Regulatory mechanisms controlling human E-cadherin gene expression. Oncogene 2006; 24:8277-90. [PMID: 16116478 DOI: 10.1038/sj.onc.1208991] [Citation(s) in RCA: 156] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In cancer cells, loss of E-cadherin gene expression caused dysfunction of the cell-cell junction system, triggering cancer invasion and metastasis. Therefore, E-cadherin is an important tumor-suppressor gene. To understand how E-cadherin gene expression is regulated in cancer cells, we have used E-cadherin-positive and -negative expressing cells to find out the possible up- or down regulating transcription factors in human E-cadherin regulatory sequences. Functional analysis of human E-cadherin regulatory sequences constructs indicated that AML1, Sp1, and p300 may play important roles in promoting E-cadherin expression. In addition, we found there are four HNF3-binding sites in human E-cadherin regulatory sequences. The exogenous HNF3 can enhance the E-cadherin promoter activity in metastatic breast cancer cells and the metastatic breast cancer cells stably transfected with HNF3 showed re-expression of E-cadherin. The HNF3 stable transfectants changed from mesenchymal-like into epithelial morphology. The transwell assays showed the re-expressed E-cadherin reduced cell motility of metastatic breast cancer cells. These results suggested HNF3 may play important roles in the upregulation of the E-cadherin promoter, with the consequent re-expression of E-cadherin, thus reducing the metastatic potential of breast cancer cells. These findings suggested HNF3 plays important roles in the upregulation of the E-cadherin gene and may be able to reduce the motility of metastatic breast cancer cells.
Collapse
Affiliation(s)
- Yan-Nan Liu
- Graduate Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | | | | | | | | | | |
Collapse
|
842
|
Bermejo-Rodríguez C, Pérez-Caro M, Pérez-Mancera PA, Sánchez-Beato M, Piris MA, Sánchez-García I. Mouse cDNA microarray analysis uncovers Slug targets in mouse embryonic fibroblasts. Genomics 2006; 87:113-8. [PMID: 16311016 DOI: 10.1016/j.ygeno.2005.09.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2005] [Revised: 09/16/2005] [Accepted: 09/22/2005] [Indexed: 11/28/2022]
Abstract
There is a need to reveal mechanisms that account for maintenance of the mesenchymal phenotype in normal development and cancer. Slug (approved gene symbol Snai2), a member of the Snail gene family of zinc-finger transcription factors, is believed to function in the maintenance of the nonepithelial phenotype. This study identified candidate Slug target genes linked to Slug gene suppression in primary mouse embryonic fibroblasts. Expression analyses were performed with a mouse cDNA microarray (Mousechip-CNIO) containing 15,000 clones. A total of 15 novel Slug target species were validated by real-time PCR or Western analyses. These included self-renewal genes (Bmi1, Nanog, Gfi1), epithelial-mesenchymal genes (Tcfe2a, Ctnb1, Sin3a, Hdac1, Hdac2, Muc1, Cldn11), survival genes (Bcl2, Bbc3), and cell cycle/damage genes (Cdkn1a, Rbl1, Mdm2). Expression patterns were studied in wild-type MEFs and Slug-deficient MEFs. Slug-complementation studies recovered aberrant gene expression in cells lacking Slug, indicating that these genes were regulated directly by Slug. These results highlight their potential roles in mediating Slug function in mesenchymal cells and may help to identify novel therapeutic biomarkers in cancers linked to Slug.
