Role of E-cadherin in Tumor Metastasis and Discovery of Compounds Targeting Metastatic Cancer Cells by Tamer T. Onder B.A. Biology B.A. History Cornell University, 2002 SUBMITTED TO THE DEPARTMENT OF BIOLOGY IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOHPY IN BIOLOGY AT THE MASSACHUSETTS INSTITUTE OF TECHNOLOGY JUNE 2008 © 2008 Massachusetts Institute of Technology. All rights reserved Signature of Author: Department of Biology Certified by: - j N, Robert A. Weinberg Professor of Biology Thesis Supervisor Accepted by: I S MASSACHU OF TEGHNot.OGy Stephen P. Bell Professor of Biology Chairperson, Graduate Committee MAY 2 9 2008 LIBRARIES 1 IARCHIVS Role of E-cadherin in Tumor Metastasis and Discovery of Compounds Targeting Metastatic Cancer Cells by Tamer T. Onder Submitted to the Department of Biology on May 23, 2008 in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy Abstract The epithelial cell adhesion molecule E-cadherin is often downregulated during carcinoma progression and metastatic spread of tumors. However, the precise mechanism and molecular basis of metastasis promotion by E-cadherin loss is not completely understood. To investigate its role in metastasis, I utilized two distinct methods of E-cadherin inhibition that distinguish between E-cadherin's cell-cell adhesion and intracellular signaling functions. While the disruption of cell-cell contacts alone does not enable metastasis in vivo, the loss of E-cadherin protein does, through induction of an epithelial-to-mesenchymal transition (EMT), invasiveness and anoikis-resistance. E-cadherin binding partner f3-catenin is necessary but not sufficient for these phenotypes. In addition, gene expression analysis shows that E-cadherin loss results in the induction of multiple transcription factors, at least one of which, Twist, is necessary for Ecadherin loss-induced metastasis. These findings indicate that E-cadherin loss in tumors contributes to metastatic dissemination by inducing wide-ranging transcriptional and functional changes. In addition to promoting metastasis, loss of E-cadherin and the accompanying EMT renders cells resistant to conventional chemotherapeutic drugs. As the cells that have undergone an EMT represent the pool of cancer cells most competent to metastasize and lead to tumor recurrence, it is of vital importance to find therapies that effectively target such cells. Paired cell lines that differ in their differentiation state were utilized to discover compounds with selective toxicity against cells that have undergone an EMT. High-throughput screening of small molecule libraries resulted in a number of compounds that specifically affect the viability of cells that have undergone an EMT while having minimal cytotoxic effects on control epithelial cells. These studies establish a proof-of-principle for discovering compounds that target highly metastatic and otherwise chemotherapy resistant cancer cells. Thesis Advisor: Robert A. Weinberg Title: Professor of Biology, MIT; Member, Whitehead Institute for Biomedical Research Biographical note TAMER ONDER Whitehead Institute for Biomedical Research 9 Cambridge Center, Room 311 Cambridge, MA 02142 (617) 258-5176 onder@mit.edu Education Cambridge, MA Massachusetts Institute of Technology Ph.D. Candidate in Department of Biology. Degree expected June 2008. Cornell University Ithaca, NY Bachelor of Arts, Biology, Magna Cum Laude, May 2002. Bachelor of Arts, History Robert College of Istanbul Research Experience Istanbul, Turkey Massachusetts Institute of Technology Cambridge, MA Ph.D. thesis, Laboratory of Dr. Robert A. Weinberg "Role of E-cadherin in Tumor Metastasis and Discovery of Compounds Targeting Metastatic Cancer Cells" (2002-2008) Cornell University Ithaca, NY Senior Honors Thesis, Laboratory of Dr.Volker Vogt "Cleavage of Rous Sarcoma Virus Gag-Pol Protein" (2001- 2002) Cornell University Ithaca, NY Undergraduate Research Assistant, Laboratory of Dr. Ross MacIntyre (2000) Publications Tamer T. Onder, Piyush B. Gupta, Sendurai A. Mani, Jing Yang, Eric S. Lander and Robert A. Weinberg. (2008) Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways. CancerResearch 68(10):3645-54. Christina Scheel, Tamer T. Onder, Antoine Karnoub and Robert A. Weinberg. (2007) Adaptation versus Selection: The Origins of Metastatic Behavior. CancerResearch 67(24):11476-9. Tamer T. Onder, Piyush B. Gupta, Eric S. Lander and Robert A. Weinberg. (2008) Identification of small molecules exhibiting specific toxicity for E-cadherin-deficient metastatic cancer cells. In preparation. Priyamvada Rai, Tamer T. Onder, Jennifer J. Young, Bo Pang, Jose McFaline, Peter C. Dedon and Robert A. Weinberg. (2008): Elimination of oxidized nucleotides is necessary to prevent onset of cellular senescence. Nature Cell Biology. In Review. Akira Orimo, Elinor Ng Eaton, Christina Scheel, Ittai Ben-Porath, Tamer T. Onder, Zhigang Wang, Andrea Richardson, and Robert A. Weinberg. (2008) SDF-1-CXCR4 autocrine signaling maintains tumor-promoting myofibroblasts in the stroma of human breast carcinomas. Submitted. Awards Department of Defense Breast Cancer Research Pre-doctoral Traineeship Award (2004-2007) Hughes Scholars Fellowship for Summer Research (2001) Teaching Experience Massachusetts Institute of Technology Cambridge, MA 7.16 Experimental Molecular Biology: Biotechnology II. Teaching Assistant (Spring 2006) 7.013 Introductory Biology. Teaching Assistant. (Fall 2004) Invited Talks International Congress of Molecular Medicine, Istanbul, March 2007 Conference Presentations Gordon Research Conference on Signaling by Adhesion Receptors, (poster) 2006, South Hadley, MA Acknowledgements I would like to begin by thanking my thesis advisor Dr. Robert A. Weinberg. Without his support and guidance, none of this would have been possible. I also want to thank him for giving me a great deal of freedom in the lab and launching me on the path to becoming an independent scientist. Last but not least, I am grateful for his mentorship and caring. I will never forget the day he came to my apartment with baskets of chicken soup as I was trying to recover from a bout of mononucleosis! I would like to thank my thesis committee members, Drs. David Sabatini and Richard Hynes for their time and advice on both scientific and career questions. I am particularly grateful to Dr. Sabatini for his help and advice during my search for a post-doctoral position. I am also grateful to Drs. David Housman and Karen Cichowski for giving the time and effort to be on my thesis defense committee. I thank members of the Weinberg Lab for providing me with a vibrant scientific environment. I thoroughly enjoyed our daily meetings and discussions. Many lab members, both current and former, have given me invaluable scientific and technical help and advice. Thank you especially to Sheila Stewart for introducing me to the Weinberg lab during my rotation, and to Jing Yang and Sendurai Mani for reagents and discussions. Ferenc Reinhardt assisted with many animal experiments and I am grateful for all his help. I am also grateful to Christine Hickey and Sumiko Williams for all the support they have given me, especially during the final stretch. I owe special gratitude to Piyush Gupta, my former baymate and fellow graduate student, with whom I had many fruitful collaborations throughout this thesis. I benefited immensely from his optimism in the face of experimental adversity. I also want to thank him for introducing me to the world of the "high throughput" way of doing science. On my first day at Whitehead orientation, I met with a fellow newcomer to the Weinberg lab and she has been one of my best friends over the years. Priya, I thank you for everything from hours spent at coffee breaks to the "thoughtful" discussions we had. You have been like a big sister to me and I consider it my luck to have known you. Although graduate school is a transforming experience, the real life-changing event for me has been meeting my fiancee Tugba Bagci. A fellow biologist and graduate student, she has been there for me throughout my years in Boston. I cannot thank her enough for her support and the meaning she gave to my life. Finally I thank my parents, Umit and Turkan Onder, for their never-ending support and caring. I can only imagine how difficult it must have been for them to live away from their only son for the past 10 years. I hope it was all worth it and I make you proud at the end. Table of Contents Abstract ................................................... B iographical note .......................................................................................................... Acknow ledgem ents...................................................................................................... T able of C ontents ......................................................................................................... 2 3 5 6 8 Chapterl : Introduction ................................................................................................ Metastasis is the primary cause of cancer-related mortality ..................................... 9 Invasion-metastasis cascade.................................................................................... 9 Multi-step nature of the invasion-metastasis cascade............................... ...... 10 ........ 14 Epithelial-to-mesenchymal transition (EMT) ....................................... EMT is a normal physiological process................................... .................................. 14 Evidence for the EMT in carcinoma progression .......................................... ...... 15 EMT in the invasion/metastasis cascade............................................ 17 E-cadherin is an important epithelial cell adhesion molecule ...................................... 22 Structure of E-cadherin ................................................................................................. 23 3-catenin and the Wnt pathway .......................................................................... 24 Mechanisms of E-cadherin loss in human tumors ...................................... .... . 25 Main Questions Addressed ..................................................................................... 28 Figures........................................... ........................................................................ 30 References ..................................................................................................................... 32 Chapter 2: Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathw ays ................................................................................................... 42 Introduction ................................................................................................................... 43 Results ................................................... 46 Characterization of in vitro phenotypes resulting from expression of E-cadherin shRNA and dominant-negative E-cadherin ....................................... ..... 46 Primary tumor formation and metastasis of E-cadherin-inhibited cells ................. 48 Functional differences in cell motility, invasion, and anoikis-resistance .................. 50 Tumor initiating capacity of E-cadherin inhibited cells ..................................... 51 Molecular mechanism of E-cadherin-loss induced EMT ..................................... 52 The role of 03-catenin in EMT and metastasis induced by E-cadherin loss ............ 53 Gene expression profiling identifies global changes resulting from E-cadherin loss ............................................................................................ 55 Expression of multiple transcriptional regulators are induced upon E-cadherin loss but not cell-cell disaggregation......................................................................57 Upregulation of Twist upon loss of E-cadherin and its functional role .................. 58 Materials and Methods.................................................................................................. 62 Figures................................................................................................................................. 68 Acknowledgements....................................................................................................... 83 R eferences .............................. ................................... . . ............................................ 84 Chapter 3: Chemoresistance and selective inhibition of cancer cells that have undergone an epithelial-to-mesenchymal transition .................................................. 99 Introduction ................................................................................................................. 100 Results........................................... ....................................................................... 106 106 Chemoresistance in E-cadherin-loss induced EMT ........................................ 108 Chemical compound screen ..................................... Validation of screen hits ........................................................... 111 113 Effects of salinomycin on cancer stem cells ..................................... 118 Figures .............................................. References ................................................................................................................... 125 Chapter 4: Conclusions and Future Directions ..................................... E-cadherin and Metastasis ..................................... Loss of E-cadherin as an inducer of EMT ..................................... Mechanism of EMT induction upon E-cadherin loss .................... .................... EMT and Chemoresistance ..................................... Chemical screen identifies compounds targeting cells in the mesenchymal state...... Potential mechanisms of action and limitations of salinomycin.............. ............... Final perspective ........................................ References ................................................................................................................... A ppendix 1................................... ................................................................ ........... A ppendix 2................................................... A ppendix 3................................................... 129 130 132 133 137 140 142 143 145 161 172 177 Chapter 1 Introduction Metastasis is the primary cause of cancer-related mortality Metastasis is the spread of cancer cells from a primary tumor to secondary sites throughout the body. While patients with primary tumors, if they are detected early, can be cured with surgical removal and chemo/radiotherapy, those that present with metastases have a much poorer prognosis. In fact, 90% of cancer-related deaths arise from metastatic growths of cancers that become resistant to therapy. Therefore, it is of vital importance to understand the molecular mechanisms that allow cancer cells to metastasize with the hope that such an understanding will enable the development of novel therapeutic approaches against metastatic and therapy-resistant cancers. Invasion-metastasis cascade The majority of cancers originate in epithelial tissues, resulting in carcinomas. During the initial steps of tumorigenesis, normal epithelial cells acquire genetic and epigenetic alterations that confer multiple traits necessary for primary tumor formation, including self-sufficiency in growth signals, extended replicative potential, and a capacity for inducing angiogenesis (Hanahan and Weinberg, 2000). However, the process of dissemination presents multiple additional challenges for the carcinoma cells residing in the confines of an epithelium. The multiple physical steps that the carcinoma cells have to complete in order to form distant metastases are referred to as the "invasion-metastasis cascade" (Weinberg, 2007). Multi-step nature of the invasion-metastasis cascade Local invasion and intravasation Within a normal epithelium, individual epithelial cells are held in place by strong cell-cell and cell-matrix adhesions and are separated from adjacent stromal cells by a dense network of glycoproteins and proteoglycans known as the basement membrane. Breaching this barrier is the first step toward malignancy for the incipient transformed cells. In fact, those tumors that have not yet done so are classified as benign or in situ and their prognosis is significantly better (Weinberg, 2007). Local invasion is a coupled process involving both extracellular matrix (ECM) degradation and cellular motility. Proteolytic degradation of the basement membrane happens mainly through the actions of matrix metalloproteases (MMPs). MMP-mediated cleavage of the ECM may also lead to the release of sequestered growth factors and chemokines that increase proliferation and motility of tumor cells (Egeblad and Werb, 2002). With local invasion cancer cells gain more direct access to blood vessels, which provide enhanced nutrients and oxygen to the growing tumor. Except in a few types of cancers, such as lobular breast carcinomas, where single cells can be observed to invade into the stroma, local invasion of most carcinomas occurs primarily through collective (en masse) invasion into nearby stroma (Friedl and Wolf, 2003). Dissemination of cancer cells throughout the body occurs mainly via the bloodstream. Lymphatic spread is also widely observed in some carcinomas, and this form of metastasis is thought to be an early prognostic indicator of tumor invasiveness (Wong and Hynes, 2006). How the individual or clusters of carcinoma cells enter the lumina of blood or lymphatic vessels is not completely understood. One molecular determinant of the intravasation process was discovered by Yang et al. from our laboratory. In that study, the transcription factor Twist was found to be essential for the intravasation and eventual lung metastasis of murine mammary carcinoma cells implanted subcutaneously (Yang et al., 2004). As will be discussed below, expression of Twist in carcinoma cells leads to an epithelial-to-mesenchymal transition (EMT) with a highly motile and invasive phenotype. Survival in transit As tumor cells enter the bloodstream they lose their adhesion to the ECM and are deprived of survival signals that originate from such interactions. Therefore, if they are to survive, tumor cells must become resistant to apoptosis that is triggered by loss of anchorage; this form of programmed cell death is called anoikis. Although the actual time that the tumor cells spend in transit may be on the order of minutes, successful establishment of anchorage at the secondary site may take far longer. In experimental systems, genes that confer resistance to anoikis, such as TrkB, XIAP and ILK, have been shown to also facilitate formation of metastases, suggesting that anoikis resistance may be a rate-limiting step in the invasion-metastasis cascade (Attwell et al., 2000; Berezovskaya et al., 2005; Douma et al., 2004; Yawata et al., 1998). Extravasation Tumor cells come to arrest in distant capillaries by a variety of means. These include physical entrapment due to the large diameter of tumor cells, specific attachment to endothelial cells via integrins and other adhesion receptors, and retention in certain tissues by chemokine receptor-ligand interactions (Gupta and Massague, 2006). At first, tumor cells may proliferate inside the lumina of the capillaries, but they eventually break out and invade into the surrounding tissue in a process called extravasation (Chambers et al., 2002). Intra-vital microscopy of experimental metastasis models indicate that many tumors can extravasate quite efficiently; in one model system 83% of intravenously injected tumor cells were found to have extravasated after 3 days (Luzzi et al., 1998). This finding suggests that this step of the invasion-metastasis cascade may not require the acquisition of other traits beyond those involved in cell motility and local invasion. Colonization and secondary tumor initiation It has been clinically and experimentally recognized that a majority of disseminated tumor cells persist in a dormant state in their new microenvironment without ever growing to form secondary tumors (Chambers et al., 2002; Naumov et al., 2002). For example, among breast cancer patients that present with only primary tumors, 30-40% were found to have cytokeratin positive- tumor cells in their bone marrow (Braun et al., 2000). However, a majority of such patients never developed clinically detectable metastases within four years. Therefore, growth at secondary sites, also known as colonization, seems to be the least efficient step in the metastatic cascade. As with primary tumor formation, successful colonization depends on both tumor-cell-intrinsic and secondary site-specific factors. Early work from Fidler and colleagues showed that a given primary tumor may harbor rare subclones with high colonizing ability when injected intravenously (Fidler and Kripke, 1977). More recent studies have suggested that only a subpopulation of cells within solid tumors, termed cancer stem cells (CSCs), have the ability to seed the formation of new tumors upon transplantation (Al-Hajj et al., 2003; Wicha, 2006). The implication of these findings is that certain tumor cells, due either to their mutational status and/or their differentiation state, are more likely to generate secondary tumors. The functional differences that allow such subpopulations of cells to be more adept at colonization are not well understood. However, the ability to elicit angiogenesis, to recruit appropriate stroma, and respond to site-specific growth factors are likely to be important factors in governing colonizing ability (Chambers et al., 2002). Resistance to therapy Although it is not considered to be a distinct biological step in the invasionmetastasis cascade, resistance to chemotherapy is intimately linked to the progression of metastatic disease. In fact, eviidence from model experimental systems indicates that migratory cells isolated from primary tumors express anti-apoptotic genes and are able to survive chemotherapy to a greater extent (Goswami et al., 2004). Most patients whose primary tumors are surgically removed also undergo a regimen of radiation and/or chemotherapy. Therefore, secondary tumors thatlater arise in such patients must have survived these initial cytotoxic treatments. When metastases become clinically apparent, patients generally undergo a second round of extensive therapy. However, these later treatments are rarely curative, meaning that metastatic tumors almost invariably become resistant to subsequent rounds of therapy. Epithelial-to-mesenchymal transition (EMT) Due to its multi-step nature, the successful completion of the metastatic process requires the acquisition of multiple traits by tumor cells. An important conceptual question in the field of metastasis research has been how a transformed epithelial cell manages to accumulate such a multitude of malignancy-associated traits. When this thesis work was started, a transdifferentiation program that naturally occurs during development termed epithelial-to-mesenchymal transition (EMT) was put forth as a possible mechanism to the simultaneous acquisition of such traits (Thiery, 2002). EMT is a normal physiological process One aspect of an EMT is the loss of epithelial-cell attributes, such as cell polarity, cell-cell adhesion, and cytokeratins, which are epithelium-specific intermediate filaments. Concomitantly, cells undergoing an EMT acquire mesenchymal phenotypes, such as spindle-shape morphology, increased motility, expression of vimentin, a mesenchymespecific intermediate filament, and N-cadherin, a cell adhesion molecule normally expressed in neurons, muscle cells and fibroblasts (Figure 1). EMT is a physiological process that occurs during various stages in embryonic development. It is used mainly to create new tissues from existing cell layers, for example during the formation of mesoderm in gastrulation and migration of the neural crest cells from the neural plate (Savagner, 2001). Later in development, EMTs can be observed during tubulogenesis and branching in the mammary gland and in wound healing, where it is necessary for the re-epithelization of the wound by adjacent keratinocytes (Grunert et al., 2003). For example, skin from knock-out mice lacking Slug, an EMT-inducing transcription factor, shows impaired wound healing and keratinocyte migration (Savagner et al., 2005). Evidence for the EMT in carcinoma progression A role for the EMT in carcinoma cell invasion was first described by Thiery and colleagues almost 20 years ago (Boyer et al., 1989a; Boyer et al., 1989b). Since then, direct demonstration of an EMT in human tumor samples has been difficult for two main reasons. First, EMTs are observed to occur in response to extracellular signals originating from the tumor stroma; therefore they are thought to happen in a minority of cells at the invasion fronts of carcinomas (Brabletz et al., 2001; Weinberg, 2007). Second, histological staining of human tumors with conventional markers cannot distinguish the mesenchymal cells that are formed during an EMT from the fibroblasts present in the tumor stroma. For these reasons, studies on EMT and carcinoma progression initially utilized in vitro cell lines, mouse models and eventually gene expression analyses of clinical samples. One of the first systems used to study the EMT was the Madin-Darby canine kidney (MDCK) epithelial cell line. The scattering behavior of these cells in response to a fibroblast-derived factor led to the identification of the hepatocyte growth factor (HGF), the ligand for the c-Met receptor tyrosine kinase (Stoker and Perryman, 1985). c-Met is known to be overexpressed or activated in certain carcinomas and can drive tumour progression in vivo (Thiery, 2002). Another cell line frequently used in EMT studies is the Eph4 mouse mammary carcinoma cell line. Studies by Beug and colleagues showed that activation of the transforming growth factor-beta (TGF-3) signaling in Ras- transformed Eph4 cells suffices to induce an EMT in vitro and metastasis in vivo (Oft et al., 1996). Subsequent to these studies that employ soluble proteins, specific transcription factors that can program the EMT were identified (see below). Since then, many studies have shown that cultured epithelial cell lines of various sources can be induced into a mesenchymal phenotype by overexpressing one of these factors (Batlle et al., 2000; Bolos et al., 2003; Cano et al., 2000; Comijn et al., 2001; Perez-Moreno et al., 2001; Wu et al., 2006; Yang et al., 2004). In addition, the expression levels of these transcription factors were evaluated in human tumors. Histological studies showed that increased expression of individual EMT-inducing transcription factors (EMT-TFs) is associated with metastatic progression, poor prognosis, and tumor dedifferentiation (Aigner et al., 2007a; Blanco et al., 2002; Elloul et al., 2005; Martin et al., 2005). For example, in breast cancers, expression of the EMT-TFs Slug and Twist is detected preferentially in primary tumors of patients who later develop metastatic disease (Martin et al., 2005). More recently, transgenic mouse models have been employed to study the EMT in carcinoma progression. For example, expression of one of the EMT-TFs, Snail, has also been linked directly to tumor recurrence in a doxycycline-inducible MMTV-Her2 mouse mammary tumor model (Moody et al., 2005). In addition, passage though an EMT has been shown to be required for metastasis of polyoma virus middle T antigen (PyVMT)-driven mouse mammary tumors (Xue et al., 2003). This was accomplished by placing the thymidine kinase gene under the control of an EMT-specific promoter (S100A4). Administration of gancyclovir to transgenic MMTV-PyV-MT animals inhibited lung metastases of mammary tumors, indicating that cells responsible for the metastases undergo an EMT at one point in tumor progression (Xue et al., 2003). Since several distinct transcription factors can lead to the same well-defined cell biological phenotypes associated with a mesenchymal state, the EMT is thought to have a core transcriptional program. In fact, gene expression profiling of cell lines expressing different EMT-inducers have revealed a set of genes that are co-regulated during the EMT (Jechlinger et al., 2003; Moreno-Bueno et al., 2006). Recent studies have taken advantage of the presence of a core EMT signature to look for evidence of an EMT in human tumor samples. Genes associated with an EMT were found to be upregulated in high-risk head-and-neck squamous cell carcinomas, metaplastic carcinomas of the breast, invasive thyroid carcinomas, and metastatic melanomas (Alonso et al., 2007; Chung et al., 2006; Lien et al., 2007; Vasko et al., 2007). In addition, gene expression of profiling of disseminated tumor cells obtained from bone marrow aspirates of breast cancer patients indicated that these micrometastatic cells express high levels of Twist (Watson et al., 2007). Interestingly, in this study the patients had already received multiple rounds of chemotherapy, suggesting that the Twist-expressing disseminated cells survived such treatment (Watson et al., 2007). Further studies utilizing laser capture microdissection to isolate tumor cells at the tumor-stroma interface are likely to yield additional evidence for an EMT in solid tumors. EMT in the invasion/metastasis cascade One of the hallmarks of the EMT is loss of epithelial cell-cell adhesion by repression of the classical adhesion molecule E-cadherin. Loss of cell-cell adhesion is thought to be a necessary step in the invasion-metastasis cascade, as it allows detachment of carcinoma cells from one another. In fact, inhibition of disaggregation by E-cadherin overexpression diminishes invasiveness of aggressive cell lines in vitro and prevents progression from adenoma to carcinoma in vivo (Frixen et al., 1991; Perl et al., 1998). The role of E-cadherin in metastasis and how its expression is regulated is discussed in more detail below. Loss of cell-cell adhesion during an EMT is associated with a concomitant increase in carcinoma cell motility and invasiveness. Motile behavior induced by an EMT is due in part to the activation of small GTPases, such as Rho (Bhowmick et al., 2001). Importantly, expression of RhoC has been shown to be necessary and sufficient for metastasis of in model systems (Clark et al., 2000). The EMT also leads to increased expression of certain MMPs, such as MMP-2 and MMP-9 (Jorda et al., 2005; Tester et al., 2000). Therefore, an EMT may allow cancer cells to invade locally and migrate through the endothelium of capillary walls to intravasate into the bloodstream. Recent studies have revealed that the induction of an EMT, in addition to promoting the early steps of the invasion-metastasis cascade, may allow cancer cells to complete the subsequent steps as well. For example, the transcription factor Snail, a potent inducer of EMT, has been shown to confer resistance to apoptosis induced by withdrawal of growth factors (Vega et al., 2004). There is also a growing body of literature indicating that the EMT can confer resistance to genotoxic stress, such as that induced by chemotherapeutic agents (Robson et al., 2006; Wang et al., 2004; Zhang et al., 2007). Therefore, cancer cells that have undergone an EMT may be better equipped to survive in the circulation. In addition, those who have already disseminated and formed micrometastases at the time of therapy may be able to withstand such treatments, eventually leading to recurrence. Colonization of distant tissues may ultimately be tied to the ability of disseminated tumor cells to self-renew or exist in a cancer stem cell (CSC)-like state. At least in breast carcinomas, the CSCs have the following important cellular properties: (1) CSCs can be enriched by sorting cells for CD44high/CD24 low antigenic profile (Al-Hajj et al., 2003); (2) CSCs can form spherical colonies in specialized suspension cultures; these colonies are termed mammospheres (Dontu et al., 2003; Dontu and Wicha, 2005); (3) CSCs can form tumors at limiting dilution with as little as 1000 cells (Al-Hajj et al., 2003; Ince et al., 2007). Recent work from our laboratory has shown that induction of an EMT in transformed mammary epithelial cells results in the acquisition of all of these CSC properties (Mani et al., 2008). In addition, induction of EMT in other model systems and clinical tumors has been linked to dedifferentiation, tumor recurrence, and decrease in relapse-free survival in patients with breast cancer (Aigner et al., 2007a; Moody et al., 2005). The hallmarks of the EMT enumerated above suggest that this transition can potentially allow the carcinoma cells to execute many of the rate-limiting steps in the invasion-metastasis cascade. Therefore, there has been great interest to identify the mechanisms that can trigger an EMT in carcinoma cells. Regulation of the EMT Extracellular signals that induce the EMT During development, EMTs are induced by a combination of extracellular morphogenic proteins, such as fibroblast growth factor (FGF), platelet-derived growth factor (PDGF), epidermal growth factor (EGF), transforming growth factor beta (TGF-P), bone morphogenetic protein (BMP) and Wnt family members (Peinado et al., 2007). In addition, carcinoma cells can be induced in vitro to undergo an EMT by HGF, IGF (insulin-like growth factor), TNF-a (tumor necrosis alpha), PDGF, EGF, TGF-P, Notch and Hedgehog signals (Huber et al., 2005). In tumors, these EMT-inducing signals are thought to originate mainly from the nearby activated stroma, although in some cases tumor cells themselves may have autocrine expression of these factors (Vincent-Salomon and Thiery, 2003). It should be noted that there is quite a bit of cell type specificity in EMT induction by these various signaling pathways. In addition, the underlying genetic makeup of carcinoma cells can collaborate with certain extracellular signals to induce the EMT, as in the case of Ras mutations and TGF-P signals (Oft et al., 1996). Transcriptional control of the EMT In addition to extracellular proteins, a number of transcription factors are necessary and/or sufficient to induce the EMT both in physiological and pathological contexts. These have originally been identified as important developmental regulators. For example, Snail belongs to a family of zinc finger-containing transcriptional repressors and plays an important role in the control of gastrulation (Barrallo-Gimeno and Nieto, 2005). Slug is a related family member that functions during neural crest migration (Nieto et al., 1994; Savagner et al., 1997). Twist is a basic-helix-loop-helix (bHLH) transcription factor necessary for mesoderm specification (Furlong et al., 2001). Among other such developmentally important EMT-inducing transcription factors (EMT-TFs) are Sipl, Goosecoid, and FoxC2 (Comijn et al., 2001; Hartwell et al., 2006; Mani et al., 2007). In addition, there are EMT-TFs that were identified based on their ability to cause repression of E-cadherin expression, which is a hallmark of the EMT. These are the zinc finger protein Zebl (also known as BEF1 and TCF8) and the bHLH proteins E47 and E2A (Eger et al., 2005; Perez-Moreno et al., 2001) (Figure 1). In the context of both physiological and pathological EMTs, the expression of EMT-TFs is regulated by various upstream signals. During development, Snail and Twist are under the control of FGF and NF-ic3 signaling pathways, a finding that is recapitulated in carcinoma cell lines (Jiang et al., 1991; Julien et al., 2007). On the other hand, Slug is regulated by the P-catenin/Wnt pathway through direct transcriptional induction in both Xenopus development and colon cancer cell lines (Conacci-Sorrell et al., 2003; Vallin et al., 2001). Recently, high-mobility group protein HMGA2 has been shown to be essential for the TGF-0 induced expression of Snail, Slug and Twist (Thuault et al., 2006). Twist is also a target of EGF-receptor signaling, where EGFR activation results in the direct binding of signal transducer and activator of transcription-3 (STAT3) to the twist promoter (Lo et al., 2007). IL-6 or Src-induced activation of STAT3 can also lead to increased Twist expression (Cheng et al., 2008). The regulation of other bHLH and ZEB factors during an EMT is almost completely unknown. While the upstream signals that induce these factors are diverse, all of the EMTTFs share a common downstream target, the classical epithelial cell adhesion molecule E- cadherin. These factors regulate E-cadherin expression either by binding directly to its promoter or through indirect mechanisms (Peinado et al., 2007). As this thesis focuses on how E-cadherin expression affects metastatic progression, the following sections will be devoted to this protein. E-cadherin is an important epithelial cell adhesion molecule E-cadherin (originally termed Uvomorulin) was first identified as the target of an antibody capable of preventing calcium-dependent compaction of mouse embryos and embryonal carcinoma cells (Hyafil et al., 1981; Peyrieras et al., 1983; Yoshida and Takeichi, 1982). Expression cloning using such antibodies allowed the isolation and characterization of the murine E-cadherin cDNA, which was then shown to be sufficient to cause cell-cell adhesion (Nagafuchi et al., 1987; Schuh et al., 1986). In addition, earlier experiments using E-cadherin antibodies demonstrated an essential role for this molecule in maintaining epithelial cell adhesion (Behrens et al., 1985; Gumbiner et al., 1988; Vestweber and Kemler, 1985). E-cadherin is the principal component of the adherens junctions between epithelial cells. E-cadherin is expressed in all epithelial tissues (Shimoyama et al., 1989). The Ecadherin knock-out phenotype is early embryonic lethal due to decompaction of the preimplantation embryo (Larue et al., 1994; Riethmacher et al., 1995) Conditional deletion of E-cadherin in adult tissues leads to disruption of adherens junctions and loss of epithelial integrity. Moreover, loss of E-cadherin results in altered epithelial cell differentiation, as evidenced by tissue- specific knock-outs in the skin and the mammary gland (Boussadia et al., 2002; Tinkle et al., 2004; Young et al., 2003). In addition to mediating cell-cell adhesion, E-cadherin also serves to establish apico-basal polarity, a hallmark of epithelial cells (McNeill et al., 1990). Therefore, E-cadherin has been viewed as a gatekeeper of epithelial cell identity (Berx and Van Roy, 2001). Structure of E-cadherin Classical cadherin molecules (E- and N-cadherin) have five extracellular cadherin repeats, a transmembrane, and a cytoplasmic intracellular domain. Cell adhesion is accomplished through homophilic interactions between cadherin molecules expressed on adjacent cells. The most amino-terminal cadherin repeat is essential for this process and contains a histidine-alanine-valine domain and tryptophan residues that mediate transcadherin binding (Blaschuk et al., 1990; Cavallaro and Christofori, 2004; Halbleib and Nelson, 2006). Classical cadherins preferentially form homophilic interactions (E-E or NN), although heterophilic binding has been observed in certain experimental settings (Prakasam et al., 2006). Early work demonstrated that the cytoplasmic domain of E-cadherin is essential for cell adhesion even though the actual physical binding between two epithelial cells occurs in the extracellular space (Ozawa et al., 1990). In its cytoplasmic domain, Ecadherin associates with a number of catenin proteins (3, y, p120 and indirectly oacatenin), all of which regulate the association of adherens junction to the underlying actin cytoskeleton (Halbleib and Nelson, 2006). In addition to having structural roles in maintaining the adherens junctions, catenins also take part in diverse signaling pathways. For example, loss of a-catenin leads to hyperactivation of the MAPK signaling, whereas p120 catenin has been shown to regulate Rho and Nf-1•p pathways (Perez-Moreno et al., 2006; Vasioukhin et al., 2001; Wildenberg et al., 2006). Among the catenins, the best studied is p-catenin, whose function as an essential component of the Wnt pathway is discussed below. D1-catenin and the Wnt pathway p-catenin initially binds E-cadherin in the endoplasmic reticulum, and this interaction is necessary for the proper post-posttranslational delivery of the latter to the plasma membrane (Chen et al., 1999). In the absence of canonical Wnt signaling, cytoplasmic p-catenin that is unbound to E-cadherin is targeted for degradation by the proteasome through its phosphorylation by casein kinase I and glycogen synthase kinase3 beta (GSK-3p) (Nelson and Nusse, 2004). The phosphorylation of p-catenin occurs on a scaffolding complex containing the axin and the adenomatous polyposis coli (APC) proteins. Activation of Wnt signaling leads to inhibition of GSK-3p, resulting in cytoplasmic accumulation and nuclear translocation of f3-catenin, which binds to its transcription cofactors, T-cell factor/lymphoid enhancer factor (TCF/LEF). TCF/LEF then activates transcription of Wnt target genes, such as ephrin receptors, CD44, cyclin D1, fibronectin and c-myc (Clevers, 2006). An unanswered question in the field has been whether a single pool of p-catenin can participate in both cell adhesion and Wnt signaling. Since E-cadherin, TCF and APC all bind to approximately the same binding groove on 3-catenin in mutually exclusive ways, the level of E-cadherin expression is likely to have an impact on the amount of 3-catenin that is available for TCF/LEF association (Huber and Weis, 