Collapse
Affiliation(s)
- Camino Bermejo-Rodríguez
- Laboratorio 13, Instituto de Biología Molecular y Celular del Cáncer, CSIC/Universidad de Salamanca, Campus Unamuno, 37007 Salamanca, Spain
| | | | | | | | | | | |
Collapse
|
843
|
Jordà M, Olmeda D, Vinyals A, Valero E, Cubillo E, Llorens A, Cano A, Fabra A. Upregulation of MMP-9 in MDCK epithelial cell line in response to expression of the Snail transcription factor. J Cell Sci 2005; 118:3371-85. [PMID: 16079281 DOI: 10.1242/jcs.02465] [Citation(s) in RCA: 187] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Overexpression of the transcription factor Snail in epithelial MDCK cells promotes the epithelial-mesenchymal transition (EMT) and the acquisition of an invasive phenotype. We report here that the expression of Snail is associated with an increase in the promoter activity and expression of the matrix metalloproteinase MMP-9. The effect of Snail silencing on MMP-9 expression corroborates this finding. Induced transcription of MMP-9 by Snail is driven by a mechanism dependent on the MAPK and phosphoinositide 3-kinase (PI3K) signalling pathways. Although other regions of the promoter were required for a complete stimulation by Snail, a minimal fragment (nucleotides -97 to +114) produces a response following an increased phosphorylation of Sp-1 and either Sp-1 or Ets-1 binding to the GC-box elements contained in this region. The expression of a dominant negative form of MEK decreased these complexes. A moderate increase in the binding of the nuclear factor kappaB (NFkappaB) to the upstream region (nucleotide -562) of the MMP-9 promoter was also observed in Snail-expressing cells. Interestingly, oncogenic H-Ras (RasV12) synergistically co-operates with Snail in the induction of MMP-9 transcription and expression. Altogether, these results indicate that MMP-9 transcription is activated in response to Snail expression and that it might explain, at least in part, the invasive properties of the Snail-expressing cells.
Collapse
Affiliation(s)
- Mireia Jordà
- Centre d'Oncologia Molecular, IDIBELL-Institut de Recerca Oncològica, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
844
|
Song J, Jie C, Polk P, Shridhar R, Clair T, Zhang J, Yin L, Keppler D. The candidate tumor suppressor CST6 alters the gene expression profile of human breast carcinoma cells: down-regulation of the potent mitogenic, motogenic, and angiogenic factor autotaxin. Biochem Biophys Res Commun 2005; 340:175-82. [PMID: 16356477 DOI: 10.1016/j.bbrc.2005.11.171] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Accepted: 11/23/2005] [Indexed: 02/06/2023]
Abstract
We recently coined CST6 as a novel candidate tumor suppressor gene for breast cancer. CST6 indeed is expressed in the normal human breast epithelium, but little or not at all in breast carcinomas and breast cancer cell lines. Moreover, ectopic expression of CST6 in human breast cancer cells suppressed cell proliferation, migration, invasion, and orthotopic tumor growth. To obtain insights into the molecular mechanism by which CST6 exhibits its pleiotropic effects on tumor cells, we compared global gene expression profiles in mock- and CST6-transfected human MDA-MB-435S cells. Out of 12,625 transcript species, 61 showed altered expression. These included genes for extracellular matrix components, cytokines, kinases, and phosphatases, as well as several key transcription factors. TaqMan PCR assays were used to confirm the microarray data for 7 out of 11 genes. One down-regulated gene product, secreted autotaxin/lyso-phospholipase D, was of particular interest because its down-regulation by CST6 could explain most of CST6's effect on the breast cancer cells. This study thus provides the first evidence that CST6 plays a role in the modulation of genes, particularly, genes that are highly relevant to breast cancer progression.
Collapse
Affiliation(s)
- Jin Song
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA
| | | | | | | | | | | | | | | |
Collapse
|
845
|
Andersen H, Mejlvang J, Mahmood S, Gromova I, Gromov P, Lukanidin E, Kriajevska M, Mellon JK, Tulchinsky E. Immediate and delayed effects of E-cadherin inhibition on gene regulation and cell motility in human epidermoid carcinoma cells. Mol Cell Biol 2005; 25:9138-50. [PMID: 16199889 PMCID: PMC1265771 DOI: 10.1128/mcb.25.20.9138-9150.2005] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The invasion suppressor protein, E-cadherin, plays a central role in epithelial cell-cell adhesion. Loss of E-cadherin expression or function in various tumors of epithelial origin is associated with a more invasive phenotype. In this study, by expressing a dominant-negative mutant of E-cadherin (Ec1WVM) in A431 cells, we demonstrated that specific inhibition of E-cadherin-dependent cell-cell adhesion led to the genetic reprogramming of tumor cells. In particular, prolonged inhibition of cell-cell adhesion activated expression of vimentin and repressed cytokeratins, suggesting that the effects of Ec1WVM can be classified as epithelial-mesenchymal transition. Both short-term and prolonged expression of Ec1WVM resulted in morphological transformation and increased cell migration though to different extents. Short-term expression of Ec1WVM up-regulated two AP-1 family members, c-jun and fra-1, but was insufficient to induce complete mesenchymal transition. AP-1 activity induced by the short-term expression of Ec1WVM was required for transcriptional up-regulation of AP-1 family members and down-regulation of two other Ec1WVM-responsive genes, S100A4 and igfbp-3. Using a dominant-negative mutant of c-Jun (TAM67) and RNA interference-mediated silencing of c-Jun and Fra-1, we demonstrated that AP-1 was required for cell motility stimulated by the expression of Ec1WVM. In contrast, Ec1WVM-mediated changes in cell morphology were AP-1-independent. Our data suggest that mesenchymal transition induced by prolonged functional inhibition of E-cadherin is a slow and gradual process. At the initial step of this process, Ec1WVM triggers a positive autoregulatory mechanism that increases AP-1 activity. Activated AP-1 in turn contributes to Ec1WVM-mediated effects on gene expression and tumor cell motility. These data provide novel insight into the tumor suppressor function of E-cadherin.
Collapse
Affiliation(s)
- Henriette Andersen
- Department of Cancer Studies and Molecular Medicine, University of Leicester, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
846
|
Rosivatz E, Becker KF, Kremmer E, Schott C, Blechschmidt K, Höfler H, Sarbia M. Expression and nuclear localization of Snail, an E-cadherin repressor, in adenocarcinomas of the upper gastrointestinal tract. Virchows Arch 2005; 448:277-87. [PMID: 16328348 DOI: 10.1007/s00428-005-0118-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2005] [Accepted: 10/18/2005] [Indexed: 01/11/2023]
Abstract
Transcriptional E-cadherin down-regulation can be mediated by Snail, a zinc finger transcription factor. To be able to examine nuclear Snail immunoreactivity in archival human cancers, we established a monoclonal antibody against the purified human Snail protein. The specificity of the selected rat antibody Sn9H2 was demonstrated by Western blot analysis using extracts from different cell lines and by immunofluorescence and immunohistochemistry of primary tissues. Subsequently, a series of 340 adenocarcinomas of the upper gastrointestinal tract, including tumours from the oesophagus (n=154), cardia (n=102) and stomach (n=84), arranged in tissue microarrays, were examined for Snail expression and were correlated to E-cadherin expression and clinico-pathological parameters. Nuclear Snail immunoreactivity was seen in 27 tumours (7.9%) and tended to be more frequent in oesophageal adenocarcinomas (11.1%) than in cardiac (6.9%) or gastric (3.6%) carcinomas (p=0.0428). In 35% of the Snail-positive cases, E-cadherin immunoreactivity was lost. No correlation was found for nuclear Snail expression and tumour grade, Lauren's classification, WHO classification, tumour stage and tumour size. The pattern of Snail expression observed with our new hybridoma, Sn9H2, which is currently the only antibody that reacts with endogenous nuclear (active) Snail, suggests only a minor role of Snail in tumours of the upper gastrointestinal tract.