2001). Some studies provide evidence for this notion; for example overexpression of E-cadherin or its cytoplasmic tail is able to sequester 3-catenin away from the nucleus and decrease Pcatenin-dependent transcription (Conacci-Sorrell et al., 2003; Gottardi et al., 2001; Kuphal and Behrens, 2006; Orsulic et al., 1999; Stockinger et al., 2001). However, other studies suggest that there are two distinct molecular forms of 3-catenin that participate either in Wnt signaling or cell adhesion (Gottardi and Gumbiner, 2004). In addition, tyrosine phosphorylation of p-catenin in response to growth factor signaling seems to play an important role in determining which of these functions it can carry out (Brembeck et al., 2004). While there is growing evidence that E-cadherin-mediated sequestration of 3-catenin can compete with Wnt signaling, it has not been clear whether loss of Ecadherin and the concomitant increase in free cytosolic P-catenin may lower the threshold of activation of the Wnt signaling pathway. Mechanisms of E-cadherin loss in human tumors In a variety of human tumors, histological examination combined with patient outcome data has revealed that reduced or absent expression of E-cadherin is associated with poor prognosis, tumor dedifferentiation, and occurrence of metastases (Schipper et al., 1991; Siitonen et al., 1996; Sulzer et al., 1998; Umbas et al., 1994). A number of genetic, epigenetic, transcriptional, and post-translational mechanisms may lead to reduced E-cadherin expression and/or activity. In this section I will briefly discuss these. Somatic alterations, such as point mutations, in-frame and frame-shift deletions, and insertions in the E-cadherin gene, have been observed mainly in lobular breast carcinomas and diffuse-type gastric carcinomas (Becker et al., 1994; Berx et al., 1995; Berx et al., 1996; Kanai et al., 1994). Importantly, these two tumor types that have E- cadherin gene mutations also exhibit loss of the wild-type E-cadherin allele, suggesting that E-cadherin loss in such tumors results from a two-hit mechanism, as seen in the classical tumor suppressor genes. In addition, germline mutations in E-cadherin are found in familial gastric cancers, and the tumor cells arising in such families sustain secondary somatic mutations in the wildtype E-cadherin allele (Guilford et al., 1998). E-cadherin mutations in other types of cancers are rarely detected, although chromosome 16q22.1, where the E-cadherin locus resides, is frequently deleted in advanced hepatocellular, prostate and breast carcinomas (Hirohashi, 1998; Sato et al., 1990; Tsuda et al., 1990). In contrast to somatic mutations, which happen predominantly in the two types of cancers mentioned above, many other tumor types exhibit reduced E-cadherin expression due to hypermethylation of the CpG islands in its promoter (Strathdee, 2002). In a number of human cell lines, E-cadherin can be re-expressed by treatment with 5azacytidine, an inhibitor of DNA methyltransferases (Graff et al., 1995; Yoshiura et al., 1995). Moreover, methylation of the E-cadherin promoter has been closely correlated with malignant progression of breast cancer (Nass et al., 2000). Another type of transcriptional repression of the E-cadherin promoter occurs via the action of specific transcription factors involved in the EMT program. This type of Ecadherin repression can also be observed during developmental EMTs, for example, at the primitive streak in the mouse embryo (Thiery, 2003). Zinc finger proteins, such Snail and Slug, accomplish E-cadherin repression by recruiting specific chromatin-remodeling complexes to the E-cadherin locus. These complexes contain the C-terminal binding protein 1 (CtBP-1), Sin3a and histone deacetylases HDAC1 and 2 (Peinado et al., 2007). The mechanism by which other EMT-TFs, such as the bHLH protein Twist, repress E- cadherin transcription remains unknown. However, the E-box sequences in the Ecadherin promoter seem to be essential for Twist-mediated repression (Yang et al., 2004). E-cadherin expression can also be modulated by post-translational mechanisms. While the extracellular domains of E-cadherin can be cleaved by proteases such as MMP7 and ADAM10, its cytoplasmic domain can be a target for proteolytic cleavage by the 7-secretase activity of Presenilin-1 (Marambaud et al., 2002; Maretzky et al., 2005; McGuire et al., 2003). All of these proteolytic cleavages result in loss of cell adhesion. Ecadherin turnover can also be modulated by endocytosis, internalization and ubiquitylation by the E3 ubiquitin ligase Hakai, or phosphorylation by receptor tyrosine kinases (Avizienyte et al., 2002; Bryant and Stow, 2004; Fujita et al., 2002; Kamei et al., 1999; Lu et al., 2003). In addition, expression of certain glycoproteins, such as MUC1 and dysadherin, has been reported to inhibit E-cadherin's adhesive functions (Ino et al., 2002; Makiguchi et al., 1996). Main Questions Addressed The mechanisms by which E-cadherin is inactivated in human tumors can be placed into two general categories: (1) those that abrogate its adhesion function while preserving its intracellular domain; and (2) those that lead to E-cadherin's substantial repression or complete absence. At the time this thesis work was initiated, there was a large body of work examining E-cadherin expression and mutational status as a prognostic indicator. However, the contribution of different types of E-cadherin loss to tumor progression and metastasis was largely undefined. Since several structural proteins associated with adherens junctions can also have intracellular signaling functions, I hypothesized that the two types of E-cadherin inactivation may have qualitatively different outcomes in terms of cancer cell invasion and metastasis. In the first scenario, loss of E-cadherin's adhesive function may enable the first step of the metastatic cascade-the disaggregation of cancer cells from one another. On the other hand, total absence of E-cadherin might lead to activation downstream signaling pathways that support the successful completion of additional steps of the invasion-metastasis cascade (Figure 2). As mentioned above, passage through an EMT has the potential to endow carcinoma cells with the ability to execute many, if not all, the steps of this cascade. Importantly, all EMT-inducing transcription factors work, at least in part, by repressing E-cadherin. Therefore, another question I addressed was whether loss of E-cadherin itself could be sufficient for the induction of the EMT. In other words, given the known correlation between loss of E-cadherin expression and the upregulation of these transcription factors in human tumors, I was interested in learning whether the loss of E- cadherin is just one manifestation of the multi-component EMT program that is activated in tumors or whether, alternatively, E-cadherin itself acts as a pleiotropic regulator of cell phenotype, enabling it to function as a master regulator of epithelial cell behavior. Figures EMT-TFs Snail Slug Twist Zebl Goosecoid EMT Epithelial cells Mesenchymal cells E-cadherin Vimentin N-cadherin Fibronectin Cytokeratins Figure 1. Schematic representation of the epithelial-to-mesenchymal transition (EMT) A subset of EMT-inducing transcription factors (EMT-TFs) and the changes they elicit in epithelial cells are listed. Wild-type DominantNegative Loss of E-cadherin p12 0 ! ao-catenin Actin Actin Signaling Signaling Figure 2. Schematic representation of the structures of wild-type and dominant-negative E-cadherin molecules. Also shown are cytoplasmic catenins that serve to link E-cadherin to the underlying cytoskeleton. Note that while the dominant-negative E-cadherin still binds and sequesters these catenins, the complete absence of E-cadherin leads to their liberation and potential involvement in downstream signaling pathways. References Aigner, K., Dampier, B., Descovich, L., Mikula, M., Sultan, A., Schreiber, M., Mikulits, W., Brabletz, T., Strand, D., Obrist, P., Sommergruber, W., Schweifer, N., Wernitznig, A., Beug, H., Foisner, R. and Eger, A. (2007) The transcription factor ZEB 1 (deltaEFl) promotes tumour cell dedifferentiation by repressing master regulators of epithelial polarity. Oncogene, 26, 6979-6988. Al-Hajj, M., Wicha, M.S., Benito-Hernandez, A., Morrison, S.J. and Clarke, M.F. (2003) Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci US A, 100, 3983-3988. Alonso, S.R., Tracey, L., Ortiz, P., Perez-Gomez, B., Palacios, J., Pollan, M., Linares, J., Serrano, S., Saez-Castillo, A.I., Sanchez, L., Pajares, R., Sanchez-Aguilera, A., Artiga, M.J., Piris, M.A. and Rodriguez-Peralto, J.L. (2007) A high-throughput study in melanoma identifies epithelial-mesenchymal transition as a major determinant of metastasis. CancerRes, 67, 3450-3460. Attwell, S., Roskelley, C. and Dedhar, S. (2000) The integrin-linked kinase (ILK) suppresses anoikis. Oncogene, 19, 3811-3815. Avizienyte, E., Wyke, A.W., Jones, R.J., McLean, G.W., Westhoff, M.A., Brunton, V.G. and Frame, M.C. (2002) Src-induced de-regulation of E-cadherin in colon cancer cells requires integrin signalling. Nat Cell Biol, 4, 632-638. Barrallo-Gimeno, A. and Nieto, M.A. (2005) The Snail genes as inducers of cell movement and survival: implications in development and cancer. Development, 132, 3151-3161. Batlle, E., Sancho, E., Franci, C., Dominguez, D., Monfar, M., Baulida, J. and Garcia De Herreros, A. (2000) The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nat Cell Biol, 2, 84-89. Becker, K.F., Atkinson, M.J., Reich, U., Becker, I., Nekarda, H., Siewert, J.R. and Hofler, H. (1994) E-cadherin gene mutations provide clues to diffuse type gastric carcinomas. Cancer Res, 54, 3845-3852. Behrens, J., Birchmeier, W., Goodman, S.L. and Imhof, B.A. (1985) Dissociation of Madin-Darby canine kidney epithelial cells by the monoclonal antibody anti-arc1: mechanistic aspects and identification of the antigen as a component related to uvomorulin. J Cell Biol, 101, 1307-1315. Berezovskaya, O., Schimmer, A.D., Glinskii, A.B., Pinilla, C., Hoffman, R.M., Reed, J.C. and Glinsky, G.V. (2005) Increased expression of apoptosis inhibitor protein XIAP contributes to anoikis resistance of circulating human prostate cancer metastasis precursor cells. Cancer Res, 65, 2378-2386. Berx, G., Cleton-Jansen, A.M., Nollet, F., de Leeuw, W.J., van de Vijver, M., Cornelisse, C. and van Roy, F. (1995) E-cadherin is a tumour/invasion suppressor gene mutated in human lobular breast cancers. Embo J, 14, 6107-6115. Berx, G., Cleton-Jansen, A.M., Strumane, K., de Leeuw, W.J., Nollet, F., van Roy, F. and Cornelisse, C. (1996) E-cadherin is inactivated in a majority of invasive human lobular breast cancers by truncation mutations throughout its extracellular domain. Oncogene, 13, 1919-1925. Berx, G. and Van Roy, F. (2001) The E-cadherin/catenin complex: an important gatekeeper in breast cancer tumorigenesis and malignant progression. Breast CancerRes, 3, 289-293. Bhowmick, N.A., Ghiassi, M., Bakin, A., Aakre, M., Lundquist, C.A., Engel, M.E., Arteaga, C.L. and Moses, H.L. (2001) Transforming growth factor-betal mediates epithelial to mesenchymal transdifferentiation through a RhoA-dependent mechanism. Mol Biol Cell, 12, 27-36. Blanco, M.J., Moreno-Bueno, G., Sarrio, D., Locascio, A., Cano, A., Palacios, J. and Nieto, M.A. (2002) Correlation of Snail expression with histological grade and lymph node status in breast carcinomas. Oncogene, 21, 3241-3246. Blaschuk, O.W., Sullivan, R., David, S. and Pouliot, Y. (1990) Identification of a cadherin cell adhesion recognition sequence. Dev Biol, 139, 227-229. Bolos, V., Peinado, H., Perez-Moreno, M.A., Fraga, M.F., Esteller, M. and Cano, A. (2003) The transcription factor Slug represses E-cadherin expression and induces epithelial to mesenchymal transitions: a comparison with Snail and E47 repressors. J Cell Sci, 116, 499-511. Boussadia, O., Kutsch, S., Hierholzer, A., Delmas, V. and Kemler, R. (2002) E-cadherin is a survival factor for the lactating mouse mammary gland. Mech Dev, 115, 5362. Boyer, B., Tucker, G.C., Valles, A.M., Franke, W.W. and Thiery, J.P. (1989a) Rearrangements of desmosomal and cytoskeletal proteins during the transition from epithelial to fibroblastoid organization in cultured rat bladder carcinoma cells. J Cell Biol, 109, 1495-1509. Boyer, B., Tucker, G.C., Valles, A.M., Gavrilovic, J. and Thiery, J.P. (1989b) Reversible transition towards a fibroblastic phenotype in a rat carcinoma cell line. Int J Cancer Suppl, 4, 69-75. Brabletz, T., Jung, A., Reu, S., Porzner, M., Hlubek, F., Kunz-Schughart, L.A., Knuechel, R. and Kirchner, T. (2001) Variable beta-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc Natl Acad Sci USA, 98, 10356-10361. Braun, S., Pantel, K., Muller, P., Janni, W., Hepp, F., Kentenich, C.R., Gastroph, S., Wischnik, A., Dimpfl, T., Kindermann, G., Riethmuller, G. and Schlimok, G. (2000) Cytokeratin-positive cells in the bone marrow and survival of patients with stage I, II, or III breast cancer. N Engl J Med, 342, 525-533. Brembeck, F.H., Schwarz-Romond, T., Bakkers, J., Wilhelm, S., Hammerschmidt, M. and Birchmeier, W. (2004) Essential role of BCL9-2 in the switch between betacatenin's adhesive and transcriptional functions. Genes Dev, 18, 2225-2230. Bryant, D.M. and Stow, J.L. (2004) The ins and outs of E-cadherin trafficking. Trends Cell Biol, 14, 427-434. Cano, A., Perez-Moreno, M.A., Rodrigo, I., Locascio, A., Blanco, M.J., del Barrio, M.G., Portillo, F. and Nieto, M.A. (2000) The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nat Cell Biol, 2, 76-83. Cavallaro, U. and Christofori, G. (2004) Cell adhesion and signalling by cadherins and Ig-CAMs in cancer. Nat Rev Cancer, 4, 118-132. Chambers, A.F., Groom, A.C. and MacDonald, I.C. (2002) Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer,2, 563-572. Chen, Y.T., Stewart, D.B. and Nelson, W.J. (1999) Coupling assembly of the Ecadherin/beta-catenin complex to efficient endoplasmic reticulum exit and basallateral membrane targeting of E-cadherin in polarized MDCK cells. J Cell Biol, 144, 687-699. Cheng, G.Z., Zhang, W., Sun, M., Wang, Q., Coppola, D., Mansour, M., Xu, L., Costanzo, C., Cheng, J.Q. and Wang, L.H. (2008) Twist is transcriptionally induced by activation of STAT3 and mediates STAT3 oncogenic function. J Biol Chem. Chung, C.H., Parker, J.S., Ely, K., Carter, J., Yi, Y., Murphy, B.A., Ang, K.K., ElNaggar, A.K., Zanation, A.M., Cmelak, A.J., Levy, S., Slebos, R.J. and Yarbrough, W.G. (2006) Gene expression profiles identify epithelial-tomesenchymal transition and activation of nuclear factor-kappaB signaling as characteristics of a high-risk head and neck squamous cell carcinoma. CancerRes, 66, 8210-8218. Clark, E.A., Golub, T.R., Lander, E.S. and Hynes, R.O. (2000) Genomic analysis of metastasis reveals an essential role for RhoC. Nature, 406, 532-535. Clevers, H. (2006) Wnt/beta-catenin signaling in development and disease. Cell, 127, 469-480. Comijn, J., Berx, G., Vermassen, P., Verschueren, K., van Grunsven, L., Bruyneel, E., Mareel, M., Huylebroeck, D. and van Roy, F. (2001) The two-handed E box binding zinc finger protein SIP1 downregulates E-cadherin and induces invasion. Mol Cell, 7, 1267-1278. Conacci-Sorrell, M., Simcha, I., Ben-Yedidia, T., Blechman, J., Savagner, P. and BenZe'ev, A. (2003) Autoregulation of E-cadherin expression by cadherin-cadherin interactions: the roles of beta-catenin signaling, Slug, and MAPK. J Cell Biol, 163, 847-857. Dontu, G., Abdallah, W.M., Foley, J.M., Jackson, K.W., Clarke, M.F., Kawamura, M.J. and Wicha, M.S. (2003) In vitro propagation and transcriptional profiling of human mammary stem/progenitor cells. Genes Dev, 17, 1253-1270. Douma, S., Van Laar, T., Zevenhoven, J., Meuwissen, R., Van Garderen, E. and Peeper, D.S. (2004) Suppression of anoikis and induction of metastasis by the neurotrophic receptor TrkB. Nature, 430, 1034-1039. Egeblad, M. and Werb, Z. (2002) New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer,2, 161-174. Eger, A., Aigner, K., Sonderegger, S., Dampier, B., Oehler, S., Schreiber, M., Berx, G., Cano, A., Beug, H. and Foisner, R. (2005) DeltaEF1 is a transcriptional repressor of E-cadherin and regulates epithelial plasticity in breast cancer cells. Oncogene, 24, 2375-2385. Elloul, S., Elstrand, M.B., Nesland, J.M., Trope, C.G., Kvalheim, G., Goldberg, I., Reich, R. and Davidson, B. (2005) Snail, Slug, and Smad-interacting protein 1 as novel parameters of disease aggressiveness in metastatic ovarian and breast carcinoma. Cancer, 103, 1631-1643. Fidler, I.J. and Kripke, M.L. (1977) Metastasis results from preexisting variant cells within a malignant tumor. Science, 197, 893-895. Friedl, P. and Wolf, K. (2003) Tumour-cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer,3, 362-374. Frixen, U.H., Behrens, J., Sachs, M., Eberle, G., Voss, B., Warda, A., Lochner, D. and Birchmeier, W. (1991) E-cadherin-mediated cell-cell adhesion prevents invasiveness of human carcinoma cells. J Cell Biol, 113, 173-185. Fujita, Y., Krause, G., Scheffner, M., Zechner, D., Leddy, H.E., Behrens, J., Sommer, T. and Birchmeier, W. (2002) Hakai, a c-Cbl-like protein, ubiquitinates and induces endocytosis of the E-cadherin complex. Nat Cell Biol, 4, 222-231. Furlong, E.E., Andersen, E.C., Null, B., White, K.P. and Scott, M.P. (2001) Patterns of gene expression during Drosophila mesoderm development. Science, 293, 16291633. Gottardi, C.J. and Gumbiner, B.M. (2004) Distinct molecular forms of beta-catenin are targeted to adhesive or transcriptional complexes. J Cell Biol, 167, 339-349. Gottardi, C.J., Wong, E. and Gumbiner, B.M. (2001) E-cadherin suppresses cellular transformation by inhibiting beta-catenin signaling in an adhesion-independent manner. J Cell Biol, 153, 1049-1060. Graff, J.R., Herman, J.G., Lapidus, R.G., Chopra, H., Xu, R., Jarrard, D.F., Isaacs, W.B., Pitha, P.M., Davidson, N.E. and Baylin, S.B. (1995) E-cadherin expression is silenced by DNA hypermethylation in human breast and prostate carcinomas. CancerRes, 55, 5195-5199. Grunert, S., Jechlinger, M. and Beug, H. (2003) Diverse cellular and molecular mechanisms contribute to epithelial plasticity and metastasis. Nat Rev Mol Cell Biol, 4, 657-665. Guilford, P., Hopkins, J., Harraway, J., McLeod, M., McLeod, N., Harawira, P., Taite, H., Scoular, R., Miller, A. and Reeve, A.E. (1998) E-cadherin germline mutations in familial gastric cancer. Nature, 392, 402-405. Gumbiner, B., Stevenson, B. and Grimaldi, A. (1988) The role of the cell adhesion molecule uvomorulin in the formation and maintenance of the epithelial junctional complex. J Cell Biol, 107, 1575-1587. Gupta, G.P. and Massague, J. (2006) Cancer metastasis: building a framework. Cell, 127, 679-695. Halbleib, J.M. and Nelson, W.J. (2006) Cadherins in development: cell adhesion, sorting, and tissue morphogenesis. Genes Dev, 20, 3199-3214. Hanahan, D. and Weinberg, R.A. (2000) The hallmarks of cancer. Cell, 100, 57-70. Hartwell, K.A., Muir, B., Reinhardt, F., Carpenter, A.E., Sgroi, D.C. and Weinberg, R.A. (2006) The Spemann organizer gene, Goosecoid, promotes tumor metastasis. ProcNatl Acad Sci US A, 103, 18969-18974. Hirohashi, S. (1998) Inactivation of the E-cadherin-mediated cell adhesion system in human cancers. Am J Pathol,153, 333-339. Huber, A.H. and Weis, W.I. (2001) The structure of the beta-catenin/E-cadherin complex and the molecular basis of diverse ligand recognition by beta-catenin. Cell, 105, 391-402. Huber, M.A., Kraut, N. and Beug, H. (2005) Molecular requirements for epithelialmesenchymal transition during tumor progression. CurrOpin Cell Biol, 17, 548558. Hyafil, F., Babinet, C. and Jacob, F. (1981) Cell-cell interactions in early embryogenesis: a molecular approach to the role of calcium. Cell, 26, 447-454. Ino, Y., Gotoh, M., Sakamoto, M., Tsukagoshi, K. and Hirohashi, S. (2002) Dysadherin, a cancer-associated cell membrane glycoprotein, down-regulates E-cadherin and promotes metastasis. ProcNatl Acad Sci U S A, 99, 365-370. Jechlinger, M., Grunert, S., Tamir, I.H., Janda, E., Ludemann, S., Waerner, T., Seither, P., Weith, A., Beug, H. and Kraut, N. (2003) Expression profiling of epithelial plasticity in tumor progression. Oncogene, 22, 7155-7169. Jiang, J., Kosman, D., Ip, Y.T. and Levine, M. (1991) The dorsal morphogen gradient regulates the mesoderm determinant twist in early Drosophila embryos. Genes Dev, 5, 1881-1891. Jorda, M., Olmeda, D., Vinyals, A., Valero, E., Cubillo, E., Llorens, A., Cano, A. and Fabra, A. (2005) Upregulation of MMP-9 in MDCK epithelial cell line in response to expression of the Snail transcription factor. J Cell Sci, 118, 3371-3385. Julien, S., Puig, I., Caretti, E., Bonaventure, J., Nelles, L., van Roy, F., Dargemont, C., de Herreros, A.G., Bellacosa, A. and Larue, L. (2007) Activation of NF-kappaB by Akt upregulates Snail expression and induces epithelium mesenchyme transition. Oncogene, 26, 7445-7456. Kamei, T., Matozaki, T., Sakisaka, T., Kodama, A., Yokoyama, S., Peng, Y.F., Nakano, K., Takaishi, K. and Takai, Y. (1999) Coendocytosis of cadherin and c-Met coupled to disruption of cell-cell adhesion in MDCK cells--regulation by Rho, Rac and Rab small G proteins. Oncogene, 18, 6776-6784. Kanai, Y., Oda, T., Tsuda, H., Ochiai, A. and Hirohashi, S. (1994) Point mutation of the E-cadherin gene in invasive lobular carcinoma of the breast. Jpn J CancerRes, 85, 1035-1039. Kuphal, F. and Behrens, J. (2006) E-cadherin modulates Wnt-dependent transcription in colorectal cancer cells but does not alter Wnt-independent gene expression in fibroblasts. Exp Cell Res, 312, 457-467. Larue, L., Ohsugi, M., Hirchenhain, J. and Kemler, R. (1994) E-cadherin null mutant embryos fail to form a trophectoderm epithelium. ProcNatl Acad Sci US A, 91, 8263-8267. Lien, H.C., Hsiao, Y.H., Lin, Y.S., Yao, Y.T., Juan, H.F., Kuo, W.H., Hung, M.C., Chang, K.J. and Hsieh, F.J. (2007) Molecular signatures of metaplastic carcinoma of the breast by large-scale transcriptional profiling: identification of genes potentially related to epithelial-mesenchymal transition. Oncogene, 26, 7859-7871. Lo, H.W., Hsu, S.C., Xia, W., Cao, X., Shih, J.Y., Wei, Y., Abbruzzese, J.L., Hortobagyi, G.N. and Hung, M.C. (2007) Epidermal growth factor receptor cooperates with signal transducer and activator of transcription 3 to induce epithelialmesenchymal transition in cancer cells via up-regulation of TWIST gene expression. CancerRes, 67, 9066-9076. Lu, Z., Ghosh, S., Wang, Z. and Hunter, T. (2003) Downregulation of caveolin-1 function by EGF leads to the loss of E-cadherin, increased transcriptional activity of beta-catenin, and enhanced tumor cell invasion. Cancer Cell, 4, 499-515. Luzzi, K.J., MacDonald, I.C., Schmidt, E.E., Kerkvliet, N., Morris, V.L., Chambers, A.F. and Groom, A.C. (1998) Multistep nature of metastatic inefficiency: dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Am J Pathol,153, 865-873. Makiguchi, Y., Hinoda, Y. and Imai, K. (1996) Effect of MUC1 mucin, an anti-adhesion molecule, on tumor cell growth. Jpn J CancerRes, 87, 505-511. Mani, S.A., Guo, W., Liao, M., Eaton, E.N., Ayyanan, A., Zhou, A., Brooks, M., Reinhard, F., Zhang, C.C., Shipitsin, M., Campbell, L.L., Polyak, K., Brisken, C., Yang, J. and Weinberg, R.A. (2008) The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell, In press. Mani, S.A., Yang, J., Brooks, M., Schwaninger, G., Zhou, A., Miura, N., Kutok, J.L., Hartwell, K., Richardson, A.L. and Weinberg, R.A. (2007) Mesenchyme Forkhead 1 (FOXC2) plays a key role in metastasis and is associated with aggressive basal-like breast cancers. ProcNatl Acad Sci U S A, 104, 10069-10074. Marambaud, P., Shioi, J., Serban, G., Georgakopoulos, A., Sarner, S., Nagy, V., Baki, L., Wen, P., Efthimiopoulos, S., Shao, Z., Wisniewski, T. and Robakis, N.K. (2002) A presenilin-1/gamma-secretase cleavage releases the E-cadherin intracellular domain and regulates disassembly of adherens junctions. Embo J, 21, 1948-1956. Maretzky, T., Reiss, K., Ludwig, A., Buchholz, J., Scholz, F., Proksch, E., de Strooper, B., Hartmann, D. and Saftig, P. (2005) ADAMIO mediates E-cadherin shedding and regulates epithelial cell-cell adhesion, migration, and beta-catenin translocation. ProcNatl Acad Sci US A, 102, 9182-9187. Martin, T.A., Goyal, A., Watkins, G. and Jiang, W.G. (2005) Expression of the transcription factors snail, slug, and twist and their clinical significance in human breast cancer. Ann Surg Oncol, 12, 488-496. McGuire, J.K., Li, Q. and Parks, W.C. (2003) Matrilysin (matrix metalloproteinase-7) mediates E-cadherin ectodomain shedding in injured lung epithelium. Am J Pathol, 162, 1831-1843. McNeill, H., Ozawa, M., Kemler, R. and Nelson, W.J. (1990) Novel function of the cell adhesion molecule uvomorulin as an inducer of cell surface polarity. Cell, 62, 309-316. Moody, S.E., Perez, D., Pan, T.C., Sarkisian, C.J., Portocarrero, C.P., Sterner, C.J., Notorfrancesco, K.L., Cardiff, R.D. and Chodosh, L.A. (2005) The transcriptional repressor Snail promotes mammary tumor recurrence. Cancer Cell, 8, 197-209. Moreno-Bueno, G., Cubillo, E., Sarrio, D., Peinado, H., Rodriguez-Pinilla, S.M., Villa, S., Bolos, V., Jorda, M., Fabra, A., Portillo, F., Palacios, J. and Cano, A. (2006) Genetic profiling of epithelial cells expressing E-cadherin repressors reveals a distinct role for Snail, Slug, and E47 factors in epithelial-mesenchymal transition. CancerRes, 66, 9543-9556. Nagafuchi, A., Shirayoshi, Y., Okazaki, K., Yasuda, K. and Takeichi, M. (1987) Transformation of cell adhesion properties by exogenously introduced E-cadherin cDNA. Nature, 329, 341-343. Nass, S.J., Herman, J.G., Gabrielson, E., Iversen, P.W., Parl, F.F., Davidson, N.E. and Graff, J.R. (2000) Aberrant methylation of the estrogen receptor and E-cadherin 5' CpG islands increases with malignant progression in human breast cancer. Cancer Res, 60, 4346-4348. Naumov, G.N., MacDonald, I.C., Weinmeister, P.M., Kerkvliet, N., Nadkarni, K.V., Wilson, S.M., Morris, V.L., Groom, A.C. and Chambers, A.F. (2002) Persistence of solitary mammary carcinoma cells in a secondary site: a possible contributor to dormancy. CancerRes, 62, 2162-2168. Nelson, W.J. and Nusse, R. (2004) Convergence of Wnt, beta-catenin, and cadherin pathways. Science, 303, 1483-1487. Nieto, M.A., Sargent, M.G., Wilkinson, D.G. and Cooke, J. (1994) Control of cell behavior during vertebrate development by Slug, a zinc finger gene. Science, 264, 835-839. Oft, M., Peli, J., Rudaz, C., Schwarz, H., Beug, H. and Reichmann, E. (1996) TGF-betal and Ha-Ras collaborate in modulating the phenotypic plasticity and invasiveness of epithelial tumor cells. Genes Dev, 10, 2462-2477. Orsulic, S., Huber, O., Aberle, H., Arnold, S. and Kemler, R. (1999) E-cadherin binding prevents beta-catenin nuclear localization and beta-catenin/LEF- 1-mediated transactivation. J Cell Sci, 112 ( Pt 8), 1237-1245. Ozawa, M., Ringwald, M. and Kemler, R. (1990) Uvomorulin-catenin complex formation is regulated by a specific domain in the cytoplasmic region of the cell adhesion molecule. Proc Natl Acad Sci US A, 87, 4246-4250. Peinado, H., Olmeda, D. and Cano, A. (2007) Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat Rev Cancer,7, 415428. Perez-Moreno, M., Davis, M.A., Wong, E., Pasolli, H.A., Reynolds, A.B. and Fuchs, E. (2006) pl20-catenin mediates inflammatory responses in the skin. Cell, 124, 631644. Perez-Moreno, M.A., Locascio, A., Rodrigo, I., Dhondt, G., Portillo, F., Nieto, M.A. and Cano, A. (2001) A new role for E12/E47 in the repression of E-cadherin expression and epithelial-mesenchymal transitions. J Biol Chem, 276, 2742427431. Perl, A.K., Wilgenbus, P., Dahl, U., Semb, H. and Christofori, G. (1998) A causal role for E-cadherin in the transition from adenoma to carcinoma. Nature, 392, 190-193. Peyrieras, N., Hyafil, F., Louvard, D., Ploegh, H.L. and Jacob, F. (1983) Uvomorulin: a nonintegral membrane protein of early mouse embryo. ProcNatl Acad Sci US A, 80, 6274-6277. Prakasam, A.K., Maruthamuthu, V. and Leckband, D.E. (2006) Similarities between heterophilic and homophilic cadherin adhesion. Proc Natl Acad Sci US A, 103, 15434-15439. Riethmacher, D., Brinkmann, V. and Birchmeier, C. (1995) A targeted mutation in the mouse E-cadherin gene results in defective preimplantation development. Proc Natl Acad Sci U S A, 92, 855-859. Robson, E.J., Khaled, W.T., Abell, K. and Watson, C.J. (2006) Epithelial-tomesenchymal transition confers resistance to apoptosis in three murine mammary epithelial cell lines. Differentiation,74, 254-264. Sato, T., Tanigami, A., Yamakawa, K., Akiyama, F., Kasumi, F., Sakamoto, G. and Nakamura, Y. (1990) Allelotype of breast cancer: cumulative allele losses promote tumor progression in primary breast cancer. CancerRes, 50, 7184-7189. Savagner, P. (2001) Leaving the neighborhood: molecular mechanisms involved during epithelial-mesenchymal transition. Bioessays, 23, 912-923. Savagner, P., Kusewitt, D.F., Carver, E.A., Magnino, F., Choi, C., Gridley, T. and Hudson, L.G. (2005) Developmental transcription factor slug is required for effective re-epithelialization by adult keratinocytes. J Cell Physiol, 202, 858-866. Savagner, P., Yamada, K.M. and Thiery, J.P. (1997) The zinc-finger protein slug causes desmosome dissociation, an initial and necessary step for growth factor-induced epithelial-mesenchymal transition. J Cell Biol, 137, 1403-1419. Schipper, J.H., Frixen, U.H., Behrens, J., Unger, A., Jahnke, K. and Birchmeier, W. (1991) E-cadherin expression in squamous cell carcinomas of head and neck: inverse correlation with tumor dedifferentiation and lymph node metastasis. CancerRes, 51, 6328-6337. Schuh, R., Vestweber, D., Riede, I., Ringwald, M., Rosenberg, U.B., Jackle, H. and Kemler, R. (1986) Molecular cloning of the mouse cell adhesion molecule uvomorulin: cDNA contains a B 1-related sequence. Proc Natl Acad Sci U S A, 83, 1364-1368. Shimoyama, Y., Hirohashi, S., Hirano, S., Noguchi, M., Shimosato, Y., Takeichi, M. and Abe, 0. (1989) Cadherin cell-adhesion molecules in human epithelial tissues and carcinomas. CancerRes, 49, 2128-2133. Siitonen, S.M., Kononen, J.T., Helin, H.J., Rantala, I.S., Holli, K.A. and Isola, J.J. (1996) Reduced E-cadherin expression is associated with invasiveness and unfavorable prognosis in breast cancer. Am J Clin Pathol, 105, 394-402. Stockinger, A., Eger, A., Wolf, J., Beug, H. and Foisner, R. (2001) E-cadherin regulates cell growth by modulating proliferation-dependent beta-catenin transcriptional activity. J Cell Biol, 154, 1185-1196. Stoker, M. and Perryman, M. (1985) An epithelial scatter factor released by embryo fibroblasts. J Cell Sci, 77, 209-223. Strathdee, G. (2002) Epigenetic versus genetic alterations in the inactivation of Ecadherin. Semin CancerBiol, 12, 373-379. Sulzer, M.A., Leers, M.P., van Noord, J.A., Bollen, E.C. and Theunissen, P.H. (1998) Reduced E-cadherin expression is associated with increased lymph node metastasis and unfavorable prognosis in non-small cell lung cancer. Am J Respir Crit Care Med, 157, 1319-1323. Tester, A.M., Ruangpanit, N., Anderson, R.L. and Thompson, E.W. (2000) MMP-9 secretion and MMP-2 activation distinguish invasive and metastatic sublines of a mouse mammary carcinoma system showing epithelial-mesenchymal transition traits. Clin Exp Metastasis, 18, 553-560. Thiery, J.P. (2002) Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer,2, 442-454. Thiery, J.P. (2003) Epithelial-mesenchymal transitions in development and pathologies. Curr Opin Cell Biol, 15, 740-746. Thuault, S., Valcourt, U., Petersen, M., Manfioletti, G., Heldin, C.H. and Moustakas, A. (2006) Transforming growth factor-beta employs HMGA2 to elicit epithelialmesenchymal transition. J Cell Biol, 174, 175-183. Tinkle, C.L., Lechler, T., Pasolli, H.A. and Fuchs, E. (2004) Conditional targeting of Ecadherin in skin: insights into hyperproliferative and degenerative responses. Proc Natl Acad Sci U S A, 101, 552-557. Tsuda, H., Zhang, W.D., Shimosato, Y., Yokota, J., Terada, M., Sugimura, T., Miyamura, T. and Hirohashi, S. (1990) Allele loss on chromosome 16 associated with progression of human hepatocellular carcinoma. Proc Natl Acad Sci U S A, 87, 6791-6794. Umbas, R., Isaacs, W.B., Bringuier, P.P., Schaafsma, H.E., Karthaus, H.F., Oosterhof, G.O., Debruyne, F.M. and Schalken, J.A. (1994) Decreased E-cadherin expression is associated with poor prognosis in patients with prostate cancer. Cancer Res, 54, 3929-3933. Vallin, J., Thuret, R., Giacomello, E., Faraldo, M.M., Thiery, J.P. and Broders, F. (2001) Cloning and characterization of three Xenopus slug promoters reveal direct regulation by Lef/beta-catenin signaling. J Biol Chem, 276, 30350-30358. Vasioukhin, V., Bauer, C., Degenstein, L., Wise, B. and Fuchs, E. (2001) Hyperproliferation and defects in epithelial polarity upon conditional ablation of alpha-catenin in skin. Cell, 104, 605-617. Vasko, V., Espinosa, A.V., Scouten, W., He, H., Auer, H., Liyanarachchi, S., Larin, A., Savchenko, V., Francis, G.L., de la Chapelle, A., Saji, M. and Ringel, M.D. (2007) Gene expression and functional evidence of epithelial-to-mesenchymal transition in papillary thyroid carcinoma invasion. Proc Natl Acad Sci U S A, 104, 2803-2808. Vega, S., Morales, A.V., Ocana, O.H., Valdes, F., Fabregat, I. and Nieto, M.A. (2004) Snail blocks the cell cycle and confers resistance to cell death. Genes Dev, 18, 1131-1143. Vestweber, D. and Kemler, R. (1985) Identification of a putative cell adhesion domain of uvomorulin. Embo J, 4, 3393-3398. Vincent-Salomon, A. and Thiery, J.P. (2003) Host microenvironment in breast cancer development: epithelial-mesenchymal transition in breast cancer development. Breast Cancer Res, 5, 101-106. Wang, X., Ling, M.T., Guan, X.Y., Tsao, S.W., Cheung, H.W., Lee, D.T. and Wong, Y.C. (2004) Identification of a novel function of TWIST, a bHLH protein, in the development of acquired taxol resistance in human cancer cells. Oncogene, 23, 474-482. Watson, M.A., Ylagan, L.R., Trinkaus, K.M., Gillanders, W.E., Naughton, M.J., Weilbaecher, K.N., Fleming, T.P. and Aft, R.L. (2007) Isolation and molecular profiling of bone marrow micrometastases identifies TWIST1 as a marker of early tumor relapse in breast cancer patients. Clin CancerRes, 13, 5001-5009. Weinberg, R.A. (2007) The biology of cancer.Garland Science, New York. Wicha, M.S. (2006) Cancer stem cells and metastasis: lethal seeds. Clin CancerRes, 12, 5606-5607. Wildenberg, G.A., Dohn, M.R., Carnahan, R.H., Davis, M.A., Lobdell, N.A., Settleman, J. and Reynolds, A.B. (2006) pl20-catenin and p190RhoGAP regulate cell-cell adhesion by coordinating antagonism between Rac and Rho. Cell, 127, 1027-1039. Wong, S.Y. and Hynes, R.O. (2006) Lymphatic or hematogenous dissemination: how does a metastatic tumor cell decide? Cell Cycle, 5, 812-817. Wu, X., Chen, H., Parker, B., Rubin, E., Zhu, T., Lee, J.S., Argani, P. and Sukumar, S. (2006) HOXB7, a homeodomain protein, is overexpressed in breast cancer and confers epithelial-mesenchymal transition. CancerRes, 66, 9527-9534. Xue, C., Plieth, D., Venkov, C., Xu, C. and Neilson, E.G. (2003) The gatekeeper effect of epithelial-mesenchymal transition regulates the frequency of breast cancer metastasis. CancerRes, 63, 3386-3394. Yang, J., Mani, S.A., Donaher, J.L., Ramaswamy, S., Itzykson, R.A., Come, C., Savagner, P., Gitelman, I., Richardson, A. and Weinberg, R.A. (2004) Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell, 117, 927-939. Yawata, A., Adachi, M., Okuda, H., Naishiro, Y., Takamura, T., Hareyama, M., Takayama, S., Reed, J.C. and Imai, K. (1998) Prolonged cell survival enhances peritoneal dissemination of gastric cancer cells. Oncogene, 16, 2681-2686. Yoshida, C. and Takeichi, M. (1982) Teratocarcinoma cell adhesion: identification of a cell-surface protein involved in calcium-dependent cell aggregation. Cell, 28, 217-224. Yoshiura, K., Kanai, Y., Ochiai, A., Shimoyama, Y., Sugimura, T. and Hirohashi, S. (1995) Silencing of the E-cadherin invasion-suppressor gene by CpG methylation in human carcinomas. Proc Natl Acad Sci U S A, 92, 7416-7419. Young, P., Boussadia, O., Halfter, H., Grose, R., Berger, P., Leone, D.P., Robenek, H., Charnay, P., Kemler, R. and Suter, U. (2003) E-cadherin controls adherens junctions in the epidermis and the renewal of hair follicles. Embo J, 22, 57235733. Zhang, X., Wang, Q., Ling, M.T., Wong, Y.C., Leung, S.C. and Wang, X. (2007) Antiapoptotic role of TWIST and its association with Akt pathway in mediating taxol resistance in nasopharyngeal carcinoma cells. Int J Cancer, 120, 1891-1898. Chapter 2 Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways Tamer T. Onder,"l Piyush B. Gupta,2'3 Sendurai A. Mani,' Jing Yang, 4 Eric S. Lander,"2 ', and Robert A. Weinberg 1 (1) Whitehead Institute for Biomedical Research, Cambridge, MA 02142 (2) Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139 (3) Broad Institute of MIT and Harvard, Cambridge, MA 02142 (4) Department of Pharmacology and Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093-0636 (5) Department of Systems Biology, Harvard Medical School, Boston, MA 02115 An abridged from of this work has been published in: Onder TT, Gupta PB, Mani SA, Yang J, Lander ES, Weinberg RA. Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways. CancerResearch. 