Collapse
Affiliation(s)
- Erika Rosivatz
- Institut für Pathologie, Technische Universität München, Trogerstrasse 18, München, Germany
| | | | | | | | | | | | | |
Collapse
|
847
|
Wilson CA, Cajulis EE, Green JL, Olsen TM, Chung YA, Damore MA, Dering J, Calzone FJ, Slamon DJ. HER-2 overexpression differentially alters transforming growth factor-beta responses in luminal versus mesenchymal human breast cancer cells. Breast Cancer Res 2005; 7:R1058-79. [PMID: 16457687 PMCID: PMC1410754 DOI: 10.1186/bcr1343] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2005] [Revised: 09/27/2005] [Accepted: 10/06/2005] [Indexed: 12/29/2022] Open
Abstract
Introduction Amplification of the HER-2 receptor tyrosine kinase has been implicated in the pathogenesis and aggressive behavior of approximately 25% of invasive human breast cancers. Clinical and experimental evidence suggest that aberrant HER-2 signaling contributes to tumor initiation and disease progression. Transforming growth factor beta (TGF-β) is the dominant factor opposing growth stimulatory factors and early oncogene activation in many tissues, including the mammary gland. Thus, to better understand the mechanisms by which HER-2 overexpression promotes the early stages of breast cancer, we directly assayed the cellular and molecular effects of TGF-β1 on breast cancer cells in the presence or absence of overexpressed HER-2. Methods Cell proliferation assays were used to determine the effect of TGF-β on the growth of breast cancer cells with normal or high level expression of HER-2. Affymetrix microarrays combined with Northern and western blot analysis were used to monitor the transcriptional responses to exogenous TGF-β1 in luminal and mesenchymal-like breast cancer cells. The activity of the core TGF-β signaling pathway was assessed using TGF-β1 binding assays, phospho-specific Smad antibodies, immunofluorescent staining of Smad and Smad DNA binding assays. Results We demonstrate that cells engineered to over-express HER-2 are resistant to the anti-proliferative effect of TGF-β1. HER-2 overexpression profoundly diminishes the transcriptional responses induced by TGF-β in the luminal MCF-7 breast cancer cell line and prevents target gene induction by a novel mechanism that does not involve the abrogation of Smad nuclear accumulation, DNA binding or changes in c-myc repression. Conversely, HER-2 overexpression in the context of the mesenchymal MDA-MB-231 breast cell line potentiated the TGF-β induced pro-invasive and pro-metastatic gene signature. Conclusion HER-2 overexpression promotes the growth and malignancy of mammary epithelial cells, in part, by conferring resistance to the growth inhibitory effects of TGF-β. In contrast, HER-2 and TGF-β signaling pathways can cooperate to promote especially aggressive disease behavior in the context of a highly invasive breast tumor model.
Collapse
Affiliation(s)
- Cindy A Wilson
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | | | - Jennifer L Green
- Department of Biology, California Institute of Technology, Pasadena, CA 91125, USA
| | - Taylor M Olsen
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | | | | | - Judy Dering
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | | | - Dennis J Slamon
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
848
|
Hay ED. The mesenchymal cell, its role in the embryo, and the remarkable signaling mechanisms that create it. Dev Dyn 2005; 233:706-20. [PMID: 15937929 DOI: 10.1002/dvdy.20345] [Citation(s) in RCA: 453] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
This review centers on the role of the mesenchymal cell in development. The creation of this cell is a remarkable process, one where a tightly knit, impervious epithelium suddenly extends filopodia from its basal surface and gives rise to migrating cells. The ensuing process of epithelial-mesenchymal transformation (EMT) creates the mechanism that makes it possible for the mesenchymal cell to become mobile, so as to leave the epithelium and move through the extracellular matrix. EMT is now recognized as a very important mechanism for the remodeling of embryonic tissues, with the power to turn an epithelial somite into sclerotome mesenchyme, and the neural crest into mesenchyme that migrates to many targets. Thus, the time has come for serious study of the underlying mechanisms and the signaling pathways that are used to form the mesenchymal cell in the embryo. In this review, I discuss EMT centers in the embryo that are ready for such serious study and review our current understanding of the mechanisms used for EMT in vitro, as well as those that have been implicated in EMT in vivo. The purpose of this review is not to describe every study published in this rapidly expanding field but rather to stimulate the interest of the reader in the study of the role of the mesenchymal cell in the embryo, where it plays profound roles in development. In the adult, mesenchymal cells may give rise to metastatic tumor cells and other pathological conditions that we will touch on at the end of the review.