2008. May 15; 68(10):3645-54. Gene expression profiling was carried out in collaboration with Piyush Gupta in Eric Lander's laboratory at the Broad Institute and Sendurai Mani in our laboratory. Experiments involving Twist were completed in collaboration with Jing Yang. Ferenc Reinhardt provided technical help in animal experiments. All other experimentation was carried out by the thesis author, Tamer Onder. Introduction As discussed in the first chapter, upon progression to high grade neoplasia, cancer cells acquire qualities that enable them to invade neighboring tissues and, ultimately, to metastasize. The steps involved in metastatic dissemination include loss of cell-cell adhesion, increased motility and invasiveness, entry into and survival in the circulation, dispersion to distant anatomical sites, extravasation, and colonization of some of those sites (Gupta and Massague, 2006). While significant progress has been made in identifying molecular alterations responsible for primary tumor growth, the signaling pathways that regulate metastatic progression are less clear. One protein prominently associated with tumor invasiveness, metastatic dissemination, and poor patient prognosis is the epithelial cell adhesion molecule Ecadherin. For example, according to one early study more than 70% of advanced in breast tumors with either lymph node or distant metastases exhibited reduced E-cadherin expression (Oka et al., 1993). Similar observations were subsequently made in prostate, lung and squamous cell carcinomas (Schipper et al., 1991; Sulzer et al., 1998; Umbas et al., 1994). Based on the function of E-cadherin as a cell-cell adhesion molecule, it was hypothesized that loss of homotypic epithelial cell adhesion is an important contributor to carcinoma progression. Initial in vitro experiments involving forced expression of E-cadherin in invasive cancer cells showed that E-cadherin acts as a suppressor of invasiveness (Frixen et al., 1991; Vleminckx et al., 1991). Subsequently, Christofori and colleagues have shown that loss of E-cadherin can be causally involved in progression from adenoma to carcinoma in an in vivo mouse model (Perl et al., 1998). In the Rip-Tag model of P-islet cell tumorigenesis, transgenic expression of E-cadherin prevented malignant progression. Moreover, expression of a truncated E-cadherin allele that acts as a dominant-negative accelerated tumor cell invasion (Perl et al., 1998). Recently, the Jonkers group also showed that combined loss of p53 and E-cadherin in the mouse mammary gland results in metastatic tumors resembling human lobular breast cancers (Derksen et al., 2006). In human tumors, loss or reduction of E-cadherin expression can be caused by somatic mutations, chromosomal deletions, proteolytic cleavage, and silencing of the CDH1 promoter (Berx et al., 1998; Hirohashi, 1998; Maretzky et al., 2005; Strathdee, 2002). While some of these events, such as point mutations, lead to loss of the adhesive function of E-cadherin, they may preserve the expression of the intracellular domain of E-cadherin. Alternatively, other mechanisms of E-cadherin loss, such as promoter hypermethylation, lead to a complete shut down of E-cadherin synthesis. Therefore, I first wished to ascertain whether there is a qualitative difference in cell phenotype following two different types of E-cadherin inactivation. In light of its well-established function in maintaining adherens junctions, Ecadherin loss ostensibly promotes metastasis by enabling the first step of the metastatic cascade-the disaggregation of cancer cells from one another. However, it has been unclear whether E-cadherin loss also supports the successful completion of additional steps of the invasion-metastasis cascade. Previous work has revealed that several structural proteins associated with adherens junctions can also mediate intracellular signaling functions (see chapter 1) (Nelson and Nusse, 2004; Perez-Moreno et al., 2006; Vasioukhin et al., 2001; Wildenberg et al., 2006). Accordingly, E-cadherin loss may result in the activation of specific downstream signal transduction pathways that, in turn, confer traits upon cancer cells facilitating completion of the later steps of metastasis. In the first part of this work, I examined whether loss of cell-cell contacts is the sole contribution to metastasis provided by E-cadherin loss. I observed that experimental inhibition of cell-adhesion alone does not lead to metastasis. On the other hand, inhibition of E-cadherin protein expression led to the activation of multiple transcriptional pathways associated with the induction of an epithelial-to-mesenchymal transition and facilitated robust metastasis. I demonstrated that at least two of these downstream pathways contribute to the later steps of metastatic dissemination. Thus, these findings reveal a complex transcriptional network that is controlled by the E-cadherin molecule. Results Characterization of in vitro phenotypes resulting from expression of E-cadherin shRNA and dominant-negative E-cadherin In the following set of experiments I took advantage of a series of human mammary epithelial cell lines to express or inhibit genes of interest. These cells were initially derived in our laboratory from primary human mammary epithelial cells (HMECs) by the sequential introduction of the genes encoding the SV40 early region and the telomerase catalytic subunit, yielding immortalized but untransformed human mammary epithelial cells (HMLE) (Elenbaas et al., 2001). I subsequently infected HMLE cells with a retrovirus carrying an oncogenic H-ras allele to create the transformed HMLER cell line. In addition, I infected the cells with a viral vector expressing the green fluorescent protein (GFP). Figure 1 depicts the scheme used in creating these and subsequent cell lines. To resolve E-cadherin's role in cell-cell adhesion from its intracellular signaling functions, I utilized two distinct methods of inhibiting E-cadherin function: (1) shRNAmediated knockdown of E-cadherin, which resulted in >90% reduction of E-cadherin protein levels (shEcad) and (2) expression of a truncated form of E-cadherin (DN-Ecad) lacking the ectodomain of the wildtype protein (Figure 2A, B). This truncation product, which retains the cytoplasmic domain, has previously been shown to act as a dominantnegative by binding and titrating cytoplasmic proteins associated with the adherens junctions, thereby abstracting them from wild-type E-cadherin molecules (Dahl et al., 1996). Importantly, the DN-Ecad construct utilized here mimics many of the in-framedeleted and point-mutated E-cadherin alleles isolated from human tumors. These E- cadherin alleles have also been shown to be deficient in cell-cell adhesion and to act in a trans-dominant manner in cell-based assays (Handschuh et al., 1999). HMLER cells expressing control shRNA (shCntrl) grew in monolayer culture as epithelial clusters with a typical cobblestone morphology, while knockdown of Ecadherin and expression of the dominant-negative protein both resulted in loss of cellcell contacts and cell scattering (Figure 2A). Similar phenotypes were observed with the immortalized HMLE cells as well (Figure 3A). While the cell populations expressing shEcad and DN-Ecad both lost cell-cell adhesion, the individual cells expressing shEcad displayed, in addition, an elongated, fibroblastic morphology (Figure 2A, 3A). To confirm that this phenotype was a specific consequence of E-cadherin ablation, I re-expressed a wild-type E-cadherin in HMLEshEcad cells. Expression of an shRNA-resistant wild-type murine E-cadherin gene reversed the observed phenotypes, causing the shE-cad cells to revert to an epithelial morphology and to regenerate a cobblestone growth pattern in monolayer culture (Figure 4A,B). The acquisition of a fibroblastic morphology by the cells expressing shEcad suggested that these cells had undergone an epithelial-to-mesenchymal transition (EMT). To determine whether, in addition to the observed morphological changes, the molecular alterations associated with an EMT occurred upon loss of E-cadherin, I assessed the status of EMT markers in control, shEcad- and DN-Ecad- expressing cells. Upon shRNAmediated loss of E-cadherin, expression of mesenchymal proteins, such as N-cadherin and vimentin, was markedly upregulated (Figure 2B, C, 3B). In contrast, none of the mesenchymal proteins was upregulated in the HMLER-DN-Ecad cells (Figure 2B). While the expression of adherens junction-associated catenins was largely maintained in the absence of E-cadherin, epithelial cytokeratins were downregulated (Figure 2B). These observations indicate that complete loss of E-cadherin protein results in an EMT, whereas inhibition of E-cadherin-mediated cell-cell adhesion causes cell scattering without the additional changes in differentiation state associated with passage through the EMT program. Primary tumor formation and metastasis of E-cadherin-inhibited cells To determine whether E-cadherin perturbation affects primary tumor growth, I injected the control HMLER cells and the two E-cadherin-inhibited derivative lines subcutaneously into immunocompromised mice. While all three cell lines generated primary tumors with comparable latencies, tumor growth rates differed slightly among the shCntrl, DN-Ecad and shEcad HMLER cells, with the latter growing most rapidly (Figure 5A). I confirmed that E-cadherin suppression and expression of the dominantnegative construct were maintained during the course of tumor growth (Figure 6A). Immunohistochemical staining of tumors with anti-E-cadherin antibodies also supported this conclusion (Figure 6B). I next determined whether metastatic dissemination to the lungs occurred when these various transformed cells were implanted in an orthotopic tissue site-the mammary fat pad. To facilitate detection of micro- and macroscopic metastases, I engineered the various introduced HMLER cells to express the green fluorescent protein (GFP). I controlled for differences in primary tumor growth rates by sacrificing animals when their tumor burden reached a pre-established threshold. Accordingly, the mean primary tumor weights at the end of the experiment were comparable across the shCntrl, shEcad and DN-Ecad cohorts (Figure 6B). In consonance with previous findings (Elenbaas et al., 2001), control HMLER cells did not form any macroscopic nodules in the lungs, and only a few GFP-positive cells could be detected as micrometastases in the lungs of animals bearing these tumors (Figure 5D). In contrast, animals bearing HMLER-shEcad tumors harbored numerous micro- and macroscopic metastases in their lungs (Figure 5D). In fact, the lung metastatic burden in animals bearing HMLER-shEcad tumors covered a substantial portion of the total lung surface after an 8 week incubation period. (Figure 5C, D). While I did find small numbers of cells in the lungs of mice bearing HMLER-DN-Ecad tumors, macroscopic nodules were never observed. These results indicate that the two modes of E-cadherin inhibition yield cancer cells with qualitatively different metastatic powers. Since the HMLER-DN-Ecad cells were incapable of forming macroscopic metastases from primary tumors growing at orthotopic sites, I also subjected these cells to a less stringent test of metastatic competence - the "experimental metastasis" model, which involves tail vein injection of tumor cells. Using this assay, HMLER-shCntrl cells rarely formed macroscopic nodules in the lungs after an 8-week incubation period (2 nodules in 1/5 mice) (Figure 5C, D). In contrast, HMLER-shEcad cells established numerous lung macrometastases, some of which invaded the rib cage. However, the DNEcad cells did not form either micro- or macroscopic lung nodules in any of the mice, thereby echoing the previous results (Figure 5D). Taken together, these observations indicated that while the loss of cell-cell contacts does not suffice to impart metastatic competence to cancer cells, the concomitant liberation of adherens junction proteins and other associated proteins from the cytoplasmic tail of E-cadherin indeed suffices to do so. Hence, in addition to promoting dissemination from primary tumor sites via the disruption of cell-cell contacts, Ecadherin loss facilitates the successful completion of one or more subsequent ratelimiting steps of the invasion-metastasis cascade. Functional differences in cell motility, invasion, and anoikis-resistance To determine the functional changes in cell behavior that occurred following Ecadherin loss, I employed several in vitro assays to characterize the control and Ecadherin-perturbed HMLER cells. More specifically, I utilized Boyden chamber assays to gauge the migratory and invasive abilities of these various cells. While control cells were minimally motile and invasive, E-cadherin loss resulted in a significant increase in both motility and invasiveness (Figure 7A, B). However, the DN-Ecad cells, while more motile than controls (albeit less so than the shEcad cells), failed to invade through Matrigel-coated membranes. Disseminating tumor cells that enter into the bloodstream lose their attachment to the extracellular matrix, resulting in induction of the form of apoptosis termed anoikis (Gupta and Massague, 2006). Moreover, expression of anti-apoptotic molecules that confer resistance to anoikis has been shown to promote metastasis (Douma et al., 2004; Martin et al., 2004). I therefore set out to evaluate the anoikis-sensitivity of the DN-Ecad, shEcad and control HMLER cells. When placed into suspension cultures, the viability of both control and DN-Ecad cells declined dramatically over a 2-day period (Figure 7C); in contrast, shEcad cultures exhibited only a minor decrease in cell number during the same time period (Figure 7C). To confirm that loss of viability was indeed a consequence of increased apoptosis, I stained cells from these cultures with annexin V. While the control and DN-Ecad suspension cultures exhibited a significant fraction of cells undergoing apoptosis, minimal annexin V-positivity was observed in the shEcad cultures (Figure 7D). Taken together, these results indicated that EMT induction upon E-cadherin loss promotes cancer cell invasiveness and enables survival in the absence of substrate attachment. Tumor initiating capacity of E-cadherin inhibited cells Colonization or establishment of distant macro-metastases, by definition, requires that the disseminated cells be capable of tumor initiation at the secondary sites. The recent discovery that tumor formation and growth are driven by a minor subpopulation of cancer cells within tumors termed cancer stem cells (CSCs), led to the hypothesis that metastatic ability of a given primary tumor may depend on the prevalence of tumorinitiating cells within that tumor (Gupta and Massague, 2006). Importantly, during the course of this thesis, research from our laboratory indicated that breast carcinoma cells that have undergone an EMT display many features of CSCs, such as tumor seeding at limiting dilutions, formation of mammospheres and high CD44, low CD24 expression (Mani et al., 2008). Since I observed that E-cadherin-inhibited HMLER cells were highly metastatic, I wished to determine whether loss of E-cadherin and the resulting EMT led to the acquisition of CSC-like properties. Limiting dilution assays indicated that the HMLERshEcad line seeded tumors with as few as 1000 cells in vivo, which was approximately two orders of magnitude less than the minimum number of cells required by the HMLERshCntrl line to seed tumors (Figure 8A). Additionally, when grown in suspension, HMLER-shEcad cells showed a -100-fold increase in mammosphere formation relative to control cells (15 spheres vs. -0.15 spheres per 100 cells; Figure 8B). These functional assays indicated that the HMLERshEcad line harbored significantly greater numbers of cells with CSC-like properties relative to the HMLERshCntrl cells. This notion was further supported by FACS analyses using antibodies against markers that are reported to enrich for breast CSC populations (Figure 8C) (AlHajj et al., 2003). Thus, the HMLER-shEcad line exhibited a -10-fold increase in the percentage of CD44high/CD24low cells compared to the control HMLERshCntrl line (Figure 8C). Taken together, these results demonstrated that loss of E-cadherin and the resulting EMT led to the acquisition of CSC-like properties. Moreover, these findings lend credence to the idea that the number of metastases generated from a primary tumor depends on the proportion of cells that have a CSC-like phenotype within that primary tumor Molecular mechanism of E-cadherin-loss induced EMT Collectively, the above experiments indicate that induction of the EMT upon loss of E-cadherin is sufficient to endow cells with essentially all the traits needed to complete the invasion-metastasis cascade. Importantly, the acquisition of these traits was not a direct consequence of the disruption of cell-cell adhesion per se, suggesting that activation of specific signaling pathways following loss of E-cadherin is important for the observed phenotypes. The role of P-catenin in EMT and metastasis induced by E-cadherin loss A critical intracellular protein associated with the E-cadherin cytoplasmic tail is 3-catenin, which has previously been implicated in the induction of EMTs in various contexts (Conacci-Sorrell et al., 2003; Eger et al., 2004; Kim et al., 2002; Liebner et al., 2004; Morali et al., 2001; Yang et al., 2006b). I therefore examined whether E-cadherin loss influenced [-catenin status and whether such change in [-catenin contributed, in turn, to the subsequent induction of EMT. While in control cells [-catenin localization was predominantly associated with cell-cell junctions, shEcad cells exhibited a diffuse cytoplasmic and nuclear localization pattern (Figure 9A). Examination of the phosphorylation status of P-catenin in shEcad cells indicated that it was largely unphosphorylated and therefore present in an active state (Figure 9A) (Nelson and Nusse, 2004). In contrast, a significant amount of [-catenin protein in control cells was phosphorylated and thus targeted for ubiquitylation and degradation (Figure 9A). It is known that the main kinase responsible for phoshorylating and thereby labeling [-catenin for subsequent ubiquitylation is GSK3-P (Clevers, 2006). In accordance, treatment of cells with a GSK3-1 inhibitor reduced the amount of phosphorylated p-catenin in whole cell lysates, suggesting that this pathway is also functional in the human mammary epithelial cell system (Figure 9A). I next determined whether the observed levels of p-catenin phosphorylation in control and shEcad cells was correlated with the activateion state of GSK-30. Indeed, I found that GSK-30 was largely in a phosphorylated and therefore inactive state in the shEcad cells, in contrast to its unphosphorylated state in control cells (Figure 9A) (Cross et al., 1995). Hence, loss of E-cadherin was sufficient to liberate P-catenin from its site of sequestration adjacent to the plasma membrane and to permit its survival in an unphosphorylated, stabilized state. To determine whether P-catenin was functionally active following E-cadherin loss, I inhibited 3-catenin expression in HMLER-shEcad cells using a lentiviral shRNA vector (shEcad+shBcat; Figure 9B). Downregulation of P-catenin resulted in a slightly reduced growth rate in vitro (Figure 9C). Importantly, 3-catenin knockdown in shEcad cells significantly diminished expression of the mesenchymal proteins N-cadherin, vimentin and fibronectin (Figure 9B). In contrast, the expression of epithelial cytokeratins in the shEcad cells did not change upon P-catenin knockdown (Figure 8B). I next assessed whether P-catenin affected cell-biological phenotypes associated with metastasis. P-catenin inhibition in shEcad cells decreased cancer cell invasiveness (Figure 9D). In addition, knockdown of 0-catenin sensitized shEcad cells to apoptosis when the cells were placed in suspension culture (Figure 9D). To assess the contribution, if any, of 3-catenin to the metastatic behavior of shEcad cells, I introduced the control shEcad and double knockdown (shEcad+shpcat) cells via the tail-vein directly into the circulation. I observed that the HMLER+shEcad+shpcat cells were significantly impaired (approxiametly 10-fold) in their ability to form lung metastases compared to the HMLER+shEcad cells (Figure 9E). Since P-catenin was necessary for various aspects of the EMT occurring following E-cadherin loss, I examined whether P-catenin activation would, on its own, suffice to induce EMT. To accomplish this I made use of an N-terminal truncation mutant of P-catenin that is refractory to GSK-30 mediated phosphorylation and subsequent degradation (Kolligs et al., 1999). Overexpression of this constitutively active p-catenin in HMLE cells failed to induce an EMT (Figure 9F). Although significant overexpression was achieved, I did not detect any increase in the expression of mesenchymal markers and no change in cell morphology (Figure 9F). Taken together, these data indicate that while p-catenin is required for changes in the expression of certain biochemical markers associated with the EMT as well as for acquisition of traits facilitating metastasis, it does not suffice on its own to induce these phenotypes. Gene expression profiling identifies global changes resulting from E-cadherin loss To obtain an unbiased view of the molecular changes resulting from the liberation of intracellular adherens junction proteins from the cytoplasmic domain of E-cadherin, we compared the gene expression profiles of the shEcad, DN-Ecad, and control cells. In order to avoid the influence of specific oncogenic lesions, we utilized the immortalized, non-transformed mammary epithelial (HMLE) cells for this analysis. Furthermore, we reasoned that a comparison between the shEcad- and the DN-Ecad- expressing cells would make it possible to identify those changes in gene expression that were not consequences of the loss of cell-cell adhesion. In addition, we incorporated into this analysis the expression profile of cells in which both E-cadherin and p-catenin were inhibited; this would allow us to evaluate the contribution of p-catenin to the gene expression changes observed following inhibition of E-cadherin. The global expression profiles revealed that the shEcad cells exhibited significantly greater overall differential gene expression relative to the control cells than did the DN-Ecad cells (Figure 10A, B; see Appendix 1 and 2 for complete lists). Nonetheless, there was an overlap between the genes that were differentially expressed in both the shEcad and DN-Ecad cells relative to the control cells (18 induced and 59 repressed; Appendix 3). This indicated that the loss of cell-cell contacts, on its own, suffices to alter expression of this subset of cellular genes. Among the genes significantly induced in the shEcad cells relative to controls were a number of mesenchymal markers known to be associated with passage through an EMT, including N-cadherin, vimentin, fibronectin, and 7 distinct collagens. In addition to these induced mesenchymal markers, 11 distinct cytokeratin genes were downregulated in the shEcad cells relative to the controls (Appendix 1). While none of the above-mentioned mesenchymal genes was induced in cells expressing DN-Ecad, we did observe downregulation of 4 of the 11 cytokeratin genes in the DN-Ecad-expressing cells. We further analyzed these changes by testing for their association with the biological processes arrayed in the Gene Ontology (GO) database (Dennis et al., 2003). Genes most significantly downregulated in the shEcad and DN-Ecad cells were, in both cases, strongly associated with cell differentiation (Figure 10C). While the genes induced in the shEcad cells were strongly associated with matrix adhesion and skeletal development, the genes induced in the DN-Ecad cells were not significantly associated with any specific biological processes listed in the GO database (Figure 10C). This indicated that downregulation of certain epithelial genes results directly from the dissolution of cell-cell contacts, and that the induction of mesenchymal characteristics required, in addition, the loss of the cytoplasmic domain of E-cadherin. To directly assess the extent to which of these various cell lines had entered into a mesenchymal state, we performed hierarchical clustering on the expression data using a global metric computed on the basis of a 225-gene set capable of discriminating between mammary epithelial cells that have or have not been induced to undergo EMT by a variety of means, including the expression of the Snail and Twist transcription factors (manuscript in preparation). This analysis revealed that the expression profiles of the control and DN-Ecad cell lines were essentially indistinguishable with respect to these EMT-associated genes and were both indicative of cells that have not undergone an EMT (Figure 10A). In contrast, the expression profile of the shEcad cells indicated that they had in fact passed through an EMT (Figure 10A). These findings reinforced our earlier conclusion that the shEcad cells, but not the DN-Ecad cells, have indeed adopted a mesenchymal cell state. Expression of multiple transcriptional regulators are induced upon E-cadherin loss but not cell-cell disaggregation To evaluate the contribution of P-catenin to the gene expression changes observed upon E-cadherin loss, we compared the expression profile of the doubly altered cells (HMLE shEcad + sh3cat) with the profile of the shEcad cells. We observed that -14% of the genes (84 genes out of 617) that were differentially expressed in shEcad cells relative to the control cells were dependent upon p-catenin for their regulation. The p-catenindependence of a number of representative genes induced or repressed by E-cadherin loss is depicted in Figure 10D. Among these p-catenin dependent genes were a subset of known Wnt target genes such as ephrin B2 receptor, cadherinl 1 and fibronectin (Batlle et al., 2002; De Langhe et al., 2005; Hadeball et al., 1998). Since 84% of the gene expression changes that occurred upon E-cadherin loss were not dependent on p-catenin function, it was likely that other transcriptional regulators were mediating the transcriptional changes following E-cadherin loss. In fact, there were 19 transcription regulators that were upregulated more than 3-fold in shEcad cells relative to control cells (Figure 10E). Of these factors, only two were upregulated in the DN-Ecad cells, indicating that induction of the remaining 17 required the loss of the cytoplasmic tail of E-cadherin. In light of the observation that p-catenin is responsible for a minority of gene expression changes that occurred upon E-cadherin loss, these findings further implicated one or more of these 17 factors as potential contributors to the EMT observed following E-cadherin loss. Upregulation of Twist upon loss of E-cadherin and its functional role The presence of the transcription factors Twist and TCF8 (ZEB-1) among the set of genes highly upregulated upon E-cadherin loss was surprising (Figure 10D). Twist had previously been shown to be essential for the metastasis of mouse mammary tumor cells and to be expressed in a high proportion of human lobular breast cancers in correlation with E-cadherin downregulation (Yang et al., 2004). Twist is functionally related to a set of transcription factors, including Snail, Slug, SIP1, ZEB-1, that are known to repress E-cadherin expression and are sufficient in various cellular contexts to induce EMTs. Based on extensive previous work, these factors are thought to act upstream of E-cadherin, and several are known to directly repress transcription of the CDH1 gene itself (Bolos et al., 2003; Cano et al., 2000; Comijn et al., 2001; Eger et al., 2005; Grooteclaes and Frisch, 2000). The identification of Twist and ZEB1 in our gene expression data suggested the opposite possibility-namely that the expression of EMT-inducing transcription factors may, under certain circumstances, be controlled by the levels of E-cadherin. We pursued this hypothesis by examining the functional consequences of Twist induction following E-cadherin loss. Analyses of Twist mRNA levels using real-time RT-PCR validated the expression array data, indicating that Twist was specifically upregulated in HMLERshEcad but not in control or the DNE-cad cells (Figure 11A). To determine whether Twist expression played a functionally important role in the EMT induced upon E-cadherin loss, we super-infected shEcad cells with a lentiviral shRNA vector targeting Twist. Introduction of this shRNA vector resulted in an approximately 5-fold decrease in Twist levels, reducing it below the basal levels present in the parental HMLER line; Figure 11B). While Twist inhibition led to a -50% reduction of N-cadherin levels, the expression of vimentin remained largely unaffected (Figure 11B). To further evaluate the functional consequences of Twist upregulation to metastasis-associated traits, we characterized the contribution of Twist to the induction of invasion and anoikis resistance following E-cadherin loss. Twist inhibition resulted in a significant reduction in the invasive behavior of shEcad cells (Figure 11C). In addition, sensitivity to induction of anoikis was partially restored upon Twist inhibition (Figure 11C). These findings indicated that Twist is an important mediator of signaling in response to E-cadherin loss and operates upstream of certain cellular functions that contribute to metastatic competence. The observed effects on motility, invasiveness and anoikis-resistance following Twist knock-down were similar to those observed upon 3catenin knock-down. Since the induction of Twist was not P-catenin-dependent, we concluded that these two factors may be acting in parallel to program the cellular changes observed following E-cadherin loss. To determine if Twist was also mediating the acquisition of metastatic powers following E-cadherin downregulation, we evaluated the metastatic ability of Twistinhibited cells using the experimental metastasis assay. While control shEcad cells were able to efficiently colonize the lung, as before, cells doubly inhibited for E-cadherin and Twist exhibited a marked decrease in the ability to do so (Figure 11D). Collectively, these results indicate that Twist is a crucial downstream effector of cellular functions in response to E-cadherin loss and is necessary for the metastasis of E-cadherin-deficient cells. Taken together, the experiments described in this chapter indicate that while loss of cell-cell adhesion may be a requisite step in the invasion-metastasis cascade, it on its own does not enable formation of metastases. On the other hand, loss of E-cadherin expression is sufficient to endow cells with metastatic powers and it does so through induction of an epithelial-to-mesenchymal transition (EMT), invasiveness and anoikisresistance. Moreover, wide-ranging transcriptional changes are observed upon E-cadherin loss, some of which depend on E-cadherin binding partner P-catenin. E-cadherin also regulates the expression levels of several additional transcription factors, one of which, Twist, is functionally important for metastasis following loss of E-cadherin. These observations indicate that E-cadherin acts a master regulator of epithelial cell identity and of carcinoma cell dissemination. Materials and Methods Cell culture. Immortalized human breast epithelial cells (HMLE) generated through the introduction of the SV40 large T (LT) antigen, and hTERT were maintained as described (Elenbaas et al. 2001). To generate tumorigenic and GFP-expressing derivatives, we infected HMLE cells with a pBabe H-ras and PRRL-GFP vector (Elenbaas et al., 2001). To measure cell growth rates, 2500 cells were seeded onto 96 well plates in triplicate. Cell viability was measured using Cell-titer-Glo (Promega) according to manufacturer's instructions. Treatment with the GSK inhibitor BIO (Calbiochem) was done for 6 hours at 1gM concentration. Plasmids. Sequences targeting the E-cadherin and 3-catenin shRNAs were taken from the website of the RNAi consortium at the Broad Institute and cloned into the PLKOpuro and -hygro vectors 1 (Stewart et al., 2003). E-cadherin and p-catenin targeting sequences were GCAGAAATTATTGGGCTCTTT and GCTTGGAATGAGACTGCT GAT, respectively. Control shRNAs were either targeted against GFP or a non- functional E-cadherin shRNA. Twist shRNA expression vector was described previously (Yang et al. 2004). Wildtype and dominant-negative mutant E-cadherin cDNAs (gifts of Dr. Gerhard Christofori) were expressed from the pWZL-Blasticidin vector. pBp-AN90pcatenin vector was used as described (Hartwell et al., 2006). 1(http://www.broad.mit.edu/genome bio/trc/ Viral Production and Infection of target cells. Viral production and infection of target cells were described previously (Stewart et al., 2003). Infected cells were selected with 2ug/ml puromycin, 200ug/ml Hygromycin, and 10ug/ml Blasticidin. Antibodies, Immunoblotting, Immunofluorescence and Histology. Antibodies utilized were E-cadherin, ax-catenin, 0-catenin, y-catenin, N-cadherin (BD Transduction), phospho-p-catenin, phospho-GSK3p, GSK3P (Cell Signaling), Vimentin V9 (NeoMarkers), Actin, Tubulin (Abcam), H-Ras (Santa Cruz), Fibronectin (Sigma), Cytokeratin 8 (Troma-1, Developmental Studies Hybridoma Bank, University of Iowa). Twist monoclonal antibody was used as previously described (Yang et al., 2004). For immunohistochemical staining anti-large T antigen (Santa Cruz) and anti-E-cadherin (Cell Signaling) antibodies were used as described (Elenbaas et al. 2001). FACS. The anti-CD44 (clone G44-26) antibody conjugated to Allophycocyanin (APC) and the anti-CD24 antibody (clone ML5) conjugated to Phycoerythrin (PE) used for FACS analysis were obtained from BD Bioscience and used according to the manufacturer's instructions. Propidium Iodide (5ug/ml) was included in the staining protocol to distinguish live cells. Anoikis assays. 75,000 cells were seeded onto 6-well ultra-low attachment plates (Coming). After 24 or 48 hours, cells were harvested and incubated at 370 C with 0.25% trypsin for 5 min to prevent cell aggregation. Viable cells were counted using trypan blue. For FACS analysis, we used the ApoAlert Annexin V-FITC Apoptosis Kit (Takara-Bio) according to manufacturer's instructions and used a FACSCalibur flow cytometer (Becton Dickinson) Motility and Invasion assays. 5 x 105 cells were resuspended in 1:1 mixture of DME:F12 and placed into uncoated or Matrigel- coated Transwell inserts containing 8pm filter (BD Falcon) in triplicate. The bottom wells contained DME:F12 media with growth factors, insulin (10 pg/mL), epidermal growth factor (10 ng/mL), and hydrocortisone (1 pg/mL). After 12 or 16 hours the cells on the upper surface of the filters were removed with a cotton swab. The filters were fixed and stained using a DiffQuick staining kit (Dade Behring) and photographed. The migrated cells were then counted. Mammosphere Assays. Mammosphere culture was performed as described (Dontu et al., 2003), except that the culture medium contained 0.9 % methyl cellulose (Stem cell technologies) to prevent cell aggregation. 102 or 103 cells were plated per well in ultralow attachment 96-well plates (Coming). The mammospheres were cultured for 7-10 days and then photographed and counted. In vivo tumorigenesis and metastasis assays. NOD-SCID mice (propagated on site) and nude mice (NCR nude; Taconic, Hudson, NY) were used in these studies, and all protocols were approved by the Massachusetts Institute of Technology Committee on Animal Care. Nude mice received 400 rad of 7-radiation using a dual 137Cesium source 1 day before tumor cell injection. Mice were anesthetized with either avertin or isoflurane. For orthotopic injections, 1 million cells in 20 pl of Matrigel (Becton Dickinson) diluted 1:2 in DME were injected into each of two mammary glands per female NOD-SCID mouse. For subcutaneous injections, lx 106 cells in 100 pl of Matrigel diluted 1:2 in DME were injected at each of two sites per nude mouse. Tumor diameters were measured biweekly using precision calipers. Primary tumor bearing animals were sacrificed when the tumors reached a diameter of 2 cm. For tail vein injections, 5 x 105 cells in 200 pl PBS were injected per NOD/SCID mouse and the mice were sacrificed 8 weeks post injections. For limiting dilution assays 106, 105 104 , and 103 of HMLER-shCntrl or HMLER-shEcad cells in 100 pl of Matrigel diluted 1:2 in DME were injected subcutaneously into NOD-SCID mice. The tumor incidence was monitored for 60 days following injection. Visualization and Quantification of GFP-Labeled Lung Metastases. Upon necropsy, lungs of injected mice were removed and examined under a Leica MZ 12 fluorescence dissection microscope. Images of lung lobes were captured at identical settings. The amount of metastatic burden was quantified using ImageJ 2 and expressed as a percentage of the total lung area captured (Rasband, 1997-2006). Microarray Hybridization, Data Collection, and Analysis. Total RNA was extracted from 3 independent culture plates for each cell type using an RNeasy Mini kit (Qiagen). Synthesis of cRNA and hybridization/scanning of microarrays were performed with Affymetrix GeneChip products as described in the GeneChip manual. Normalization of the raw gene expression data, quality-control checks and subsequent analyses were 2 http://rsb.info.nih.gov/ii/ performed using the open-source R-project statistical software (http://www.r-project.org/) (Team, 2007) in conjunction with Bioconductor packages. Raw data files (.CEL) were converted into probe-set values using RMA normalization. Hierarchical clustering with complete linkage was performed on the probe-set data using the Euclidean metric. The probes used for hierarchical clustering and heatmap generation were identified on the basis of their consistent up-regulation and down-regulation across a set of isogenic lines (HMLE) that were induced into EMT using a variety of inducers: Twist-, TGF-P3-, Snailor Goosecoid- overexpression. The identified probes constitute an EMT expression signature whose targets are >3-fold up-regulated or down-regulated in each of the cell lines induced into EMT relative to vector control cells that were not induced into EMT. The microarray data have been deposited in NCBIs Gene Expression Omnibus and are accessible through GEO Series accession number GSE9691. SYBR-Green Real-Time RT-PCR. RT-PCR analysis of Twist were carried out as described (Yang et al., 2004). Twist GGAGTCCGCAGTCTTACGAG, and its TCTGGAGGACCTGGTAGAGG. GAPDH GACCCCTTCATTGACCTCAAC, and RT-PCR RT-PCR RT-PCR its RT-PCR forward primer is reverse primer is forward primer is reverse primer is CTTCTCCATGGTGGTGAAGA. Total RNA extracted as described above was reverse transcribed with Hexanucleotide Mix (Roche). The resulting cDNAs were used for PCR using SYBR-Green Master PCR mix (Applied Biosystem) in triplicates. PCR and data collection were performed on iCycler (BioRad). All quantitations were normalized to an endogenous control GAPDH. The relative quantitation value for each target gene compared to the calibrator for that target is expressed as 2 -(Ct"c) (Ct and Cc are the mean threshold cycle differences after normalizing to GAPDH). Figures Primary Human Mammary Epithelial Cells (HMEC) + Telomerase - hTert HME + Large T, small T antigen HMLE +GFP + oncogenic H-Ras HMLER Jc-q-----. 4' shCntrl shE-cad DN-Ecad Figure 1. Schematic representation of the generation of HMLER-shCntrl, shEcad, and DN-Ecad cell lines. HMLE cell line was created by Elenbaas et al. (Elenbaas et al., 2001). All subsequent genetic manipulations were performed during the course of this thesis work as described. Figure 2. Characterization of in vitro phenotypes resulting from expression of E-cadherin shRNA and dominant-negative E-cadherin HMLE R shCntri *o shEcad po W-DN-Ecad 0 DU ap a Uj U I N-cadherin E-cadherin - Vimentin -catenin - - ~&catenin - a-catenin DN-Ecad - CK-8 A C E-cadherin Vimentin .e- N-cadherin shCntrl shEcad Figure 2. Characterization of in vitro phenotypes resulting from expression of Ecadherin shRNA and dominant-negative E-cadherin. (A) The morphology of HMLER-shCntrl, shEcad, and DN-Ecad cell lines as grown under regular tissue culture conditions. (B) Expression levels of the endogenous and dominant-negative mutant E-cadherin (Arrowhead), p-catenin, '-catenin, a catenin, Cytokeratin-8 and mesenchymal proteins N-cadherin and vimentin in HMLER-shCntrl, shEcad, and DN-Ecad cells examined by immunoblotting. 3-actin is used as a loading control. (C) Immunofluorescence staining of E-cadherin (green), vimentin (red) and N-cadherin (Green) in the HMLER cells expressing either shCntrl or shEcad. The blue signal represents the nuclear DNA staining by DAPI. Figure 3. Morphology of shCntrl, shEcad and DN-Ecad expressing HMLE cells A B HMLE shCntrl shEcad LUULJ H-Ras I 0-catenin N-cadheri n ~ N-cadheri n Vimentin DN-Ecad _jJ ".W E-cadherin shEcad ijiliJ -.J 2 shCntrl Actin ~ ~ ";~ -~9-~s~A· I -WO Figure 3. Morphology of immortalized HMLE cells expressing shCntrl, shEcad, and DN-Ecad. (A) The morphology of HMLE-shCntrl, shEcad, and DN-Ecad cell lines as grown under regular tissue culture conditions. (B) Expression levels of H-ras, E-cadherin, P-catenin, N-cadherin and vimentin in shCntrl- or shEcad- expressing immortalized (HMLE) and transformed (HMLER) breast epithelial cells examined by immunoblotting. p-actin is used as a loading control. A B shEcad LL shEcad pWB-GFP E-cadherin II N-caanenrn pWB-Ecad shEcad *iN. Vimentin Actin wo Figure 4. Re-expression of E-cadherin restores the epithelial phenotype in shEcad cells. (A) Morphology of immortalized HMLE-shEcad cells expressing control vector (pWB) or murine E-cadherin expression vector (pWB-Ecad). (B) Expression levels of E-cadherin, N-cadherin and vimentin in HMLE-shCntrl or HMLE-shEcad cells expressing control vector (pWB) or murine E-cadherin expression vector (pWB-Ecad). p-actin is used as a loading control. disaggregation Figure 5. Loss of E-cadherin is sufficient for metastasis whereas cellular isnot C B A 25 -4- MO-*w - s•Ead 25 15 _ ;5 10 9 16 l 0 10 20 30 40 50 ii Days Tail-vein Orthotopic GFP LargeT 1·. LargeT GFP shCrtrl S :·' T~-,· I: shEcad -;i-_ I- -. - - ------ DN-Ecad ** ;;:;:,· ;·· Figure 5. Loss of E-cadherin is sufficient for metastasis whereas cellular disaggregation is not. (A) Growth patterns of primary subcutaneous tumors formed by the HMLER-shCntrl, shEcad and DN-Ecad cells. Each data point represents the mean ± s.d. (standard deviation) of 8 primary tumors. (B) Final primary tumor weights of HMLER-shCntrl, shEcad, and DN-Ecad tumors grown subcutaneously. Each bar represents the mean ± s.d. of 8 primary tumors. (C) Quantification of total lung metastasis burden in mice bearing either orthotopic (implanted in mammary gland) primary tumors of HMLER-shCntrl, shEcad, and DNEcad cells or 8-weeks after tail-vein injection of these lines. Each bar represents the mean ± s.d. of 5 mice analyzed per group (* p<0.001). (D) Representative fluorescence images of mouse lung lobes bearing either orthotopic primary tumors of HMLER-shCntrl, shEcad, and DN-Ecad cells or 8-weeks after tailvein injection of these lines. GFP signal denotes the presence of tumor cells (Left Panels). Arrows point to occasional tumor cells detected in the lungs of control and DN-Ecad tumor bearing animals. Representative immunohistochemical staining of sections with anti-Large T antibody from the same set of lungs. Brown nuclear staining denotes the tumor cells. N, lung tissue; M, metastatic nodule (Right Panels). Figure 6. Maintenance of E-cadherin knock-down and DN-Ecad expression in primary tumors and tumor-derived cell lines A shEcad shCntrl .E Qa EE E E H-H DN-Ecad 0-- EE O.H E-cadherin Tubulin DN-Ecad - shCntrl shEcad Large T E-cadherin Figure 6. Maintenance of E-cadherin knock-down and DN-Ecad expression in primary tumors and tumor-derived cell lines. (A) Expression levels of E-cadherin and dominant-negative mutant E-cadherin (Arrowhead) in parental and tumor derived HMLER-shCntrl, shEcad, and DN-Ecad cells. Tubulin is used as loading controls. (B)Representative serial tumor sections stained for E-cadherin and Large T antigen to mark the tumor cells (T). Arrowhead in lower left panel points to membranous Ecadherin staining in shCntrl tumors whereas no such staining is observed in shEcad tumors. Note that the staining in the mouse skin adjacent to the shEcad tumor serves as a positive control. Figure 7.Loss of E-cadherin leads to increased invasiveness and resistance to anoikis A B shCntrl Motilitv AInvasion DN-Ecad shEcad I t I i C r r, LIATM CD UA) U, w CO E 0i ----- StIW -·-- LJt1*Cad M -- · I i `- -ii 5Gc O E g r WJ~ j C) bP OOM I N&Sd OEM ~ I f Vnnexi V - Sh V CN& Figure 7. Loss of E-cadherin leads to increased invasiveness and resistance to anoikis. (A) HMLER-shCntrl, shEcad, and DN-Ecad cells were induced to move or invade through uncoated or Matrigel-coated transwell membranes. After 12 (uncoated) or 16 (Matrigel-coated) hours the migrated cells were fixed, stained, photographed. Representative photographs of Transwell membranes showing stained migrated cells from either motility or invasion experiments. (B) Quantification of motility and invasion experiments (A) performed by counting the number of migrated cells. Assays were done in triplicate, and the mean ± s.d. are shown (* p<0.01 compared to shCntrl). (C) HMLER-shCntrl, shEcad, and DN-Ecad cells were put in suspension and cultured on ultra-low attachment plates in triplicate. After the indicated times, viable cells were counted using trypan blue. Each bar represents the mean + s.d of the remaining viable cells expressed as a percentage of the initial seeding. (D) Representative FACS histograms indicating the percentage of apoptotic HMLERshCntrl, shEcad, and DN-Ecad cells in suspension as determined by binding of FITCconjugated Annexin-V. The FACS analyses were done 24 hours after suspension culture. Each bar on the right graph represents the mean + s.d of the Annexin V positive cells expressed as a percentage of total cells (*p<0.001). Figure 8. HMLER-shEcad cells have cancer stem cell-like properties Tumorigenicity- HMLER shCntrl shEcad 1x10 6 8/10 10110 1x1 05 3/4 4/4 1x10 4 1/4 4/4 1x103 0/4 4/4 B II QlEcid, ... ..... # of amrnospheres/ 100 cells shEcad shCntrl i~~k I )O ~ ~ ·- ·1~ CD24 Figure 8. HMLER-shEcad cells have cancer stem cell-like properties (A) Subcutaneous injections of serial dilutions of HMLER cells expressing either control shRNA (shCntrl) or shRNA targeting E-cadherin (shEcad). The proportion of injection sites that developed tumors within 60 days is shown. (B) Number of mammospheres formed from an initial seeding of 100 cells per well. On the right are representative photographs of wells showing increased numbers of mammospheres in HMLER-shEcad seeded wells. (C) Antigenic profile of HMLER-shCntrl and HMLER-shEcad cells as determined by FACS analysis with APC-conjugated CD44 and PE-conjugated CD24 antibodies. Note that CD44h" /CD24Low population is reported to enrich for breast CSCs (Al-Hajj et al., 2003). Figure 9. p-catenin is activated upon E-cadherin loss and is necessary for E-cadherinloss-induced EMT A B shEcad $.catenin + Dapi #-catenin U DMSO GSKi E-cadherin shCntrl p--0 p- at . r:E}}2; . ?) : ,catenin N-cadherin I p-GSK3-, Fibronectin GSK3-, shEcad Vimentin CK-8 Actin - r- 0-.9 aC 0 1 Days 2 3 Ssnostiksa stican Anoikis Invasion cc - shEcad -0 n, LU m 1-catenin E-cadherin shEcad + N-cad herin - shBcat Vimentin Actin ... O Figure 9. P-catenin is activated upon E-cadherin loss and is necessary for Ecadherin induced EMT (A) Immunofluorescence staining for p-catenin (Green) in shCntrl and shEcad cells showing differential localization. Right panels show the nuclear DAPI (blue) staining of the same cells. White arrows indicate the nuclear p-catenin staining observed in the shEcad cells. Immunoblots showing phospho-P-catenin, total 3-catenin, phosho-GSK3p and total GSK levels in either untreated shCntrl and shEcad cells or in the same set of cells treated with DMSO or GSK inhibitor BIO (1pM) for 6 hours. (B) Expression levels of E-cadherin, 3-catenin, E-cadherin, N-cadherin and vimentin in HMLER-shCntrl, shEcad, and double-knock down cells (shEcad+shp-cat) cells examined by immunoblotting. 3-actin is used as a loading control. (C) In vitro growth rate of HMLER shEcad and shEcad + shBcat cell lines as determined by Cell-titer-glo assay. (D) Left graph: HMLER-shCntrl, shEcad, and shEcad + shp-cat cells were induced to invade through Matrigel-coated transwell membranes. After 16 hours, the invaded cells were fixed, stained, photographed and counted. Assays were done in triplicate, and the mean ± s.d. are shown relative to shCntrl cell invasion (*p<0.01 compared to shEcad). Right graph: The percentage of apoptotic HMLER-shCntrl, shEcad, and shEcad + shp-cat cells in suspension were determined using binding of FITC-conjugated Annexin-V. The FACS analyses were done 24 hours after suspension culture. Each bar represents the mean ± s.d of the Annexin V- positive cells expressed as a percentage of total cells (*p<0.01 compared to shEcad). (E) Representative fluorescence images of mouse lung lobes 8-weeks after tail-vein injection of shEcad or shEcad + shp-cat cells. GFP signal denotes the presence of tumor cells (Left Panels). Quantification of total lung metastasis burden in the same sets of mice. Each bar represents the mean ± s.d. of 7 mice analyzed (Right Panels) (*p<0.001). (F) Overexpression of active B-catenin is not sufficient to cause the EMT.Expression levels of P-catenin, E-cadherin, N-cadherin and vimentin in HMLER cells expressing either empty vector (pbp) or truncated p-catenin cDNA (pbp AN-pcat, indicated by arrowhead). HMLER-shEcad cell lysate was included to serve as a positive control for Ncadherin and vimentin. p-actin was used as a loading control. Figure 10. Gene expression profiling identifies molecular changes induced upon Ecadherin loss mill m M -" u (I U) LU LO iii 0 M Z u, Figure 11. Twist is upregulated upon E-cadherin loss and plays an essential role in Ecadherin-loss-induced metastasis B A shEcad - - C L· z EQ '5 5, ZJ• . Twist 0 - C_ (V EU 0.1 GUL0 > N-cadherin -- Vimentin 0r Actin C C 0 v_ "_ Acin ~C I? r go" VECO 04EM~a Invasion D R ,; + r, "t ;ap " Anoikis 30 shEcad 20) -1 ' shEcad + shTwist Zl 0 1 A 9).1 •T0)JJ Figure 11. Twist is upregulated upon loss of E-cadherin and plays functionally important role in E-cadherin loss-induced EMT and metastasis (A) The relative levels of Twist mRNA measured by real-time RT-PCR in the HMLER shEcad and DN-Ecad cells compared to shCntrl cells. Each bar represents the mean + SEM of the PCRs in triplicate. (B) Expression levels of Twist, vimentin and N-cadherin in HMLER-shCntrl, shEcad, and double-knock down cells (shEcad + shTwist). 13-actin is used as a loading control. (C) Right graph: HMLER-shCntrl, shEcad, and shEcad + shTwist cells were induced to invade through Matrigel-coated transwell membranes. After 16 hours, the invaded cells were fixed, stained, photographed and counted. Assays were done in triplicate, and the mean ± s.d. are shown relative to shCntrl cell invasion (*p<0.01 compared to shEcad). Left graph: The percentage of apoptotic HMLER-shCntrl, shEcad, and shEcad + shTwist cells in suspension were determined using binding of FITC-conjugated Annexin-V. The FACS analyses were done 24 hours after suspension culture. Each bar represents the mean ± s.d of the Annexin V- positive cells expressed as a percentage of total cells (*p<0.01 compared to shEcad). (D) Representative fluorescence images of mouse lung lobes 8-weeks after tail-vein injection of shEcad or shEcad + shTwist cells. GFP signal denotes the presence of tumor cells (Left Panels). Quantification of total lung metastasis burden in the same sets of mice is shown on the right panels. Each bar represents the mean ± s.d. of 5 mice analyzed (*p<0.0 1). Figure 10. Gene expression profiling identifies molecular changes induced upon Ecadherin loss (A) Hierarchical clustering of shEcad, DN-Ecad, shEcad+shBcat and shCntrl cells. Distances between samples were computed using the Euclidean metric applied to a 225probe subset corresponding to genes highly induced or repressed upon EMT (71 genes induced, 84 genes repressed). (B) Differentially expressed genes in shEcad and DN-Ecad cells relative to control shGFP cells. The numbers of genes either induced or repressed that surpass the indicated threshold of differential expression (1.5x, 2x, 2.5x, 3x log 2-fold change) are depicted. (C) Gene ontology analysis of differentially induced or repressed genes in shEcad and DN-Ecad cells with respect to enrichment of genes with assignments to specific biological processes. Fold enrichment and the number of genes in a particular biological process are shown. Note that no significant enrichment of genes for any biological process was observed in DN-Ecad induced genes (indicated by *). (D) Dependence on 3-catenin of gene expression changes in shEcad cells. Depicted are representative genes, either induced or repressed in shEcad cells, whose regulation is dependent on (6 genes on the left) or independent of 3-catenin activity (6 genes on the right). Each dot represents the value for an individual replicate expression value (colors correspond to samples) for the gene listed on the x-axis. The dotted line indicates the 1.5 log2 fold-change threshold. (E) Induction of multiple transcription regulators in shEcad cells. Each dot represents the value for an individual replicate expression value (colors correspond to samples) for the gene listed on the x-axis. The dotted line indicates the 1.5 log2 fold-change threshold. Note that TCF8 (Zeb 1) and Twist are among the highly upregualted genes in this category. Figure 11. Twist is upregulated upon E-cadherin loss and plays an essential role in Ecadherin-loss-induced metastasis A B C shEcad -- Y z 6, ft W EuWi Twist N-cadherin n3*16 >bin 61. Vimentin *1 G --- C C , ... f .. . 4: "_, Actin St I Ata, CNon d Invasion D shEcad Anoikis 255 C .3 'S shEcad + shTwist -4 J :1 o Figure 11. Twist is upregulated upon loss of E-cadherin and plays functionally important role in E-cadherin loss-induced EMT and metastasis (A) The relative levels of Twist mRNA measured by real-time RT-PCR in the HMLER shEcad and DN-Ecad cells compared to shCntrl cells. Each bar represents the mean + SEM of the PCRs in triplicate. (B) Expression levels of Twist, vimentin and N-cadherin in HMLER-shCntrl, shEcad, and double-knock down cells (shEcad + shTwist). p-actin is used as a loading control. (C) Right graph: HMLER-shCntrl, shEcad, and shEcad + shTwist cells were induced to invade through Matrigel-coated transwell membranes. After 16 hours, the invaded cells were fixed, stained, photographed and counted. Assays were done in triplicate, and the mean ± s.d. are shown relative to shCntrl cell invasion (*p<0.01 compared to shEcad). Left graph: The percentage of apoptotic HMLER-shCntrl, shEcad, and shEcad + shTwist cells in suspension were determined using binding of FITC-conjugated Annexin-V. The FACS analyses were done 24 hours after suspension culture. Each bar represents the mean ± s.d of the Annexin V- positive cells expressed as a percentage of total cells (*p<0.01 compared to shEcad). (D) Representative fluorescence images of mouse lung lobes 8-weeks after tail-vein injection of shEcad or shEcad + shTwist cells. GFP signal denotes the presence of tumor cells (Left Panels). Quantification of total lung metastasis burden in the same sets of mice is shown on the right panels. Each bar represents the mean + s.d. of 5 mice analyzed (*p<0.01). Acknowledgements I would like to thank Priyamvada Rai, Tugba Bagci, Richard O. Hynes, David M. Sabatini, and members of R.A.W.'s laboratory for helpful comments and discussions throughout the course of these experiments. I also thank Drs. Gerhard Christofori and Kimberly Hartwell for reagents, Ahmet Acar and Ferenc Reinhart for technical help, and Casey Gates and the gene expression platform at the Broad Institute for data collection and helpful advice. Ludwig Center for Molecular Oncology at MIT and the W.M. Keck Foundation Biological imaging facility at the Whitehead Institute is also acknowledged. References Aigner, K., Dampier, B., Descovich, L., Mikula, M., Sultan, A., Schreiber, M., Mikulits, W., Brabletz, T., Strand, D., Obrist, P., Sommergruber, W., Schweifer, N., Wernitznig, A., Beug, H., Foisner, R. and Eger, A. (2007a) The transcription factor ZEB1 (deltaEF1) promotes tumour cell dedifferentiation by repressing master regulators of epithelial polarity. Oncogene. Aigner, K., Dampier, B., Descovich, L., Mikula, M., Sultan, A., Schreiber, M., Mikulits, W., Brabletz, T., Strand, D., Obrist, P., Sommergruber, W., Schweifer, N., Wernitznig, A., Beug, H., Foisner, R. and Eger, A. (2007b) The transcription factor ZEB 1 (deltaEFl) promotes tumour cell dedifferentiation by repressing master regulators of epithelial polarity. Oncogene, 26, 6979-6988. Al-Hajj, M., Wicha, M.S., Benito-Hernandez, A., Morrison, S.J. and Clarke, M.F. (2003) Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci US A, 100, 3983-3988. Alonso, S.R., Tracey, L., Ortiz, P., Perez-Gomez, B., Palacios, J., Pollan, M., Linares, J., Serrano, S., Saez-Castillo, A.I., Sanchez, L., Pajares, R., Sanchez-Aguilera, A., Artiga, M.J., Piris, M.A. and Rodriguez-Peralto, J.L. (2007) A high-throughput study in melanoma identifies epithelial-mesenchymal transition as a major determinant of metastasis. CancerRes, 67, 3450-3460. Arslan, P., Montecucco, C., Celi, D. and Pozzan, T. (1981) Effect of monovalent cation ionophores on lymphocyte cellular metabolism. Biochim Biophys Acta, 643, 177181. Attwell, S., Roskelley, C. and Dedhar, S. (2000) The integrin-linked kinase (ILK) suppresses anoikis. Oncogene, 19, 3811-3815. Avizienyte, E., Wyke, A.W., Jones, R.J., McLean, G.W., Westhoff, M.A., Brunton, V.G. and Frame, M.C. (2002) Src-induced de-regulation of E-cadherin in colon cancer cells requires integrin signalling. Nat Cell Biol, 4, 632-638. Barrallo-Gimeno, A. and Nieto, M.A. (2005) The Snail genes as inducers of cell movement and survival: implications in development and cancer. Development, 132, 3151-3161. Batlle, E., Henderson, J.T., Beghtel, H., van den Born, M.M., Sancho, E., Huls, G., Meeldijk, J., Robertson, J., van de Wetering, M., Pawson, T. and Clevers, H. (2002) Beta-catenin and TCF mediate cell positioning in the intestinal epithelium by controlling the expression of EphB/ephrinB. Cell, 111, 251-263. Batlle, E., Sancho, E., Franci, C., Dominguez, D., Monfar, M., Baulida, J. and Garcia De Herreros, A. (2000) The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nat Cell Biol, 2, 84-89. Becker, K.F., Atkinson, M.J., Reich, U., Becker, I., Nekarda, H., Siewert, J.R. and Hofler, H. (1994) E-cadherin gene mutations provide clues to diffuse type gastric carcinomas. CancerRes, 54, 3845-3852. Behrens, J., Birchmeier, W., Goodman, S.L. and Imhof, B.A. (1985) Dissociation of Madin-Darby canine kidney epithelial cells by the monoclonal antibody anti-arc1: mechanistic aspects and identification of the antigen as a component related to uvomorulin. J Cell Biol, 101, 1307-1315. Berezovskaya, O., Schimmer, A.D., Glinskii, A.B., Pinilla, C., Hoffman, R.M., Reed, J.C. and Glinsky, G.V. (2005) Increased expression of apoptosis inhibitor protein XIAP contributes to anoikis resistance of circulating human prostate cancer metastasis precursor cells. CancerRes, 65, 2378-2386. Berx, G., Becker, K.F., Hofler, H. and van Roy, F. (1998) Mutations of the human Ecadherin (CDH1) gene. Hum Mutat, 12, 226-237. Berx, G., Cleton-Jansen, A.M., Nollet, F., de Leeuw, W.J., van de Vijver, M., Cornelisse, C. and van Roy, F. (1995) E-cadherin is a tumour/invasion suppressor gene mutated in human lobular breast cancers. Embo J, 14, 6107-6115. Berx, G., Cleton-Jansen, A.M., Strumane, K., de Leeuw, W.J., Nollet, F., van Roy, F. and Cornelisse, C. (1996) E-cadherin is inactivated in a majority of invasive human lobular breast cancers by truncation mutations throughout its extracellular domain. Oncogene, 13, 1919-1925. Berx, G. and Van Roy, F. (2001) The E-cadherin/catenin complex: an important gatekeeper in breast cancer tumorigenesis and malignant progression. Breast Cancer Res, 3, 289-293. Bhowmick, N.A., Ghiassi, M., Bakin, A., Aakre, M., Lundquist, C.A., Engel, M.E., Arteaga, C.L. and Moses, H.L. (2001) Transforming growth factor-betal mediates epithelial to mesenchymal transdifferentiation through a RhoA-dependent mechanism. Mol Biol Cell, 12, 27-36. Blanco, M.J., Moreno-Bueno, G., Sarrio, D., Locascio, A., Cano, A., Palacios, J. and Nieto, M.A. (2002) Correlation of Snail expression with histological grade and lymph node status in breast carcinomas. Oncogene, 21, 3241-3246. Blaschuk, O.W., Sullivan, R., David, S. and Pouliot, Y. (1990) Identification of a cadherin cell adhesion recognition sequence. Dev Biol, 139, 227-229. Bolos, V., Peinado, H., Perez-Moreno, M.A., Fraga, M.F., Esteller, M. and Cano, A. (2003) The transcription factor Slug represses E-cadherin expression and induces epithelial to mesenchymal transitions: a comparison with Snail and E47 repressors. J Cell Sci, 116, 499-511. Boussadia, O., Kutsch, S., Hierholzer, A., Delmas, V. and Kemler, R. (2002) E-cadherin is a survival factor for the lactating mouse mammary gland. Mech Dev, 115, 5362. Boyer, B., Tucker, G.C., Valles, A.M., Franke, W.W. and Thiery, J.P. (1989a) Rearrangements of desmosomal and cytoskeletal proteins during the transition from epithelial to fibroblastoid organization in cultured rat bladder carcinoma cells. J Cell Biol, 109, 1495-1509. Boyer, B., Tucker, G.C., Valles, A.M., Gavrilovic, J. and Thiery, J.P. (1989b) Reversible transition towards a fibroblastic phenotype in a rat carcinoma cell line. Int J Cancer Suppl, 4, 69-75. Brabletz, T., Jung, A., Reu, S., Porzner, M., Hlubek, F., Kunz-Schughart, L.A., Knuechel, R. and Kirchner, T. (2001) Variable beta-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc Natl Acad Sci USA, 98, 10356-10361. Braun, S., Pantel, K., Muller, P., Janni, W., Hepp, F., Kentenich, C.R., Gastroph, S., Wischnik, A., Dimpfl, T., Kindermann, G., Riethmuller, G. and Schlimok, G. (2000) Cytokeratin-positive cells in the bone marrow and survival of patients with stage I, II, or III breast cancer. N Engl J Med, 342, 525-533. Brembeck, F.H., Schwarz-Romond, T., Bakkers, J., Wilhelm, S., Hammerschmidt, M. and Birchmeier, W. (2004) Essential role of BCL9-2 in the switch between betacatenin's adhesive and transcriptional functions. Genes Dev, 18, 2225-2230. Bryant, D.M. and Stow, J.L. (2004) The ins and outs of E-cadherin trafficking. Trends Cell Biol, 14, 427-434. Cano, A., Perez-Moreno, M.A., Rodrigo, I., Locascio, A., Blanco, M.J., del Barrio, M.G., Portillo, F. and Nieto, M.A. (2000) The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nat Cell Biol, 2, 76-83. Cavallaro, U. and Christofori, G. (2004) Cell adhesion and signalling by cadherins and Ig-CAMs in cancer. Nat Rev Cancer, 4, 118-132. Chabner, B.A. and Roberts, T.G., Jr. (2005) Timeline: Chemotherapy and the war on cancer. Nat Rev Cancer,5, 65-72. Chambers, A.F., Groom, A.C. and MacDonald, I.C. (2002) Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer,2, 563-572. Chen, Y.T., Stewart, D.B. and Nelson, W.J. (1999) Coupling assembly of the Ecadherin/beta-catenin complex to efficient endoplasmic reticulum exit and basallateral membrane targeting of E-cadherin in polarized MDCK cells. J Cell Biol, 144, 687-699. Cheng, G.Z., Chan, J., Wang, Q., Zhang, W., Sun, C.D. and Wang, L.H. (2007) Twist transcriptionally up-regulates AKT2 in breast cancer cells leading to increased migration, invasion, and resistance to paclitaxel. CancerRes, 67, 1979-1987. Cheng, G.Z., Zhang, W., Sun, M., Wang, Q., Coppola, D., Mansour, M., Xu, L., Costanzo, C., Cheng, J.Q. and Wang, L.H. (2008) Twist is transcriptionally induced by activation of STAT3 and mediates STAT3 oncogenic function. J Biol Chem. Chung, C.H., Parker, J.S., Ely, K., Carter, J., Yi, Y., Murphy, B.A., Ang, K.K., ElNaggar, A.K., Zanation, A.M., Cmelak, A.J., Levy, S., Slebos, R.J. and Yarbrough, W.G. (2006) Gene expression profiles identify epithelial-tomesenchymal transition and activation of nuclear factor-kappaB signaling as characteristics of a high-risk head and neck squamous cell carcinoma. CancerRes, 66, 8210-8218. Clare, S.E., Nakhlis, F. and Panetta, J.C. (2000) Molecular biology of breast cancer metastasis. The use of mathematical models to determine relapse and to predict response to chemotherapy in breast cancer. Breast CancerRes, 2, 430-435. Clark, E.A., Golub, T.R., Lander, E.S. and Hynes, R.O. (2000) Genomic analysis of metastasis reveals an essential role for RhoC. Nature, 406, 532-535. Clevers, H. (2006) Wnt/beta-catenin signaling in development and disease. Cell, 127, 469-480. Comijn, J., Berx, G., Vermassen, P., Verschueren, K., van Grunsven, L., Bruyneel, E., Mareel, M., Huylebroeck, D. and van Roy, F. (2001) The two-handed E box binding zinc finger protein SIP1 downregulates E-cadherin and induces invasion. Mol Cell, 7, 1267-1278. Conacci-Sorrell, M., Simcha, I., Ben-Yedidia, T., Blechman, J., Savagner, P. and BenZe'ev, A. (2003) Autoregulation of E-cadherin expression by cadherin-cadherin interactions: the roles of beta-catenin signaling, Slug, and MAPK. J Cell Biol, 163, 847-857. Cross, D.A., Alessi, D.R., Cohen, P., Andjelkovich, M. and Hemmings, B.A. (1995) Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature, 378, 785-789. Dahl, U., Sjodin, A. and Semb, H. (1996) Cadherins regulate aggregation of pancreatic beta-cells in vivo. Development, 122, 2895-2902. Daniele, R.P., Holian, S.K. and Nowell, P.C. (1978) A potassium ionophore (Nigericin) inhibits stimulation of human lymphocytes by mitogens. J Exp Med, 147, 571-581. De Langhe, S.P., Sala, F.G., Del Moral, P.M., Fairbanks, T.J., Yamada, K.M., Warburton, D., Bums, R.C. and Bellusci, S. (2005) Dickkopf-1 (DKK1) reveals that fibronectin is a major target of Wnt signaling in branching morphogenesis of the mouse embryonic lung. Dev Biol, 277, 316-331. Dean, M., Fojo, T. and Bates, S. (2005) Tumour stem cells and drug resistance. Nat Rev Cancer, 5, 275-284. Dennis, G., Jr., Sherman, B.T., Hosack, D.A., Yang, J., Gao, W., Lane, H.C. and Lempicki, R.A. (2003) DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol, 4, P3. Derksen, P.W., Liu, X., Saridin, F., van der Gulden, H., Zevenhoven, J., Evers, B., van Beijnum, J.R., Griffioen, A.W., Vink, J., Krimpenfort, P., Peterse, J.L., Cardiff, R.D., Berns, A. and Jonkers, J. (2006) Somatic inactivation of E-cadherin and p53 in mice leads to metastatic lobular mammary carcinoma through induction of anoikis resistance and angiogenesis. Cancer Cell, 10, 437-449. Diehn, M. and Clarke, M.F. (2006) Cancer stem cells and radiotherapy: new insights into tumor radioresistance. J Natl CancerInst, 98, 1755-1757. Dontu, G., Abdallah, W.M., Foley, J.M., Jackson, K.W., Clarke, M.F., Kawamura, M.J. and Wicha, M.S. (2003) In vitro propagation and transcriptional profiling of human mammary stem/progenitor cells. Genes Dev, 17, 1253-1270. Douma, S., Van Laar, T., Zevenhoven, J., Meuwissen, R., Van Garderen, E. and Peeper, D.S. (2004) Suppression of anoikis and induction of metastasis by the neurotrophic receptor TrkB. Nature, 430, 1034-1039. Egeblad, M. and Werb, Z. (2002) New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer, 2, 161-174. Eger, A., Aigner, K., Sonderegger, S., Dampier, B., Oehler, S., Schreiber, M., Berx, G., Cano, A., Beug, H. and Foisner, R. (2005) DeltaEF1 is a transcriptional repressor of E-cadherin and regulates epithelial plasticity in breast cancer cells. Oncogene, 24, 2375-2385. Eger, A., Stockinger, A., Park, J., Langkopf, E., Mikula, M., Gotzmann, J., Mikulits, W., Beug, H. and Foisner, R. (2004) beta-Catenin and TGFbeta signalling cooperate to maintain a mesenchymal phenotype after FosER-induced epithelial to mesenchymal transition. Oncogene, 23, 2672-2680. Elenbaas, B., Spirio, L., Koerner, F., Fleming, M.D., Zimonjic, D.B., Donaher, J.L., Popescu, N.C., Hahn, W.C. and Weinberg, R.A. (2001) Human breast cancer cells generated by oncogenic transformation of primary mammary epithelial cells. Genes Dev, 15, 50-65. Elloul, S., Elstrand, M.B., Nesland, J.M., Trope, C.G., Kvalheim, G., Goldberg, I., Reich, R. and Davidson, B. (2005) Snail, Slug, and Smad-interacting protein 1 as novel parameters of disease aggressiveness in metastatic ovarian and breast carcinoma. Cancer, 103, 1631-1643. Fidler, I.J. and Kripke, M.L. (1977) Metastasis results from preexisting variant cells within a malignant tumor. Science, 197, 893-895. Finn, R.S., Dering, J., Ginther, C., Wilson, C.A., Glaspy, P., Tchekmedyian, N. and Slamon, D.J. (2007) Dasatinib, an orally active small molecule inhibitor of both the src and abl kinases, selectively inhibits growth of basal-type/"triple-negative" breast cancer cell lines growing in vitro. Breast CancerRes Treat, 105, 319-326. Frank, N.Y., Margaryan, A., Huang, Y., Schatton, T., Waaga-Gasser, A.M., Gasser, M., Sayegh, M.H., Sadee, W. and Frank, M.H. (2005) ABCB5-mediated doxorubicin transport and chemoresistance in human malignant melanoma. CancerRes, 65, 4320-4333. Friedl, P. and Wolf, K. (2003) Tumour-cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer, 3, 362-374. Frixen, U.H., Behrens, J., Sachs, M., Eberle, G., Voss, B., Warda, A., Lochner, D. and Birchmeier, W. (1991) E-cadherin-mediated cell-cell adhesion prevents invasiveness of human carcinoma cells. J Cell Biol, 113, 173-185. Fujita, Y., Krause, G., Scheffner, M., Zechner, D., Leddy, H.E., Behrens, J., Sommer, T. and Birchmeier, W. (2002) Hakai, a c-Cbl-like protein, ubiquitinates and induces endocytosis of the E-cadherin complex. Nat Cell Biol, 4, 222-231. Furlong, E.E., Andersen, E.C., Null, B., White, K.P. and Scott, M.P. (2001) Patterns of gene expression during Drosophila mesoderm development. Science, 293, 16291633. Furlong, I.J., Ascaso, R., Lopez Rivas, A. and Collins, M.K. (1997) Intracellular acidification induces apoptosis by stimulating ICE-like protease activity. J Cell Sci, 110 ( Pt 5), 653-661. Furlong, I.J., Lopez Mediavilla, C., Ascaso, R., Lopez Rivas, A. and Collins, M.K. (1998) Induction of apoptosis by valinomycin: mitochondrial permeability transition causes intracellular acidification. Cell Death Differ, 5, 214-221. Giannakakou, P., Sackett, D.L., Kang, Y.K., Zhan, Z., Buters, J.T., Fojo, T. and Poruchynsky, M.S. (1997) Paclitaxel-resistant human ovarian cancer cells have mutant beta-tubulins that exhibit impaired paclitaxel-driven polymerization. J Biol Chem, 272, 17118-17125. Gottardi, C.J. and Gumbiner, B.M. (2004) Distinct molecular forms of beta-catenin are targeted to adhesive or transcriptional complexes. J Cell Biol, 167, 339-349. Gottardi, C.J., Wong, E. and Gumbiner, B.M. (2001) E-cadherin suppresses cellular transformation by inhibiting beta-catenin signaling in an adhesion-independent manner. J Cell Biol, 153, 1049-1060. Gradl, D., Kuhl, M. and Wedlich, D. (1999) The Wnt/Wg signal transducer beta-catenin controls fibronectin expression. Mol Cell Biol, 19, 5576-5587. Graff, J.R., Herman, J.G., Lapidus, R.G., Chopra, H., Xu, R., Jarrard, D.F., Isaacs, W.B., Pitha, P.M., Davidson, N.E. and Baylin, S.B. (1995) E-cadherin expression is silenced by DNA hypermethylation in human breast and prostate carcinomas. Cancer Res, 55, 5195-5199. Grooteclaes, M.L. and Frisch, S.M. (2000) Evidence for a function of CtBP in epithelial gene regulation and anoikis. Oncogene, 19, 3823-3828. Gros, P., Ben Neriah, Y.B., Croop, J.M. and Housman, D.E. (1986) Isolation and expression of a complementary DNA that confers multidrug resistance. Nature, 323, 728-731. Grunert, S., Jechlinger, M. and Beug, H. (2003) Diverse cellular and molecular mechanisms contribute to epithelial plasticity and metastasis. Nat Rev Mol Cell Biol, 4, 657-665. Guilford, P., Hopkins, J., Harraway, J., McLeod, M., McLeod, N., Harawira, P., Taite, H., Scoular, R., Miller, A. and Reeve, A.E. (1998) E-cadherin germline mutations in familial gastric cancer. Nature, 392, 402-405. Gumbiner, B., Stevenson, B. and Grimaldi, A. (1988) The role of the cell adhesion molecule uvomorulin in the formation and maintenance of the epithelial junctional complex. J Cell Biol, 107, 1575-1587. Gumireddy, K., Sun, F., Klein-Szanto, A.J., Gibbins, J.M., Gimotty, P.A., Saunders, A.J., Schultz, P.G. and Huang, Q.(2007) In vivo selection for metastasis promoting genes in the mouse. ProcNatl Acad Sci US A, 104, 6696-6701. Gupta, G.P. and Massague, J. (2006) Cancer metastasis: building a framework. Cell, 127, 679-695. Hadeball, B., Borchers, A. and Wedlich, D. (1998) Xenopus cadherin- 11 (Xcadherin-11) expression requires the Wg/Wnt signal. Mech Dev, 72, 101-113. Halbleib, J.M. and Nelson, W.J. (2006) Cadherins in development: cell adhesion, sorting, and tissue morphogenesis. Genes Dev, 20, 3199-3214. Hanahan, D. and Weinberg, R.A. (2000) The hallmarks of cancer. Cell, 100, 57-70. Handschuh, G., Candidus, S., Luber, B., Reich, U., Schott, C., Oswald, S., Becke, H., Hutzler, P., Birchmeier, W., Hofler, H. and Becker, K.F. (1999) Tumourassociated E-cadherin mutations alter cellular morphology, decrease cellular adhesion and increase cellular motility. Oncogene, 18, 4301-4312. Hartwell, K.A., Muir, B., Reinhardt, F., Carpenter, A.E., Sgroi, D.C. and Weinberg, R.A. (2006) The Spemann organizer gene, Goosecoid, promotes tumor metastasis. ProcNatl Acad Sci U S A, 103, 18969-18974. Hirohashi, S. (1998) Inactivation of the E-cadherin-mediated cell adhesion system in human cancers. Am J Pathol,153, 333-339. Howe, L.R., Watanabe, O., Leonard, J. and Brown, A.M. (2003) Twist is up-regulated in response to Wntl and inhibits mouse mammary cell differentiation. CancerRes, 63, 1906-1913. Huber, A.H. and Weis, W.I. (2001) The structure of the beta-catenin/E-cadherin complex and the molecular basis of diverse ligand recognition by beta-catenin. Cell, 105, 391-402. Huber, M.A., Kraut, N. and Beug, H. (2005) Molecular requirements for epithelialmesenchymal transition during tumor progression. Curr Opin Cell Biol, 17, 548558. Hyafil, F., Babinet, C. and Jacob, F. (1981) Cell-cell interactions in early embryogenesis: a molecular approach to the role of calcium. Cell, 26, 447-454. Ino, Y., Gotoh, M., Sakamoto, M., Tsukagoshi, K. and Hirohashi, S. (2002) Dysadherin, a cancer-associated cell membrane glycoprotein, down-regulates E-cadherin and promotes metastasis. ProcNatl Acad Sci U S A, 99, 365-370. Inoue, A., Seidel, M.G., Wu, W., Kamizono, S., Ferrando, A.A., Bronson, R.T., Iwasaki, H., Akashi, K., Morimoto, A., Hitzler, J.K., Pestina, T.I., Jackson, C.W., Tanaka, R., Chong, M.J., McKinnon, P.J., Inukai, T., Grosveld, G.C. and Look, A.T. (2002) Slug, a highly conserved zinc finger transcriptional repressor, protects hematopoietic progenitor cells from radiation-induced apoptosis in vivo. Cancer Cell, 2, 279-288. Inukai, T., Inoue, A., Kurosawa, H., Goi, K., Shinjyo, T., Ozawa, K., Mao, M., Inaba, T. and Look, A.T. (1999) SLUG, a ces-1-related zinc finger transcription factor gene with antiapoptotic activity, is a downstream target of the E2A-HLF oncoprotein. Mol Cell, 4, 343-352. Jechlinger, M., Grunert, S., Tamir, I.H., Janda, E., Ludemann, S., Waerner, T., Seither, P., Weith, A., Beug, H. and Kraut, N. (2003) Expression profiling of epithelial plasticity in tumor progression. Oncogene, 22, 7155-7169. Jemal, A., Siegel, R., Ward, E., Hao, Y., Xu, J., Murray, T. and Thun, M.J. (2008) Cancer statistics, 2008. CA CancerJ Clin, 58, 71-96. Jiang, J., Kosman, D., Ip, Y.T. and Levine, M. (1991) The dorsal morphogen gradient regulates the mesoderm determinant twist in early Drosophila embryos. Genes Dev, 5, 1881-1891. Jorda, M., Olmeda, D., Vinyals, A., Valero, E., Cubillo, E., Llorens, A., Cano, A. and Fabra, A. (2005) Upregulation of MMP-9 in MDCK epithelial cell line in response to expression of the Snail transcription factor. J Cell Sci, 118, 3371-3385. Julien, S., Puig, I., Caretti, E., Bonaventure, J., Nelles, L., van Roy, F., Dargemont, C., de Herreros, A.G., Bellacosa, A. and Larue, L. (2007) Activation of NF-kappaB by Akt upregulates Snail expression and induces epithelium mesenchyme transition. Oncogene, 26, 7445-7456. Kajita, M., McClinic, K.N. and Wade, P.A. (2004) Aberrant expression of the transcription factors snail and slug alters the response to genotoxic stress. Mol Cell Biol, 24, 7559-7566. Kajiyama, H., Shibata, K., Terauchi, M., Yamashita, M., Ino, K., Nawa, A. and Kikkawa, F. (2007) Chemoresistance to paclitaxel induces epithelial-mesenchymal transition and enhances metastatic potential for epithelial ovarian carcinoma cells. Int J Oncol, 31, 277-283. Kamei, T., Matozaki, T., Sakisaka, T., Kodama, A., Yokoyama, S., Peng, Y.F., Nakano, K., Takaishi, K. and Takai, Y. (1999) Coendocytosis of cadherin and c-Met coupled to disruption of cell-cell adhesion in MDCK cells--regulation by Rho, Rac and Rab small G proteins. Oncogene, 18, 6776-6784. Kanai, Y., Oda, T., Tsuda, H., Ochiai, A. and Hirohashi, S. (1994) Point mutation of the E-cadherin gene in invasive lobular carcinoma of the breast. Jpn J CancerRes, 85, 1035-1039. Kim, C.F., Jackson, E.L., Woolfenden, A.E., Lawrence, S., Babar, I., Vogel, S., Crowley, D., Bronson, R.T. and Jacks, T. (2005) Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell, 121, 823-835. Kim, K., Lu, Z. and Hay, E.D. (2002) Direct evidence for a role of beta-catenin/LEF-1 signaling pathway in induction of EMT. Cell Biol Int, 26, 463-476. Klapholz-Brown, Z., Walmsley, G.G., Nusse, Y.M., Nusse, R. and Brown, P.O. (2007) Transcriptional program induced by wnt protein in human fibroblasts suggests mechanisms for cell cooperativity in defining tissue microenvironments. PLoS ONE, 2, e945. Kobayashi, S., Boggon, T.J., Dayaram, T., Janne, P.A., Kocher, O., Meyerson, M., Johnson, B.E., Eck, M.J., Tenen, D.G. and Halmos, B. (2005) EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N Engl J Med, 352, 786792. Kolligs, F.T., Hu, G., Dang, C.V. and Fearon, E.R. (1999) Neoplastic transformation of RK3E by mutant beta-catenin requires deregulation of Tcf/Lef transcription but not activation of c-myc expression. Mol Cell Biol, 19, 5696-5706. Kuphal, F. and Behrens, J. (2006) E-cadherin modulates Wnt-dependent transcription in colorectal cancer cells but does not alter Wnt-independent gene expression in fibroblasts. Exp Cell Res, 312, 457-467. Lapidot, T., Sirard, C., Vormoor, J., Murdoch, B., Hoang, T., Caceres-Cortes, J., Minden, M., Paterson, B., Caligiuri, M.A. and Dick, J.E. (1994) A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature, 367, 645648. Larue, L., Ohsugi, M., Hirchenhain, J. and Kemler, R. (1994) E-cadherin null mutant embryos fail to form a trophectoderm epithelium. Proc Natl Acad Sci U S A, 91, 8263-8267. Li, C., Heidt, D.G., Dalerba, P., Burant, C.F., Zhang, L., Adsay, V., Wicha, M., Clarke, M.F. and Simeone, D.M. (2007) Identification of pancreatic cancer stem cells. CancerRes, 67, 1030-1037. Liebner, S., Cattelino, A., Gallini, R., Rudini, N., Iurlaro, M., Piccolo, S. and Dejana, E. (2004) Beta-catenin is required for endothelial-mesenchymal transformation during heart cushion development in the mouse. J Cell Biol, 166, 359-367. Lien, H.C., Hsiao, Y.H., Lin, Y.S., Yao, Y.T., Juan, H.F., Kuo, W.H., Hung, M.C., Chang, K.J. and Hsieh, F.J. (2007) Molecular signatures of metaplastic carcinoma of the breast by large-scale transcriptional profiling: identification of genes potentially related to epithelial-mesenchymal transition. Oncogene, 26, 7859-7871. Liu, Y., El-Naggar, S., Darling, D.S., Higashi, Y. and Dean, D.C. (2008) Zeb1 links epithelial-mesenchymal transition and cellular senescence. Development, 135, 579-588. Lo, H.W., Hsu, S.C., Xia, W., Cao, X., Shih, J.Y., Wei, Y., Abbruzzese, J.L., Hortobagyi, G.N. and Hung, M.C. (2007) Epidermal growth factor receptor cooperates with signal transducer and activator of transcription 3 to induce epithelialmesenchymal transition in cancer cells via up-regulation of TWIST gene expression. CancerRes, 67, 9066-9076. Lombaerts, M., van Wezel, T., Philippo, K., Dierssen, J.W., Zimmerman, R.M., Oosting, J., van Eijk, R., Eilers, P.H., van de Water, B., Cornelisse, C.J. and Cleton-Jansen, A.M. (2006) E-cadherin transcriptional downregulation by promoter methylation but not mutation is related to epithelial-to-mesenchymal transition in breast cancer cell lines. Br J Cancer, 94, 661-671. Lu, Z., Ghosh, S., Wang, Z. and Hunter, T. (2003) Downregulation of caveolin-1 function by EGF leads to the loss of E-cadherin, increased transcriptional activity of beta-catenin, and enhanced tumor cell invasion. CancerCell, 4, 499-515. Luzzi, K.J., MacDonald, I.C., Schmidt, E.E., Kerkvliet, N., Morris, V.L., Chambers, A.F. and Groom, A.C. (1998) Multistep nature of metastatic inefficiency: dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Am J Pathol, 153, 865-873. Maestro, R., Dei Tos, A.P., Hamamori, Y., Krasnokutsky, S., Sartorelli, V., Kedes, L., Doglioni, C., Beach, D.H. and Hannon, G.J. (1999) Twist is a potential oncogene that inhibits apoptosis. Genes Dev, 13, 2207-2217. Makiguchi, Y., Hinoda, Y. and Imai, K. (1996) Effect of MUC1 mucin, an anti-adhesion molecule, on tumor cell growth. Jpn J CancerRes, 87, 505-511. Mani, S.A., Guo, W., Liao, M., Eaton, E.N., Ayyanan, A., Zhou, A., Brooks, M., Reinhard, F., Zhang, C.C., Shipitsin, M., Campbell, L.L., Polyak, K., Brisken, C., Yang, J. and Weinberg, R.A. (2008) The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell, In press. Mani, S.A., Yang, J., Brooks, M., Schwaninger, G., Zhou, A., Miura, N., Kutok, J.L., Hartwell, K., Richardson, A.L. and Weinberg, R.A. (2007) Mesenchyme Forkhead 1 (FOXC2) plays a key role in metastasis and is associated with aggressive basal-like breast cancers. ProcNatl Acad Sci U S A, 104, 10069-10074. Marambaud, P., Shioi, J., Serban, G., Georgakopoulos, A., Sarner, S., Nagy, V., Baki, L., Wen, P., Efthimiopoulos, S., Shao, Z., Wisniewski, T. and Robakis, N.K. (2002) A presenilin-1/gamma-secretase cleavage releases the E-cadherin intracellular domain and regulates disassembly of adherens junctions. Embo J, 21, 1948-1956. Maretzky, T., Reiss, K., Ludwig, A., Buchholz, J., Scholz, F., Proksch, E., de Strooper, B., Hartmann, D. and Saftig, P. (2005) ADAM10 mediates E-cadherin shedding and regulates epithelial cell-cell adhesion, migration, and beta-catenin translocation. Proc Natl Acad Sci U S A, 102, 9182-9187. Martin, S.S., Ridgeway, A.G., Pinkas, J., Lu, Y., Reginato, M.J., Koh, E.Y., Michelman, M., Daley, G.Q., Brugge, J.S. and Leder, P. (2004) A cytoskeleton-based functional genetic screen identifies Bcl-xL as an enhancer of metastasis, but not primary tumor growth. Oncogene, 23, 4641-4645. Martin, T.A., Goyal, A., Watkins, G. and Jiang, W.G. (2005) Expression of the transcription factors snail, slug, and twist and their clinical significance in human breast cancer. Ann Surg Oncol, 12, 488-496. McGuire, J.K., Li, Q. and Parks, W.C. (2003) Matrilysin (matrix metalloproteinase-7) mediates E-cadherin ectodomain shedding in injured lung epithelium. Am J Pathol, 162, 1831-1843. McNeill, H., Ozawa, M., Kemler, R. and Nelson, W.J. (1990) Novel function of the cell adhesion molecule uvomorulin as an inducer of cell surface polarity. Cell, 62, 309-316. Metzstein, M.M. and Horvitz, H.R. (1999) The C. elegans cell death specification gene ces-1 encodes a snail family zinc finger protein. Mol Cell, 4, 309-319. Mitani, M., Yamanishi, T. and Miyazaki, Y. (1975) Salinomycin: a new monovalent cation ionophore. Biochem Biophys Res Commun, 66, 1231-1236. Mitani, M., Yamanishi, T., Miyazaki, Y. and Otake, N. (1976) Salinomycin effects on mitochondrial ion translocation and respiration. Antimicrob Agents Chemother, 9, 655-660. Moody, S.E., Perez, D., Pan, T.C., Sarkisian, C.J., Portocarrero, C.P., Sterner, C.J., Notorfrancesco, K.L., Cardiff, R.D. and Chodosh, L.A. (2005) The transcriptional repressor Snail promotes mammary tumor recurrence. Cancer Cell, 8, 197-209. Morali, O.G., Delmas, V., Moore, R., Jeanney, C., Thiery, J.P. and Larue, L. (2001) IGFII induces rapid beta-catenin relocation to the nucleus during epithelium to mesenchyme transition. Oncogene, 20, 4942-4950. Moreno-Bueno, G., Cubillo, E., Sarrio, D., Peinado, H., Rodriguez-Pinilla, S.M., Villa, S., Bolos, V., Jorda, M., Fabra, A., Portillo, F., Palacios, J. and Cano, A. (2006) Genetic profiling of epithelial cells expressing E-cadherin repressors reveals a distinct role for Snail, Slug, and E47 factors in epithelial-mesenchymal transition. CancerRes, 66, 9543-9556. Nagafuchi, A., Shirayoshi, Y., Okazaki, K., Yasuda, K. and Takeichi, M. (1987) Transformation of cell adhesion properties by exogenously introduced E-cadherin cDNA. Nature, 329, 341-343. Nass, S.J., Herman, J.G., Gabrielson, E., Iversen, P.W., Parl, F.F., Davidson, N.E. and Graff, J.R. (2000) Aberrant methylation of the estrogen receptor and E-cadherin 5' CpG islands increases with malignant progression in human breast cancer. Cancer Res, 60, 4346-4348. Naumov, G.N., MacDonald, I.C., Weinmeister, P.M., Kerkvliet, N., Nadkarni, K.V., Wilson, S.M., Morris, V.L., Groom, A.C. and Chambers, A.F. (2002) Persistence of solitary mammary carcinoma cells in a secondary site: a possible contributor to dormancy. CancerRes, 62, 2162-2168. Nelson, W.J. and Nusse, R. (2004) Convergence of Wnt, beta-catenin, and cadherin pathways. Science, 303, 1483-1487. Nieto, M.A., Sargent, M.G., Wilkinson, D.G. and Cooke, J. (1994) Control of cell behavior during vertebrate development by Slug, a zinc finger gene. Science, 264, 835-839. Oft, M., Peli, J., Rudaz, C., Schwarz, H., Beug, H. and Reichmann, E. (1996) TGF-betal and Ha-Ras collaborate in modulating the phenotypic plasticity and invasiveness of epithelial tumor cells. Genes Dev, 10, 2462-2477. Oka, H., Shiozaki, H., Kobayashi, K., Inoue, M., Tahara, H., Kobayashi, T., Takatsuka, Y., Matsuyoshi, N., Hirano, S., Takeichi, M. and et al. (1993) Expression of Ecadherin cell adhesion molecules in human breast cancer tissues and its relationship to metastasis. CancerRes, 53, 1696-1701. Orsulic, S., Huber, O., Aberle, H., Arnold, S. and Kemler, R. (1999) E-cadherin binding prevents beta-catenin nuclear localization and beta-catenin/LEF- 1-mediated transactivation. J Cell Sci, 112 ( Pt 8), 1237-1245. Ozawa, M., Ringwald, M. and Kemler, R. (1990) Uvomorulin-catenin complex formation is regulated by a specific domain in the cytoplasmic region of the cell adhesion molecule. ProcNatl Acad Sci U S A, 87, 4246-4250. Peinado, H., Olmeda, D. and Cano, A. (2007) Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat Rev Cancer,7, 415428. Perez-Moreno, M., Davis, M.A., Wong, E., Pasolli, H.A., Reynolds, A.B. and Fuchs, E. (2006) p120-catenin mediates inflammatory responses in the skin. Cell, 124, 631644. Perez-Moreno, M.A., Locascio, A., Rodrigo, I., Dhondt, G., Portillo, F., Nieto, M.A. and Cano, A. (2001) A new role for E12/E47 in the repression of E-cadherin expression and epithelial-mesenchymal transitions. J Biol Chem, 276, 2742427431. Perl, A.K., Wilgenbus, P., Dahl, U., Semb, H. and Christofori, G. (1998) A causal role for E-cadherin in the transition from adenoma to carcinoma. Nature, 392, 190-193. Perrais, M., Chen, X., Perez-Moreno, M. and Gumbiner, B.M. (2007) E-cadherin homophilic ligation inhibits cell growth and epidermal growth factor receptor signaling independently of other cell interactions. Mol Biol Cell, 18, 2013-2025. Peyrieras, N., Hyafil, F., Louvard, D., Ploegh, H.L. and Jacob, F. (1983) Uvomorulin: a nonintegral membrane protein of early mouse embryo. ProcNatl Acad Sci U S A, 80, 6274-6277. Pham, C.G., Bubici, C., Zazzeroni, F., Knabb, J.R., Papa, S., Kuntzen, C. and Franzoso, G. (2007) Upregulation of Twist-1 by NF-kappaB blocks cytotoxicity induced by chemotherapeutic drugs. Mol Cell Biol, 27, 