Collapse
Affiliation(s)
- Elizabeth D Hay
- Harvard Medical School, Department of Cell Biology, Boston, Massachusetts 02115, USA.
| |
Collapse
|
849
|
Kondo M, Cubillo E, Tobiume K, Shirakihara T, Fukuda N, Suzuki H, Shimizu K, Takehara K, Cano A, Saitoh M, Miyazono K. A role for Id in the regulation of TGF-beta-induced epithelial-mesenchymal transdifferentiation. Cell Death Differ 2005; 11:1092-101. [PMID: 15181457 DOI: 10.1038/sj.cdd.4401467] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Epithelial-mesenchymal transdifferentiation (EMT) is a critical morphogenic event that occurs during embryonic development and during the progression of various epithelial tumors. EMT can be induced by transforming growth factor (TGF)-beta in mouse NMuMG mammary epithelial cells. Here, we demonstrate a central role of helix-loop-helix factors, E2A and inhibitor of differentiation (Id) proteins, in TGF-beta-induced EMT. Epithelial cells ectopically expressing E2A adopt a fibroblastic phenotype and acquire migratory/invasive properties, concomitant with the suppression of E-cadherin expression. Id proteins interacted with E2A proteins and antagonized E2A-dependent suppression of the E-cadherin promoter. Levels of Id proteins were dramatically decreased by TGF-beta. Moreover, NMuMG cells overexpressed Id2 showed partial resistance to TGF-beta-induced EMT. Id proteins thus inhibit the action of E2A proteins on the expression of E-cadherin, but after TGF-beta stimulation, E2A proteins are present in molar excess of the Id proteins, thus over-riding their inhibitory function and leading to EMT.
Collapse
Affiliation(s)
- M Kondo
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
850
|
Schmidt CR, Gi YJ, Patel TA, Coffey RJ, Beauchamp RD, Pearson AS. E-cadherin is regulated by the transcriptional repressor SLUG during Ras-mediated transformation of intestinal epithelial cells. Surgery 2005; 138:306-12. [PMID: 16153441 DOI: 10.1016/j.surg.2005.06.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2005] [Revised: 06/01/2005] [Accepted: 06/05/2005] [Indexed: 12/17/2022]
Abstract
BACKGROUND Loss of the cell membrane protein E-cadherin is a critical event during Ras-mediated transformation of intestinal epithelial cells. The purpose of our study is to determine if activation of the transcriptional repressor SLUG is an important component of the mechanism of Ras-induced loss of E-cadherin. METHODS Rat intestinal epithelial (RIE) cells were engineered to express mutated human Ha-Ras(Val12) complementary DNA (H-Ras cells). Cell morphology was examined by light microscopy. RNA and protein expression were measured by semiquantitative polymerase chain reaction and Western blot analyses, respectively. Short interfering RNA with 2 different oligos was used to knock down the expression of SLUG. RESULTS Oncogenic ras induces upregulation of the transcriptional repressor SLUG and subsequent downregulation of the junctional protein E-cadherin. Gene silencing of SLUG by short interfering RNA allows E-cadherin to be reexpressed. E-cadherin protein reexpression allows partial rescue of the transformed phenotype. CONCLUSION These data suggest a mechanism whereby Ras signaling causes an upregulation of transcriptional repressors and subsequent downregulation of E-cadherin as a malignant phenotype is propagated.
Collapse
Affiliation(s)
- Carl R Schmidt
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, USA
| | | | | | | | | | | |
Collapse
|