3920-3935. Phillips, T.M., McBride, W.H. and Pajonk, F. (2006) The response of CD24(/low)/CD44+ breast cancer-initiating cells to radiation. J Natl CancerInst, 98, 1777-1785. Prakasam, A.K., Maruthamuthu, V. and Leckband, D.E. (2006) Similarities between heterophilic and homophilic cadherin adhesion. ProcNatl Acad Sci US A, 103, 15434-15439. Qian, X., Karpova, T., Sheppard, A.M., McNally, J. and Lowy, D.R. (2004) E-cadherinmediated adhesion inhibits ligand-dependent activation of diverse receptor tyrosine kinases. Embo J, 23, 1739-1748. Rasband, W.S. (1997-2006) ImageJ U. S. National Institutes of Health, Bethesda, Maryland, USA, http://rsb.info.nih.gov/ij/, 1997-2006. Ricci-Vitiani, L., Lombardi, D.G., Pilozzi, E., Biffoni, M., Todaro, M., Peschle, C. and De Maria, R. (2007) Identification and expansion of human colon-cancerinitiating cells. Nature, 445, 111-115. Riethmacher, D., Brinkmann, V. and Birchmeier, C. (1995) A targeted mutation in the mouse E-cadherin gene results in defective preimplantation development. Proc Natl Acad Sci US A, 92, 855-859. Robson, E.J., Khaled, W.T., Abell, K. and Watson, C.J. (2006) Epithelial-tomesenchymal transition confers resistance to apoptosis in three murine mammary epithelial cell lines. Differentiation,74, 254-264. Roche-Lestienne, C., Soenen-Cornu, V., Grardel-Duflos, N., Lai, J.L., Philippe, N., Facon, T., Fenaux, P. and Preudhomme, C. (2002) Several types of mutations of the Abl gene can be found in chronic myeloid leukemia patients resistant to STI571, and they can pre-exist to the onset of treatment. Blood, 100, 1014-1018. Sato, T., Tanigami, A., Yamakawa, K., Akiyama, F., Kasumi, F., Sakamoto, G. and Nakamura, Y. (1990) Allelotype of breast cancer: cumulative allele losses promote tumor progression in primary breast cancer. CancerRes, 50, 7184-7189. Savagner, P. (2001) Leaving the neighborhood: molecular mechanisms involved during epithelial-mesenchymal transition. Bioessays, 23, 912-923. Savagner, P., Kusewitt, D.F., Carver, E.A., Magnino, F., Choi, C., Gridley, T. and Hudson, L.G. (2005) Developmental transcription factor slug is required for effective re-epithelialization by adult keratinocytes. J Cell Physiol, 202, 858-866. Savagner, P., Yamada, K.M. and Thiery, J.P. (1997) The zinc-finger protein slug causes desmosome dissociation, an initial and necessary step for growth factor-induced epithelial-mesenchymal transition. J Cell Biol, 137, 1403-1419. Schatton, T., Murphy, G.F., Frank, N.Y., Yamaura, K., Waaga-Gasser, A.M., Gasser, M., Zhan, Q., Jordan, S., Duncan, L.M., Weishaupt, C., Fuhlbrigge, R.C., Kupper, T.S., Sayegh, M.H. and Frank, M.H. (2008) Identification of cells initiating human melanomas. Nature, 451, 345-349. Scheel, C., Onder, T., Karnoub, A. and Weinberg, R.A. (2007) Adaptation versus selection: the origins of metastatic behavior. CancerRes, 67, 11476-11479; discussion 11479-11480. Schipper, J.H., Frixen, U.H., Behrens, J., Unger, A., Jahnke, K. and Birchmeier, W. (1991) E-cadherin expression in squamous cell carcinomas of head and neck: inverse correlation with tumor dedifferentiation and lymph node metastasis. CancerRes, 51, 6328-6337. Schuh, R., Vestweber, D., Riede, I., Ringwald, M., Rosenberg, U.B., Jackle, H. and Kemler, R. (1986) Molecular cloning of the mouse cell adhesion molecule uvomorulin: cDNA contains a B 1-related sequence. ProcNatl Acad Sci U S A, 83, 1364-1368. Shah, A.N., Summy, J.M., Zhang, J., Park, S.I., Parikh, N.U. and Gallick, G.E. (2007) Development and characterization of gemcitabine-resistant pancreatic tumor cells. Ann Surg Oncol, 14, 3629-3637. Shimoyama, Y., Hirohashi, S., Hirano, S., Noguchi, M., Shimosato, Y., Takeichi, M. and Abe, 0. (1989) Cadherin cell-adhesion molecules in human epithelial tissues and carcinomas. CancerRes, 49, 2128-2133. Siitonen, S.M., Kononen, J.T., Helin, H.J., Rantala, I.S., Holli, K.A. and Isola, J.J. (1996) Reduced E-cadherin expression is associated with invasiveness and unfavorable prognosis in breast cancer. Am J Clin Pathol,105, 394-402. Singh, S.K., Hawkins, C., Clarke, I.D., Squire, J.A., Bayani, J., Hide, T., Henkelman, R.M., Cusimano, M.D. and Dirks, P.B. (2004) Identification of human brain tumour initiating cells. Nature, 432, 396-401. Stewart, S.A., Dykxhoorn, D.M., Palliser, D., Mizuno, H., Yu, E.Y., An, D.S., Sabatini, D.M., Chen, I.S., Hahn, W.C., Sharp, P.A., Weinberg, R.A. and Novina, C.D. (2003) Lentivirus-delivered stable gene silencing by RNAi in primary cells. Rna, 9, 493-501. Stockinger, A., Eger, A., Wolf, J., Beug, H. and Foisner, R. (2001) E-cadherin regulates cell growth by modulating proliferation-dependent beta-catenin transcriptional activity. J Cell Biol, 154, 1185-1196. Stoker, M. and Perryman, M. (1985) An epithelial scatter factor released by embryo fibroblasts. J Cell Sci, 77, 209-223. Strathdee, G. (2002) Epigenetic versus genetic alterations in the inactivation of Ecadherin. Semin CancerBiol, 12, 373-379. Sulzer, M.A., Leers, M.P., van Noord, J.A., Bollen, E.C. and Theunissen, P.H. (1998) Reduced E-cadherin expression is associated with increased lymph node metastasis and unfavorable prognosis in non-small cell lung cancer. Am J Respir Crit Care Med, 157, 1319-1323. Team, R.D.C. (2007) R: A Language and Environment for Statistical Computing. R Foundation for Statistical Computing, Vienna, Austria. Teicher, B.A., Ikebe, M., Ara, G., Keyes, S.R. and Herbst, R.S. (1997) Transforming growth factor-beta 1 overexpression produces drug resistance in vivo: reversal by decorin. In Vivo, 11, 463-472. Tester, A.M., Ruangpanit, N., Anderson, R.L. and Thompson, E.W. (2000) MMP-9 secretion and MMP-2 activation distinguish invasive and metastatic sublines of a mouse mammary carcinoma system showing epithelial-mesenchymal transition traits. Clin Exp Metastasis, 18, 553-560. Thiery, J.P. (2002) Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer, 2, 442-454. Thiery, J.P. (2003) Epithelial-mesenchymal transitions in development and pathologies. Curr Opin Cell Biol, 15, 740-746. Thomson, S., Buck, E., Petti, F., Griffin, G., Brown, E., Ramnarine, N., Iwata, K.K., Gibson, N. and Haley, J.D. (2005) Epithelial to mesenchymal transition is a determinant of sensitivity of non-small-cell lung carcinoma cell lines and xenografts to epidermal growth factor receptor inhibition. CancerRes, 65, 94559462. Thuault, S., Valcourt, U., Petersen, M., Manfioletti, G., Heldin, C.H. and Moustakas, A. (2006) Transforming growth factor-beta employs HMGA2 to elicit epithelialmesenchymal transition. J Cell Biol, 174, 175-183. Tinkle, C.L., Lechler, T., Pasolli, H.A. and Fuchs, E. (2004) Conditional targeting of Ecadherin in skin: insights into hyperproliferative and degenerative responses. Proc Natl Acad Sci US A, 101, 552-557. Tsuda, H., Zhang, W.D., Shimosato, Y., Yokota, J., Terada, M., Sugimura, T., Miyamura, T. and Hirohashi, S. (1990) Allele loss on chromosome 16 associated with progression of human hepatocellular carcinoma. ProcNatl Acad Sci U S A, 87, 6791-6794. Umbas, R., Isaacs, W.B., Bringuier, P.P., Schaafsma, H.E., Karthaus, H.F., Oosterhof, G.O., Debruyne, F.M. and Schalken, J.A. (1994) Decreased E-cadherin expression is associated with poor prognosis in patients with prostate cancer. CancerRes, 54, 3929-3933. Vallin, J., Thuret, R., Giacomello, E., Faraldo, M.M., Thiery, J.P. and Broders, F. (2001) Cloning and characterization of three Xenopus slug promoters reveal direct regulation by Lef/beta-catenin signaling. J Biol Chem, 276, 30350-30358. Valsesia-Wittmann, S., Magdeleine, M., Dupasquier, S., Garin, E., Jallas, A.C., Combaret, V., Krause, A., Leissner, P. and Puisieux, A. (2004) Oncogenic cooperation between H-Twist and N-Myc overrides failsafe programs in cancer cells. Cancer Cell, 6, 625-630. Vasioukhin, V., Bauer, C., Degenstein, L., Wise, B. and Fuchs, E. (2001) Hyperproliferation and defects in epithelial polarity upon conditional ablation of alpha-catenin in skin. Cell, 104, 605-617. Vasko, V., Espinosa, A.V., Scouten, W., He, H., Auer, H., Liyanarachchi, S., Larin, A., Savchenko, V., Francis, G.L., de la Chapelle, A., Saji, M. and Ringel, M.D. (2007) Gene expression and functional evidence of epithelial-to-mesenchymal transition in papillary thyroid carcinoma invasion. Proc Natl Acad Sci U S A, 104, 2803-2808. Vega, S., Morales, A.V., Ocana, O.H., Valdes, F., Fabregat, I. and Nieto, M.A. (2004) Snail blocks the cell cycle and confers resistance to cell death. Genes Dev, 18, 1131-1143. Vestweber, D. and Kemler, R. (1985) Identification of a putative cell adhesion domain of uvomorulin. Embo J, 4, 3393-3398. Vincent-Salomon, A. and Thiery, J.P. (2003) Host microenvironment in breast cancer development: epithelial-mesenchymal transition in breast cancer development. Breast CancerRes, 5, 101-106. Vleminckx, K., Vakaet, L., Jr., Mareel, M., Fiers, W. and van Roy, F. (1991) Genetic manipulation of E-cadherin expression by epithelial tumor cells reveals an invasion suppressor role. Cell, 66, 107-119. Wang, X., Ling, M.T., Guan, X.Y., Tsao, S.W., Cheung, H.W., Lee, D.T. and Wong, Y.C. (2004) Identification of a novel function of TWIST, a bHLH protein, in the development of acquired taxol resistance in human cancer cells. Oncogene, 23, 474-482. Watanabe, N., Kawaguchi, M. and Kobayashi, Y. (1998) Activation of interleukin-lbetaconverting enzyme by nigericin is independent of apoptosis. Cytokine, 10, 645653. Watson, M.A., Ylagan, L.R., Trinkaus, K.M., Gillanders, W.E., Naughton, M.J., Weilbaecher, K.N., Fleming, T.P. and Aft, R.L. (2007) Isolation and molecular profiling of bone marrow micrometastases identifies TWIST1 as a marker of early tumor relapse in breast cancer patients. Clin CancerRes, 13, 5001-5009. Weinberg, R.A. (2007) The biology of cancer.Garland Science, New York. Wicha, M.S. (2006) Cancer stem cells and metastasis: lethal seeds. Clin CancerRes, 12, 5606-5607. Wildenberg, G.A., Dohn, M.R., Carnahan, R.H., Davis, M.A., Lobdell, N.A., Settleman, J. and Reynolds, A.B. (2006) p120-catenin and p190RhoGAP regulate cell-cell adhesion by coordinating antagonism between Rac and Rho. Cell, 127, 1027-1039. Wong, S.Y. and Hynes, R.O. (2006) Lymphatic or hematogenous dissemination: how does a metastatic tumor cell decide? Cell Cycle, 5, 812-817. Wu, W.S., Heinrichs, S., Xu, D., Garrison, S.P., Zambetti, G.P., Adams, J.M. and Look, A.T. (2005) Slug antagonizes p53-mediated apoptosis of hematopoietic progenitors by repressing puma. Cell, 123, 641-653. Wu, X., Chen, H., Parker, B., Rubin, E., Zhu, T., Lee, J.S., Argani, P. and Sukumar, S. (2006) HOXB7, a homeodomain protein, is overexpressed in breast cancer and confers epithelial-mesenchymal transition. CancerRes, 66, 9527-9534. Xue, C., Plieth, D., Venkov, C., Xu, C. and Neilson, E.G. (2003) The gatekeeper effect of epithelial-mesenchymal transition regulates the frequency of breast cancer metastasis. CancerRes, 63, 3386-3394. Yagoda, N., von Rechenberg, M., Zaganjor, E., Bauer, A.J., Yang, W.S., Fridman, D.J., Wolpaw, A.J., Smukste, I., Peltier, J.M., Boniface, J.J., Smith, R., Lessnick, S.L., Sahasrabudhe, S. and Stockwell, B.R. (2007) RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature, 447, 864-868. Yang, A.D., Fan, F., Camp, E.R., van Buren, G., Liu, W., Somcio, R., Gray, M.J., Cheng, H., Hoff, P.M. and Ellis, L.M. (2006a) Chronic oxaliplatin resistance induces epithelial-to-mesenchymal transition in colorectal cancer cell lines. Clin Cancer Res, 12, 4147-4153. Yang, J., Mani, S.A., Donaher, J.L., Ramaswamy, S., Itzykson, R.A., Come, C., Savagner, P., Gitelman, I., Richardson, A. and Weinberg, R.A. (2004) Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell, 117, 927-939. Yang, L., Lin, C. and Liu, Z.R. (2006b) P68 RNA helicase mediates PDGF-induced epithelial mesenchymal transition by displacing Axin from beta-catenin. Cell, 127, 139-155. Yang, W.S. and Stockwell, B.R. (2008) Synthetic Lethal Screening Identifies Compounds Activating Iron-Dependent, Nonapoptotic Cell Death in OncogenicRAS-Harboring Cancer Cells. Chem Biol, 15, 234-245. Yauch, R.L., Januario, T., Eberhard, D.A., Cavet, G., Zhu, W., Fu, L., Pham, T.Q., Soriano, R., Stinson, J., Seshagiri, S., Modrusan, Z., Lin, C.Y., O'Neill, V. and Amler, L.C. (2005) Epithelial versus mesenchymal phenotype determines in vitro sensitivity and predicts clinical activity of erlotinib in lung cancer patients. Clin Cancer Res, 11, 8686-8698. Yawata, A., Adachi, M., Okuda, H., Naishiro, Y., Takamura, T., Hareyama, M., Takayama, S., Reed, J.C. and Imai, K. (1998) Prolonged cell survival enhances peritoneal dissemination of gastric cancer cells. Oncogene, 16, 2681-2686. Yoshida, C. and Takeichi, M. (1982) Teratocarcinoma cell adhesion: identification of a cell-surface protein involved in calcium-dependent cell aggregation. Cell, 28, 217-224. Yoshiura, K., Kanai, Y., Ochiai, A., Shimoyama, Y., Sugimura, T. and Hirohashi, S. (1995) Silencing of the E-cadherin invasion-suppressor gene by CpG methylation in human carcinomas. ProcNatl Acad Sci U S A, 92, 7416-7419. Young, P., Boussadia, O., Halfter, H., Grose, R., Berger, P., Leone, D.P., Robenek, H., Charnay, P., Kemler, R. and Suter, U. (2003) E-cadherin controls adherens junctions in the epidermis and the renewal of hair follicles. Embo J, 22, 57235733. Yu, F., Yao, H., Zhu, P., Zhang, X., Pan, Q., Gong, C., Huang, Y., Hu, X., Su, F., Lieberman, J. and Song, E. (2007) let-7 regulates self renewal and tumorigenicity of breast cancer cells. Cell, 131, 1109-1123. Zhang, X., Wang, Q., Ling, M.T., Wong, Y.C., Leung, S.C. and Wang, X. (2007) Antiapoptotic role of TWIST and its association with Akt pathway in mediating taxol resistance in nasopharyngeal carcinoma cells. Int J Cancer, 120, 1891-1898. Zhuo, W.L., Wang, Y., Zhuo, X.L., Zhang, Y.S. and Chen, Z.T. (2008) Short interfering RNA directed against TWIST, a novel zinc finger transcription factor, increases A549 cell sensitivity to cisplatin via MAPK/mitochondrial pathway. Biochem Biophys Res Commun. Chapter 3 Chemoresistance and selective inhibition of cancer cells that have undergone an epithelial-tomesenchymal transition Tamer T. Onder,12•' 5 Piyush B. Gupta,2'3 ' Eric S. Lander,''1 4 and Robert A. Weinberg 1 (1) Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142 (2) Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139 (3) Broad Institute of MIT and Harvard, Cambridge, MA 02142 (4) Department of Systems Biology, Harvard Medical School, Boston, MA 02115 (5)These authors contributed equally to this work The high throughput screening and follow-up validation studies were performed in close collaboration with Piyush Gupta from the Lander Lab. Initial observation and characterization of the chemoresistance of mesenchymal cells were performed by the thesis author. Introduction Cancer is the leading cause of death in the US in people under 85 years of age (Jemal et al., 2008). More than 90% of these cancer-related deaths are due to metastatic disease that cannot be cured or controlled with the current treatments of surgery, radiation and chemotherapy. During the past two decades there have been only small decreases in the death rates from cancer (decrease of 1.5% per year in males from 1992 to 2002 and 0.8% per year in females from 1994 to 2002) (Jemal et al., 2008). In fact, most of the observed decreases are due to reduction in tobacco use with respect to lung cancers and early detection in colorectal, breast and prostate cancers (Weinberg, 2007). The failure to decrease cancer-related mortality is ultimately due to the inefficiency of current chemotherapeutic drugs. Although treatment is effective in increasing 5-year survival rates, a large percentage of patients who receive treatment still succumb to metastastatic relapse. For example, survival rates at 10 years for women receiving multiple chemotherapy regiments is just above 50% (Clare et al., 2000). Although in vitro and in vivo studies show that cytotoxic chemotherapy drugs are effective in killing cancer cells, a majority of them fail to eradicate tumor cells once they become metastatic. The main reason for this inefficiency is that tumor cells acquire resistance to killing by such drugs. There are a number of alternative mechanisms by which tumors become chemoresistant. Since tumors are often heterogeneous cell populations with genomic instability and genetic diversity, treatment with anti-cancer agents constitutes a selective pressure for the outgrowth of subclones with pre-existing resistance mechanisms already in place (Chabner and Roberts, 2005). One of the mechanisms of acquired resistance is 100 the alteration of proteins targeted by the drug. For example, paclitaxel (a microtubuledestabilizing chemotherapeutic) resistance is associated with the expression of a mutant form of beta-tubulin that binds this compound with less affinity (Giannakakou et al., 1997). Such mechanisms have also been observed with more recently developed targeted therapies. For example, in the case of treatment of chronic myelogenous leukemia (CML) with imatinib (Gleevec), resistance occurs due to the expansion of clones harboring mutated bcr-abl alleles. Importantly, cells harboring such mutant and drug resistant alleles are present before drug treatment (Roche-Lestienne et al., 2002). A similar pattern is also observed in non-small-cell lung cancers treated with small molecule inhibitors of the epidermal growth factor receptor (EGFR) (Kobayashi et al., 2005). In many cases, tumor cells that become resistant to a particular cytotoxic agent display cross-resistance to unrelated drugs as well. This phenomenon is known as multiple drug resistance (MDR). MDR is associated with decreased intracellular drug accumulation and/or inactivation of downstream apoptotic machinery. The first gene conferring MDR was cloned over 20 years ago by the Housman laboratory and codes for a p-glycoprotein efflux pump (Gros et al., 1986). Since then a family of such proteins have been isolated and shown to act as ATP-dependent transporters of small molecules (Dean et al., 2005). In addition, MDR can be caused by alterations in the apoptotic pathway, such as mutations in the tumor suppressor p53 gene and overexpression of antiapoptotic Bcl2 protein (Weinberg, 2007). Regardless of how it arises, overcoming MDR remains one of the biggest challenges to curing or controlling metastatic cancers. 101 Cancer stem cells and chemoresistance The discovery of cancer stem cells (CSC) has added a new level of complexity to treatment of tumors. Recent evidence shows that within a primary tumor only a subset of the cancer cells are able to propagate the tumor, whereas the bulk rest are unable to do so. Although the presence of such CSCs has long been recognized in hematological malignancies, they were only recently identified in a variety of solid tumors, such as cancers of the breast, colon, pancreas, lung, brain and melanoma (Al-Hajj et al., 2003; Kim et al., 2005; Lapidot et al., 1994; Li et al., 2007; Ricci-Vitiani et al., 2007; Schatton et al., 2008; Singh et al., 2004). CSCs can be prospectively identified based on expression of specific cell-surface markers using fluorescence-activated cell sorting (FACS). Such experiments have shown that within primary tumors only a small percentage of cells are CSCs. Because the standard approach to identification of candidate chemotherapeutic compounds relies on inhibition of overall tumor cell growth in short term assays, it is quite possible that assays of this type would fail entirely to identify potential therapies targeting minority CSC populations. Recent reports have indicated that CSCs are more resistant to a wide spectrum of cancer treatments in current use. For example, CD 4 4 high CD24ol w CSCs isolated from breast carcinoma lines are resistant to gamma-irradiation and epirubicin, a commonly used chemotherapeutic drug (Phillips et al., 2006; Yu et al., 2007). Importantly, breast cancers from patients who received chemotherapy were found to be substantially enriched in CD 4 4 high CD24 low cells, suggesting that treatment may in fact lead to the relative outgrowth of CSCs within a given tumor (Yu et al., 2007). In addition, glioma CSCs, identified based on CD133 positivity, are reported to be 102 resistant to DNA- damaging agents (Bao et al., 2006; Diehn and Clarke, 2006). CSCs may also be multidrug resistant due to expression of high levels of ATP- binding-cassette (ABC) transporters implicated in conferring MDR. A recent study show that melanoma stem cells can be identified based on ABCB5 expression, a known chemoresistance mediator (Frank et al., 2005; Schatton et al., 2008). It is unclear at this stage whether it is possible to identify treatments that target CSCs, or whether it might not be the case that CSCs are inevitably resistant to all forms of therapeutic intervention as a result of an intrinsic resistance to apoptosis. The epithelial-to-mesenchymal transition as a determinant of chemoresistance It has become increasingly evident that an epithelial-to-mesenchymal transition (EMT) can render cells resistant to various forms of apoptosis. Activation of TGF-P signaling in mouse mammary carcinoma cells leads to an EMT that is accompanied by resistance to UV-induced cell death (Robson et al., 2006). TGF-P treatment has also been reported to lead to resistance to cyclophosphamide and cisplatin, two commonly used chemotherapeutic drugs (Teicher et al., 1997). Recent reports suggest that the EMT can also confer resistance to the newer generation of targeted therapies. Gene expression profiling of 42 non-small-cell lung cancer (NSCLC) cell lines whose erlotinib (an EGFR inhibitor) sensitivity was predetermined, revealed that resistant cell lines displayed a strong multi-gene signature indicative of an EMT (Yauch et al., 2005). An independent report confirmed that NSCLC cell lines that have a mesenchymal phenotype are insensitive to the growth inhibitory effects of EGFR inhibition in vitro and in xenografts 103 (Thomson et al., 2005). These findings have recently been extended to pancreatic and colorectal cancer cell lines as well (Buck et al., 2007). How passage through an EMT enables cells to withstand apoptotic stimuli is not completely understood. However, accumulating evidence suggests that cell survival is an intrinsic part of the program activated by the EMT-inducing transcription factors (EMTTFs). The first connection between EMT-TFs and cell survival was made in Caenorhabditiselegans where a Snail superfamily member, CES1, was shown to block the programmed cell death of certain neurons (Metzstein and Horvitz, 1999). Subsequently, Snail was shown to confer resistance to cell death induced by the withdrawal of serum and by pro-apoptotic signals, such TNF-a.(Vega et al., 2004) The related family member Slug also has an anti-apoptotic function, protecting hematopoetic progenitor cells from radiation-induced death (Inoue et al., 2002). Furthermore, upregulation of Slug in human leukaemias is associated with cell survival, and Slug overexpression can confer cytokine independence to a IL-3-dependent murine pro-B cell line to a similar extent as Bcl-2 or Bcl-xL (Inukai et al., 1999). Other EMT-TFs also have anti-apoptotic functions. For example, Twist was idenitifed by the Hannon laboratory in a functional screen for genes that could counteract the proapoptotic effects of the myc oncogene (Maestro et al., 1999). The clinical relevance of this finding was confirmed by the observation that Twist is overexpressed in N-myc-amplified neuroblastomas and inhibits the ARF/p53 pathway involved in the myc-dependent apoptosis (Valsesia-Wittmann et al., 2004). Furthermore, the ability of NF-icB to block cytotoxicity induced by chemotherapeutic drugs also depends on Twist expression (Pham et al., 2007). Twist is essential for cisplatin resistance of lung 104 carcinoma cell lines, as experimental Twist inhibition sensitizes these cells to cisplatin. (Zhuo et al., 2008). Moreover, overexpression of Twist decreases the sensitivity of various cancer cell lines to paclitaxel, another cytotoxic agent (Cheng et al., 2007; Wang et al., 2004; Zhang et al., 2007). The link between EMT and chemoresistance has important implications for cancer progression and treatment. The EMT may be one process that couples metastatic dissemination with the development of chemoresistance. As cells that have undergone an EMT represent the pool of cancer cells most competent to metastasize and lead to tumor recurrence, it is of vital importance to find therapies that effectively target such cells. In experiments described in this chapter I establish that cell lines rendered metastatic through E-cadherin-loss-induced EMT are indeed resistant to a wide range of chemotherapeutic drugs. These cell lines were then utilized as a basis for a highthroughput chemical library screen to identify compounds that specifically target cells that have undergone an EMT. 105 Results Chemoresistance in E-cadherin-loss induced EMT We first wished to confirm that passage through an EMT confers resistance to conventional chemotherapeutic drugs. As described in detail in Chapter 2, inhibition of E-cadherin results in a stable mesenchymal differentiation of the experimentally transformed HMLER human breast cancer cell line. While control shRNA expressing cells (HMLER-shGFP) display epithelial phenotypes, such as cobblestone growth morphology and cytokeratin expression, cells expressing an shRNA vector against Ecadherin (HMLER-shEcad) undergo an EMT characterized by the loss of these epithelial phenotypes and the acquisition of mesenchymal ones in their stead, such as fibroblastic morphology and expression of vimentin. We took advantage of these experimentally derived cell lines to address whether the EMT program results in chemoresistance. Accordingly, we treated cultured HMLER-shGFP and HMLER-shEcad cells for a period of 3 days with either doxorubicin or paclitaxel, doing so at various drug concentrations. Both of these drugs are commonly used chemotherapeutic drugs. While doxorubicin is a DNA-damaging agent, paclitaxel is a microtubule-depolymerizing compound. Examination of the dose-response curves indicated that, in the case of both drugs, the IC50 for the HMLER-shEcad cells was significantly higher than the IC50 for the HMLER-shGFP cells. This IC50 measurement reflects the dose of applied drug at which a therapeutic response, such as death of treated cells, is observed. Hence, a higher IC50 indicates an increased resistance of cells to one or another drug. Thus, compared to control cells, HMLER-shEcad cells were -3-fold more resistant to paclitaxel and -10fold more resistant to doxorubicin (Figure 1A). 106 Increased drug resistance could be due to differential effects of the transforming oncogene in the two cell populations or; alternatively, it could be a more general property of mesenchymal cells or cells that have undergone an EMT. Therefore, we studied HMLE cells, which are immortalized but non-tumorigenic breast epithelial cells that differ from HMLER cells in that they lack an introduced HrasV12 oncogene. Importantly, as described in Chapter 2 and similarly to HMLER cells, loss of E-cadherin in HMLE cells also results in mesenchymal differentiation. We treated HMLE-shGFP and HMLE-shEcad cells for a period of three days with a series of chemical compounds at three different concentrations . These compounds included the established chemotherapy drugs doxorubicin, paclitaxel, actinomycin D, and camptothecin, as well as a broad spectrum kinase inhibitor, staurosporine. The HMLEshEcad cells displayed an increased resistance to each of the examined drugs, relative to the HMLE-shGFP cells (Figure 1B). While the extent of resistance depended on the particular drug examined, the increased resistance was observed for all tested compounds. This observation indicated that the mesenhymal HMLE-shEcad cells are intrinsically resistant to cell death compared to their epithelial counterparts, irrespective of the death-inducing agent used. In addition, the above results indicated that epithelial and mesenchymal populations of cells exhibit differential sensitivities to a variety of chemical compounds independent of neoplastic transformation. Therefore, the primary reason for the differential sensitivity to cell death exhibited by the transformed HMLERshEcad and HMLER-shGFP cells is the divergent differentiation states of these lines. As mentioned in Chapter 1, in the tumor context the EMT can be induced by soluble signals originating from the tumor stroma (Brabletz et al., 2001; Weinberg, 2007). 107 In general, a minority of carcinoma cells is in close contact with the surrounding stromal cells and is thought to have undergone an EMT. Therefore, tumors are likely to be heterogeneous cell populations composed of a majority of cells in the epithelial differentiation state and a minority of cells that have acquired a mesenchymal phenotype. To determine the effect of conventional chemotherapy drugs on a mixture of epithelial and mesenchymal cells, we co-mixed GFP-labeled HMLE-shEcad cells with unlabeled HMLE-shGFP cells at a ratio of 1:20, and treated the resulting co-culture with paclitaxel (Figure 2). This mixture provided an internally controlled method to evaluate whether differential drug sensitivity was also manifest under conditions in which mesenchymal cells co-existed with epithelial cells and constituted, at the same time, a small minority of the overall cell population. After 3 days of paclitaxel (10nM) treatment, FACS analysis revealed a 3-fold increase in the proportion of mesenchymal HMLEshEcad cells relative to DMSO-treated co-cultures (Figure 2). A more modest increase in HMLE-shEcad cells was also observed with a lower dose of paclitaxel (2.5nM). Together, these results indicated that paclitaxel treatment of heterogeneous cell populations can result in the selective outgrowth of cells that have undergone an EMT. Chemical compound screen Collectively, the above observations indicated that the EMT is not only associated with an increase in metastatic ability but it is also accompanied by an increased resistance to chemotherapy-induced cell death. Therefore, we speculated that compounds that inhibit the viability of mesenchymal cells arising from an EMT would preferentially target cancer cell subpopulations capable of seeding tumors and metastasizing. We postulated 108 that the HMLE-shGFP and HMLE-shEcad cell lines could be exploited for the purpose of discovering compounds that selectively target the cells in the mesenchymal differentiation state. Accordingly, we developed conditions to use an ATP-based luminescence viability assay with 384-well plates in high throughput screening (see Materials and Methods). In this assay, the quantity of luminescence emitted by the cells is a measure of their viability. We screened a library collection of -15,000 compounds in this initial proof-of-concept screen. These libraries included collections of compounds with known bioactivity, natural extracts, agents known to affect histone deacetylases, and several commercial libraries that encompassed a diverse array of potential therapeutic compounds (see Materials and Methods). To enable rigorous plate-based normalization, each compound plate carried internal control wells (-10% of total) that contained only DMSO, the solvent in which the compounds were dissolved. Each compound plate was pin-transferred into 2 replicates for each of the HMLE-shEcad and HMLE-shGFP cell lines. Drug treatment was initiated one day following seeding and the cell viability was measured 3 days following drug treatment (Figure 3A-schematic). We conducted our screen in duplicate for each of the HMLE-shGFP and HMLE-shEcad cell lines. Using the measurements from the plate-normalized DMSO control wells, we constructed the distribution of luminescence intensities for each cell line in the absence of compound, but in the presence of control solvent. Each measurement for both control and test wells constituted an average of the two replicates for a cell line. Relative to these baseline distributions, we calculated a Z-score that reflected the biological effect of every compound on the cells being tested, one for each of the HMLE-shGFP and HMLE- 109 shEcad cell lines (Figure 3B). The computed Z-scores did not display a positional bias across drug plate, column, or row number, suggesting that there were no systematic biases in the analysis of the data normalized relative to the internal control DMSO wells (data not shown). Identification of inhibitory compounds using this method indicated that the vast majority of the compounds inhibited the viability of both the HMLE-shGFP and HMLEshEcad cells. Since we were interested in compounds exhibiting preferential lethality toward the cells that had been induced to undergo an EMT, we identified those compounds that inhibited HMLE-shEcad cell viability but did not surpass the threshold of significance of inhibiting HMLE-shGFP viability. About 10% of the compounds in the screened collections inhibited the viability of HMLE-shEcad cells, but the vast majority of these compounds (98%) also inhibited the control cells. Only -0.2% of total library exhibited selective toxicity toward the HMLEshEcad cells. Various established drugs used for cancer chemotherapy were included in the compound plates. Less than 1% of these chemotherapeutic agents scored as being positive in our assay, in that they failed to affect preferentially the cells that have undergone an EMT. This suggested that compounds targeting rapidly proliferating cells were not selectively enriched in our assay. In fact, the HMLE-shGFP cells have a slightly higher rate of proliferation than the HLME-shEcad cells (data not shown). Thus, there was no enrichment of positive hits in the subset of established chemotherapy drugs relative to the total compounds screened. 110 Validation of screen hits Based on their ready availability, we decided to pursue eight of the identified compounds that were selectively toxic for HMLE-shEcad cells for further study; these compounds were salinomycin, etoposide, acridine, cetylpyridinium chloride, resorcinol, clotrimazole, abamectin, and nigericin. For each of these compounds, we evaluated the effects on cell viability across a range of doses, doing so in parallel for sibling cell lines that had either been induced into or not induced to pass through an EMT. Three compounds (etoposide, salinomycin, abamectin) showed moderate to strong selectivity (IC50 is -10-fold lower for HMLEshEcad cells vs HMLE-shCntrl cells); one compound showed weaker selectivity (nigericin; -7-fold) (Figure 4A). The remaining 4 compounds did not exhibit specific effects on viability upon follow-up, and therefore failed to be validated (data not shown). To determine if the specificity of the validated compounds for the HMLE-shEcad cells was a consequence of the mesenchymal differentiation state rather than the particular genetic perturbation used, we also tested the effects of these compounds on cells induced to undergo EMT due to Twist overexpression (HMLE-Twist). In fact, the dose-response curves of each of the tested compounds for HMLE-Twist cells were virtually identical to those observed for the HMLE-shEcad cells (Figure 4B). This observation strongly suggested that compounds exhibiting specific activity in this assay were targeting distinct states of differentiation rather than the particular inducing agents themselves. In order to examine the effects of salinomycin and abamectin treatment on heterogenous cell populations, we co-mixed GFP-labeled HMLE cells with unlabeled 111 HMLE-Twist cells and treated the resulting co-cultures with test compounds. As a positive control for this experiment, we treated co-cultures with blasticidin, which selects for the epithelial HMLE-GFP cells harboring a blasticidin-resistance gene. We observed that treatment with salinomycin, abamectin or blasticidin resulted in an increase in the GFP-positive fraction relative to DMSO-treated controls, indicating that the unlabelled mesenchymal cells were depleted from the population in response to treatment (Figure 5). This stood in contrast to our earlier findings demonstrating that paclitaxel treatment resulted in a selection for mesenchymal cells (Figure 2). Hence, the chemical compounds identified through our screen, in contrast to a commonly used chemotherapeutic agent, resulted in the selective killing of cells that have undergone an EMT. While these compounds were identified as selective inhibitors of immortalized human mammary epithelial cells (HMLE-shEcad) that had undergone an EMT, it was not clear whether they would also exhibit any selective effect on the corresponding tumorigenic cells (HMLER-shEcad). To examine this notion, we treated HMLER-shCntrl and HMLER-shEcad cells with the three most selective compounds for three days and generated dose response curves as before. While salinomycin continued to exhibit selective toxicity against mesenchymal cells (-6 fold difference in IC50, 1.5PM for shEcad vs. 9 pLM for shCntrl cells), the two other compounds, abamectin and etoposide, inhibited the viability of both cell lines to similar extents, indicating that they were no longer differentially targeting one or the other cell populations (Figure 6). This finding suggests that, for a subset of the compounds, oncogenic transformation masks the sensitivity conferred by the differentiation state of the cells. 112 Effects of salinomycin on cancer stem cells Recent studies from our laboratory indicate that induction of an EMT by overexpression of Twist or Snail endows breast carcinoma cells with many of the properties of cancer stem cells, such as tumor seeding at limiting dilution, mammosphere formation, and a CD 4 4 high/ CD24low antigenic profile (Mani et al., 2008). We had demonstrated that these observations also hold true for cells induced into an EMT via inhibition of E-cadherin (HMLERshEcad; Chapter 2, Figure 8). Importantly, HMLERshEcad cells are resistant to a variety of chemotherapeutic compounds similar to what is observed with prospectively identified CSCs from human patients and tumor cell lines (Yu et al., 2007). Since the above mentioned screen identified compounds that are preferentially cytotoxic to HMLERshEcad cells that have undergone an EMT, we speculated that these compounds will also have a cytotoxic effect on the CD 4 4 high/ CD2410l CSC populations that exist in breast carcinoma cell lines. To test this notion, we analyzed using FACS several human breast cancer cell lines for the expression of markers (CD44high/CD24 low) reported to enrich for cancer stem cells: the MDA-MB-231 and SUM159 cells contained greater than 90% CD44high/CD241ow cells; in contrast, the T47D cell line displayed <.1% CD44high/CD241ow cells (Figure 7A). Interestingly, the MDA-MB-231 and SUM159 lines both have a mesenchymal and highly metastatic phenotype, whereas the T47D line is epithelial and poorly-metastatic. We treated these lines with salinomycin and observed that the CSCenriched cell lines SUM159 and MDA-MB-231 were more sensitive across a range of doses relative to the CSC-poor T47D cells (Figure 7B). These data suggest that salinomycin sensitivity of breast cancer cell lines correlates with the prevalence of CSC- 113 like cells in such lines. However, further experimentation is needed to determine whether salinomycin treatment functionally decreases tumor-initiating ability. Taken together, the experiments described in this chapter establish a proof-ofprinciple for discovering compounds that target highly metastatic and otherwise chemotherapy-resistant cancer cells. Although resistant to a wide range of currently used drugs, cells that have undergone an EMT may have unique vulnerabilities that could be uncovered using the approach outlined above. In addition, this approach could potentially be useful to target tumors and tumor initiating cells that have a mesenchymal phenotype. 114 Materials and Methods Cell culture. Immortalized (HMLE) or transformed (HMLER) breast epithelial cells that express either control shRNA (shCntrl) or shRNA targeting E-cadherin (shEcad) were generated and maintained as described in Chapter 2. HMLE-Twist cells were also described previously (Yang et al., 2004). To create GFP ihyphen expressing lines, we infected the HMLE and HMLE-shEcad cells with a pWZL-GFP retrovirus carrying the blasticidin resistance gene using standard procedures (Elenbaas et al., 2001). Characterization of resistance to cytotoxic agents. Doxorubicin, paclitaxel, actinomycin D, campthotecin and staurosporine were purchased from Sigma and dissolved in dimethyl sulfoxide (DMSO). 5000 cells in 100 ul of medium were plated per well in 96-well plates. 24 hrs after seeding, compounds at the indicated concentration were added to wells (5 wells per each concentration). DMSO treatment was used as negative control. Cell viability was measured after 72 hrs using the CellTiter96 AQueous Assay (MTT) (Promega) according to manufacturer's instructions. Dose response curves were generated with GraphPad Prism software (GraphPad Software, Inc.). For the co-mix experiments, unlabelled and GFP-labeled cells were mixed at the indicated ratios and seeded onto 6-well plates. Triplicate wells were then treated with DMSO or the compounds (paclitaxel, salinomycin, abamectin and blasticidin) for 48 hrs. The cells were washed with PBS, trypsinized and analyzed by FACS for GFP positivity. 115 Chemical Screen Information about the general automated HTS protocol is available at http://www.broad.harvard.edulchembio/index.html. The assay was initiated by plating 40 iL of medium containing 1000 cells/well into white 384-well opaque-bottom plates (Nunc, Rochester, NY) using an automated plate filler (Bio-Tek [tFiller; Winsooki, VT) and allowing the cells to adhere for 24 hours. 100 nL of compound stock solutions in DMSO was transferred from stock plates into the 384-well assay plates using an automated pin-based compound transfer robot (CyBio CyBi-Well vario; Woburn, MA). For most compounds, the final concentration in each well was calculated to be 10uM. The screen was performed in duplicate. For negative controls, entire DMSO-treated control plates were employed, in addition to DMSO-only control wells that were incorporated into each compound assay plate. The cells were assayed for luciferase activity with the addition 20 ul of CellTiter-Glo Luminescent Cell Viability Assay solution (Promega). The luminescent signal from each plate was detected using an automated plate reader (Perkin-Elmer Envision 1; Wellesley, MA). The compound plate numbers for screened plates were 2158-2167, 2099-2105, 2290-2297, 2403-2407, Biokinl-2. The primary data were analyzed using the commercial software package SpotFire (SpotFire, Inc., Somerville, MA). Validation of hits Individual compounds were purchased from Sigma and dissolved in DMSO with the exception of nigericin, which was dissolved in 100% ethnanol. Activity of the 116 compounds were quantified by generating dose-response curves for HMLE-shCntrl, HMLE-shEcad and HMLE-Twist under the same cell density and culture conditions described for the initial screen, using the 384-well plate based system. 117 Figures A HMLER R shGFP shEcad * HMLER-shGFP * HMLER-shEcad * HMLER-srGFP a ,I1"1 10-12 10~ o HMLER-shEcad a 1010 r Doorubidin [l4 10 f 10' 1tm lor?0I ir law" R===t~rrt'NI mHMLE-shGFP E HMLE-shEcad mW I noxr On Dxru~II "so , Doxorubicin iU I St 1LO"y DI c Iti Actinomycin D Paci 1 ID Itx Paclitaxel Camm 2IrM 1132or I IM Campthotecin ie i1a" S IStaurorosporine Chemotherapy Drugs Figure 1. Chemoresistance of cells that have undergone an EMT (A) Mesenchymally transdifferentiated cancer cells (HMLER-shEcad) (red) display increased resistance to doxorubicin and paclitaxel-induced cell death relative to control HMLER-shCntrl cells (gray). Dose response curves were generated by treating cells with the indicated doses (X-axis) for 3 days. Cell viability was measured using the MTT assay and values are represented as a fraction of DMSO treated control wells whose value was arbitrarily set at 1. Dotted line indicates 50% survival, corresponding to the IC50 for that compound. (B) Immortalized, non-tumorigenic breast epithelial cells (HMLE) in a mesenchymal state (shEcad) exhibit increased resistance to a variety of drug treatments compared to control immortalized breast epithelial cells (shGFP). Values are presented as a percentage of DMSO treated controls, which was arbitrarily set at 100%. Data were expressed as the mean ± SD. 118 HMLE-shGFP I Admix 5% GFP + cells HMLE-shEca d-pWBGFP Day 1: Treat mix cultures Day 4 : Facs for GFP AL, ---TDmso -I" 1 nM 2.5nM Paclitaxel Figure 2. Treatment with Paclitaxel leads to selective expansion of mesenchymal cells. GFP-labeled immortalized epithelial cells in the mesenchymal state (HMLE-shEcadpWBGFP) were admixed to control epithelial cells (HMLE-shGFP) at ratio of 1:20. The resulting mixed cultures were treated with paclitaxel at the indicated concentrations. Treatment for 3 days with (2.5nM, 10nM) paclitaxel resulted in an increase (-30%, 300%) in the fraction of mesenchymally transdifferentiated HMLE-shEcad cells relative to dmso-treated controls. 119 \ L HMLEsh6FP HMLEshEEcad ChII Lurrminescence Cel Viabiity Assay sNatmra 3Day .... ..>Pr:•.. inc ubation Extracds 8ioeadires Commercial - Libraes 1 Chom"c"l 1 Analze Data -15000 compoun 76, 00 asays r :t for selective toxicy screened otal WOMANrrau o ibU %ICle n ei rg74111ui 9~.r, " 9 H :, 9.) 9·4,~ * 9- i.. i 9~ 90 -1 - -- . - . . Z-scare A Z-Kare A Figure 3. The chemical screen (A) Schematic of the screen design and protocol. (B) (top left) Histogram of normalized values for the viability of each tested compound for control epithelial HMLEshGFP cells. (bottom left) Scatter plot of normalized Zscores for the viability of each tested compound for mesenchymal HMLEshEcad. The xand y-axes represent the two independent replicates for the screen. Red dots represent DMSO-containing wells; blue dots are compound wells (top right) The red region in the histogram represents compounds that exhibit toxicity for control cells. Thresholding out these compounds produces the scatter plot in (bottom right), enabling the identification of compounds that selectivity kill mesenchymally transdifferentiated epithelial cells (yellow dots). 120 A Ur > ,nB E-Z_di .fe-s'ad 1.2 I.5 a ano 4 .10 10 104 104 ' 10-6 Salinomycin [M] B Etoposide [M] -15 115. I " Abamectin [M] I.' 1 Nigericin [M] " SshCr•il ELTWAn -" I" 1i. *11- It............ (0 in- .. .. . .... .. . 10 10 10 A 10 A Salinomycin [M] 10 ,I 10 f i5DNa 10'a 104 10 Etoposide [M] 107 102 10- 10- 106 Abamectin [M] 104 Nigericin [M] Figure 4. Validation of compounds hits (A) Dose-response curves for HMLE-shCntrl cells (green circles) and HMLE-shEcad cells (red triangles) treated with compounds that tested positively in the primary screen. Dose response curves were generated by treating cells with the indicated doses (X-axis) for 3 days. Cell viability values are represented as a fraction of DMSO treated control wells whose value was arbitrarily set at 1. Dotted line indicates 50% survival, corresponding to the IC50 for that compound. (B) Chemical compounds toxicities are independent of the genetic element used to induce the EMT. Dose-response curves of the viability of HMLE-shCntrl cells (green squares) and mammary epithelial cells that were induced to undergo an EMT through expression of the Twist transcription factor (HMLE-Twist) (blue triangles). 121 L ti i ~ g r ""` "" Dmso Salinomycin 5uM Abamectin 1.25uM a Blasticidin HMLE-GFP and HMLE-Twist mix 10 0 Dmso 1.25uM 5uM Salinomycin O.25uM 1.25uM Blast Abamectin Figure 5. Treatment with hit compounds can selectively deplete mesenchymal cells from a heterogenous cell population. GFP-labeled immortalized mammary epithelial cells (HMLE-GFP) were mixed with with unlabeled Twist expressing cells (HMLE-Twist) at a ratio of 1:10. The mixed cultures were treated with either salinomycin, abamectin, blasticidin (blast) or DMSO at the indicated concentrations for 3 days, and subsequently imaged and analyzed using FACS. Quantification of the percent of GFP-positive cells is shown. Blasticidin treatment is a positive control for the selection of GFP-positive cells, which harbor a blasticidin resistance gene. 122 L Jill I'L• •~L...•'.I FI-MLER P-Ecad i4 Salinomycin [M] SI-MLER shQrl * I-LERshEcad ORLB. 11 .8 1g3- 143* 10' i0 Etoposide [M] F-MLER CQIrl F-MLER shEad .4 Abamectin [M] Figure 6. Effects of transformation on compound selectivity Dose-response curves of control (HMLER-shCntrl, green) and mesenchymal (HMLERshEcad, red) tumorigenic mammary epithelial cells treated with salinomycin, etoposide and abamectin. 123 Cell line % CD44 + CD24- Suml59 92.6 % 89.9% MDA-MB-231 TAW 7D I 7I L. .. S40 1I (U SLmrn59 T47D IV3A-231 1.50) 5 -L I.. .... u, us.. 1- 106- . 1 1064 Salinomycin [M] Figure 7. Salinomycin sensitivity correlates with the prevalence of CD44high/CD24low cells. (A) Suml59, MDA-MB-231 and T47D breast cancer cell lines were analyzed by FACS using anti-CD44 and anti-CD24 antibodies. (B) Dose response curves for Sum159 (red), MDA-MB-231 (black), and T47D (green) cells treated with salinomycin for 72 hours. 124 References Al-Hajj, M., Wicha, M.S., Benito-Hernandez, A., Morrison, S.J. and Clarke, M.F. (2003) Prospective identification of tumorigenic breast cancer cells. ProcNatl Acad Sci US A, 100, 3983-3988. Arslan, P., Montecucco, C., Celi, D. and Pozzan, T. (1981) Effect of monovalent cation ionophores on lymphocyte cellular metabolism. Biochim Biophys Acta, 643, 177181. Brabletz, T., Jung, A., Reu, S., Porzner, M., Hlubek, F., Kunz-Schughart, L.A., Knuechel, R. and Kirchner, T. (2001) Variable beta-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. ProcNatl Acad Sci US A, 98, 10356-10361. Chabner, B.A. and Roberts, T.G., Jr. (2005) Timeline: Chemotherapy and the war on cancer. Nat Rev Cancer,5, 65-72. Cheng, G.Z., Chan, J., Wang, Q., Zhang, W., Sun, C.D. and Wang, L.H. (2007) Twist transcriptionally up-regulates AKT2 in breast cancer cells leading to increased migration, invasion, and resistance to paclitaxel. CancerRes, 67, 1979-1987. Clare, S.E., Nakhlis, F. and Panetta, J.C. (2000) Molecular biology of breast cancer metastasis. The use of mathematical models to determine relapse and to predict response to chemotherapy in breast cancer. Breast CancerRes, 2, 430-435. Daniele, R.P., Holian, S.K. and Nowell, P.C. (1978) A potassium ionophore (Nigericin) inhibits stimulation of human lymphocytes by mitogens. J Exp Med, 147, 571-581. Dean, M., Fojo, T. and Bates, S. (2005) Tumour stem cells and drug resistance. Nat Rev Cancer, 5, 275-284. Diehn, M. and Clarke, M.F. (2006) Cancer stem cells and radiotherapy: new insights into tumor radioresistance. J Natl CancerInst, 98, 1755-1757. Elenbaas, B., Spirio, L., Koerner, F., Fleming, M.D., Zimonjic, D.B., Donaher, J.L., Popescu, N.C., Hahn, W.C. and Weinberg, R.A. (2001) Human breast cancer cells generated by oncogenic transformation of primary mammary epithelial cells. Genes Dev, 15, 50-65. Frank, N.Y., Margaryan, A., Huang, Y., Schatton, T., Waaga-Gasser, A.M., Gasser, M., Sayegh, M.H., Sadee, W. and Frank, M.H. (2005) ABCB5-mediated doxorubicin transport and chemoresistance in human malignant melanoma. CancerRes, 65, 4320-4333. Furlong, I.J., Ascaso, R., Lopez Rivas, A. and Collins, M.K. (1997) Intracellular acidification induces apoptosis by stimulating ICE-like protease activity. J Cell Sci, 110 ( Pt 5), 653-661. Furlong, I.J., Lopez Mediavilla, C., Ascaso, R., Lopez Rivas, A. and Collins, M.K. (1998) Induction of apoptosis by valinomycin: mitochondrial permeability transition causes intracellular acidification. Cell Death Differ, 5, 214-221. Giannakakou, P., Sackett, D.L., Kang, Y.K., Zhan, Z., Buters, J.T., Fojo, T. and Poruchynsky, M.S. (1997) Paclitaxel-resistant human ovarian cancer cells have mutant beta-tubulins that exhibit impaired paclitaxel-driven polymerization. J Biol Chem, 272, 17118-17125. 125 Gros, P., Ben Neriah, Y.B., Croop, J.M. and Housman, D.E. (1986) Isolation and expression of a complementary DNA that confers multidrug resistance. Nature, 323, 728-731. Inoue, A., Seidel, M.G., Wu, W., Kamizono, S., Ferrando, A.A., Bronson, R.T., Iwasaki, H., Akashi, K., Morimoto, A., Hitzler, J.K., Pestina, T.I., Jackson, C.W., Tanaka, R., Chong, M.J., McKinnon, P.J., Inukai, T., Grosveld, G.C. and Look, A.T. (2002) Slug, a highly conserved zinc finger transcriptional repressor, protects hematopoietic progenitor cells from radiation-induced apoptosis in vivo. Cancer Cell, 2, 279-288. Inukai, T., Inoue, A., Kurosawa, H., Goi, K., Shinjyo, T., Ozawa, K., Mao, M., Inaba, T. and Look, A.T. (1999) SLUG, a ces-1-related zinc finger transcription factor gene with antiapoptotic activity, is a downstream target of the E2A-HLF oncoprotein. Mol Cell, 4, 343-352. Jemal, A., Siegel, R., Ward, E., Hao, Y., Xu, J., Murray, T. and Thun, M.J. (2008) Cancer statistics, 2008. CA CancerJ Clin, 58, 71-96. Kim, C.F., Jackson, E.L., Woolfenden, A.E., Lawrence, S., Babar, I., Vogel, S., Crowley, D., Bronson, R.T. and Jacks, T. (2005) Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell, 121, 823-835. Kobayashi, S., Boggon, T.J., Dayaram, T., Janne, P.A., Kocher, O., Meyerson, M., Johnson, B.E., Eck, M.J., Tenen, D.G. and Halmos, B. (2005) EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N Engl J Med, 352, 786792. Lapidot, T., Sirard, C., Vormoor, J., Murdoch, B., Hoang, T., Caceres-Cortes, J., Minden, M., Paterson, B., Caligiuri, M.A. and Dick, J.E. (1994) A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature, 367, 645648. Li, C., Heidt, D.G., Dalerba, P., Burant, C.F., Zhang, L., Adsay, V., Wicha, M., Clarke, M.F. and Simeone, D.M. (2007) Identification of pancreatic cancer stem cells. CancerRes, 67, 1030-1037. Maestro, R., Dei Tos, A.P., Hamamori, Y., Krasnokutsky, S., Sartorelli, V., Kedes, L., Doglioni, C., Beach, D.H. and Hannon, G.J. (1999) Twist is a potential oncogene that inhibits apoptosis. Genes Dev, 13, 2207-2217. Mani, S.A., Guo, W., Liao, M., Eaton, E.N., Ayyanan, A., Zhou, A., Brooks, M., Reinhard, F., Zhang, C.C., Shipitsin, M., Campbell, L.L., Polyak, K., Brisken, C., Yang, J. and Weinberg, R.A. (2008) The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell, In press. Metzstein, M.M. and Horvitz, H.R. (1999) The C. elegans cell death specification gene ces-1 encodes a snail family zinc finger protein. Mol Cell, 4, 309-319. Mitani, M., Yamanishi, T. and Miyazaki, Y. (1975) Salinomycin: a new monovalent cation ionophore. Biochem Biophys Res Commun, 66, 1231-1236. Mitani, M., Yamanishi, T., Miyazaki, Y. and Otake, N. (1976) Salinomycin effects on mitochondrial ion translocation and respiration. Antimicrob Agents Chemother,9, 655-660. Pham, C.G., Bubici, C., Zazzeroni, F., Knabb, J.R., Papa, S., Kuntzen, C. and Franzoso, G. (2007) Upregulation of Twist-i by NF-kappaB blocks cytotoxicity induced by chemotherapeutic drugs. Mol Cell Biol, 27, 3920-3935. 126 Phillips, T.M., McBride, W.H. and Pajonk, F. (2006) The response of CD24(/low)/CD44+ breast cancer-initiating cells to radiation. J Natl CancerInst, 98, 1777-1785. Ricci-Vitiani, L., Lombardi, D.G., Pilozzi, E., Biffoni, M., Todaro, M., Peschle, C. and De Maria, R. (2007) Identification and expansion of human colon-cancerinitiating cells. Nature, 445, 111-115. Robson, E.J., Khaled, W.T., Abell, K. and Watson, C.J. (2006) Epithelial-tomesenchymal transition confers resistance to apoptosis in three murine mammary epithelial cell lines. Differentiation,74, 254-264. Roche-Lestienne, C., Soenen-Cornu, V., Grardel-Duflos, N., Lai, J.L., Philippe, N., Facon, T., Fenaux, P. and Preudhomme, C. (2002) Several types of mutations of the Abl gene can be found in chronic myeloid leukemia patients resistant to STI571, and they can pre-exist to the onset of treatment. Blood, 100, 1014-1018. Schatton, T., Murphy, G.F., Frank, N.Y., Yamaura, K., Waaga-Gasser, A.M., Gasser, M., Zhan, Q., Jordan, S., Duncan, L.M., Weishaupt, C., Fuhlbrigge, R.C., Kupper, T.S., Sayegh, M.H. and Frank, M.H. (2008) Identification of cells initiating human melanomas. Nature, 451, 345-349. Singh, S.K., Hawkins, C., Clarke, I.D., Squire, J.A., Bayani, J., Hide, T., Henkelman, R.M., Cusimano, M.D. and Dirks, P.B. (2004) Identification of human brain tumour initiating cells. Nature, 432, 396-401. Teicher, B.A., Ikebe, M., Ara, G., Keyes, S.R. and Herbst, R.S. (1997) Transforming growth factor-beta 1 overexpression produces drug resistance in vivo: reversal by decorin. In Vivo, 11, 463-472. Thomson, S., Buck, E., Petti, F., Griffin, G., Brown, E., Ramnarine, N., Iwata, K.K., Gibson, N. and Haley, J.D. (2005) Epithelial to mesenchymal transition is a determinant of sensitivity of non-small-cell lung carcinoma cell lines and xenografts to epidermal growth factor receptor inhibition. CancerRes, 65, 94559462. Valsesia-Wittmann, S., Magdeleine, M., Dupasquier, S., Garin, E., Jallas, A.C., Combaret, V., Krause, A., Leissner, P. and Puisieux, A. (2004) Oncogenic cooperation between H-Twist and N-Myc overrides failsafe programs in cancer cells. Cancer Cell, 6, 625-630. Vega, S., Morales, A.V., Ocana, O.H., Valdes, F., Fabregat, I. and Nieto, M.A. (2004) Snail blocks the cell cycle and confers resistance to cell death. Genes Dev, 18, 1131-1143. Wang, X., Ling, M.T., Guan, X.Y., Tsao, S.W., Cheung, H.W., Lee, D.T. and Wong, Y.C. (2004) Identification of a novel function of TWIST, a bHLH protein, in the development of acquired taxol resistance in human cancer cells. Oncogene, 23, 474-482. Watanabe, N., Kawaguchi, M. and Kobayashi, Y. (1998) Activation of interleukin-lbetaconverting enzyme by nigericin is independent of apoptosis. Cytokine, 10, 645653. Weinberg, R.A. (2007) The biology of cancer. Garland Science, New York. Yagoda, N., von Rechenberg, M., Zaganjor, E., Bauer, A.J., Yang, W.S., Fridman, D.J., Wolpaw, A.J., Smukste, I., Peltier, J.M., Boniface, J.J., Smith, R., Lessnick, S.L., Sahasrabudhe, S. and Stockwell, B.R. (2007) RAS-RAF-MEK-dependent 127 oxidative cell death involving voltage-dependent anion channels. Nature, 447, 864-868. Yang, W.S. and Stockwell, B.R. (2008) Synthetic Lethal Screening Identifies Compounds Activating Iron-Dependent, Nonapoptotic Cell Death in OncogenicRAS-Harboring Cancer Cells. Chem Biol, 15, 234-245. Yauch, R.L., Januario, T., Eberhard, D.A., Cavet, G., Zhu, W., Fu, L., Pham, T.Q., Soriano, R., Stinson, J., Seshagiri, S., Modrusan, Z., Lin, C.Y., O'Neill, V. and Amler, L.C. (2005) Epithelial versus mesenchymal phenotype determines in vitro sensitivity and predicts clinical activity of erlotinib in lung cancer patients. Clin CancerRes, 11, 8686-8698. Yu, F., Yao, H., Zhu, P., Zhang, X., Pan, Q., Gong, C., Huang, Y., Hu, X., Su, F., Lieberman, J. and Song, E. (2007) let-7 regulates self renewal and tumorigenicity of breast cancer cells. Cell, 131, 1109-1123. Zhang, X., Wang, Q., Ling, M.T., Wong, Y.C., Leung, S.C. and Wang, X. (2007) Antiapoptotic role of TWIST and its association with Akt pathway in mediating taxol resistance in nasopharyngeal carcinoma cells. Int J Cancer, 120, 1891-1898. Zhuo, W.L., Wang, Y., Zhuo, X.L., Zhang, Y.S. and Chen, Z.T. (2008) Short interfering RNA directed against TWIST, a novel zinc finger transcription factor, increases A549 cell sensitivity to cisplatin via MAPK/mitochondrial pathway. Biochem Biophys Res Commun. 128 Chapter 4 Conclusions and Future Directions 129 This thesis focused on two issues. First, I addressed the role of cell-cell adhesion and E-cadherin in the epithelial-to-mesenchymal transition (EMT) and metastatic progression. I was able to accomplish this by creating new cell line models deficient in cell-cell adhesion and E-cadherin expression and analyzing their phenotypes in detail. In the second part of this thesis, I examined the link between the EMT and development of chemoresistance. I took advantage of the cell lines created in the first part to perform a high-throughput small molecule screen. Through this screen, I identified compounds that selectively target cell populations that have a mesenchymal phenotype. In this chapter, I will discuss the implications of my findings and potential future directions. E-cadherin and Metastasis Experiments described in Chapter 2, indicate that the loss of E-cadherin has wideranging transcriptional and functional consequences for human breast epithelial cells. First of all, E-cadherin loss, achieved through shRNA-mediated knock-down, is sufficient to confer metastatic ability upon breast cancer cells that are otherwise essentially nonmetastatic. However, the acquisition of metastatic ability by cancer cells following Ecadherin loss is not attributable solely to the disruption of intercellular adhesion contacts, as demonstrated by utilizing a dominant-negative E-cadherin mutant. Instead, it is the additional loss of E-cadherin protein that provokes an EMT, attended by increased cellular motility, invasiveness and resistance to apoptosis. Thus, E-cadherin can act as a central modulator of the cell-biological phenotypes and the molecular factors that govern metastatic dissemination of carcinomas. 130 The mechanisms by which E-cadherin is inactivated in human tumors can be placed into two general categories: (1) those that result in the production of a nonfunctional protein and (2) those that lead to E-cadherin's complete absence. The present findings suggest that point mutations in the extracellular domain, which preserve Ecadherin's cytoplasmic tail and act analogously to the dominant negative mutant, are not likely to result in an EMT or to afford functional traits that allow completion of the later steps of metastasis (Berx et al., 1998). In contrast, complete elimination of E-cadherin expression, as shown by RNAi experiments, results in the activation of the malignancyassociated traits listed above. Therefore, the complete absence of E-cadherin in human carcinomas, occurring via nonsense mutations or locus repression/loss, is likely to result in highly metastatic tumors. Stated differently, loss of E-cadherin, in combination with certain additional oncogenic lesions, results in the acquisition of multiple functional traits that contribute to the completion of several rate-limiting steps in the invasion-metastasis cascade. The oncogenic lesions that cooperate with the loss of E-cadherin and lead to an EMT remains to be determined. It has been shown that loss of E-cadherin in normal murine mammary gland leads to induction of apoptosis in alveolar cells (Boussadia et al., 2002). In other tissues such as the skin, loss of E-cadherin results in loss of tissue integrity (Tinkle et al., 2004). Therefore, it is likely that the metastasis-promoting effects of E-cadherin loss can only become apparent after transformation or immortalization of cells that sustain such a loss. In fact, tumor formation and metastasis is observed in mice lacking E-cadherin in the mammary gland only when p53 is also deleted (Derksen et al., 2006). This is consistent with my observations that E-cadherin loss results in an EMT in 131 HMLE cells that lack both functional Rb and p53 pathways. On the other hand, preliminary experiments indicate that HMEC cells that are immortalized only through telomerase expression undergo cell death in response to loss of E-cadherin (data not shown). Taken together these observations suggest that dismantling of tumor suppressor pathways are necessary to counter the apoptotic stimuli that originate from loss of homotypic cell-cell adhesion. Loss of E-cadherin as an inducer of EMT Transcriptional repression of E-cadherin has generally been recognized to be one of the last downstream events in the EMT process. In fact, a number of EMT-inducing transcription factors (EMT-TFs) have been identified specifically because of their ability to repress E-cadherin (Comijn et al., 2001; Perez-Moreno et al., 2001). In this study, I show that loss of E-cadherin itself can be an EMT-inducing stimulus. This suggests that, in certain contexts, E-cadherin functions as an upstream controller of the EMT program. Importantly, truncation of the extracellular domain and loss of cell-cell adhesion is not sufficient to cause an EMT. This observation is in line with recent data from established breast cancer cell lines. Expression profiling of 27 cell lines with different mechanisms of E-cadherin inactivation revealed that the cell lines that harbor CDH1 mutations but retain cell-surface protein expression have an epithelial gene expression signature (Lombaerts et al., 2006). On the other hand, cell lines that have no detectable protein expression due to promoter hypermethylation have a mesenchymal phenotype with high expression of EMT-TFs such as Slug, and Sipl (Lombaerts et al., 2006). These observations together with my experimental results are consistent with the notion that constitutive loss of E- 132 cadherin expression in human tumors occurring either through promoter hypermethylation or chromosomal deletions lead to a mesenchymal phenotype. The present findings also have implications about how EMT may be induced in human tumors. Since EMT in development is induced by soluble morphogenic signals, it is hypothesized that, in human carcinomas, EMT-inducing signals do not originate in the cancer cells themselves but come from other cell types in the tumor stroma (Scheel et al., 2007). Due to restricted exposure to the soluble factors, EMT is thought to be induced transiently in carcinomas, enabling cells to leave the primary tumor and then revert back to an epithelial state in distant tissues. In fact, there is evidence for this notion in human colon carcinomas (Brabletz et al., 2001). However, my findings in this thesis indicate that loss of E-cadherin, on its own, may lead to an EMT, and therefore, suggest that epi/genetic alterations within the cancer cell genomes can lead to cell-autonomous induction of an EMT. Moreover, in cases where E-cadherin is permanently lost, transdifferentiation to a mesenchymal state would be stably maintained in secondary tumors as well (Scheel et al., 2007). Mechanism of EMT induction upon E-cadherin loss In the human mammary epithelial cell system, the retention of the cytoplasmic tail of E-cadherin prevented EMT induction. Therefore, in search for mediators of EMT induction, I initially focused on proteins known to bind this cytoplasmic domain, specifically 3-catenin. 3-catenin has previously been implicated in the induction of EMTs in various contexts (Conacci-Sorrell et al., 2003; Eger et al., 2004; Kim et al., 2002; Liebner et al., 2004; Morali et al., 2001; Yang et al., 2006b). In addition, 1-catenin has 133 been identified in an in vivo functional screen as a metastasis-promoting gene (Gumireddy et al., 2007). I observed that loss of E-cadherin resulted in the release of P-catenin from adherens junctions. Importantly, this pool of P-catenin was stabilized due to inactivation of GSK3P in shEcad cells. How E-cadherin modulates GSK3P activity remains an open question. The data presented in chapter 2 showed that GSK3P is highly phosphorylated and therefore inactive in the E-cadherin negative cells. In addition to canonical Wnts, several mitogenic signaling pathways are known to inactivate GSK30, mainly through increased Akt activity. Therefore, it is possible that loss of E-cadherin leads to a greater activation of receptor tyrosine kinases such as EGFR, which then lowers the activity of GSK3P. In fact, some studies indicate that E-cadherin inhibits ligand-dependent activation of receptor tyrosine kinases by decreasing receptor mobility and ligand binding affinity (Perrais et al., 2007; Qian et al., 2004). Therefore, an important future direction would be to gauge the activity of various mitogenic signaling pathways and their downstream effectors such as Akt in cells that have undergone an EMT. In most studies, activation and nuclear accumulation of P-catenin is correlated with induction of EMT, but no causal link has been established (Eger et al., 2004; Lu et al., 2003). I wished to directly assess the functional role of 0-catenin through loss-offunction experiments. Consequently, I inhibited p-catenin expression in E-cadherinnegative mesenchymal cells and observed that the cell biological traits associated with metastasis, such as invasiveness and anoikis resistance, were inhibited. However, gene expression profiling indicated loss of 3-catenin did not suffice to fully reverse the global EMT state caused by E-cadherin loss. In fact, this analysis revealed that while some 134 known p-catenin targets such as fibronectin and ephrin receptor B2 were regulated by loss of E-cadherin, other targets such as myc and cyclinD were not (Batlle et al., 2002; Clevers, 2006; Gradl et al., 1999; Klapholz-Brown et al., 2007). These data indicate that liberation of P-catenin due to loss of E-cadherin may not be sufficient to induce the entire spectrum of Wnt target genes. Moreover, these observations suggest that while p-catenin is essential for the maintenance of EMT, it only regulates a subset of the EMT program. Although I found 0-catenin to be necessary for several aspects of E-cadherin-lossinduced EMT, it was not sufficient to cause these phenotypes. Therefore, I concluded that additional signals beyond those conveyed by P-catenin must be important in transducing the downstream effects of E-cadherin loss. Others have reported that the adherens junction-associated proteins a- and p120- catenin may also modulate intracellular signaling pathways (Vasioukhin et al., 2001; Wildenberg et al., 2006). For example, loss of a-catenin leads to hyperactivation of the MAPK signaling, whereas p120 catenin has been shown to regulate Rho and Nf-icp pathways (Perez-Moreno et al., 2006; Vasioukhin et al., 2001; Wildenberg et al., 2006). However, I have been unable to find evidence for the activation of signaling pathways by either of these two proteins following E-cadherin loss (data not shown). To be able to understand the molecular pathways important for E-cadherin lossinduced EMT, I undertook to identify the spectrum of transcriptional changes resulting from E-cadherin loss. Expression profiling revealed that 19 transcription factors were highly induced following E-cadherin loss, all of which did not ostensibly require 0catenin for their regulation (ie. their expression levels were not changed upon P-catenin inhibition). Interestingly, Twist and ZEB-1, both previously known to cause E-cadherin 135 repression, were among the transcription factors upregulated following E-cadherin loss (Eger et al., 2005; Yang et al., 2004). Twist had previously been shown to be essential for the metastasis of mouse mammary tumor cells and to be expressed in a high proportion of human lobular breast cancers (Yang et al., 2004). More than 80% of lobular breast cancers show loss-of-heterozygosity at the CDH1 locus, and have no detectable Ecadherin expression (Strathdee, 2002). Therefore, it is tempting to speculate that epi/genetic loss of E-cadherin is the primary reason why lobular cancers express high levels of Twist. Re-expression of E-cadherin in the few available lobular breast cancer lines could address this notion. I next determined that Twist is functionally important for metastasis induced by loss of E-cadherin. In addition, Twist was essential for cancer cell invasiveness and resistance to anoikis. However, Twist inhibition did not result in a complete reversion to an epithelial phenotype. While N-cadherin levels decreased, Vimentin expression was still maintained in the absence of Twist. This suggests that Twist, on its own, regulates a portion of the EMT program and that there are yet other signals that are important for the maintenance of the EMT. Another unanswered question is how Twist expression is turned on during EMT induction. In the mouse mammary gland, Wnt overexpression can induce Twist, and murine twist promoter has putative TCF/LEF binding sites. However, these sites are not conserved in the human twist promoter (Howe et al., 2003). In addition, my experiments showed that f-catenin inhibition does not alter Twist levels in mesenchymal cells, suggesting that Twist is not downstream of P-catenin in the EMT program in human mammary epithelial cells. During development and in some carcinoma cell lines, 136 activation of NF-,ib signaling upregulates Twist expression (Jiang et al., 1991; Pham et al., 2007). In addition, enhanced EGF-STAT3 signaling has been shown to directly activate Twist transcription (Lo et al., 2007). In the future, it would be important to closely examine whether these signaling pathways are activated upon loss of E-cadherin. I found that Zebl, another EMT-TF, was highly upregulated in E-cadherinnegative cells. Zebl expression in human breast cancers is closely linked to tumor dedifferentiation (Aigner et al., 2007b). The contribution of Zebl to the induction of EMT and metastasis following E-cadherin loss is currently unknown and warrants further study. The induction of multiple EMT-TFs upon E-cadherin loss suggests that E-cadherin represses its own repressors. In fact, reporter assays indicate that E-cadherin promoter activity is repressed in E-cadherin inhibited cells (data not shown). Taken together, these observations suggest that loss of E-cadherin yields a feed-forward signaling loop whereby the EMT-induced mesenchymal state, once established, is stabilized and perhaps maintained. Thus, E-cadherin can act as a central modulator of the cell-biological phenotypes and the molecular factors that govern metastatic dissemination of carcinomas. EMT and Chemoresistance During the course of experiments described in Chapter 2, I observed that cells that have undergone an EMT become resistant to apoptosis induced by loss of anchorage. Subsequently, I wished to determine whether EMT conferred resistance to other forms of apoptotic stimuli as well. Given the link between metastatic recurrence and emergence of chemotherapy resistance in human tumors, I specifically wanted to know whether passage through an EMT resulted in resistance to chemotherapy-induced cytotoxicity. 137 I observed that human mammary carcinoma cells induced to undergo an EMT by inhibition of E-cadherin were significantly resistant to cell death following treatment with chemotherapeutic drugs. Importantly, the utilized drugs act via diverse mechanisms, such as intercalation into DNA (doxorubicin) or destabilization of microtubules (paclitaxel), suggesting that EMT confers a multi-drug-resistant (MDR)-like phenotype. The observed resistance was also independent of transformation and seemed to be a direct consequence of the mesenchymal differentiation state. Although there is little information about how the mesenchymal state confers chemoresistance, accumulating evidence suggests that the actions of EMT-TFs are crucial to block cell death. In cases where EMT-TFs protect against genotoxic stress, such as those elicited by gamma irradiation and drugs like doxorubicin, they are thought counteract p53-mediated induction of pro-apoptotic genes (Inoue et al., 2002; Kajita et al., 2004). For example, in irradiated hematopoetic cells, Slug antagonizes p53-mediated transcription of PUMA, a pro-apoptotic BH3-only protein, through direct binding to the puma promoter (Wu et al., 2005). In addition, cells that pass through an EMT have slower cell cycles, potentially allowing cells to repair any sustained damage. In fact, Snail is known to repress Cyclin D2, while other EMT-TFs such as Zeb-1 can induce p21WAF1/CIP1, thereby leading to slower progression through the cell cycle (Liu et al., 2008; Vega et al., 2004). In the cell system utilized throughout this thesis, the p53 pathway is nonfunctional due to expression of the Large-T antigen in the human mammary epithelial cells. Therefore, the observed resistance to cell death is likely to occur via p53independent mechanisms. In fact, there is some evidence that EMT-TFs can act 138 independently of p53 to block programmed cell death. For example, Twist has recently been shown to mediate resistance to paclitaxel through activation of Akt signaling (Cheng et al., 2007; Zhang et al., 2007). In addition, Twist can interfere with downstream apoptotic machinery through modulation of Bcl-2 phosphorylation (Pham et al., 2007). In the future it would be important to investigate whether such mechanisms are operative in E-cadherin-inhibited mesenchymal cells. Since mesenchymal cells have an MDR-like phenotype, it would also be interesting to examine whether cells that have undergone an EMT express higher level of ABC transporters that are known mediators of MDR. The observation that EMT confers chemoresistance has important implications for cancer therapy. First, it provides a mechanistic link between metastatic dissemination and the development of secondary tumors that are chemoresistant. Second, it suggests that chemotherapy treatment may actually lead to the preferential survival and expansion of cancer cells that have a mesenchymal and more invasive phenotype. In fact, there is growing evidence that long term exposure of cancer cell lines to cytotoxic agents promotes the outgrowth of variants with mesenchymal properties (Kajiyama et al., 2007; Shah et al., 2007; Wang et al., 2004; Yang et al., 2006a). Third, it suggests that, in cases where chemotherapy resistance is due to passage through an EMT, agents that can interfere with the EMT program may re-sensitize tumor cells to treatment. In support of this notion, a recent study reported that short interfering RNAs directed against Twist can sensitize lung carcinoma cells to cisplatin (Zhuo et al., 2008). 139 Chemical screen identifies compounds targeting cells in the mesenchymal state Cells that have undergone an EMT represent a double challenge to the prevention and management of tumor progression. Not only are they more invasive and likely to form secondary tumors, they are also resistant to a variety current therapeutics. Therefore, it is of vital importance to find therapies that effectively target such cells. Since Ecadherin-loss resulted in the induction of an EMT and mimicked the chemoresistant phenotype observed in other systems, I postulated that the stable cell lines created during the initial part of this thesis work could be used as tools for high throughput screening of chemical libraries. Experiments in this portion of the thesis were carried out in collaboration with Piyush Gupta from the Lander laboratory. The use of the abovementioned cell lines afforded two main advantages. First, chemoresistance and EMT induction was achieved through a single manipulation (Ecadherin inhibition), thereby creating almost isogenic cell lines that differ only by virtue of being either in the epithelial or mesenchymal differentiation state. Therefore, we could perform the chemical library screen essentially twice using cells that have undergone an EMT and their epithelial counterparts. This approach allowed us to identify those compounds that exhibited specific toxicity against cells in the mesenchymal differentiation state. Second, EMT could be induced in immortalized cells, thereby allowing us to find EMT-specific rather than transformation-specific agents. However, the use of transformed cells should be considered as an alternative approach in the future, because the presence of different oncogenes in cells that have undergone an EMT may yield additional compounds. 140 We have identified one compound, salinomycin, as being selectively toxic to carcinoma cells that have undergone an EMT. We also observed that among established breast cancer cell lines salinomycin sensitivity correlates with the prevalence of cells with CD44high/CD241ow antigenic profile. These markers are reported to enrich for breast CSCs (Al-Hajj et al., 2003). Therefore, it is critical to test whether salinomycin treatment affects CSC properties, such as tumor formation at limiting dilution. Given our laboratory's observations that passage through an EMT leads to a CSC-like state, it is tempting to speculate that approaches described in this thesis may be utilized to find compounds targeting CSC populations. We do not know at this point by which mechanism salinomycin exerts its cytotoxic effect. Salinomycin is a polyether antibiotic that belongs to a class of compounds called ionophores. Ionophores are hydrophobic molecules that selectively bind to a given metal ion and increase its cell permeability by shielding the charge of the ion. This enables the ions to penetrate the hydrophobic interior of the lipid bilayer. Salinomycin specifically binds potassium ions (Mitani et al., 1975). Early reports indicated that salinomycin causes rapid release of K' from mitochondria, loss of mitochondrial membrane potential, and eventual inhibition of oxidative phosphorylation (Mitani et al., 1976). Other potassium ionophores, such as valinomycin and nigericin, also interfere with mitochondrial function (Arslan et al., 1981; Daniele et al., 1978; Furlong et al., 1998). It is interesting to note that nigericin, like salinomycin, exhibited differential toxicity to HMLE-shEcad cells and was an initial hit in the screen. It would be important to know whether other potassium-selective ionophores, such as valinomycin, 141 would also be selectively toxic (valinomycin was not in the chemical libraries we screened). Potential mechanisms of action and limitations of salinomycin Given the known effects of potassium ionophores on mitochondria and oxidative respiration, it is possible that their preferential toxicity to mesenchymal cells may due to fundamental differences in the mode of ATP production pre- and post-EMT. For example, if the mesenchymal cells primarily rely on oxidative phosphorylation by mitochondria for ATP production, they would be more sensitive to salinomycin compared to epithelial cells that mainly employ glycolysis. A prediction of this hypothesis is that mesenchymal cells will be more sensitive to inhibition of aerobic respiration by low oxygen tension, whereas epithelial cells would be more susceptible to inhibition of glycolysis. Preliminary experiments did not reveal significant differences between epithelial cells and their counterparts that have undergone an EMT with respect to cell viability under conditions of hypoxia or inhibition of glycolysis(data not shown). However, further examination is needed to establish whether differences in the mode of ATP production are the basis of salinomycin selectivity. Therefore, the reason why potassium ionophores may preferentially inhibit the viability of cells in the mesenchymal state remains to be determined. Another unresolved issue is the mechanism by which salinomycin inhibits cell viability. Our preliminary experiments indicate that salinomycin treatment causes cell death rather than cell cycle arrest. Whether the observed cell death upon salinomycin treatment is due to the activation of the classical apoptotic machinery remains to be 142 determined. While some studies indicate that treatment with potassium ionophores causes caspase activation, others have found that bcl-2 overexpression or caspase inhibitors cannot rescue ionophore-induced cell death (Furlong et al., 1997; Watanabe et al., 1998). It is possible that interference with mitochondrial function and membrane permeability activate multiple cell death mechanisms. In fact, recent chemical screens identified a class of compounds that act on the mitochondria and cause cell death via a novel iron-dependent oxidative mechanism (Yagoda et al., 2007; Yang and Stockwell, 2008). In the future it would be important to determine what cell death mechanisms are activated upon salinomycin treatment in cells that have undergone an EMT. Although in vitro treatment of salinomycin is effective against cancer cells that have undergone an EMT, it is not possible to use this compound in vivo. Micromolar concentrations would have to be achieved in order to be effective against human breast cancer cells implanted into immunocompromised mice. At such concentrations, salinomycin is known to be highly toxic (Lagas et al., 2008). Therefore, measuring the efficacy of salinomycin against cells that have undergone an EMT in an in vivo xenograft model is not feasible and remains to be a limitation of this compound. Final perspective All in all, this study demonstrates a practical approach to the identification of agents exhibiting specific toxicity for cancer cells that have undergone an EMT. Moreover, it indicates that even though passage through an EMT is associated with resistance to cell death, it is possible to find compounds that selectively kill cells in the mesenchymal differentiation state. In fact, a recent study demonstrated that basal-type 143 and mesenchymal breast cancer cell lines are specifically sensitive to a small molecule inhibitor of src and abl kinases (Finn et al., 2007). These findings suggest that the induction of EMT may bring unique vulnerabilities to carcinoma cells. These vulnerabilities can be exploited through high throughput screening approaches such as those described here and form the basis for future therapeutics. 144 References Aigner, K., Dampier, B., Descovich, L., Mikula, M., Sultan, A., Schreiber, M., Mikulits, W., Brabletz, T., Strand, D., Obrist, P., Sommergruber, W., Schweifer, N., Wernitznig, A., Beug, H., Foisner, R. and Eger, A. (2007a) The transcription factor ZEB1 (deltaEF1) promotes tumour cell dedifferentiation by repressing master regulators of epithelial polarity. Oncogene, 26, 6979-6988. Aigner, K., Dampier, B., Descovich, L., Mikula, M., Sultan, A., Schreiber, M., Mikulits, W., Brabletz, T., Strand, D., Obrist, P., Sommergruber, W., Schweifer, N., Wernitznig, A., Beug, H., Foisner, R. and Eger, A. (2007b) The transcription factor ZEB 1 (deltaEFl) promotes tumour cell dedifferentiation by repressing master regulators of epithelial polarity. Oncogene. Al-Hajj, M., Wicha, M.S., Benito-Hernandez, A., Morrison, S.J. and Clarke, M.F. (2003) Prospective identification of tumorigenic breast cancer cells. ProcNatl Acad Sci USA, 100, 3983-3988. Alonso, S.R., Tracey, L., Ortiz, P., Perez-Gomez, B., Palacios, J., Pollan, M., Linares, J., Serrano, S., Saez-Castillo, A.I., Sanchez, L., Pajares, R., Sanchez-Aguilera, A., Artiga, M.J., Piris, M.A. and Rodriguez-Peralto, J.L. (2007) A high-throughput study in melanoma identifies epithelial-mesenchymal transition as a major determinant of metastasis. CancerRes, 67, 3450-3460. Arslan, P., Montecucco, C., Celi, D. and Pozzan, T. (1981) Effect of monovalent cation ionophores on lymphocyte cellular metabolism. Biochim Biophys Acta, 643, 177181. Attwell, S., Roskelley, C. and Dedhar, S. (2000) The integrin-linked kinase (ILK) suppresses anoikis. Oncogene, 19, 3811-3815. Avizienyte, E., Wyke, A.W., Jones, R.J., McLean, G.W., Westhoff, M.A., Brunton, V.G. and Frame, M.C. (2002) Src-induced de-regulation of E-cadherin in colon cancer cells requires integrin signalling. Nat Cell Biol, 4, 632-638. Bao, S., Wu, Q., McLendon, R.E., Hao, Y., Shi, Q., Hjelmeland, A.B., Dewhirst, M.W., Bigner, D.D. and Rich, J.N. (2006) Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature, 444, 756-760. Barrallo-Gimeno, A. and Nieto, M.A. (2005) The Snail genes as inducers of cell movement and survival: implications in development and cancer. Development, 132, 3151-3161. Batlle, E., Henderson, J.T., Beghtel, H., van den Born, M.M., Sancho, E., Huls, G., Meeldijk, J., Robertson, J., van de Wetering, M., Pawson, T. and Clevers, H. (2002) Beta-catenin and TCF mediate cell positioning in the intestinal epithelium by controlling the expression of EphB/ephrinB. Cell, 111, 251-263. Batlle, E., Sancho, E., Franci, C., Dominguez, D., Monfar, M., Baulida, J. and Garcia De Herreros, A. (2000) The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nat Cell Biol, 2, 84-89. Becker, K.F., Atkinson, M.J., Reich, U., Becker, I., Nekarda, H., Siewert, J.R. and Hofler, H. (1994) E-cadherin gene mutations provide clues to diffuse type gastric carcinomas. CancerRes, 54, 3845-3852. Behrens, J., Birchmeier, W., Goodman, S.L. and Imhof, B.A. (1985) Dissociation of Madin-Darby canine kidney epithelial cells by the monoclonal antibody anti-arc- 145 1: mechanistic aspects and identification of the antigen as a component related to uvomorulin. J Cell Biol, 101, 1307-1315. Berezovskaya, O., Schimmer, A.D., Glinskii, A.B., Pinilla, C., Hoffman, R.M., Reed, J.C. and Glinsky, G.V. (2005) Increased expression of apoptosis inhibitor protein XIAP contributes to anoikis resistance of circulating human prostate cancer metastasis precursor cells. CancerRes, 65, 2378-2386. Berx, G., Becker, K.F., Hofler, H. and van Roy, F. (1998) Mutations of the human Ecadherin (CDH1) gene. Hum Mutat, 12, 226-237. Berx, G., Cleton-Jansen, A.M., Nollet, F., de Leeuw, W.J., van de Vijver, M., Cornelisse, C. and van Roy, F. (1995) E-cadherin is a tumour/invasion suppressor gene mutated in human lobular breast cancers. Embo J, 14, 6107-6115. Berx, G., Cleton-Jansen, A.M., Strumane, K., de Leeuw, W.J., Nollet, F., van Roy, F. and Cornelisse, C. (1996) E-cadherin is inactivated in a majority of invasive human lobular breast cancers by truncation mutations throughout its extracellular domain. Oncogene, 13, 1919-1925. Berx, G. and Van Roy, F. (2001) The E-cadherin/catenin complex: an important gatekeeper in breast cancer tumorigenesis and malignant progression. Breast CancerRes, 3, 289-293. Bhowmick, N.A., Ghiassi, M., Bakin, A., Aakre, M., Lundquist, C.A., Engel, M.E., Arteaga, C.L. and Moses, H.L. (2001) Transforming growth factor-betal mediates epithelial to mesenchymal transdifferentiation through a RhoA-dependent mechanism. Mol Biol Cell, 12, 27-36. Blanco, M.J., Moreno-Bueno, G., Sarrio, D., Locascio, A., Cano, A., Palacios, J. and Nieto, M.A. (2002) Correlation of Snail expression with histological grade and lymph node status in breast carcinomas. Oncogene, 21, 3241-3246. Blaschuk, O.W., Sullivan, R., David, S. and Pouliot, Y. (1990) Identification of a cadherin cell adhesion recognition sequence. Dev Biol, 139, 227-229. Bolos, V., Peinado, H., Perez-Moreno, M.A., Fraga, M.F., Esteller, M. and Cano, A. (2003) The transcription factor Slug represses E-cadherin expression and induces epithelial to mesenchymal transitions: a comparison with Snail and E47 repressors. J Cell Sci, 116, 499-511. Boussadia, O., Kutsch, S., Hierholzer, A., Delmas, V. and Kemler, R. (2002) E-cadherin is a survival factor for the lactating mouse mammary gland. Mech Dev, 115, 5362. Boyer, B., Tucker, G.C., Valles, A.M., Franke, W.W. and Thiery, J.P. (1989a) Rearrangements of desmosomal and cytoskeletal proteins during the transition from epithelial to fibroblastoid organization in cultured rat bladder carcinoma cells. J Cell Biol, 109, 1495-1509. Boyer, B., Tucker, G.C., Valles, A.M., Gavrilovic, J. and Thiery, J.P. (1989b) Reversible transition towards a fibroblastic phenotype in a rat carcinoma cell line. Int J CancerSuppl, 4, 69-75. Brabletz, T., Jung, A., Reu, S., Porzner, M., Hlubek, F., Kunz-Schughart, L.A., Knuechel, R. and Kirchner, T. (2001) Variable beta-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. ProcNatl Acad Sci US A, 98, 10356-10361. 146 Braun, S., Pantel, K., Muller, P., Janni, W., Hepp, F., Kentenich, C.R., Gastroph, S., Wischnik, A., Dimpfl, T., Kindermann, G., Riethmuller, G. and Schlimok, G. (2000) Cytokeratin-positive cells in the bone marrow and survival of patients with stage I, II, or III breast cancer. N Engl J Med, 342, 525-533. Brembeck, F.H., Schwarz-Romond, T., Bakkers, J., Wilhelm, S., Hammerschmidt, M. and Birchmeier, W. (2004) Essential role of BCL9-2 in the switch between betacatenin's adhesive and transcriptional functions. Genes Dev, 18, 2225-2230. Bryant, D.M. and Stow, J.L. (2004) The ins and outs of E-cadherin trafficking. Trends Cell Biol, 14, 427-434. Buck, E., Eyzaguirre, A., Barr, S., Thompson, S., Sennello, R., Young, D., Iwata, K.K., Gibson, N.W., Cagnoni, P. and Haley, J.D. (2007) Loss of homotypic cell adhesion by epithelial-mesenchymal transition or mutation limits sensitivity to epidermal growth factor receptor inhibition. Mol CancerTher, 6, 532-541. Cano, A., Perez-Moreno, M.A., Rodrigo, I., Locascio, A., Blanco, M.J., del Barrio, M.G., Portillo, F. and Nieto, M.A. (2000) The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nat Cell Biol, 2, 76-83. Cavallaro, U. and Christofori, G. (2004) Cell adhesion and signalling by cadherins and Ig-CAMs in cancer. Nat Rev Cancer,4, 118-132. Chabner, B.A. and Roberts, T.G., Jr. (2005) Timeline: Chemotherapy and the war on cancer. Nat Rev Cancer, 5, 65-72. Chambers, A.F., Groom, A.C. and MacDonald, I.C. (2002) Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer,2, 563-572. Chen, Y.T., Stewart, D.B. and Nelson, W.J. (1999) Coupling assembly of the Ecadherin/beta-catenin complex to efficient endoplasmic reticulum exit and basallateral membrane targeting of E-cadherin in polarized MDCK cells. J Cell Biol, 144, 687-699. Cheng, G.Z., Chan, J., Wang, Q., Zhang, W., Sun, C.D. and Wang, L.H. (2007) Twist transcriptionally up-regulates AKT2 in breast cancer cells leading to increased migration, invasion, and resistance to paclitaxel. CancerRes, 67, 1979-1987. Cheng, G.Z., Zhang, W., Sun, M., Wang, Q., Coppola, D., Mansour, M., Xu, L., Costanzo, C., Cheng, J.Q. and Wang, L.H. (2008) Twist is transcriptionally induced by activation of STAT3 and mediates STAT3 oncogenic function. J Biol Chem. Chung, C.H., Parker, J.S., Ely, K., Carter, J., Yi, Y., Murphy, B.A., Ang, K.K., ElNaggar, A.K., Zanation, A.M., Cmelak, A.J., Levy, S., Slebos, R.J. and Yarbrough, W.G. (2006) Gene expression profiles identify epithelial-tomesenchymal transition and activation of nuclear factor-kappaB signaling as characteristics of a high-risk head and neck squamous cell carcinoma. CancerRes, 66, 8210-8218. Clare, S.E., Nakhlis, F. and Panetta, J.C. (2000) Molecular biology of breast cancer metastasis. The use of mathematical models to determine relapse and to predict response to chemotherapy in breast cancer. Breast CancerRes, 2, 430-435. Clark, E.A., Golub, T.R., Lander, E.S. and Hynes, R.O. (2000) Genomic analysis of metastasis reveals an essential role for RhoC. Nature, 406, 532-535. 147 Clevers, H. (2006) Wnt/beta-catenin signaling in development and disease. Cell, 127, 469-480. Comijn, J., Berx, G., Vermassen, P., Verschueren, K., van Grunsven, L., Bruyneel, E., Mareel, M., Huylebroeck, D. and van Roy, F. (2001) The two-handed E box binding zinc finger protein SIP1 downregulates E-cadherin and induces invasion. Mol Cell, 7, 1267-1278. Conacci-Sorrell, M., Simcha, I., Ben-Yedidia, T., Blechman, J., Savagner, P. and BenZe'ev, A. (2003) Autoregulation of E-cadherin expression by cadherin-cadherin interactions: the roles of beta-catenin signaling, Slug, and MAPK. J Cell Biol, 163, 847-857. Cross, D.A., Alessi, D.R., Cohen, P., Andjelkovich, M. and Hemmings, B.A. (1995) Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature, 378, 785-789. Dahl, U., Sjodin, A. and Semb, H. (1996) Cadherins regulate aggregation of pancreatic beta-cells in vivo. Development, 122, 2895-2902. Daniele, R.P., Holian, S.K. and Nowell, P.C. (1978) A potassium ionophore (Nigericin) inhibits stimulation of human lymphocytes by mitogens. J Exp Med, 147, 571-581. De Langhe, S.P., Sala, F.G., Del Moral, P.M., Fairbanks, T.J., Yamada, K.M., Warburton, D., Bums, R.C. and Bellusci, S. (2005) Dickkopf-1 (DKK1) reveals that fibronectin is a major target of Wnt signaling in branching morphogenesis of the mouse embryonic lung. Dev Biol, 277, 316-331. Dean, M., Fojo, T. and Bates, S. (2005) Tumour stem cells and drug resistance. Nat Rev Cancer,5, 275-284. Dennis, G., Jr., Sherman, B.T., Hosack, D.A., Yang, J., Gao, W., Lane, H.C. and Lempicki, R.A. (2003) DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol, 4, P3. Derksen, P.W., Liu, X., Saridin, F., van der Gulden, H., Zevenhoven, J., Evers, B., van Beijnum, J.R., Griffioen, A.W., Vink, J., Krimpenfort, P., Peterse, J.L., Cardiff, R.D., Berns, A. and Jonkers, J. (2006) Somatic inactivation of E-cadherin and p53 in mice leads to metastatic lobular mammary carcinoma through induction of anoikis resistance and angiogenesis. CancerCell, 10, 437-449. Diehn, M. and Clarke, M.F. (2006) Cancer stem cells and radiotherapy: new insights into tumor radioresistance. J Natl Cancer Inst, 98, 1755-1757. Dontu, G., Abdallah, W.M., Foley, J.M., Jackson, K.W., Clarke, M.F., Kawamura, M.J. and Wicha, M.S. (2003) In vitro propagation and transcriptional profiling of human mammary stem/progenitor cells. Genes Dev, 17, 1253-1270. Dontu, G. and Wicha, M.S. (2005) Survival of mammary stem cells in suspension culture: implications for stem cell biology and neoplasia. J Mammary Gland Biol Neoplasia, 10, 75-86. Douma, S., Van Laar, T., Zevenhoven, J., Meuwissen, R., Van Garderen, E. and Peeper, D.S. (2004) Suppression of anoikis and induction of metastasis by the neurotrophic receptor TrkB. Nature, 430, 1034-1039. Egeblad, M. and Werb, Z. (2002) New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer,2, 161-174. Eger, A., Aigner, K., Sonderegger, S., Dampier, B., Oehler, S., Schreiber, M., Berx, G., Cano, A., Beug, H. and Foisner, R. (2005) DeltaEF1 is a transcriptional repressor 148 of E-cadherin and regulates epithelial plasticity in breast cancer cells. Oncogene, 24, 2375-2385. Eger, A., Stockinger, A., Park, J., Langkopf, E., Mikula, M., Gotzmann, J., Mikulits, W., Beug, H. and Foisner, R. (2004) beta-Catenin and TGFbeta signalling cooperate to maintain a mesenchymal phenotype after FosER-induced epithelial to mesenchymal transition. Oncogene, 23, 2672-2680. Elenbaas, B., Spirio, L., Koerner, F., Fleming, M.D., Zimonjic, D.B., Donaher, J.L., Popescu, N.C., Hahn, W.C. and Weinberg, R.A. (2001) Human breast cancer cells generated by oncogenic transformation of primary mammary epithelial cells. Genes Dev, 15, 50-65. Elloul, S., Elstrand, M.B., Nesland, J.M., Trope, C.G., Kvalheim, G., Goldberg, I., Reich, R. and Davidson, B. (2005) Snail, Slug, and Smad-interacting protein 1 as novel parameters of disease aggressiveness in metastatic ovarian and breast carcinoma. Cancer,103, 1631-1643. Fidler, I.J. and Kripke, M.L. (1977) Metastasis results from preexisting variant cells within a malignant tumor. Science, 197, 893-895. Finn, R.S., Dering, J., Ginther, C., Wilson, C.A., Glaspy, P., Tchekmedyian, N. and Slamon, D.J. (2007) Dasatinib, an orally active small molecule inhibitor of both the src and abl kinases, selectively inhibits growth of basal-type/"triple-negative" breast cancer cell lines growing in vitro. Breast CancerRes Treat, 105, 319-326. Frank, N.Y., Margaryan, A., Huang, Y., Schatton, T., Waaga-Gasser, A.M., Gasser, M., Sayegh, M.H., Sadee, W. and Frank, M.H. (2005) ABCB5-mediated doxorubicin transport and chemoresistance in human malignant melanoma. CancerRes, 65, 4320-4333. Friedl, P. and Wolf, K. (2003) Tumour-cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer,3, 362-374. Frixen, U.H., Behrens, J., Sachs, M., Eberle, G., Voss, B., Warda, A., Lochner, D. and Birchmeier, W. (1991) E-cadherin-mediated cell-cell adhesion prevents invasiveness of human carcinoma cells. J Cell Biol, 113, 173-185. Fujita, Y., Krause, G., Scheffner, M., Zechner, D., Leddy, H.E., Behrens, J., Sommer, T. and Birchmeier, W. (2002) Hakai, a c-Cbl-like protein, ubiquitinates and induces endocytosis of the E-cadherin complex. Nat Cell Biol, 4, 222-231. Furlong, E.E., Andersen, E.C., Null, B., White, K.P. and Scott, M.P. (2001) Patterns of gene expression during Drosophila mesoderm development. Science, 293, 16291633. Furlong, I.J., Ascaso, R., Lopez Rivas, A. and Collins, M.K. (1997) Intracellular acidification induces apoptosis by stimulating ICE-like protease activity. J Cell Sci, 110 ( Pt 5), 653-661. Furlong, I.J., Lopez Mediavilla, C., Ascaso, R., Lopez Rivas, A. and Collins, M.K. (1998) Induction of apoptosis by valinomycin: mitochondrial permeability transition causes intracellular acidification. Cell Death Differ, 5, 214-221. Giannakakou, P., Sackett, D.L., Kang, Y.K., Zhan, Z., Buters, J.T., Fojo, T. and Poruchynsky, M.S. (1997) Paclitaxel-resistant human ovarian cancer cells have mutant beta-tubulins that exhibit impaired paclitaxel-driven polymerization. J Biol Chem, 272, 17118-17125. 149 Goswami, S., Wang, W., Wyckoff, J.B. and Condeelis, J.S. (2004) Breast cancer cells isolated by chemotaxis from primary tumors show increased survival and resistance to chemotherapy. CancerRes, 64, 7664-7667. Gottardi, C.J. and Gumbiner, B.M. (2004) Distinct molecular forms of beta-catenin are targeted to adhesive or transcriptional complexes. J Cell Biol, 167, 339-349. Gottardi, C.J., Wong, E. and Gumbiner, B.M. (2001) E-cadherin suppresses cellular transformation by inhibiting beta-catenin signaling in an adhesion-independent manner. J Cell Biol, 153, 1049-1060. Gradl, D., Kuhl, M. and Wedlich, D. (1999) The Wnt/Wg signal transducer beta-catenin controls fibronectin expression. Mol Cell Biol, 19, 5576-5587. Graff, J.R., Herman, J.G., Lapidus, R.G., Chopra, H., Xu, R., Jarrard, D.F., Isaacs, W.B., Pitha, P.M., Davidson, N.E. and Baylin, S.B. (1995) E-cadherin expression is silenced by DNA hypermethylation in human breast and prostate carcinomas. CancerRes, 55, 5195-5199. Grooteclaes, M.L. and Frisch, S.M. (2000) Evidence for a function of CtBP in epithelial gene regulation and anoikis. Oncogene, 19, 3823-3828. Gros, P., Ben Neriah, Y.B., Croop, J.M. and Housman, D.E. (1986) Isolation and expression of a complementary DNA that confers multidrug resistance. Nature, 323, 728-731. Grunert, S., Jechlinger, M. and Beug, H. (2003) Diverse cellular and molecular mechanisms contribute to epithelial plasticity and metastasis. Nat Rev Mol Cell Biol, 4, 657-665. Guilford, P., Hopkins, J., Harraway, J., McLeod, M., McLeod, N., Harawira, P., Taite, H., Scoular, R., Miller, A. and Reeve, A.E. (1998) E-cadherin germline mutations in familial gastric cancer. Nature, 392, 402-405. Gumbiner, B., Stevenson, B. and Grimaldi, A. (1988) The role of the cell adhesion molecule uvomorulin in the formation and maintenance of the epithelial junctional complex. J Cell Biol, 107, 1575-1587. Gumireddy, K., Sun, F., Klein-Szanto, A.J., Gibbins, J.M., Gimotty, P.A., Saunders, A.J., Schultz, P.G. and Huang, Q. (2007) In vivo selection for metastasis promoting genes in the mouse. ProcNatl Acad Sci US A, 104, 6696-6701. Gupta, G.P. and Massague, J. (2006) Cancer metastasis: building a framework. Cell, 127, 679-695. Hadeball, B., Borchers, A. and Wedlich, D. (1998) Xenopus cadherin-11 (Xcadherin-11) expression requires the Wg/Wnt signal. Mech Dev, 72, 101-113. Halbleib, J.M. and Nelson, W.J. (2006) Cadherins in development: cell adhesion, sorting, and tissue morphogenesis. Genes Dev, 20, 3199-3214. Hanahan, D. and Weinberg, R.A. (2000) The hallmarks of cancer. Cell, 100, 57-70. Handschuh, G., Candidus, S., Luber, B., Reich, U., Schott, C., Oswald, S., Becke, H., Hutzler, P., Birchmeier, W., Hofler, H. and Becker, K.F. (1999) Tumourassociated E-cadherin mutations alter cellular morphology, decrease cellular adhesion and increase cellular motility. Oncogene, 18, 4301-4312. Hartwell, K.A., Muir, B., Reinhardt, F., Carpenter, A.E., Sgroi, D.C. and Weinberg, R.A. (2006) The Spemann organizer gene, Goosecoid, promotes tumor metastasis. Proc Natl Acad Sci US A, 103, 18969-18974. 150 Hirohashi, S. (1998) Inactivation of the E-cadherin-mediated cell adhesion system in human cancers. Am J Pathol, 153, 333-339. Howe, L.R., Watanabe, O., Leonard, J. and Brown, A.M. (2003) Twist is up-regulated in response to Wntl and inhibits mouse mammary cell differentiation. CancerRes, 63, 1906-1913. Huber, A.H. and Weis, W.I. (2001) The structure of the beta-catenin/E-cadherin complex and the molecular basis of diverse ligand recognition by beta-catenin. Cell, 105, 391-402. Huber, M.A., Kraut, N. and Beug, H. (2005) Molecular requirements for epithelialmesenchymal transition during tumor progression. Curr Opin Cell Biol, 17, 548558. Hyafil, F., Babinet, C. and Jacob, F. (1981) Cell-cell interactions in early embryogenesis: a molecular approach to the role of calcium. Cell, 26, 447-454. Ince, T.A., Richardson, A.L., Bell, G.W., Saitoh, M., Godar, S., Karnoub, A.E., Iglehart, J.D. and Weinberg, R.A. (2007) Transformation of different human breast epithelial cell types leads to distinct tumor phenotypes. CancerCell, 12, 160-170. Ino, Y., Gotoh, M., Sakamoto, M., Tsukagoshi, K. and Hirohashi, S. (2002) Dysadherin, a cancer-associated cell membrane glycoprotein, down-regulates E-cadherin and promotes metastasis. Proc Natl Acad Sci US A, 99, 365-370. Inoue, A., Seidel, M.G., Wu, W., Kamizono, S., Ferrando, A.A., Bronson, R.T., Iwasaki, H., Akashi, K., Morimoto, A., Hitzler, J.K., Pestina, T.I., Jackson, C.W., Tanaka, R., Chong, M.J., McKinnon, P.J., Inukai, T., Grosveld, G.C. and Look, A.T. (2002) Slug, a highly conserved zinc finger transcriptional repressor, protects hematopoietic progenitor cells from radiation-induced apoptosis in vivo. Cancer Cell, 2, 279-288. Inukai, T., Inoue, A., Kurosawa, H., Goi, K., Shinjyo, T., Ozawa, K., Mao, M., Inaba, T. and Look, A.T. (1999) SLUG, a ces-1-related zinc finger transcription factor gene with antiapoptotic activity, is a downstream target of the E2A-HLF oncoprotein. Mol Cell, 4, 343-352. Jechlinger, M., Grunert, S., Tamir, I.H., Janda, E., Ludemann, S., Waerner, T., Seither, P., Weith, A., Beug, H. and Kraut, N. (2003) Expression profiling of epithelial plasticity in tumor progression. Oncogene, 22, 7155-7169. Jemal, A., Siegel, R., Ward, E., Hao, Y., Xu, J., Murray, T. and Thun, M.J. (2008) Cancer statistics, 2008. CA Cancer J Clin, 58, 71-96. Jiang, J., Kosman, D., Ip, Y.T. and Levine, M. (1991) The dorsal morphogen gradient regulates the mesoderm determinant twist in early Drosophila embryos. Genes Dev, 5, 1881-1891. Jorda, M., Olmeda, D., Vinyals, A., Valero, E., Cubillo, E., Llorens, A., Cano, A. and Fabra, A. (2005) Upregulation of MMP-9 in MDCK epithelial cell line in response to expression of the Snail transcription factor. J Cell Sci, 118, 3371-3385. Julien, S., Puig, I., Caretti, E., Bonaventure, J., Nelles, L., van Roy, F., Dargemont, C., de Herreros, A.G., Bellacosa, A. and Larue, L. (2007) Activation of NF-kappaB by Akt upregulates Snail expression and induces epithelium mesenchyme transition. Oncogene, 26, 7445-7456. 151 Kajita, M., McClinic, K.N. and Wade, P.A. (2004) Aberrant expression of the transcription factors snail and slug alters the response to genotoxic stress. Mol Cell Biol, 24, 7559-7566. Kajiyama, H., Shibata, K., Terauchi, M., Yamashita, M., Ino, K., Nawa, A. and Kikkawa, F. (2007) Chemoresistance to paclitaxel induces epithelial-mesenchymal transition and enhances metastatic potential for epithelial ovarian carcinoma cells. Int J Oncol, 31, 277-283. Kamei, T., Matozaki, T., Sakisaka, T., Kodama, A., Yokoyama, S., Peng, Y.F., Nakano, K., Takaishi, K. and Takai, Y. (1999) Coendocytosis of cadherin and c-Met coupled to disruption of cell-cell adhesion in MDCK cells--regulation by Rho, Rac and Rab small G proteins. Oncogene, 18, 6776-6784. Kanai, Y., Oda, T., Tsuda, H., Ochiai, A. and Hirohashi, S. (1994) Point mutation of the E-cadherin gene in invasive lobular carcinoma of the breast. Jpn J CancerRes, 85, 1035-1039. Kim, C.F., Jackson, E.L., Woolfenden, A.E., Lawrence, S., Babar, I., Vogel, S., Crowley, D., Bronson, R.T. and Jacks, T. (2005) Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell, 121, 823-835. Kim, K., Lu, Z. and Hay, E.D. (2002) Direct evidence for a role of beta-catenin/LEF-1 signaling pathway in induction of EMT. Cell Biol Int, 26, 463-476. Klapholz-Brown, Z., Walmsley, G.G., Nusse, Y.M., Nusse, R. and Brown, P.O. (2007) Transcriptional program induced by wnt protein in human fibroblasts suggests mechanisms for cell cooperativity in defining tissue microenvironments. PLoS ONE, 2, e945. Kobayashi, S., Boggon, T.J., Dayaram, T., Janne, P.A., Kocher, O., Meyerson, M., Johnson, B.E., Eck, M.J., Tenen, D.G. and Halmos, B. (2005) EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N Engl J Med, 352, 786792. Kolligs, F.T., Hu, G., Dang, C.V. and Fearon, E.R. (1999) Neoplastic transformation of RK3E by mutant beta-catenin requires deregulation of Tcf/Lef transcription but not activation of c-myc expression. Mol Cell Biol, 19, 5696-5706. Kuphal, F. and Behrens, J. (2006) E-cadherin modulates Wnt-dependent transcription in colorectal cancer cells but does not alter Wnt-independent gene expression in fibroblasts. Exp Cell Res, 312, 457-467. Lagas, J.S., Sparidans, R.W., van Waterschoot, R.A., Wagenaar, E., Beijnen, J.H. and Schinkel, A.H. (2008) P-glycoprotein limits oral availability, brain penetration, and toxicity of an anionic drug, the antibiotic salinomycin. Antimicrob Agents Chemother, 52, 1034-1039. Lapidot, T., Sirard, C., Vormoor, J., Murdoch, B., Hoang, T., Caceres-Cortes, J., Minden, M., Paterson, B., Caligiuri, M.A. and Dick, J.E. (1994) A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature, 367, 645648. Larue, L., Ohsugi, M., Hirchenhain, J. and Kemler, R. (1994) E-cadherin null mutant embryos fail to form a trophectoderm epithelium. ProcNatl Acad Sci US A, 91, 8263-8267. 152 Li, C., Heidt, D.G., Dalerba, P., Burant, C.F., Zhang, L., Adsay, V., Wicha, M., Clarke, M.F. and Simeone, D.M. (2007) Identification of pancreatic cancer stem cells. CancerRes, 67, 1030-1037. Liebner, S., Cattelino, A., Gallini, R., Rudini, N., Iurlaro, M., Piccolo, S. and Dejana, E. (2004) Beta-catenin is required for endothelial-mesenchymal transformation during heart cushion development in the mouse. J Cell Biol, 166, 359-367. Lien, H.C., Hsiao, Y.H., Lin, Y.S., Yao, Y.T., Juan, H.F., Kuo, W.H., Hung, M.C., Chang, K.J. and Hsieh, F.J. (2007) Molecular signatures of metaplastic carcinoma of the breast by large-scale transcriptional profiling: identification of genes potentially related to epithelial-mesenchymal transition. Oncogene, 26, 7859-7871. Liu, Y., El-Naggar, S., Darling, D.S., Higashi, Y. and Dean, D.C. (2008) Zebl links epithelial-mesenchymal transition and cellular senescence. Development, 135, 579-588. Lo, H.W., Hsu, S.C., Xia, W., Cao, X., Shih, J.Y., Wei, Y., Abbruzzese, J.L., Hortobagyi, G.N. and Hung, M.C. (2007) Epidermal growth factor receptor cooperates with signal transducer and activator of transcription 3 to induce epithelialmesenchymal transition in cancer cells via up-regulation of TWIST gene expression. CancerRes, 67, 9066-9076. Lombaerts, M., van Wezel, T., Philippo, K., Dierssen, J.W., Zimmerman, R.M., Oosting, J., van Eijk, R., Eilers, P.H., van de Water, B., Cornelisse, C.J. and Cleton-Jansen, A.M. (2006) E-cadherin transcriptional downregulation by promoter methylation but not mutation is related to epithelial-to-mesenchymal transition in breast cancer cell lines. Br J Cancer,94, 661-671. Lu, Z., Ghosh, S., Wang, Z. and Hunter, T. (2003) Downregulation of caveolin-1 function by EGF leads to the loss of E-cadherin, increased transcriptional activity of beta-catenin, and enhanced tumor cell invasion. CancerCell, 4, 499-515. Luzzi, K.J., MacDonald, I.C., Schmidt, E.E., Kerkvliet, N., Morris, V.L., Chambers, A.F. and Groom, A.C. (1998) Multistep nature of metastatic inefficiency: dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Am J Pathol, 153, 865-873. Maestro, R., Dei Tos, A.P., Hamamori, Y., Krasnokutsky, S., Sartorelli, V., Kedes, L., Doglioni, C., Beach, D.H. and Hannon, G.J. (1999) Twist is a potential oncogene that inhibits apoptosis. Genes Dev, 13, 2207-2217. Makiguchi, Y., Hinoda, Y. and Imai, K. (1996) Effect of MUCI mucin, an anti-adhesion molecule, on tumor cell growth. Jpn J CancerRes, 87, 505-511. Mani, S.A., Guo, W., Liao, M., Eaton, E.N., Ayyanan, A., Zhou, A., Brooks, M., Reinhard, F., Zhang, C.C., Shipitsin, M., Campbell, L.L., Polyak, K., Brisken, C., Yang, J. and Weinberg, R.A. (2008) The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell, In press. Mani, S.A., Yang, J., Brooks, M., Schwaninger, G., Zhou, A., Miura, N., Kutok, J.L., Hartwell, K., Richardson, A.L. and Weinberg, R.A. (2007) Mesenchyme Forkhead 1 (FOXC2) plays a key role in metastasis and is associated with aggressive basal-like breast cancers. Proc Natl Acad Sci US A, 104, 10069-10074. Marambaud, P., Shioi, J., Serban, G., Georgakopoulos, A., Sarner, S., Nagy, V., Baki, L., Wen, P., Efthimiopoulos, S., Shao, Z., Wisniewski, T. and Robakis, N.K. (2002) 153 A presenilin-1/gamma-secretase cleavage releases the E-cadherin intracellular domain and regulates disassembly of adherens junctions. Embo J, 21, 1948-1956. Maretzky, T., Reiss, K., Ludwig, A., Buchholz, J., Scholz, F., Proksch, E., de Strooper, B., Hartmann, D. and Saftig, P. (2005) ADAM10 mediates E-cadherin shedding and regulates epithelial cell-cell adhesion, migration, and beta-catenin translocation. Proc Natl Acad Sci U S A, 102, 9182-9187. Martin, S.S., Ridgeway, A.G., Pinkas, J., Lu, Y., Reginato, M.J., Koh, E.Y., Michelman, M., Daley, G.Q., Brugge, J.S. and Leder, P. (2004) A cytoskeleton-based functional genetic screen identifies Bcl-xL as an enhancer of metastasis, but not primary tumor growth. Oncogene, 23, 4641-4645. Martin, T.A., Goyal, A., Watkins, G. and Jiang, W.G. (2005) Expression of the transcription factors snail, slug, and twist and their clinical significance in human breast cancer. Ann Surg Oncol, 12, 488-496. McGuire, J.K., Li, Q. and Parks, W.C. (2003) Matrilysin (matrix metalloproteinase-7) mediates E-cadherin ectodomain shedding in injured lung epithelium. Am J Pathol, 162, 1831-1843. McNeill, H., Ozawa, M., Kemler, R. and Nelson, W.J. (1990) Novel function of the cell adhesion molecule uvomorulin as an inducer of cell surface polarity. Cell, 62, 309-316. Metzstein, M.M. and Horvitz, H.R. (1999) The C. elegans cell death specification gene ces-1 encodes a snail family zinc finger protein. Mol Cell, 4, 309-319. Mitani, M., Yamanishi, T. and Miyazaki, Y. (1975) Salinomycin: a new monovalent cation ionophore. Biochem Biophys Res Commun, 66, 1231-1236. Mitani, M., Yamanishi, T., Miyazaki, Y. and Otake, N. (1976) Salinomycin effects on mitochondrial ion translocation and respiration. Antimicrob Agents Chemother, 9, 655-660. Moody, S.E., Perez, D., Pan, T.C., Sarkisian, C.J., Portocarrero, C.P., Sterner, C.J., Notorfrancesco, K.L., Cardiff, R.D. and Chodosh, L.A. (2005) The transcriptional repressor Snail promotes mammary tumor recurrence. Cancer Cell, 8, 197-209. Morali, O.G., Delmas, V., Moore, R., Jeanney, C., Thiery, J.P. and Larue, L. (2001) IGFII induces rapid beta-catenin relocation to the nucleus during epithelium to mesenchyme transition. Oncogene, 20, 4942-4950. Moreno-Bueno, G., Cubillo, E., Sarrio, D., Peinado, H., Rodriguez-Pinilla, S.M., Villa, S., Bolos, V., Jorda, M., Fabra, A., Portillo, F., Palacios, J. and Cano, A. (2006) Genetic profiling of epithelial cells expressing E-cadherin repressors reveals a distinct role for Snail, Slug, and E47 factors in epithelial-mesenchymal transition. CancerRes, 66, 9543-9556. Nagafuchi, A., Shirayoshi, Y., Okazaki, K., Yasuda, K. and Takeichi, M. (1987) Transformation of cell adhesion properties by exogenously introduced E-cadherin cDNA. Nature, 329, 341-343. Nass, S.J., Herman, J.G., Gabrielson, E., Iversen, P.W., Parl, F.F., Davidson, N.E. and Graff, J.R. (2000) Aberrant methylation of the estrogen receptor and E-cadherin 5' CpG islands increases with malignant progression in human breast cancer. Cancer Res, 60, 4346-4348. Naumov, G.N., MacDonald, I.C., Weinmeister, P.M., Kerkvliet, N., Nadkarni, K.V., Wilson, S.M., Morris, V.L., Groom, A.C. and Chambers, A.F. (2002) Persistence 154 of solitary mammary carcinoma cells in a secondary site: a possible contributor to dormancy. CancerRes, 62, 2162-2168. Nelson, W.J. and Nusse, R. (2004) Convergence of Wnt, beta-catenin, and cadherin pathways. Science, 303, 1483-1487. Nieto, M.A., Sargent, M.G., Wilkinson, D.G. and Cooke, J. (1994) Control of cell behavior during vertebrate development by Slug, a zinc finger gene. Science, 264, 835-839. Oft, M., Peli, J., Rudaz, C., Schwarz, H., Beug, H. and Reichmann, E. (1996) TGF-betal and Ha-Ras collaborate in modulating the phenotypic plasticity and invasiveness of epithelial tumor cells. Genes Dev, 10, 2462-2477. Oka, H., Shiozaki, H., Kobayashi, K., Inoue, M., Tahara, H., Kobayashi, T., Takatsuka, Y., Matsuyoshi, N., Hirano, S., Takeichi, M. and et al. (1993) Expression of Ecadherin cell adhesion molecules in human breast cancer tissues and its relationship to metastasis. CancerRes, 53, 1696-1701. Orsulic, S., Huber, O., Aberle, H., Arnold, S. and Kemler, R. (1999) E-cadherin binding prevents beta-catenin nuclear localization and beta-catenin/LEF- 1-mediated transactivation. J Cell Sci, 112 ( Pt 8), 1237-1245. Ozawa, M., Ringwald, M. and Kemler, R. (1990) Uvomorulin-catenin complex formation is regulated by a specific domain in the cytoplasmic region of the cell adhesion molecule. Proc Natl Acad Sci U S A, 87, 4246-4250. Peinado, H., Olmeda, D. and Cano, A. (2007) Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat Rev Cancer,7, 415428. Perez-Moreno, M., Davis, M.A., Wong, E., Pasolli, H.A., Reynolds, A.B. and Fuchs, E. (2006) pl20-catenin mediates inflammatory responses in the skin. Cell, 124, 631644. Perez-Moreno, M.A., Locascio, A., Rodrigo, I., Dhondt, G., Portillo, F., Nieto, M.A. and Cano, A. (2001) A new role for E12/E47 in the repression of E-cadherin expression and epithelial-mesenchymal transitions. J Biol Chem, 276, 2742427431. Perl, A.K., Wilgenbus, P., Dahl, U., Semb, H. and Christofori, G. (1998) A causal role for E-cadherin in the transition from adenoma to carcinoma. Nature, 392, 190-193. Perrais, M., Chen, X., Perez-Moreno, M. and Gumbiner, B.M. (2007) E-cadherin homophilic ligation inhibits cell growth and epidermal growth factor receptor signaling independently of other cell interactions. Mol Biol Cell, 18, 2013-2025. Peyrieras, N., Hyafil, F., Louvard, D., Ploegh, H.L. and Jacob, F. (1983) Uvomorulin: a nonintegral membrane protein of early mouse embryo. Proc Natl Acad Sci U S A, 80, 6274-6277. Pham, C.G., Bubici, C., Zazzeroni, F., Knabb, J.R., Papa, S., Kuntzen, C. and Franzoso, G. (2007) Upregulation of Twist-1 by NF-kappaB blocks cytotoxicity induced by chemotherapeutic drugs. Mol Cell Biol, 27, 3920-3935. Phillips, T.M., McBride, W.H. and Pajonk, F. (2006) The response of CD24(/low)/CD44+ breast cancer-initiating cells to radiation. J Natl Cancerlnst, 98, 1777-1785. 155 Prakasam, A.K., Maruthamuthu, V. and Leckband, D.E. (2006) Similarities between heterophilic and homophilic cadherin adhesion. Proc Natl Acad Sci U S A, 103, 15434-15439. Qian, X., Karpova, T., Sheppard, A.M., McNally, J. and Lowy, D.R. (2004) E-cadherinmediated adhesion inhibits ligand-dependent activation of diverse receptor tyrosine kinases. Embo J, 23, 1739-1748. Rasband, W.S. (1997-2006) ImageJ U. S. National Institutes of Health, Bethesda, Maryland, USA, http://rsb.info.nih.gov/ii/, 1997-2006. Ricci-Vitiani, L., Lombardi, D.G., Pilozzi, E., Biffoni, M., Todaro, M., Peschle, C. and De Maria, R. (2007) Identification and expansion of human colon-cancerinitiating cells. Nature, 445, 111-115. Riethmacher, D., Brinkmann, V. and Birchmeier, C. (1995) A targeted mutation in the mouse E-cadherin gene results in defective preimplantation development. Proc Natl Acad Sci U S A, 92, 855-859. Robson, E.J., Khaled, W.T., Abell, K. and Watson, C.J. (2006) Epithelial-tomesenchymal transition confers resistance to apoptosis in three murine mammary epithelial cell lines. Differentiation,74, 254-264. Roche-Lestienne, C., Soenen-Cornu, V., Grardel-Duflos, N., Lai, J.L., Philippe, N., Facon, T., Fenaux, P. and Preudhomme, C. (2002) Several types of mutations of the Abl gene can be found in chronic myeloid leukemia patients resistant to STI571, and they can pre-exist to the onset of treatment. Blood, 100, 1014-1018. Sato, T., Tanigami, A., Yamakawa, K., Akiyama, F., Kasumi, F., Sakamoto, G. and Nakamura, Y. (1990) Allelotype of breast cancer: cumulative allele losses promote tumor progression in primary breast cancer. CancerRes, 50, 7184-7189. Savagner, P. (2001) Leaving the neighborhood: molecular mechanisms involved during epithelial-mesenchymal transition. Bioessays, 23, 912-923. Savagner, P., Kusewitt, D.F., Carver, E.A., Magnino, F., Choi, C., Gridley, T. and Hudson, L.G. (2005) Developmental transcription factor slug is required for effective re-epithelialization by adult keratinocytes. J Cell Physiol, 202, 858-866. Savagner, P., Yamada, K.M. and Thiery, J.P. (1997) The zinc-finger protein slug causes desmosome dissociation, an initial and necessary step for growth factor-induced epithelial-mesenchymal transition. J Cell Biol, 137, 1403-1419. Schatton, T., Murphy, G.F., Frank, N.Y., Yamaura, K., Waaga-Gasser, A.M., Gasser, M., Zhan, Q., Jordan, S., Duncan, L.M., Weishaupt, C., Fuhlbrigge, R.C., Kupper, T.S., Sayegh, M.H. and Frank, M.H. (2008) Identification of cells initiating human melanomas. Nature, 451, 345-349. Scheel, C., Onder, T., Karnoub, A. and Weinberg, R.A. (2007) Adaptation versus selection: the origins of metastatic behavior. CancerRes, 67, 11476-11479; discussion 11479-11480. Schipper, J.H., Frixen, U.H., Behrens, J., Unger, A., Jahnke, K. and Birchmeier, W. (1991) E-cadherin expression in squamous cell carcinomas of head and neck: inverse correlation with tumor dedifferentiation and lymph node metastasis. CancerRes, 51, 6328-6337. Schuh, R., Vestweber, D., Riede, I., Ringwald, M., Rosenberg, U.B., Jackle, H. and Kemler, R. (1986) Molecular cloning of the mouse cell adhesion molecule 156 uvomorulin: cDNA contains a B 1-related sequence. ProcNatl Acad Sci U S A, 83, 1364-1368. Shah, A.N., Summy, J.M., Zhang, J., Park, S.I., Parikh, N.U. and Gallick, G.E. (2007) Development and characterization of gemcitabine-resistant pancreatic tumor cells. Ann Surg Oncol, 14, 3629-3637. Shimoyama, Y., Hirohashi, S., Hirano, S., Noguchi, M., Shimosato, Y., Takeichi, M. and Abe, 0. (1989) Cadherin cell-adhesion molecules in human epithelial tissues and carcinomas. CancerRes, 49, 2128-2133. Siitonen, S.M., Kononen, J.T., Helin, H.J., Rantala, I.S., Holli, K.A. and Isola, J.J. (1996) Reduced E-cadherin expression is associated with invasiveness and unfavorable prognosis in breast cancer. Am J Clin Pathol, 105, 394-402. Singh, S.K., Hawkins, C., Clarke, I.D., Squire, J.A., Bayani, J., Hide, T., Henkelman, R.M., Cusimano, M.D. and Dirks, P.B. (2004) Identification of human brain tumour initiating cells. Nature, 432, 396-401. Stewart, S.A., Dykxhoorn, D.M., Palliser, D., Mizuno, H., Yu, E.Y., An, D.S., Sabatini, D.M., Chen, I.S., Hahn, W.C., Sharp, P.A., Weinberg, R.A. and Novina, C.D. (2003) Lentivirus-delivered stable gene silencing by RNAi in primary cells. Rna, 9, 493-501. Stockinger, A., Eger, A., Wolf, J., Beug, H. and Foisner, R. (2001) E-cadherin regulates cell growth by modulating proliferation-dependent beta-catenin transcriptional activity. J Cell Biol, 154, 1185-1196. Stoker, M. and Perryman, M. (1985) An epithelial scatter factor released by embryo fibroblasts. J Cell Sci, 77, 209-223. Strathdee, G. (2002) Epigenetic versus genetic alterations in the inactivation of Ecadherin. Semin CancerBiol, 12, 373-379. Sulzer, M.A., Leers, M.P., van Noord, J.A., Bollen, E.C. and Theunissen, P.H. (1998) Reduced E-cadherin expression is associated with increased lymph node metastasis and unfavorable prognosis in non-small cell lung cancer. Am J Respir Crit Care Med, 157, 1319-1323. Team, R.D.C. (2007) R: A Language and Environment for Statistical Computing. R Foundation for Statistical Computing, Vienna, Austria. Teicher, B.A., Ikebe, M., Ara, G., Keyes, S.R. and Herbst, R.S. (1997) Transforming growth factor-beta 1 overexpression produces drug resistance in vivo: reversal by decorin. In Vivo, 11, 463-472. Tester, A.M., Ruangpanit, N., Anderson, R.L. and Thompson, E.W. (2000) MMP-9 secretion and MMP-2 activation distinguish invasive and metastatic sublines of a mouse mammary carcinoma system showing epithelial-mesenchymal transition traits. Clin Exp Metastasis, 18, 553-560. Thiery, J.P. (2002) Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer, 2, 442-454. Thiery, J.P. (2003) Epithelial-mesenchymal transitions in development and pathologies. Curr Opin Cell Biol, 15, 740-746. Thomson, S., Buck, E., Petti, F., Griffin, G., Brown, E., Ramnarine, N., Iwata, K.K., Gibson, N. and Haley, J.D. (2005) Epithelial to mesenchymal transition is a determinant of sensitivity of non-small-cell lung carcinoma cell lines and 157 xenografts to epidermal growth factor receptor inhibition. CancerRes, 65, 94559462. Thuault, S., Valcourt, U., Petersen, M., Manfioletti, G., Heldin, C.H. and Moustakas, A. (2006) Transforming growth factor-beta employs HMGA2 to elicit epithelialmesenchymal transition. J Cell Biol, 174, 175-183. Tinkle, C.L., Lechler, T., Pasolli, H.A. and Fuchs, E. (2004) Conditional targeting of Ecadherin in skin: insights into hyperproliferative and degenerative responses. Proc Natl Acad Sci US A, 101, 552-557. Tsuda, H., Zhang, W.D., Shimosato, Y., Yokota, J., Terada, M., Sugimura, T., Miyamura, T. and Hirohashi, S. (1990) Allele loss on chromosome 16 associated with progression of human hepatocellular carcinoma. ProcNatl Acad Sci U S A, 87, 6791-6794. Umbas, R., Isaacs, W.B., Bringuier, P.P., Schaafsma, H.E., Karthaus, H.F., Oosterhof, G.O., Debruyne, F.M. and Schalken, J.A. (1994) Decreased E-cadherin expression is associated with poor prognosis in patients with prostate cancer. CancerRes, 54, 3929-3933. Vallin, J., Thuret, R., Giacomello, E., Faraldo, M.M., Thiery, J.P. and Broders, F. (2001) Cloning and characterization of three Xenopus slug promoters reveal direct regulation by Lef/beta-catenin signaling. J Biol Chem, 276, 30350-30358. Valsesia-Wittmann, S., Magdeleine, M., Dupasquier, S., Garin, E., Jallas, A.C., Combaret, V., Krause, A., Leissner, P. and Puisieux, A. (2004) Oncogenic cooperation between H-Twist and N-Myc overrides failsafe programs in cancer cells. Cancer Cell, 6, 625-630. Vasioukhin, V., Bauer, C., Degenstein, L., Wise, B. and Fuchs, E. (2001) Hyperproliferation and defects in epithelial polarity upon conditional ablation of alpha-catenin in skin. Cell, 104, 605-617. Vasko, V., Espinosa, A.V., Scouten, W., He, H., Auer, H., Liyanarachchi, S., Larin, A., Savchenko, V., Francis, G.L., de la Chapelle, A., Saji, M. and Ringel, M.D. (2007) Gene expression and functional evidence of epithelial-to-mesenchymal transition in papillary thyroid carcinoma invasion. ProcNatl Acad Sci U S A, 104, 2803-2808. Vega, S., Morales, A.V., Ocana, O.H., Valdes, F., Fabregat, I. and Nieto, M.A. (2004) Snail blocks the cell cycle and confers resistance to cell death. Genes Dev, 18, 1131-1143. Vestweber, D. and Kemler, R. (1985) Identification of a putative cell adhesion domain of uvomorulin. Embo J, 4, 3393-3398. Vincent-Salomon, A. and Thiery, J.P. (2003) Host microenvironment in breast cancer development: epithelial-mesenchymal transition in breast cancer development. Breast CancerRes, 5, 101-106. Vleminckx, K., Vakaet, L., Jr., Mareel, M., Fiers, W. and van Roy, F. (1991) Genetic manipulation of E-cadherin expression by epithelial tumor cells reveals an invasion suppressor role. Cell, 66, 107-119. Wang, X., Ling, M.T., Guan, X.Y., Tsao, S.W., Cheung, H.W., Lee, D.T. and Wong, Y.C. (2004) Identification of a novel function of TWIST, a bHLH protein, in the development of acquired taxol resistance in human cancer cells. Oncogene, 23, 474-482. 158 Watanabe, N., Kawaguchi, M. and Kobayashi, Y. (1998) Activation of interleukin-1betaconverting enzyme by nigericin is independent of apoptosis. Cytokine, 10, 645653. Watson, M.A., Ylagan, L.R., Trinkaus, K.M., Gillanders, W.E., Naughton, M.J., Weilbaecher, K.N., Fleming, T.P. and Aft, R.L. (2007) Isolation and molecular profiling of bone marrow micrometastases identifies TWIST1 as a marker of early tumor relapse in breast cancer patients. Clin CancerRes, 13, 5001-5009. Weinberg, R.A. (2007) The biology of cancer.Garland Science, New York. Wicha, M.S. (2006) Cancer stem cells and metastasis: lethal seeds. Clin CancerRes, 12, 5606-5607. Wildenberg, G.A., Dohn, M.R., Carnahan, R.H., Davis, M.A., Lobdell, N.A., Settleman, J. and Reynolds, A.B. (2006) p120-catenin and pl90RhoGAP regulate cell-cell adhesion by coordinating antagonism between Rac and Rho. Cell, 127, 1027-1039. Wong, S.Y. and Hynes, R.O. (2006) Lymphatic or hematogenous dissemination: how does a metastatic tumor cell decide? Cell Cycle, 5, 812-817. Wu, W.S., Heinrichs, S., Xu, D., Garrison, S.P., Zambetti, G.P., Adams, J.M. and Look, A.T. (2005) Slug antagonizes p53-mediated apoptosis of hematopoietic progenitors by repressing puma. Cell, 123, 641-653. Wu, X., Chen, H., Parker, B., Rubin, E., Zhu, T., Lee, J.S., Argani, P. and Sukumar, S. (2006) HOXB7, a homeodomain protein, is overexpressed in breast cancer and confers epithelial-mesenchymal transition. CancerRes, 66, 9527-9534. Xue, C., Plieth, D., Venkov, C., Xu, C. and Neilson, E.G. (2003) The gatekeeper effect of epithelial-mesenchymal transition regulates the frequency of breast cancer metastasis. Cancer Res, 63, 3386-3394. Yagoda, N., von Rechenberg, M., Zaganjor, E., Bauer, A.J., Yang, W.S., Fridman, D.J., Wolpaw, A.J., Smukste, I., Peltier, J.M., Boniface, J.J., Smith, R., Lessnick, S.L., Sahasrabudhe, S. and Stockwell, B.R. (2007) RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature, 447, 864-868. Yang, A.D., Fan, F., Camp, E.R., van Buren, G., Liu, W., Somcio, R., Gray, M.J., Cheng, H., Hoff, P.M. and Ellis, L.M. (2006a) Chronic oxaliplatin resistance induces epithelial-to-mesenchymal transition in colorectal cancer cell lines. Clin Cancer Res, 12, 4147-4153. Yang, J., Mani, S.A., Donaher, J.L., Ramaswamy, S., Itzykson, R.A., Come, C., Savagner, P., Gitelman, I., Richardson, A. and Weinberg, R.A. (2004) Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell, 117, 927-939. Yang, L., Lin, C. and Liu, Z.R. (2006b) P68 RNA helicase mediates PDGF-induced epithelial mesenchymal transition by displacing Axin from beta-catenin. Cell, 127, 139-155. Yang, W.S. and Stockwell, B.R. (2008) Synthetic Lethal Screening Identifies Compounds Activating Iron-Dependent, Nonapoptotic Cell Death in OncogenicRAS-Harboring Cancer Cells. Chem Biol, 15, 234-245. Yauch, R.L., Januario, T., Eberhard, D.A., Cavet, G., Zhu, W., Fu, L., Pham, T.Q., Soriano, R., Stinson, J., Seshagiri, S., Modrusan, Z., Lin, C.Y., O'Neill, V. and Amler, L.C. (2005) Epithelial versus mesenchymal phenotype determines in vitro 159 sensitivity and predicts clinical activity of erlotinib in lung cancer patients. Clin Cancer Res, 11, 8686-8698. Yawata, A., Adachi, M., Okuda, H., Naishiro, Y., Takamura, T., Hareyama, M., Takayama, S., Reed, J.C. and Imai, K. (1998) Prolonged cell survival enhances peritoneal dissemination of gastric cancer cells. Oncogene, 16, 2681-2686. Yoshida, C. and Takeichi, M. (1982) Teratocarcinoma cell adhesion: identification of a cell-surface protein involved in calcium-dependent cell aggregation. Cell, 28, 217-224. Yoshiura, K., Kanai, Y., Ochiai, A., Shimoyama, Y., Sugimura, T. and Hirohashi, S. (1995) Silencing of the E-cadherin invasion-suppressor gene by CpG methylation in human carcinomas. Proc Natl Acad Sci U S A, 92, 7416-7419. Young, P., Boussadia, O., Halfter, H., Grose, R., Berger, P., Leone, D.P., Robenek, H., Charnay, P., Kemler, R. and Suter, U. (2003) E-cadherin controls adherens junctions in the epidermis and the renewal of hair follicles. Embo J, 22, 57235733. Yu, F., Yao, H., Zhu, P., Zhang, X., Pan, Q., Gong, C., Huang, Y., Hu, X., Su, F., Lieberman, J. and Song, E. (2007) let-7 regulates self renewal and tumorigenicity of breast cancer cells. Cell, 131, 1109-1123. Zhang, X., Wang, Q., Ling, M.T., Wong, Y.C., Leung, S.C. and Wang, X. (2007) Antiapoptotic role of TWIST and its association with Akt pathway in mediating taxol resistance in nasopharyngeal carcinoma cells. Int J Cancer, 120, 1891-1898. Zhuo, W.L., Wang, Y., Zhuo, X.L., Zhang, Y.S. and Chen, Z.T. (2008) Short interfering RNA directed against TWIST, a novel zinc finger transcription factor, increases A549 cell sensitivity to cisplatin via MAPK/mitochondrial pathway. Biochem Biophys Res Commun. 160 Appendix 1 Probes and the corresponding genes upregulated more than 1.5 log-fold in shEcad cells Symbol ProbelD FBLN5 GREM1 GREM1 COL3A1 COL3A1 COL1A2 DCN COL1A2 COL3A1 FBN1 NID1 DCN PTX3 MAP1B TCF8 ENPP2 CDH2 COL1A1 POSTN ANPEP TCF8 RGS4 RGS4 DCN C5orf 13 C5orf13 CDH11 COL1A1 PRRX1 COL6A1 IGSF4B GNG11 RHOBTB3 TFPI FBN1 PRG1 NR2F1 AGTR1 BIN1 LTBP1 PDGFRL LTBP1 SPOCK 203088_at 218468_sat 218469_at 215076 s at 201852_xat 202403_s_at 201893_xat 202404_s_at 211161_sat 202766 s at 202007_at 211813 x_at 206157_at 212233_at 212764_at 209392_at 203440_at 202310_s_at 210809_s_at 202888 s at 212758 s at 204337_at 204338_s_at 211896 s at 201309 x at 201310_s_at 207173 x at 202311 s at 205991 s at 213428_sat 213948 x at 204115_at 202975_s_at 213258_at 202765 s at 201858 s at 209505_at 205357_s_at 214439 x at 202729_sat 205226_at 202728_s_at 202363_at shEcad vs shCntri loqfold change 8.5255 8.3169 8.1805 7.7299 7.5006 7.4536 7.059 6.9349 6.796 6.5269 6.1547 5.9537 5.8939 5.7621 5.7218 5.6529 5.637 5.6185 5.5791 5.4274 5.3506 5.3408 5.2446 5.2339 5.1119 5.0792 5.0275 5.0017 4.9379 4.8373 4.8178 4.7323 4.6948 4.6916 4.6542 4.6237 4.5814 4.5462 4.5256 4.5083 4.4753 4.4565 4.3719 Symbol PCDH9 LOC51334 NEBL DLC1 TOX MME BIN1 BIN1 RGL1 IGFBP4 ENPP2 TFPI KCNMA1 CLEC3B RHOBTB3 KCNMA1 STC2 CDH2 MAR3 NID1 PTGFR TGFBR3 PRKCA SDC2 NEBL COL1Al PCOLCE BIN1 PPP1R3C TBL1X TRPA1 TFPI HRASLS3 COL6A2 FLJ20701 FLJ10094 CYFIP2 DOCK10 MLPH PVRL3 KAL1 CDH11 MME 161 ProbelD 219737 s at 220014_at 203961 _at 210762 s at 204529 s at 203434 s at 202931 x at 210201 x at 209568_s_at 201508_at 210839 s at 210664 s at 221583 s at 205200_at 202976_s_at 221584 s at 203438_at 203441 s at 213256_at 202008_s_at 207177_at 204731 at 213093_at 212154_at 203962_sat 217430 x at 202465_at 210202 s at 204284_at 213400_s_at 217590_s_at 209676_at 209581_at 209156 s at 219093_at 219501 _at 215785 s at 219279_at 218211 s at 213325_at 205206_at 207172_s at 203435_sat shEcad vs shCntrl loqfold change 4.2958 4.2883 4.285 4.2619 4.2601 4.2254 4.223 4.1982 4.1962 4.1851 4.1777 4.1761 4.1568 4.1464 4.1164 4.114 4.1061 4.0338 4.0263 4.0035 3.9869 3.9797 3.9304 3.9258 3.9088 3.8942 3.8924 3.8882 3.873 3.8617 3.8253 3.7594 3.7399 3.7356 3.6949 3.6914 3.6637 3.6455 3.6164 3.5922 3.57 3.5632 3.5508 SDC2 WNT5B FBLN1 PRG1 PSCDBP OLFML3 ITGBL1 LRIG1 COL5A2 MMP2 MYL9 EML1 STC2 RGS4 BGN AGTR1 NPTX2 ANXA6 COL5A2 BGN CTGF CYP1B1 212158_at 221029_s_at 202995_sat 201859_at 209606_at 218162_at 214927_at 211596_sat 221730_at 201069_at 201058 s at 204797 s at 203439_s_at 204339 s at 213905_x_at 208016_sat 213479_at 200982_s_at 221729_at 201261_x_at 209101 _at 202437_sat 4.3594 3.4653 3.4641 3.4613 3.4607 3.4409 3.4318 3.4263 3.4211 3.3894 3.3783 3.3636 3.3635 3.3399 3.3073 3.2925 3.2837 3.2812 3.2402 3.1998 3.1784 3.173 DPYSL3 201430 s at 3.1664 PLEKHC1 MAP1B SEP6 ITGBL1 GRB10 TWIST1 ENPP1 PNMA2 RPS6KA2 PPAP2B KIAA0367 PLEKHC1 ZBTB16 JAM3 ARMCX2 LIN7A CYP1B1 SYT11 ROR1 AOX1 TCF8 209209_s_at 214577_at 212414_s_at 205422_s_at 209409 at 213943 at 205066 s at 209598 at 212912_at 212226_sat 212805_at 214212_x_at 205883_at 212813_at 203404_at 206440_at 202436_s_at 209198_s_at 205805 s at 205082 s at 210875 s at 213169_at 202435 s at 213488_at 213429_at 204620 s at 3.1654 3.1537 3.1482 3.1269 3.1258 3.1236 3.1159 3.1025 3.094 3.0796 3.0685 3.0675 3.0585 3.022 3.02 3.013 3.0027 2.9614 2.948 2.9376 2.9259 2.9256 2.9067 2.8942 2.8915 2.8839 CYPIB1 SNED1 CSPG2 PODXL KCNJ6 CHN1 TNFAIP6 SGNE1 COL6A3 DDR2 AOX1 RHOBTB3 IGSF4B DUSP1 MAGEH1 CSPG2 FBLN1 GADD45B CITED2 TSC22D3 DPYSL3 PPAP2B GADD45B KIAA0367 ATP6VOE2 L LOC22136 2 C5orf 13 REPS2 TSC22D3 PMP22 THY1 CITED2 TRAM2 RGS17 PPARG NID2 GRB10 PPAP2B ZFHX4 APXL NNMT TAGLN BEXL1 SEP6 TRAM2 TNFAIP6 RPS6KA2 EPHB2 CREB3L1 CDH18 DCN 162 201578_at 210454_s_at 212624 s at 206026 s at 203889_at 201438_at 205168_at 205083_at 216048_sat 221921 s at 201041 s at 218573_at 221731_x_at 202994 s at 209304_x_at 209357_at 208763_ sat 201431 s at 212230_at 207574 s at 212806_at 213587_sat 3.5237 2.7577 2.7265 2.7238 2.7164 2.7031 2.7007 2.6934 2.6925 2.6887 2.6673 2.6508 2.6478 2.6348 2.6341 2.6283 2.5896 2.5757 2.5756 2.5547 2.5471 2.5228 213248_at 2.5213 222344_at 205645_at 207001 x at 210139_s_at 208850_s_at 207980_s_at 202368 sat 220334_at 208510_s_at 204114_at 210999_s_at 209355 s at 219779_at 204967_at 202238_sat 205547 s at 215440 s at 214298 x at 202369 s at 206025_s_at 204906_at 218380_at 209589 s at 213059_at 206280 at 209335_at 2.5179 2.5137 2.5086 2.4862 2.4787 2.4619 2.4611 2.4521 2.4455 2.443 2.4224 2.4008 2.3933 2.387 2.3753 2.365 2.3606 2.3526 2.3508 2.35 2.3453 2.3413 2.3275 2.3086 2.3072 2.3029 TAF9L FN1 FY MLLT3 IGFBP3 CHST2 THY1 HMOX1 AP1S2 THY1 DDAH1 FLJ10357 PCAF SATB2 NR2F1 ITGA10 FGFR1 C14orf 132 TRBV3-1 ANKRD25 FBLN1 GLRX NNMT PTGER4 DNAJB4 IL17RC RIMS1 PTD015 SIX2 DUSP1 MOXD1 SLITRK5 SEMA5A CSPG2 UGDH SERPINE1 CYBRD1 PTD015 C10orf56 SPARC COL6A1 BIN1 SERPINF1 EPHB2 MYBL1 HAS2 MMP19 FLOT1 AP1S2 NAP1L3 221618 s at 214701 s at 208335 s at 204917 s at 212143 s at 203921_at 213869 x at 203665 at 203300 x at 208851 s at 209094_at 58780 s at 203845_at 213435_at 209506 s at 206766_at 211535 s at 218820_at 211796 s at 218418 s at 201787_at 209276 s at 202237_at 204897_at 203811 s at 221926 s at 216184 s at 221600 s at 206510 at 201044 x at 209708_at 214930_at 205405_at 211571 s at 203343_at 202627 s at 217889 s at 221599 at 212423 at 212667 at 212091 s at 214643 x at 202283_at 211165 x at 213906_at 206432 at 204575 s at 210142 x at 203299 s at 204749 at 2.3015 2.2525 2.2483 2.2483 2.2408 2.2324 2.2306 2.2214 2.2108 2.2091 2.2042 2.181 2.1804 2.1758 2.1707 2.1667 2.1659 2.1195 2.0973 2.0969 2.0949 2.085 2.0833 2.0798 2.0786 2.0772 2.059 2.0505 2.0482 2.048 2.0426 2.0407 2.0361 2.0221 2.0221 2.0202 2.0171 2.0043 2.0003 1.9817 1.9724 1.9536 1.9509 1.9487 1.9469 1.9418 1.936 1.9346 1.7461 1.7393 IGSF4B C14orf45 EML1 ABLIM3 ASMTL FAM50A DNAJB4 LTBP2 BLMH CCNE2 GSTA4 P8 SERPINE1 LMCD1 CDKN2C ALS2CR3 DSCR1 L1 MICAL2 FLOT1 TPM1 GALNT10 KLF9 ADAM12 FLJ22471 GULP1 TPM1 SLC7A2 WNT5A IGFBP3 WIP149 RHOBTB1 DOCK4 MITF SLC22A4 ABCA8 FTL ITGA7 MICAL2 COL5A1 ICK WASPIP GULP1 SYNE1 ACTA2 DAB2 BPGM CDK5RAP2 ALS2CR3 STARD13 163 211677 x at 220173 at 204796_at 205730 s at 36553_at 203262 s at 203810_at 204682 at 202179_at 211814 s at 202967_at 209230 s at 202628 s at 218574 s at 204159_at 202125 s at 203498_at 212472_at 208749 x at 206117 at 212256 at 203542 s at 202952 s at 218175_at 204235 s at 206116 s at 207626 s at 205990 s at 210095 s at 203827 at 212651_at 205003 at 207233 s at 205896_at 204719 at 213187 x at 216331 _at 212473 s at 212488 at 204569 at 202664 at 204237_at 209447 at 200974 at 201280 s at 203502_at 218031 s at 220935 s at 202124 s at 213103 at 1.9324 1.9304 1.921 1.9203 1.9185 1.9182 1.9164 1.913 1.9106 1.9074 1.8988 1.8972 1.8898 1.8886 1.8861 1.8845 1.8783 1.8709 1.8679 1.8502 1.8495 1.8482 1.848 1.8475 1.84 1.8341 1.8309 1.8212 1.8193 1.8116 1.8102 1.8099 1.8037 1.7989 1.7957 1.7896 1.7879 1.7868 1.7856 1.7806 1.7791 1.7714 1.7709 1.7656 1.7644 1.7578 1.7491 1.7471 1.5833 1.5808 GRB10 GULP1 ARL3 MAGED2 RECK PRAF2 ARHGAP22 PPM1H GALNT1 0 DMN PLOD2 LOC283537 ZNF22 ASMTL PRKCA UQCRB PRR3 C14orf58 COL8A1 TCEA2 GLRB FZD2 KDELR3 LOX NRP1 KDELR3 CMKOR1 GK SV2A EPS8 IGFBP7 FLOT1 LEPR ARID5B GYPC MARCKS PCOLCE2 SRR HOXA5 KLF4 GLRB ZNF22 209410_s_at 215913_s_at 220999_s_at 218330 s at 202641_at 208682_s_at 205407_at 203456_at 206298_at 212686_at 207357_sat 212730_at 202619 s at 214719_at 218005_at 209394_at 215195_at 209065_at 204795_at 219316_s_at 214587_at 203919_at 205280_at 210220_at 207265 sat 215446 s at 210510_s_at 204017_at 212977_at 215977 x at 203069_at 202609_at 201162_at 208748_s_at 209894_at 212614_at 202947 s at 201668_x_at 219295 s at 219205_at 213844_at 220266 s at 205279_s_at 218006 s at 1.7384 1.7375 1.7345 1.7324 1.7192 1.7112 1.7095 1.7086 1.7063 1.7038 1.7033 1.7032 1.6983 1.6975 1.6956 1.6866 1.6862 1.6852 1.684 1.6808 1.678 1.6771 1.6725 1.6697 1.6632 1.6617 1.6592 1.6563 1.6455 1.6444 1.6432 1.6423 1.629 1.6267 1.6259 1.6213 1.6194 1.6188 1.6176 1.6172 1.6096 1.5983 1.5893 1.5887 FLJ14627 FGFR1 FLJ20323 SDC3 Cl 0orf56 DGKI MAGED1 FLJ10357 ALDH6A1 CRIP2 SEP11 COL6A1 REPIN1 C6orf145 DAB2 HRMT1 L1 H2AFY TBL1X PTGER4 EDG2 FKBP5 NEBL FAT4 RNASE4 164 221909_at 210973_sat 211724_x_at 202898_at 212419_at 206806_at 209014_at 220326_sat 221588_x_at 208978_at 214293_at 212937 s at 219041 s at 212923_s at 201279_s at 202098 s at 214500 at 201867 s at 204896 s at 204036_at 212829_at 204560_at 207279 s at 219427_at 213397 x at 1.5775 1.5773 1.5754 1.5689 1.5671 1.5665 1.5658 1.5623 1.5614 1.5575 1.549 1.5474 1.5431 1.5427 1.5419 1.5408 1.535 1.5302 1.5282 1.5264 1.5233 1.5212 1.5195 1.5102 1.5049 Probes and the corresponding genes downregulated more than 1.5 log-fold in shEcad cells Symbol ProbelD SERPINB2 KLK10 SPRR1B SPRR1A S100A8 SLPI KRT6B KLK7 SPRR1A P13 ALDH1A3 CDH1 IL1A KRT15 TACSTD1 FXYD3 SPRR2D FST FST SERPINB13 P13 EVA1 ANXA3 TGFA 204614_at 209792_s_at 205064_at 213796at 202917_s at 203021_at 213680_at 205778_at 214549_x_at 41469_at 203180_at 201131_s_at 210118_s_at 204734_at 201839_s at 202489 s at 208539_x at 204948_s_at 207345_at 217272_s at 203691_at 203780_at 209369_at 205016_at 217767_at 222242 s at 219121 _sat 213285_at 205403_at 218186_at 214456_x at 205067_at 205014_at 204379_s at 204751 x at 209863_s at 212268_at 205916_at 206023_at 219476_at 209800_at 218677_at 202712 s at 205680_at C3 KLK5 RBM35A TMEM30B IL1 R2 RAB25 SAA2 IL1 B FGFBP1 FGFR3 DSC2 TP73L SERPINB1 S100A7 NMU C1orf116 KRT16 S100A14 CKMT1 B MMP10 shEcad vs shCntrl log-fold change -8.0717 -7.5327 -7.4097 -7.2153 -7.2083 -7.0663 -7.0042 -6.9354 -6.8801 -6.8045 -6.6936 -6.6337 -6.6207 -6.4087 -6.3193 -5.9706 -5.7607 -5.742 -5.6254 -5.621 -5.6163 -5.4362 -5.3846 -5.3115 -5.2584 -5.119 -5.1177 -5.0971 -5.0927 -5.0546 -5.0394 -5.0166 -5.0029 -4.9797 -4.9795 -4.9643 -4.9641 -4.956 -4.9272 -4.9244 -4.8836 -4.8681 -4.8513 -4.8207 165 Symbol ProbelD KLK8 IL1B DSG3 GJB3 GJB3 DSC3 EVA1 DSC3 CDH3 SPINT1 SCNN1A CDH1 SFRP1 TP73L CCND2 KRT13 CA2 SERPINB1 AQP3 KRT6B SAA2 IL1 RN TSPAN1 DSC2 ZBED2 CCND2 COL17A1 DST SERPINB7 RBM35B GJB5 SEMA3A PPL TRIM29 IL1 R2 ARTN ST14 EREG TMEM16A FLJ12684 IRF6 ARTN INHBA CXCL1 206125 s at 39402_at 205595_at 205490 x at 215243 s at 206033_s_at 203779_s_at 206032_at 203256_at 202826 at 203453_at 201130 s at 202037 s at 211194 s at 200951 s at 207935_sat 209301_at 213572 s at 39248_at 209126 x at 208607 s at 212657 s at 209114_at 204750_s_at 219836_at 200953 s at 204636_at 204455_at 206421 s at 219395_at 206156_at 206805_at 203407_at 202504_at 211372 s at 210237_at 202005_at 205767_at 218804_at 219987_at 202597_at 207675 x at 210511 s at 204470_at shEcad vs shCntrl logfold change -4.7629 -4.7398 -4.7293 -4.7065 -4.6965 -4.6921 -4.6783 -4.6599 -4.5706 -4.5679 -4.5591 -4.514 -4.4983 -4.4762 -4.4681 -4.4411 -4.4283 -4.4214 -4.3964 -4.3788 -4.3689 -4.3471 -4.3434 -4.3339 -4.2963 -4.2958 -4.2939 -4.2654 -4.2502 -4.2357 -4.2244 -4.1803 -4.1802 -4.1737 -4.1643 -4.158 -4.1383 -4.1329 -4.1232 -4.0924 -4.085 -4.0846 -4.0806 -4.0754 TNFAIP3 CSTA CD24 S100A9 VSNL1 LAMB3 CDS1 ST14 CD24 TNFAIP3 SPINT2 SFRP1 FLRT3 MAPK13 SERPINB13 CD24 SLAC2-B MFAP5 SLC2A9 TACSTD2 VSNL1 KCNK1 ABLIM1 CEACAM6 TPD52L1 HS3ST2 CD24 SERPINA1 CD24 CYP27B1 CD24 GPR56 PTGS2 PCDH7 DDR1 MMP9 KCNK1 KRT8 NRCAM TNFRSF6B DST JAG2 PLA2G4A SCEL IGSF3 ARTN JUP VGLL1 SDC1 KRT6A 202643_s_at 204971_at 266_s_at 203535_at 203798_sat 209270_at 205709_s_at 216905_s_at 208651_x at 202644_s_at 210715_s_at 202036_sat 219250_s_at 210058_at 211361 s at 209772_s at 214734_at 213764 s at 219991 _at 202286_s_at 203797_at 204679_at 200965_sat 211657_at 203786_sat 219697_at 216379_xat 202833_s at 209771_x_at 205676_at 208650_s at 212070_at 204748_at 205534_at 210749 x at 203936_s at 204678_s at 209008_xat 204105_s at 206467_x at 216918_sat 32137_at 210145_at 206884_s at 202421_at 216052_x_at 201015_sat 215729 s at 201286_at 209125_at -4.0657 -4.0473 -4.037 -4.0267 -4.0208 -4.0207 -4.0154 -4.0118 -4.011 -4.0056 -3.9916 -3.9871 -3.9723 -3.9676 -3.9426 -3.9378 -3.9259 -3.9144 -3.9037 -3.8993 -3.8677 -3.8465 -3.8298 -3.7978 -3.7828 -3.7793 -3.7742 -3.7566 -3.7566 -3.7529 -3.741 -3.7311 -3.724 -3.7072 -3.6849 -3.6812 -3.6525 -3.6445 -3.6417 -3.6272 -3.6262 -3.6194 -3.6193 -3.616 -3.6127 -3.6079 -3.5903 -3.5821 -3.187 -3.1851 166 WISP3 CEACAM6 MFAP5 CLCA2 LAMA3 MAP7 ST6GALNAC2 PKP3 HS3ST1 ARHGAP8 KRT6A MST1R SOX15 CLCA2 SNX10 TSPAN13 KIAA0746 CTSL2 F11R CLCA2 PRR5 CLDN7 JAG2 ARHGEF3 Cl0orf10 DDR1 MMP1 TPD52L1 FRMD4B LAMC2 MFAP5 MTSS1 TGFA FEZ1 KRT5 ANXA8 LCN2 FGFR2 CNTN1 ITGB6 GPRC5A RAB38 ITGB4 DDR1 SLC6A15 PRRG4 KRT18 MAPK13 MIA EDG7 210861 s at 203757 s at 213765_at 206165 s at 203726 s at 202890_at 204542_at 209873_s_at 205466_s_at 37117_at 214580 x at 205455_at 206122_at 217528_at 218404_at 217979_at 212314_at 210074_at 221664_sat 206166 s at 205980 s at 202790_at 209784_s_at 218501 _at 209183 s at 208779_x_at 204475_at 210372 s at 213056_at 207517_at 209758 s at 203037_s at 205015_s at 203562_at 201820_at 203074_at 212531 _at 203638 s at 211203 s at 208083 s at 203108_at 219412_at 204990 s at 207169 x at 206376_at 207291 _at 201596_x at 210059 s at 206560 s at 220816_at -3.58 -3.5544 -3.5389 -3.5318 -3.5096 -3.5069 -3.5046 -3.4894 -3.4885 -3.4827 -3.4803 -3.4788 -3.4785 -3.4718 -3.467 -3.461 -3.4417 -3.4204 -3.4129 -3.4108 -3.4074 -3.4059 -3.3989 -3.3836 -3.3761 -3.3719 -3.3695 -3.3566 -3.3527 -3.3308 -3.3306 -3.324 -3.3226 -3.3156 -3.3148 -3.3148 -3.3045 -3.302 -3.2895 -3.2884 -3.2842 -3.2708 -3.2688 -3.2506 -3.2303 -3.2143 -3.2058 -3.1947 -2.9577 -2.9522 GRHL2 PERP OCLN ALOX15B MALL KRT17 FXYD3 FGFR2 IL8 TMEM40 PTHLH SDC1 GPR87 MAP7 CASP1 LYPD3 PTHLH C10orf116 LISCH7 P2RX5 MAP7 AP1 M2 ITGB4 TMPRSS11E DSP KRT17 KIAA0040 PTPRZ1 NDRG1 EPN3 IF130 SERPINA1 ARL7 EHF Clorf116 COBLL1 PRSS8 CAMK2B ITGB6 MYO1D CYB5R2 BMP2 IL18 HOOK1 PTHLH ARHGEF4 LAD1 SLC39A8 219388_at 217744_s_at 209925_at 206714_at 209373_at 218764_at 205157_s_at 202488_s_at 208228s_at 202859_x at 219503_s_at 211756_at 201287 s at 219936_sat 202889_x_at 211368_s at 204952_at 210355_at 203571 s at 208190 s at 210448_s at 215471 s at 218261 at 204989 s at 220431_at 200606_at 212236_x at 203143_sat 204469_at 200632_sat 220318_at 201422_at 211429_ sat 202207_at 219850_s at 219856_at 203641_s at 202525_at 209956_sat 208084_at 212338_at 220230_s_at 205290_s at 213929_at 206295_at 219976_at 206300_s at 205109_sat 216641_sat 209267_s at -3.1843 -3.1795 -3.1688 -3.1622 -3.1618 -3.1519 -3.1434 -3.1367 -3.1343 -3.1341 -3.1329 -3.1277 -3.1196 -3.0864 -3.0855 -3.0746 -3.0696 -3.0671 -3.0648 -3.0621 -3.0565 -3.047 -3.044 -3.0392 -3.039 -3.0369 -3.0288 -3.028 -3.026 -3.0254 -3.0253 -3.0218 -3.0193 -3.0179 -3.0117 -3.0062 -3.0017 -3.0008 -2.9882 -2.9867 -2.9764 -2.9722 -2.968 -2.9656 -2.9641 -2.9637 -2.6713 -2.6695 -2.6693 -2.6649 167 IL8 CAMK2B DDR1 LPHN2 ATP12A ST6GALNAC5 CNTNAP2 LAD1 ICAM1 SYK ARL7 LAMC2 CASP1 ERBB3 IFIH1 EPS8L1 CCL20 NAV3 CXCL2 SLC1A3 CLCA2 ZNF185 CCND2 ITGA2 GPM6B COBLL1 CENTD1 BACE2 ABCA12 SFRP1 SOX9 ST6GAL1 C20orf42 CELSR1 KLF5 PCDH7 AREG CA9 CXADR EFNA1 FLJ21511 GPM6B IL1 F9 MCOLN3 SNCA AMPD3 SERPINB13 APOC1 GALNT6 FZD3 211506_s_at 210404 x at 1007 sat 206953 s at 207367_at 220979 s_at 215145_s_at 203287_at 202637 s at 207540 s at 202206_at 202267_at 209970 x at 202454 s at 219209_at 91826_at 205476_at 204823_at 209774 x at 202800_at 206164_at 203585_at 200952_s_at 205032_at 209169_at 203642 s at 213618_at 217867 x at 215465_at 202035 s at 202936 s at 201998 at 60474_at 41660_at 209211_at 205535 s at 205239_at 205199_at 203917_at 202023_at 220723 s at 209168_at 220322_at 220484_at 204466 s at 207992 s at 211362 s at 204416 x at 219956_at 219683_at -2.9502 -2.9363 -2.9327 -2.9306 -2.9275 -2.9232 -2.922 -2.8918 -2.8913 -2.8889 -2.8886 -2.8866 -2.8825 -2.8806 -2.8781 -2.8772 -2.8663 -2.8646 -2.8508 -2.842 -2.8367 -2.8347 -2.8135 -2.8122 -2.8085 -2.7934 -2.7851 -2.783 -2.7785 -2.77 -2.7474 -2.7459 -2.7443 -2.7333 -2.7262 -2.7208 -2.7179 -2.7178 -2.7145 -2.7139 -2.7075 -2.6897 -2.6885 -2.6853 -2.6822 -2.6769 -2.3753 -2.3719 -2.3662 -2.3656 ITGA6 KIAA0746 XDH TGFA ARHGAP25 SOX9 LEPREL1 ARHGAP25 DUOX1 KLF5 MYO5C SLC39A8 TINAGL1 CREG1 CDC2L2 KRT14 CA12 CTSC MBP TFCP2L1 SLC7A5 KIBRA AKR1C2 CA12 CST6 IVL UPP1 LGALS7 P2RY2 TNFSF10 PITPNC1 CASP1 KIBRA TIMP3 ARL7 PTGES MCTP2 SORL1 EPHA1 GRB14 CASP1 HOMER1 Clorf106 MGST2 ZNF165 ARG2 EPB41 L4B LYN DMD CDCP1 215177_s_at 212311_at 210301 _at 211258_s_at 38149_at 202935_sat 218717_s_at 204882_at 219597_s_at 209212 s at 218966_at 219869 s at 219058_x_at 201200_at 210473_s_at 209351_at 204508_s_at 201487_at 210136_at 219735_s at 201195_ s _at 216074_x at 209699_x_at 203963_at 206595_at 214599_at 203234_at 206400_at 206277_at 202688_at 219155_at 211366_x_at 213085_s at 201147_s at 202208_sat 210367_s_at 220603_s at 212560_at 205977_s_at 206204_at 211367 s at 213793_s at 219010_at 204168_at 206683_at 203946_s at 220161_s at 202626 s at 203881_s at 218451_at -2.6535 -2.6529 -2.6468 -2.6412 -2.6367 -2.6291 -2.6225 -2.6175 -2.6131 -2.6122 -2.5998 -2.599 -2.5933 -2.5842 -2.5773 -2.5688 -2.5675 -2.547 -2.5457 -2.5117 -2.4963 -2.4874 -2.4775 -2.4733 -2.4664 -2.4623 -2.457 -2.451 -2.4449 -2.4372 -2.4367 -2.4346 -2.4305 -2.4287 -2.4239 -2.4235 -2.4224 -2.409 -2.4074 -2.3965 -2.3904 -2.3838 -2.3836 -2.3758 -2.2031 -2.2023 -2.2021 -2.1997 -2.1964 -2.193 168 MOSC1 C20orf42 ADRB2 CD44 ABCG2 L1CAM ITGA6 C6orf105 AP1G1 CA12 F3 SLC31A2 AP1 M2 SERPINB13 AKR1C1 TRIM29 SLC6A8 KIAA1815 CXCL3 RNF128 TMEM22 HES1 KCNG1 GPM6B FAT2 MPPE1 TNFRSF21 NFE2L3 C 1orf42 GCLC LYN BIK CSH2 MPPE1 GALNT14 FOS S100A8 SLC5A1 IRX4 SPAG1 SLC6A10 AKR1C1 MOBKL2B VDR HBEGF LYN KRTHB1 FGD6 LIMK2 FLJ20273 218865_at 218796_at 206170_at 217523_at 209735_at 204584_at 201656_at 215100_at 215867_x_at 210735_s_at 204363_at 204204_at 65517_at 216258 s at 204151 _xat 211002 s at 210854_x_at 218342 s at 207850_at 219263_at 219569 s at 203394 sat 214595_at 209170_s_at 208153_s_at 214071 _at 214581 _xat 204702 s at 220620_at 202922_at 210754 s at 205780_at 212737_at 213924_at 219271 _at 209189_at 214370_at 206628_at 220225_at 210117_at 215812 s at 216594_xat 219265_at 204254 s at 38037_at 202625_at 213711_at 219901 _at 202193_at 218035 s at -2.3647 -2.3518 -2.3515 -2.3486 -2.345 -2.3411 -2.3405 -2.3374 -2.3348 -2.3279 -2.3253 -2.3245 -2.3156 -2.3142 -2.3112 -2.3103 -2.3101 -2.3097 -2.3084 -2.2968 -2.2931 -2.285 -2.284 -2.2833 -2.2746 -2.2689 -2.2674 -2.2672 -2.2661 -2.2655 -2.2651 -2.2641 -2.2632 -2.2583 -2.2465 -2.2456 -2.2432 -2.2408 -2.2365 -2.2332 -2.2312 -2.2277 -2.2269 -2.2098 -2.0319 -2.0312 -2.0263 -2.0259 -2.0259 -2.0204 BDKRB2 CLDN4 MAP3K9 RHOD GPM6B HBEGF IL RN IL4R STAC SERPINA3 CD1 D CA12 ITM2A SLC6A8 ELMO3 PARP12 DSU CLDN1 MAST4 KLK11 PLXNB1 GCLC GALNT3 EDN1 NRG1 SLC7A8 S100P SFN PSTPIP2 AQP3 TFPI2 GNA15 IGSF4 THBD MAOA RLN2 CORO1A STEAP1 CYP4F11 STAP2 PTPNS1 ANK3 EPAS1 ROBO1 DUSP6 OAS3 MYO1 B SEMA3C CDK5R1 205870_at 201428_at 213927_at 209885_at 209167_at 203821 _at 216243 s at 203233_at 205743_at 202376_at 205789_at 214164_x at 212444_at 202747_s at 213843_x_at 219411_at 218543_s_at 219648_at 218182_s at 40016_gat 205470_s_at 215807_s_at 202923_s_at 203397_sat 218995_s_at 206343_s_at 216604 s at 204351_at 209260_at 219938_sat 39249_at 209277_at 205349_at 209030_s_at 203887_s_at 204388_sat 214519_s_at 209083_at 205542_at 206153_at 221610_s_at 202897_at 206385 s at 200878_at 213194_at 208891_at 218400_at 212364_at 203789_s at 204995_at -2.1912 -2.1895 -2.1833 -2.1829 -2.182 -2.1753 -2.1707 -2.1625 -2.1585 -2.1547 -2.1538 -2.1517 -2.1452 -2.1426 -2.14 -2.1339 -2.1333 -2.1314 -2.1298 -2.1282 -2.1193 -2.1157 -2.1145 -2.1083 -2.1041 -2.1037 -2.0926 -2.0883 -2.0834 -2.0818 -2.0774 -2.0706 -2.0669 -2.0663 -2.066 -2.0623 -2.0609 -2.0592 -2.0511 -2.0509 -2.046 -2.0386 -1.9012 -1.9009 -1.899 -1.8986 -1.8964 -1.8945 -1.888 -1.887 169 HPSE KLK10 SRCAP SH2D3A TIMP3 ICAM1 TNFSF10 KCNJ15 DUSP6 SLCl 2A8 CCNA1 PTAFR PAMCI VDR AKR1C2 POPDC3 SLC6A8 LOC254531 SORL1 MTUS1 SLC27A3 CNTNAP2 DUSP6 TIMP3 C2orf31 SLC2A3 CNTNAP2 GM2A B3GNT3 CD83 IGSF4 LRRC1 RIMS3 PDPN FLJ20366 BIRC3 RIPK4 CAMK2N1 TIAF1 TNFRSF21 D4S234E SLC3A2 AQP3 PTPRE HOXA1 CYB561 DUSP10 LY75 MARK1 SRCAP 219403_s_at 215808_at 213667_at 219513 sat 201150_ s at 202638 s at 202687 s at 210119_at 208892_s_at 219874_at 205899_at 211661 _xat 210335_at 204255_ sat 211653_x_at 219926_at 202219_at 40472_at 203509_at 212096_s_at 222217 s at 219300_s_at 208893_sat 201148 s at 221245_s_at 202499_s_at 219301 s at 35820_at 204856_at 204440_at 209031 _at 218816_at 204730_at 204879_at 218692_at 210538 s at 221215 s at 218309_at 202039_at 218856_at 209569_x_at 200924 s at 203747_at 221840_at 214639 s at 209164_s_at 221563_at 205668_at 221047_sat 38766_at -2.0146 -2.0139 -2.0127 -2.0123 -2.0097 -2.0055 -2.0035 -2.0023 -1.9934 -1.9911 -1.9898 -1.9895 -1.9858 -1.9857 -1.9829 -1.9756 -1.9707 -1.9683 -1.9654 -1.9616 -1.9544 -1.9506 -1.9492 -1.9455 -1.9452 -1.942 -1.9389 -1.9388 -1.9381 -1.9367 -1.9341 -1.9321 -1.9288 -1.9271 -1.9268 -1.9197 -1.9156 -1.9131 -1.9119 -1.9072 -1.9066 -1.9053 -1.7673 -1.764 -1.7623 -1.7599 -1.7592 -1.7585 -1.7535 -1.7521 CELSR2 Cl0orf10 FLJ20130 TIMP3 ITGB4 NRG1 Cl9orf21 IL1 3RA2 RHOD LGR4 IL15RA F11R KIAA0040 KIAA0888 FAM69A DAPP1 ABLIM1 S100A2 MYO1E TREM2 SLC7A8 SNCA KIAA0247 SDC4 TBC1D4 EMR2 PIP5K2C MAST4 HERC6 PELI1 PRKCZ CYB5R1 SEMA3C IER3 ARHGEF5 DHRS1 TCF7L1 DAF PLAU CAMK2B SLC20A2 TFRC PLK2 ZNF574 ALS2CL FLNB CELSR2 KCNN4 CSF3 IL1 RAP 36499_at 209182_s at 220520_s_at 201149_s_at 211905 s at 206237_s at 212925_at 206172_at 31846_at 218326_sat 207375_s_at 222354_at 203144_s_at 213954_at 216044_x_at 219290_x_at 210461 _s_at 204268_at 203072_at 219725_at 202752_x_at 207827_x_at 202181 _at 202071_at 203387_s at 207610_s_at 218942_at 210958_s_at 219352_at 218319_at 202178_at 202263_at 203788_s at 201631 _sat 204765_at 213279_at 221016_s_at 201925_s_at 205479_s_at 211483_x at 202744_at 207332_s at 201939_at 221844_x at 222333_at 208613_s at 204029_at 204401 _at 207442_at 210233_at -1.8833 -1.8828 -1.8787 -1.8739 -1.8721 -1.8691 -1.8647 -1.8611 -1.8605 -1.8593 -1.8579 -1.8577 -1.8572 -1.8566 -1.8549 -1.8511 -1.8451 -1.8326 -1.8299 -1.8197 -1.8139 -1.8079 -1.8064 -1.8064 -1.8057 -1.8034 -1.8023 -1.8022 -1.8001 -1.7998 -1.7989 -1.7985 -1.7971 -1.7914 -1.79 -1.7887 -1.7815 -1.7798 -1.7782 -1.769 -1.6485 -1.6453 -1.6444 -1.6422 -1.6412 -1.6403 -1.6368 -1.6368 -1.6344 -1.6342 170 KIAA1 026 PTPRF FLJ20035 CYB561 KIAA1 609 ANKRD15 EDG4 CBLC QPCT AGRN HDAC9 EPS8L2 NCF2 CD9 C20orf 19 TNFRSF25 PLAU FAM69A DDX58 LTB TAGLN3 TLR2 ISG20 TMPRSS4 PTPN3 GOS2 ATP2B4 PAK6 ATP2B4 FGFR2 ADAM8 CCNG2 CSF2 LOC254531 NRCAM CEACAM1 ARHGDIB IGSF4 CD82 SEMA3B AKAP1 AJAP1 ZDHHC13 EPLIN GNAL BHLHB3 LRP12 RPS6KA1 FHOD3 213478_at 200637_sat 218986_sat 209163_at 221843 s at 212812_at 213005 s at 206723 s at 220638 s at 205174 s at 212285 s at 205659_at 218180 s_at 209949_at 201005 at 219961_sat 219423_xat 211668 s at 213689_ x at 218943 s at 207339 s at 204743_at 204924_at 204698_at 218960_at 203997_at 213524 s at 212135_s at 219461_at 212136_at 203639 s at 205180_s at 202770 s at 210229 s at 213078_xat 216959_x_at 209498_at 201288_at 209032_sat 203904 x at 203071 _at 201675_at 215789_s_at 219296_at 217892_sat 206355_at 221530 s at 220253_sat 203379 at 218980_at -1.7459 -1.7458 -1.7428 -1.7424 -1.7385 -1.7362 -1.7324 -1.7306 -1.7292 -1.7281 -1.7255 -1.7235 -1.7194 -1.7187 -1.7177 -1.7148 -1.7141 -1.7136 -1.7094 -1.7079 -1.7078 -1.7064 -1.7063 -1.7003 -1.6954 -1.6947 -1.6882 -1.6857 -1.6832 -1.6752 -1.6688 -1.666 -1.6635 -1.6632 -1.66 -1.66 -1.6558 -1.653 -1.6525 -1.6524 -1.527 -1.5261 -1.5204 -1.5179 -1.5146 -1.5145 -1.5116 -1.5109 -1.5083 -1.5013 ZHX2 PTPRF RIMS2 GM2A AGRN MYO1B DSCAM CTSH SMPDL3B AIM1 ZC3H12A DSG2 TFPI2 SEMA3F PRKCH HLA-DOB NBEAL2 SERPINB3 TMEM51 GM2A RSNL2 INPP4B HSPA5BP1 OVOL2 HES1 RAPGEF5 RAB17 SLC7A8 HOOK2 SLC39A8 TP73L CYP1A1 CORO2A GCH1 RHBDL6 KCNJ15 EVIl 203556 at 200635 s at 206137 at 209727 at 217419 x at 212365_at 211484 s at 202295 s at 205309 at 212543 at 218810 at 217901 at 209278 s at 35666 at 206099 at 205671 s at 212443 at 209719 x at 218815 s at 33646_g_at 219944 205376 218834 211778 203395 204681 218931 216092 218780 216504 at at s at s at s at s at at s at at s at 211834 s at 205749 at 205538_at 204224 s at 219202_at 211806 s at 221884 at -1.6336 -1.6258 -1.6253 -1.6248 -1.6132 -1.6123 -1.6085 -1.6042 -1.6022 -1.5998 -1.5985 -1.5952 -1.5944 -1.5894 -1.5862 -1.5844 -1.584 -1.5826 -1.5789 -1.5726 -1.5707 -1.5695 -1.5661 -1.5638 -1.5615 -1.5612 -1.561 -1.5609 -1.5608 -1.5583 -1.5549 -1.5526 -1.5522 -1.5516 -1.5419 -1.5417 -1.5407 171 B4GALT4 PTPRF TRIM34 210540 s at 200636 s at 221044 s at -1.5012 -1.5005 -1.5355 Appendix 2 Probes and the corresponding genes upregulated more than 1.5 log-fold in DN-Ecad cells Symbol ProbelD GDF15 TSC22D3 TSC22D3 RNASE4 ATF3 CYP4F1 1 AKR1C1 FBN2 AKR1C2 SSBP1 KDELR3 P8 PER1 AKR1C1 INPP5D CTH HCP5 PTGS1 LEPR PRSS8 CTH KDELR3 TXNIP BLNK AKR1C2 ANG FTL BHLHB3 CALML4 LGALS7 LHX6 FBXO2 C1QL1 221577_x_at 208763_s_at 207001_x_at 213397_x_at 202672_s_at 206153_at 204151_x_at 215717_s_at 209699_x_at 214060_at 204017_at 209230 sat 202861_at 216594_x_at 203332_s_at 217127_at 206082_at 205127_at 209894_at 202525_at 206085 sat 207265_sat 201010_s_at 207655_s_at 211653_x_at 205141 _at 213187_x_at 221530_sat 64408 s_at 206400_at 219884_at 219305_xat 205575_at 203919_at 205158_at 210233_at 206832 sat 219153 s at 36829 at 214790_at 207339_s_at 203438_at TCEA2 RNASE4 IL1 RAP SEMA3F FLJ13710 PER1 SENP6 LTB STC2 DN-Ecad vs shCntrl logfold change 4.1944 EPHB3 3.9408 ZNF198 3.7186 EPHX1 3.1831 CRELD1 3.1734 TXNIP 3.0435 TXNIP 2.9432 GABARAPL3 2.8419 PER2 2.7631 MGC4504 2.6847 H1FX 2.6659 STC2 2.6504 CCNA1 2.5846 ALDH6A1 2.5782 PFAAP5 2.5519 CALML4 2.5324 DNAJB9 2.5295 GPC1 2.5056 CYP1B1 2.5029 PNRC1 2.4523 NR1D2 2.4451 BTG2 2.4338 EML2 2.3868 ALDH6A1 2.3624 FTL 2.3226 DBP 2.3182 AKR1C2 2.2974 C1 orf38 2.2631 CRIP2 2.2382 BCL6 2.2194 ABCG1 2.2014 SLC39A7 2.1947 LAPTM5 2.1872 MAGED2 2.1745 CABC1 2.1719 CELSR2 2.1675 FLJ40092 2.1453 CPT1B 2.143 ALDH6A1 2.1385 TNFSF7 2.1233 Cep152 2.1183 KLF9 2.1117 MAGED1 172 1438_at 210282_at 202017_at 203368_at 201008 sat 201009_s_at 211458_s_at 205251 _at 219270_at 204805_s_at 203439_s_at 205899_at 221589 s at 214753_at 221879_at 202843_at 202755_s_at 202437 s at 209034_at 209750_at 201236_s_at 204398 s at 221588_x_at 212788 x at 209782_s_at 217626_at 210785_s_at 208978_at 203140_at 204567 s at 202667 s at 201721_s_at 208682_s_at 218168 sat 204029_at 213605 s at 210069_at 204290_s_at 206508_at 215170_s_at 203543_s_at 209014_at 2.1061 2.0994 2.0975 2.0948 2.0851 2.0652 2.0647 2.0641 2.0597 2.0426 2.035 2.0262 2.0076 1.9902 1.9777 1.9752 1.9661 1.9619 1.9592 1.9457 1.9384 1.9042 1.9016 1.9006 1.8997 1.8986 1.8979 1.8917 1.8899 1.8889 1.8446 1.8392 1.8239 1.8219 1.8212 1.8185 1.8113 1.8091 1.7891 1.7891 1.7802 1.7789 ALOX15B Clorf38 GOLGA4 STCH KDELR3 KLF9 GPX3 AKR1B10 LRP5 PHGDH CTTN PCK2 ASNS GOLGA2 KIAA0323 USP52 ABHD4 TP53AP1 ZNF451 NINJ1 ZNF184 CSRP2 PGM3 ZNF516 DATF1 LOC81558 LRDD DLGAP4 FLJ22639 ZNF226 POMZP3 MAGED2 CD44 EGFR TGFB1 PRKCA RRAS CYP4F3 C9orf 16 KIAA0657 SMA4 SEC24D NR1D1 KCNMA1 EPHB3 PRSS16 GCLC BTN3A3 CARM1 206714_at 207571_x_at 215203_at 202557_at 207264_at 203542 s at 201348_at 206561 s at 209468_at 201397_at 214073_at 202847_at 205047_s_at 204384_at 212356_at 203117 s at 218581_at 209917 s at 215012_at 203045_at 213452_at 211126_sat 221788_at 203604_at 213213_at 221249_s_at 221640 s at 202570_s_at 220399 at 219603 s at 210910_s_at 213627_at 210916 s at 201984_s_at 203085_s_at 213093_at 212647_at 206515_at 204480 s at 213946 s at 215599_at 202375_at 31637_s_at 221584 s at 204600_at 208165 s at 202922_at 38241 at 221939_at 2.1082 1.7762 1.7734 1.7591 1.7589 1.7578 1.7546 1.753 1.7498 1.7479 1.7473 1.7472 1.7427 1.74 1.7353 1.7313 1.7304 1.7236 1.72 1.7163 1.7163 1.7117 1.7105 1.7088 1.7052 1.7037 1.7006 1.6998 1.6959 1.6908 1.6887 1.6875 1.6867 1.6831 1.6765 1.6747 1.6721 1.6659 1.6656 1.6617 1.6564 1.6557 1.6482 1.6449 1.6394 1.6394 1.6346 1.6316 1.6284 173 EFS WIP149 C1orf66 PPFIBP1 FLJ20254 PDE4DIP LEPRE1 GABARAPL1 MDM2 EFEMP2 APOE MMP28 IL15 YIPF2 MST1 GFPT1 PFAAP5 POMZP3 HCFC1R1 IGFBP2 LOC440996 DKFZP586A0522 ZBTB16 VDP C17orf39 FLJ13841 DKFZP586H2123 HCFC1R1i PDE4DIP DAF CXorf12 AKR1C3 HERC1 PRKAG2 CDKN1B WIG1 IGSF4C KLHL24 204400_at 213836_s_at 218914_at 214375_at 217899_at 213388_at 220750_s_at 208869_s at 217373_x at 206580_s_at 203381_s at 219909_at 205992 s at 219075_at 216320_x_at 202722_s at 221899_at 204148 s at 45714_at 202718_at 222380 s at 207761 s at 205883_at 222316 at 220058_at 219995_s_at 213661 _at 218537_at 212390_at 201925_s_at 204340_at 209160_at 218306 s at 215231 _at 209112_at 219628_at 215259_s_at 221985_at 1.7773 1.6116 1.6106 1.6084 1.6053 1.6019 1.6007 1.6002 1.6001 1.5989 1.5933 1.589 1.5869 1.5852 1.5833 1.5823 1.5822 1.5783 1.5757 1.5741 1.5735 1.5652 1.5627 1.5618 1.5536 1.5473 1.5465 1.545 1.5442 1.542 1.5399 1.539 1.537 1.5346 1.5296 1.5088 1.5075 1.5054 Probes and the corresponding genes down-regulated more than 1.5 log-fold in DN-Ecad cells Symbol ProbelD SPRR2D MMP10 CXCL1 FST IL1R2 IL8 IL8 KRT13 IL1 R2 HS3ST2 CEACAM6 CEACAM6 GPRC5A CALM1 EIF5 CALM1 IGFBP3 SERPINB2 VGLL1 SKP2 P13 TP73L GLIPR1 KLK7 CYB5R2 S100A7 HSPH1 CCNE2 MCM10 FST P13 KRTHA4 IL1F9 NUP98 PHLDA1 MFAP5 PTGS2 DKK1 CXCL3 PHTF2 LCMT2 ILF3 MAP4 208539 x at 205680 at 204470 at 207345 at 205403 at 202859 x at 211506 s at 207935 s at 211372 s at 219697 at 211657 at 203757 s at 203108 at 211985 s at 208290 s at 211984 at 210095 s at 204614 at 215729 s at 203626 s at 203691 at 211194 s at 204222 s at 205778 at 220230 s at 205916 at 208744 x at 211814 s at 220651 s at 204948 s at 41469_at 206969_at 220322_at 210793 s at 217997_at 213765_at 204748_at 204602_at 207850 at 217097 s at 204012 s at 208931 s at 200835 s at DN-Ecad vs shCntrl loqfold chanqe -4.3198 -4.2073 -4.0769 -4.0606 -4.0554 -3.9569 -3.921 -3.9183 -3.7659 -3.3521 -3.3036 -3.2004 -3.0806 -3.0647 -3.0519 -3.0442 -3.0149 -2.9858 -2.9829 -2.9714 -2.9026 -2.859 -2.8157 -2.8134 -2.808 -2.7237 -2.7057 -2.697 -2.6952 -2.6801 -2.664 -2.6427 -2.6016 -2.6009 -2.5848 -2.5762 -2.5541 -2.5467 -2.5315 -2.5245 -2.4783 -2.0206 -2.0137 174 CCNE2 TMEM16A BCLAF1 B3GNT1 ENC1 SCEL ARL7 FLJ2151 1 TMPRSS11E C12orf4 EIF5 ENC1 EXOSC2 IL6ST MGC14376 KRT15 SFPQ LCN2 CDC27 DNAJA1 OSMR CXCL2 ADAMTS1 ARHGAP25 POP1 EDN1 IL7R G3BP PTHLH DSC2 DRIM MFAP5 DHX9 MFAP5 ARHGAP25 EIF5 PTHLH DUSP6 EPN3 EIF5 UTP14A HNRPM SOX9 205034_at 218804_at 201101 sat 219326 s at 201340 s at 206884 s at 202206_at 220723 s at 220431_at 218374 s at 208708 x at 201341 _at 214507 s at 204864 s at 214696_at 204734 at 201585 s at 212531 at 217878 s at 200880_at 205729_at 209774 x at 222162 s at 204882 at 213449_at 218995 s at 205798_at 201514 s at 206300 s at 204751 x at 209725_at 209758 s at 212107 s at 213764 s at 38149_at 208706 s at 210355_at 208893 s at 220318_at 208705 s at 221513 s at 200072 s at 202935 s at -2.4477 -2.3826 -2.3719 -2.3171 -2.3116 -2.3114 -2.3046 -2.29 -2.2847 -2.2585 -2.249 -2.2372 -2.2268 -2.2199 -2.2186 -2.2046 -2.1846 -2.1846 -2.1747 -2.1725 -2.1697 -2.1596 -2.1594 -2.1571 -2.1331 -2.126 -2.1137 -2.1122 -2.0935 -2.0817 -2.0775 -2.0762 -2.0727 -2.0715 -2.0603 -2.0511 -2.0479 -2.0452 -2.038 -2.0349 -2.0307 -1.7603 -1.7505 HSPA8 UTP14A SPAG1 NUP50 TMEM2 NAV3 SERPINB13 ST6GALNAC5 IFIT5 ATP12A SERPINB1 SFPQ DCBLD2 JAG1 DDX18 TMPO CSF3 HSPA8 SERPINB13 FOSL1 CANX PHLDA1 LGALS8 HRB2 MDSRP Cl orf42 GLIPR1 EIF4G1 BAT2D1 B3GALT3 EML4 CSF2 PPP2R1B PTHLH HMGCS1 IGFBP3 FEN1 LOC51136 CDC6 UGCG Clorf116 SERPINB8 ZNF365 BRIP1 ILl B NCBP1 PHLDA1 MCM4 MAX 11-Sep 210338_s_at 221514_at 210117_at 218294_s_at 218113_at 204823_at 216258_s_at 220979_s_at 203595_s_at 207367_at 213572_s at 201586_s_at 213865_at 209098_s at 205763_sat 209754_s at 207442_at 208687_x at 211362_s_at 204420_at 208853 s at 218000_sat 210732_s at 214085_x at 218587_s at 220620_at 204221_x at 208624_s at 211947_s at 211812_s at 220386_s at 210229_s at 202883_s_at 211756_at 205822_s at 212143_s at 204768 s at 221194_s_at 203967_at 221765_at 219856_at 206034_at 206448_at 221703 at 39402_at 209520_sat 217999_s at 222037_at 210734 x at 201308_s at -2.0124 -2.004 -1.9953 -1.9791 -1.9725 -1.9709 -1.9641 -1.9609 -1.9533 -1.9519 -1.948 -1.9475 -1.9457 -1.9426 -1.9341 -1.9261 -1.9234 -1.919 -1.9037 -1.8928 -1.8924 -1.8906 -1.8892 -1.8747 -1.8684 -1.8638 -1.8539 -1.8512 -1.8494 -1.8483 -1.8393 -1.83 -1.8237 -1.8203 -1.8191 -1.8186 -1.8169 -1.8152 -1.7993 -1.7962 -1.7907 -1.7817 -1.7795 -1.7702 -1.7665 -1.7664 -1.5762 -1.5701 -1.5656 -1.5637 175 G3BP2 UMPS SFRS3 WTAP EXOSC2 FBXO9 IVL EHF DHX9 METAP2 DDX1 8 SETMAR UMPS SMARCC1 C14orf169 SUB1 SOX9 STARD13 ETS1 LARP4 ATP13A3 KLK10 MYO5C SP100 HCCS KRT8 EMR2 ZMPSTE24 RRS1 CBFB CDC6 HNRPU Cl0orf10 FLJ20516 TOE1 ARL7 TXNDC CASP8 MOSCi TGFA DDX3X HSPH1 HIST1H4C SLC16A1 IL6ST LOC56902 206383 s at 215165 x at 208673 s at 210285_xat 209527_at 210638 s at 214599_at 219850_s_at 212105 s at 213899_at 208896_at 206554 x at 202706 s at 201072 s at 219526_at 221727_at 202936_s_at 213103_at 214447_at 214155 s at 219558_at 215808_at 218966_at 210218 s at 203745_at 209008_xat 207610_s_at 202939_at 209567_at 206788 s at 203968 s at 200593 s at 209183 s at 219258_at 204080_at 202208_sat 208097 s at 207686 s at 218865_at 211258 s at 201211 s at 206976 s at 205967_at 202236_sat 212196_at 203622 s at -1.7421 -1.7319 -1.73 -1.7297 -1.7263 -1.7225 -1.7207 -1.7163 -1.712 -1.7109 -1.6964 -1.6918 -1.6917 -1.6892 -1.6877 -1.6856 -1.6813 -1.6795 -1.6752 -1.6748 -1.672 -1.6714 -1.6712 -1.6577 -1.6532 -1.6501 -1.6478 -1.6441 -1.6402 -1.6284 -1.6228 -1.6152 -1.6121 -1.6089 -1.5997 -1.5982 -1.5971 -1.596 -1.5923 -1.5902 -1.5866 -1.5863 -1.5852 -1.58 -1.5785 -1.5775 PPP2R1B TSEN2 PHLDA1 KLK10 KLF10 CHRNA5 KLK11 ARL7 RFWD3 MEST P2RY2 TOR1AIP1 KIF5B SERPINB13 IMP3 SERPINB1 RBM12 SLC43A3 BRCA2 SFRS2 HSMPP8 GLT28D1 PAFAH1B1 SYNCRIP DDX52 HBEGF SLC43A3 SCYL2 202886 s at 219581 at 217996 at 209792 s at 202393 sat 206533 at 205470 s at 202207 at 218564_at 202016 at 206277 at 216100 s at 201992 s at 217272 s at 221688 s at 212268 at 212168 at 213113 s at 214727 at 200754 x at 221771 s at 219015 s at 211547 s at 209025 s at 210320 s at 38037_at 210692 s at 221220 s at -1.5621 -1.5551 -1.5512 -1.5463 -1.5455 -1.5402 -1.5381 -1.535 -1.5346 -1.5323 -1.5304 -1.5299 -1.5294 -1.529 -1.5273 -1.5268 -1.5243 -1.5231 -1.52 -1.515 -1.5134 -1.5125 -1.5123 -1.5117 -1.5092 -1.5014 -1.5004 -1.5004 176 Appendix 3 Genes differentially expressed more than 1.5 log-fold in both DN-Ecad and shEcad cells Upregulated ALDH6A1 CRIP2 CYP1B1 FTL KCNMA1 KDELR3 KLF9 LEPR MAGED1 MAGED2 P8 PRKCA RNASE4 STC2 TCEA2 TSC22D3 WIP149 ZBTB16 Downregulated ARHGAP25 ARL7 ATP12A C10orf10 Clorf116 C1 orf42 CEACAM6 CSF2 CSF3 CXCL1 CXCL2 CXCL3 CYB5R2 DSC2 DUSP6 EDN1 EHF EMR2 EPN3 FLJ21511 FST GPRC5A HBEGF HS3ST2 IL1B IL1F9 IL1 R2 IL8 IVL KLK10 KLK11 KLK7 KRT13 KRT15 KRT8 LCN2 MFAP5 MMP10 MOSC1 MYO5C NAV3 P2RY2 P13 PTGS2 PTHLH 177 S100A7 SCEL SERPINB1 SERPINB13 SERPINB2 SOX9 SPAG1 SPRR2D ST6GALNAC5 TGFA TMEM16A TMPRSS11E TP73L VGLL1 178